1
|
Liu Y, Jiang Y, Ma T, Dong W, Yang P, Peng L, Wang B, Wu C, Li Z, Zhang H, Sun Y, Niu Y, Ding Y. Cardiomyocyte-specific activation of the sarcomere-localized Dnajb6b chaperone causes cardiomyopathy and heart failure through upregulated sarcoplasmic reticulum stress. Life Sci 2025; 374:123711. [PMID: 40360088 DOI: 10.1016/j.lfs.2025.123711] [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: 02/09/2025] [Revised: 05/01/2025] [Accepted: 05/09/2025] [Indexed: 05/15/2025]
Abstract
AIMS Despite abundant expression of DNAJB6 gene in the heart, its roles in cardiac diseases remain underexplored. We aimed to investigate the function of its zebrafish (Danio rerio) ortholog, the dnajb6b gene, in cardiomyopathy and heart failure. MATERIALS AND METHODS Both loss-of-function mutation and gain-of-function transgenic approaches were employed in zebrafish. High frequency echocardiography was performed to evaluate cardiac function indices in adult zebrafish. 4-phenylbutyric acid (4-PBA) was used to pharmacologically inhibit sarcoplasmic reticulum (SR) stress in zebrafish. Western blot was carried out to determine expression of DNAJB6 isoforms in human patients' heart tissues. KEY FINDINGS Global loss-of-function mutations affecting both the sarcomere-localized short (Dnajb6b[S]) and nucleus-localized long (Dnajb6b[L]) isoforms appeared phenotypically normal. In contrast, cardiomyocyte-specific overexpression of a truncated, sarcomere-localized Dnajb6b(L) isoform (Dnajb6b[∆L]) led to severe cardiomyopathy and heart failure phenotypes. Mechanistically, Dnajb6b responded to sarcoplasmic reticulum (SR) stress and activation of Dnajb6b(∆L) resulted in elevated SR stress, accumulation of ubiquitinated protein aggregation, and aberrant activation of autophagy. 4-PBA treatment partially rescued cardiac dysfunction and extended the lifespan of zebrafish with cardiomyocyte-specific activation of Dnajb6b(∆L). Finally, elevated expression of both DNAJB6(S) and DNAJB6(L) isoforms was detected in failing human hearts, supporting their clinical relevance. SIGNIFICANCE Gain-of-function mutation in Dnajb6b(∆L) isoform causes cardiomyopathy and heart failure, likely mediated by elevated SR stress. This study enhances our understanding of Dnajb6's role in cardiac proteostasis and highlights its potential as a therapeutic target for the treatment of cardiomyopathy and heart failure.
Collapse
Affiliation(s)
- Yuting Liu
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China
| | - Yajie Jiang
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China
| | - Taiwei Ma
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China
| | - Wenjing Dong
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China
| | - Peng Yang
- Cardiovascular Surgery Department, Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Lixia Peng
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China
| | - Baokun Wang
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China
| | - Chuanhong Wu
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China
| | - Zhiqiang Li
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China
| | - Hong Zhang
- Cardiovascular Surgery Department, Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Yuanchao Sun
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China
| | - Yujuan Niu
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China
| | - Yonghe Ding
- The Affiliated Hospital of Qingdao University & Biomedical Sciences Institute, Qingdao Medical College of Qingdao University, Qingdao 266021, China.
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Shah Z, Wang C, Ullah H, You H, Philonenko ES, Regan OV, Volchkov P, Dai Y, Yu J, Samokhvalov IM. RUNX1 is a key inducer of human hematopoiesis controlling non-hematopoietic mesodermal development. Stem Cells 2025; 43:sxaf019. [PMID: 40220285 DOI: 10.1093/stmcls/sxaf019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 03/21/2025] [Indexed: 04/14/2025]
Abstract
The RUNX1/AML1 transcription factor is one of the key regulators of definitive hematopoietic development in mice. However, its role in early human hematopoiesis remains poorly investigated. In this study, we integrated a tdTomato reporter cassette into the RUNX1 locus of human pluripotent stem cells (hPSCs) to monitor and block the expression of the gene during hPSC differentiation. This approach demonstrated that expression of RUNX1 starts early in mesodermal specification focusing later on hemogenic endothelium (HE) and nascent hematopoietic cells. Lack of RUNX1 halted the development of CD43+ and CD235-CD45+ hematopoietic cells, preventing the production of clonogenic hematopoietic progenitors including the multilineage ones. The abrogation of RUNX1 resulted in the failure of definitive lineages, specifically T and NK cells. Remarkably, we instead observed the accumulation of RUNX1-null HE cells at the stage of blood cell generation. Moreover, the loss of the gene biased the development toward the lineage of CD43-CD146+CD90+CD73+ mesenchymal cells. RNA-seq analysis of RUNX1-null cells revealed the downregulation of top-level hematopoietic transcription factor genes and the reciprocal upregulation of genes associated with non-hematopoietic cells of mesodermal origin. Forced expression of RUNX1c in differentiating RUNX1-null hPSCs effectively rescued the development of CD45+ myeloid cells and megakaryocytes. Our data demonstrate that RUNX1 is a top hematopoietic inducer that simultaneously controls the expansion of non-hematopoietic lineages.
Collapse
Affiliation(s)
- Zahir Shah
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, People's Republic of China
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, United States
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, United States
| | - Cuihua Wang
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, People's Republic of China
| | - Hanif Ullah
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, People's Republic of China
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, United States
| | - Hao You
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, People's Republic of China
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-Sen University, Guangzhou 510080, People's Republic of China
| | - Elena S Philonenko
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, People's Republic of China
| | - Olga V Regan
- Department of Fundamental Medicine, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Pavel Volchkov
- Department of Fundamental Medicine, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Yong Dai
- The first affiliated hospital, School of Medicine, Anhui University of Science and Technology, Huainan, Anhui 232001, People's Republic of China
| | - Jianhua Yu
- Division of Hematology and Oncology, Department of Medicine, Chao Family Comprehensive Cancer Center, University of California, Irvine, Orange, CA 92697, United States
| | - Igor M Samokhvalov
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cells and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, People's Republic of China
| |
Collapse
|
5
|
Osborn MJ, Panda S, Reineke TM, Tolar J, Nyström A. Progress in skin gene therapy: From the inside and out. Mol Ther 2025; 33:2065-2081. [PMID: 40077969 DOI: 10.1016/j.ymthe.2025.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/04/2025] [Accepted: 03/07/2025] [Indexed: 03/14/2025] Open
Abstract
The skin is the largest organ of the body and forms and serves as the barrier for preventing external material from accessing and damaging internal organs. As the outward interface to the environment, it is accessible for the application of therapeutic agents and cellular and gene therapy represent attractive and promising options to treat severe genetic conditions for which palliation has long been the main stay. However, because of its barrier function, transit across and to the subdermal compartment can be challenging. This commentary examines the current approaches of cell and gene therapies for genetic skin disorders. We write this from a local and systemic "outside and inside." perspective. Delivery from the outside encompasses topical, intradermal, and transdermal strategies for cell and vector delivery and ex vivo cell expansion and grafting. The inside approach details systemic delivery via infusion of cells or agents toward providing benefit to the skin. We use recessive dystrophic epidermolysis bullosa (RDEB) as a representative and paradigmatic disease to showcase these approaches as a means to highlight potential broader applicability to other conditions.
Collapse
Affiliation(s)
- Mark J Osborn
- Medical School, Department of Pediatrics, Division of Blood and Marrow Transplant and Cellular and Gene Therapy, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Sidharth Panda
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jakub Tolar
- Medical School, Department of Pediatrics, Division of Blood and Marrow Transplant and Cellular and Gene Therapy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Alexander Nyström
- Department of Dermatology, Medical Faculty, Medical Center, University of Freiburg, 79106 Freiburg, Germany.
| |
Collapse
|
6
|
Doddamani D, Carlson DF, McTeir L, Taylor L, Nandi S, Davey MG, McGrew MJ, Glover JD. PRDM14 is essential for vertebrate gastrulation and safeguards avian germ cell identity. Dev Biol 2025; 521:129-137. [PMID: 39938772 DOI: 10.1016/j.ydbio.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 01/30/2025] [Accepted: 02/08/2025] [Indexed: 02/14/2025]
Abstract
The zinc finger transcription factor PRDM14, part of the PR domain containing protein family, is critical for mammalian primordial germ cell (PGC) specification, epigenetic reprogramming and maintaining naïve pluripotency in stem cells. However, PRDM14's role in other species is not well understood. In chicken, PRDM14 is broadly expressed in the early embryo, before becoming restricted to the forming neural plate, migratory PGCs, and later, in the adult testes. To investigate the role of PRDM14 we generated two independent targeted chicken lines and bred homozygous knockout embryos. Strikingly, we found that gastrulation was disrupted in PRDM14-/- embryos, which lacked a definitive primitive streak. Transcriptomic and in situ hybridisation analyses revealed a broad loss of anterior primitive streak marker genes, coupled with downregulation of the multifunctional antagonists CHRD and CER1, and expansion of the NODAL expression domain. Further analysis of PRDM14-/- embryos revealed PGCs were still specified but significantly reduced in number, and PRDM14-/- PGCs could not be propagated in vitro. Knockdown studies in vitro confirmed that PRDM14 is essential for PGC survival and antagonises FGF-induced somatic differentiation, similar to PRDM14's role in mammalian stem cells. Taken together, our results show that in chicken, PRDM14 plays a multifunctional and essential role during embryonic development.
Collapse
Affiliation(s)
- Dadakhalandar Doddamani
- The Roslin Institute, R(D)SVS, The University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK; ICMR-Regional Medical Research Centre, Port Blair, Andaman and Nicobar Islands, India
| | | | - Lynn McTeir
- The Roslin Institute, R(D)SVS, The University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK
| | - Lorna Taylor
- The Roslin Institute, R(D)SVS, The University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK
| | - Sunil Nandi
- The Roslin Institute, R(D)SVS, The University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK
| | - Megan G Davey
- The Roslin Institute, R(D)SVS, The University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK
| | - Mike J McGrew
- The Roslin Institute, R(D)SVS, The University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK
| | - James D Glover
- The Roslin Institute, R(D)SVS, The University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK.
| |
Collapse
|
7
|
Nakamae K, Ide S, Ohnuki N, Nakagawa Y, Okuhara K, Bono H. PtWAVE: a high-sensitive deconvolution software of sequencing trace for the detection of large indels in genome editing. BMC Bioinformatics 2025; 26:114. [PMID: 40301714 PMCID: PMC12039204 DOI: 10.1186/s12859-025-06139-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 04/08/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Tracking of Insertions and DEletions (TIDE) analysis, which computationally deconvolves capillary sequencing data derived from the DNA of bulk or clonal cell populations to estimate the efficiency of targeted mutagenesis by programmable nucleases, has played a significant role in the field of genome editing. However, the detection range covered by conventional TIDE analysis is limited. Range extension for deconvolution is required to detect larger deletions and insertions (indels) derived from genome editing in TIDE analysis. However, extending the deconvolution range introduces uncertainty into the deconvolution process. Moreover, the accuracy and sensitivity of TIDE analysis tools for large deletions (> 50 bp) remain poorly understood. RESULTS In this study, we introduced a new software called PtWAVE that can detect a wide range of indel sizes, up to 200 bp. PtWAVE also offers options for variable selection and fitting algorithms to prevent uncertainties in the model. We evaluated the performance of PtWAVE by using in vitro capillary sequencing data that mimicked DNA sequencing, including large deletions. Furthermore, we confirmed that PtWAVE can stably analyze trace sequencing data derived from actual genome-edited samples. CONCLUSIONS PtWAVE demonstrated superior accuracy and sensitivity compared to the existing TIDE analysis tools for DNA samples, including large deletions. PtWAVE can accelerate genome editing applications in organisms and cell types in which large deletions often occur when programmable nucleases are applied.
Collapse
Affiliation(s)
- Kazuki Nakamae
- Genome Editing Innovation Center, Hiroshima University, 3-10-23 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-0046, Japan.
- PtBio Inc., Hiroshima, 739-0046, Japan.
| | - Saya Ide
- Genome Editing Innovation Center, Hiroshima University, 3-10-23 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-0046, Japan
- PtBio Inc., Hiroshima, 739-0046, Japan
| | - Nagaki Ohnuki
- Genome Editing Innovation Center, Hiroshima University, 3-10-23 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-0046, Japan
- PtBio Inc., Hiroshima, 739-0046, Japan
| | | | - Keisuke Okuhara
- PtBio Inc., Hiroshima, 739-0046, Japan
- Graduate School of Integrated Sciences for Life, Hiroshima University, 3-10-23 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-0046, Japan
| | - Hidemasa Bono
- Genome Editing Innovation Center, Hiroshima University, 3-10-23 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-0046, Japan.
- Graduate School of Integrated Sciences for Life, Hiroshima University, 3-10-23 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-0046, Japan.
| |
Collapse
|
8
|
He S, Liu Y, Zhang Z, Cai M, Hao Y, Hu H. Gene Editing in Ganoderma lucidum: Development, Challenges, and Future Prospects. J Fungi (Basel) 2025; 11:310. [PMID: 40278130 PMCID: PMC12029067 DOI: 10.3390/jof11040310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/05/2025] [Accepted: 04/09/2025] [Indexed: 04/26/2025] Open
Abstract
As an emerging and innovative technology, gene-editing technology has been widely applied in crop breeding, human disease treatment, animal model research, drug and vaccine development, and microbial engineering. We mainly introduce the development of gene-editing technology, the application of clustered regularly interspaced short palindromic repeat/Cas9 (CRISPR/Cas9) in Ganoderma lucidum breeding, the current challenges and optimization strategies in the use of gene-editing technology in Ganoderma breeding, as well as the current status of gene-editing technology in Ganoderma breeding. Finally, the future research directions and innovative strategies that gene editing may explore in Ganoderma breeding are prospects given the existing background, future research directions, and innovative strategies that gene editing may explore in Ganoderma breeding prospects.
Collapse
Affiliation(s)
- Shiqi He
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (S.H.); (Y.L.); (Z.Z.); (M.C.); (Y.H.)
| | - Yuanchao Liu
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (S.H.); (Y.L.); (Z.Z.); (M.C.); (Y.H.)
| | - Zhi Zhang
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (S.H.); (Y.L.); (Z.Z.); (M.C.); (Y.H.)
- Guangdong Yuewei Biotechnology Co., Ltd., Shaoguan 512029, China
| | - Manjun Cai
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (S.H.); (Y.L.); (Z.Z.); (M.C.); (Y.H.)
| | - Yufan Hao
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (S.H.); (Y.L.); (Z.Z.); (M.C.); (Y.H.)
| | - Huiping Hu
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (S.H.); (Y.L.); (Z.Z.); (M.C.); (Y.H.)
- Guangdong Yuewei Biotechnology Co., Ltd., Shaoguan 512029, China
| |
Collapse
|
9
|
Salaudeen AL, Mateyko N, de Boer CG. RAPID-DASH: Single-Day Assembly of Guide RNA Arrays for Multiplexed CRISPR-Cas9 Applications. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.09.648054. [PMID: 40291718 PMCID: PMC12027327 DOI: 10.1101/2025.04.09.648054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Guide RNA (gRNA) arrays can enable targeting multiple genomic loci simultaneously using CRISPR-Cas9. In this study, we present a streamlined and efficient method to rapidly construct gRNA arrays with up to 10 gRNA units in a single day. We demonstrate that gRNA arrays maintain robust functional activity across all positions, and can incorporate libraries of gRNAs, combining scalability and multiplexing. Our approach will streamline combinatorial perturbation research by enabling the economical and rapid construction, testing, and iteration of gRNA arrays.
Collapse
|
10
|
Ottaviano G, Qasim W. Current landscape of vector safety and genotoxicity after hematopoietic stem or immune cell gene therapy. Leukemia 2025:10.1038/s41375-025-02585-8. [PMID: 40200078 DOI: 10.1038/s41375-025-02585-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 03/10/2025] [Accepted: 03/21/2025] [Indexed: 04/10/2025]
Abstract
Malignant transformation of gene modified haematopoietic stem cells caused anxiety following adverse events in early clinical trials using gamma-retroviral vectors (γRV) to correct haematopoietic stem cells (HSC) in monogenic immune disorders. Adoption of HIV-derived lentiviral vectors (LV) with SIN (self-inactivating) configurations greatly reduced risks and subsequently hundreds of patients have been dosed with HSC gene therapy for blood, immune and metabolic conditions. Nevertheless, as experience builds, it's now well recognised that vector integration can drive clonal expansions and these may carry long term safety risks. Documented cases of haematological malignancy after SIN-LV gene therapy have recently emerged, in particular where heterologous retroviral promoters were employed and there are concerns around certain insulator elements and other possible contributors to clonal expansions. Similarly, tens of thousands of subjects have now received engineered T cell products, and longstanding dogma that mature T cells cannot be transformed is being questioned, with reports of a small number of malignant transformation events and wider concerns around secondary malignancies in some groups of patients. We summarize current clinical information and revisit genotoxicity risks following ex-vivo gene modification of HSC and T cells.
Collapse
Affiliation(s)
- Giorgio Ottaviano
- Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy.
- Molecular and Cellular Immunology, University College London, London, UK.
| | - Waseem Qasim
- Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Molecular and Cellular Immunology, University College London, London, UK
| |
Collapse
|
11
|
Mohanta R, Maiti P, Sharangi AB, Roy S, Hazra S, Chakraborty S, Ghorai S. Directed mutagenesis in fruit crops. 3 Biotech 2025; 15:104. [PMID: 40177007 PMCID: PMC11958931 DOI: 10.1007/s13205-025-04268-8] [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: 01/02/2025] [Accepted: 03/07/2025] [Indexed: 04/05/2025] Open
Abstract
Fruit crops are rich source of important vitamins, minerals, and dietary fibres. They are essential for global agriculture with respect to nutritional security. Globally, there is a rapid decline in the genetic base of fruit crops warranting breeding strategies to overcome the challenge. Applied mutagenesis has emerged as a viable approach for the focused enhancement of fruit crops utilizing precise genetic alterations to increase a variety of desirable characteristics. However, traditional mutagenesis using physical and chemical mutagens are majorly random in nature. Directed mutagenesis with advancements in genetic engineering and molecular technology allows precise manipulation of genes, which facilitates the efficient and precise knockout of target genes and the targeted insertion or modification of specific DNA sequences within the genome via homologous recombination (HR)-mediated gene replacement. This review presents an in-depth exploration of several directed mutagenesis techniques including CRISPR-Cas9, TILLING, TALEN, MutMap, and MutMap + emphasizing their transformative applications in fruit crops. It also discusses about space mutagenesis. These advanced techniques empower researchers to precisely introduce specific mutations into the genome, skilfully altering gene expression and reshaping protein function with remarkable precision. This review highlights successful examples of directed mutagenesis in a variety of fruit crops such as apples, grapes, citrus, and strawberries and elucidates the impact of directed mutagenesis on traits such as fruit size, colour, flavour, shelf-life, and resistance to diseases and environmental stresses.
Collapse
Affiliation(s)
- Rajdeep Mohanta
- Department of Agriculture, Brainware University, Barasat, Kolkata, 700125 West Bengal India
| | - Payal Maiti
- Department of Post-Harvest Management, Bidhan Chandra Krishi Viswavidyalaya, Mohanpur, 741252 West Bengal India
| | - Amit Baran Sharangi
- Department of Plantation Spices Medicinal & Aromatic Crops, Bidhan Chandra Krishi Viswavidyalaya, Mohanpur, 741252 West Bengal India
| | - Sourav Roy
- Department of Agriculture, Brainware University, Barasat, Kolkata, 700125 West Bengal India
| | - Soham Hazra
- Department of Agriculture, Brainware University, Barasat, Kolkata, 700125 West Bengal India
| | - Souvik Chakraborty
- Department of Post-Harvest Management, Bidhan Chandra Krishi Viswavidyalaya, Mohanpur, 741252 West Bengal India
| | - Subhadwip Ghorai
- Department of Agriculture, Brainware University, Barasat, Kolkata, 700125 West Bengal India
| |
Collapse
|
12
|
He J, Liu J, Yue Y, Wang L, Liu Z, Xi G, An L, Tian J, Wang Y. Genome Editing in Mouse Embryo Using the CRISPR/Cas12i3 System. Int J Mol Sci 2025; 26:3036. [PMID: 40243700 PMCID: PMC11988942 DOI: 10.3390/ijms26073036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/15/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
The CRISPR/Cas system is a sizable family that is currently a popular and efficient gene editing tool. Cas12i3, as a member of the Type V-I family, has the characteristics of recognizing T-rich PAM sequences and being guided by shorter crRNA and has higher gene editing efficiency than Cas9 in rice. However, as a potential tool in accelerating the breeding process, the application of Cas12i3 in mammalian embryos has not yet been reported. Our study systematically evaluated the feasibility of applying CRISPR/Cas12i3 to gene editing in mouse embryos, with the core pluripotency regulator gene Nanog as the target. We successfully constructed a Nanog loss-of-function mouse embryo model using CRISPR/Cas12i3. At the targeted Nanog locus, its editing efficiency exceeded that of the Cas9 system under matched experimental conditions; no off-target phenomenon was detected. Moreover, the Cas12i3 system exhibited no side effect on mouse embryo development and proliferation of blastocyst cells. Finally, we obtained healthy chimeric gene-edited offspring by optimizing the concentration of the Cas12i3 mixture. These results confirm the feasibility and safety of CRISPR/Cas12i3 for gene editing in mammals, which provides a reliable tool for one-step generation of gene-edited animals for applications in biology, medical research, and large livestock breeding.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yinjuan Wang
- Frontiers Science Center for Molecular Design Breeding (MOE), Key Laboratory of Animal Genetics, Breeding and Reproduction of the Ministry of Agriculture and Rural Affairs, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (J.H.); (J.L.); (Y.Y.); (L.W.); (Z.L.); (G.X.); (L.A.); (J.T.)
| |
Collapse
|
13
|
Baldini A, Battaglia F, Perrella G. The generation of novel epialleles in plants: the prospective behind re-shaping the epigenome. FRONTIERS IN PLANT SCIENCE 2025; 16:1544744. [PMID: 40190658 PMCID: PMC11968746 DOI: 10.3389/fpls.2025.1544744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 02/07/2025] [Indexed: 04/09/2025]
Abstract
Chromatin organization is a relevant layer of control of gene expression during plant development. Chromatin states strictly depend on associated features such as DNA methylation, histone modifications and histone variants. Thus, epigenome editing has become of primary interest to alter gene expression without disrupting genomic sequences. Different tools have been developed to address this challenge, starting with modular Zinc Finger Proteins (ZFPs) and Transcription Activator Like Effectors (TALEs). However, the discovery of CRISPR/Cas9 system and the adaptability of technologies based on enzymatically dead Cas9 (dCas9) have paved the way towards a reliable and adaptable epigenome editing in a great variety of organisms. In this review, we will focus on the application of targeted epigenome editing technologies in plants, summarizing the most updated advances in this field. The promising results obtained by altering the expression state of targets involved in flowering time and abiotic stress resistance are crucial not only for elucidating the molecular interactions that underly chromatin dynamics, but also for future applications in breeding programs as an alternative route to genetic manipulation towards the achievement of higher quality crops particularly in terms of nutritional properties, yield and tolerance.
Collapse
Affiliation(s)
| | | | - Giorgio Perrella
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
14
|
Danti L, Lundin K, Nedeczey-Ruzsák P, Tuuri T, Tapanainen JS. FOXL2 drives the differentiation of supporting gonadal cells in early ovarian development. Reprod Biol Endocrinol 2025; 23:44. [PMID: 40102860 PMCID: PMC11917015 DOI: 10.1186/s12958-025-01377-0] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/03/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Forkhead box L2 (FOXL2) is a transcription factor from the forkhead box family primarily expressed in the pituitary, ovaries, and eyelids. Human mutations in FOXL2 cause blepharophimosis, ptosis, epicanthus and inversus syndrome (BPES), which can be associated with primary ovarian insufficiency, and is indirectly linked with differences of sex development (DSD). Animal studies have shown the crucial role that FOXL2 plays in the development, function, and maintenance of the ovary as well as in sex determination. However, the specific role of FOXL2 in early human somatic cell ovarian development is largely unknown. METHODS In this study, we utilised CRISPR/Cas9 genome activation and a previously published in-house 14-day gonadal differentiation protocol to study the role of FOXL2. RESULTS Our results demonstrate that FOXL2 downregulates coelomic epithelial markers GATA4 and LHX9, female gonadal markers RSPO1 and WNT4, and male gonadal markers SOX9, NR0B1 and DHH. The differentially expressed genes were mostly associated with Kyoto encyclopaedia of genes and genomes (KEGG) pathways relating to cell adhesion molecules and gene ontology (GO) pathways relating to extracellular matrix and junction formation. Furthermore, a comparative analysis with existing single cell RNA sequencing data from human in vivo-derived samples elucidated that FOXL2 initiates the downregulation of coelomic epithelial genes GATA4, LHX9 and UPK3B at day 6. By day 8, the genes ARX and GATA2 are transiently upregulated by FOXL2 induction and then downregulated as the genes LGR5, TSPAN8, OSR1 and TAC1 become upregulated. CONCLUSIONS These findings suggest that FOXL2 facilitates the exit of differentiating cells from the coelomic epithelium and initially drives them towards a transitional identity before progressing into early supporting gonadal-like cells. The findings of this study significantly advance our understanding of normal gonadal development which can be used as a basis to elucidate pathological gonadal development underlying BPES.
Collapse
Affiliation(s)
- Laura Danti
- Department of Obstetrics and Gynaecology, University of Helsinki and Helsinki University Hospital, P.O. Box 140, Helsinki, 00029 HUS, Finland
- Department of Medicine Huddinge (MedH), Biosciences and Nutrition Unit, Karolinska Institutet, Huddinge, Sweden
| | - Karolina Lundin
- Department of Obstetrics and Gynaecology, University of Helsinki and Helsinki University Hospital, P.O. Box 140, Helsinki, 00029 HUS, Finland.
| | - Petra Nedeczey-Ruzsák
- Department of Obstetrics and Gynaecology, University of Helsinki and Helsinki University Hospital, P.O. Box 140, Helsinki, 00029 HUS, Finland
| | - Timo Tuuri
- Department of Obstetrics and Gynaecology, University of Helsinki and Helsinki University Hospital, P.O. Box 140, Helsinki, 00029 HUS, Finland
| | - Juha S Tapanainen
- Department of Obstetrics and Gynaecology, University of Helsinki and Helsinki University Hospital, P.O. Box 140, Helsinki, 00029 HUS, Finland
- Department of Obstetrics and Gynaecology, HFR - Cantonal Hospital of Fribourg and University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
15
|
Dicipulo R, Selland LG, Carpenter RG, Waskiewicz AJ. Functional role for Taz during hindbrain ventricle morphogenesis. PLoS One 2025; 20:e0313262. [PMID: 40080483 PMCID: PMC11906067 DOI: 10.1371/journal.pone.0313262] [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: 12/13/2023] [Accepted: 10/21/2024] [Indexed: 03/15/2025] Open
Abstract
The brain ventricle system, composed of the ventricular cavities and the cerebral spinal fluid within, performs critical functions including circulation of nutrients, removal of wastes, and cushioning of neural tissues. Development of the hindbrain ventricle requires a series of factors that coordinate its initial formation and subsequent inflation. Previous work has demonstrated that the transcriptional co-activator Taz (also known as WW domain-containing transcription regulator protein 1, Wwtr1), a component of Hippo signalling, is active at hindbrain rhombomere boundaries where it is regulated by mechanotransduction and promotes proliferation. Here, we demonstrate that Taz is also a critical regulator of hindbrain ventricle development. Zebrafish embryos that lack Taz protein fail to undergo initial midline separation of the hindbrain ventricle. Furthermore, the ventricle phenotype is a result of disorganized cytoskeletal F-actin and apicobasal polarity components. In addition, we have demonstrated that the hindbrain rhombomere boundaries are a location of active Wnt-Hippo crosstalk. Through our work, we propose a model where Taz protein is stabilized at rhombomere boundaries and promotes proper cell polarity necessary for formation of the brain ventricle.
Collapse
Affiliation(s)
- Renée Dicipulo
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Lyndsay G. Selland
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Rowan G. Carpenter
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Andrew J. Waskiewicz
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
- Women & Children’s Health Research Institute, University of Alberta, Edmonton, Canada
| |
Collapse
|
16
|
O'Donnell CW, Farelli JD, Belaghzal H, Chen J, Beech L, Sullivan J, Morrison-Smith C, Siecinski S, Katz A, Mildrum S, Gurnani M, Dhanania P, Webb CR, Castello Coatti G, Rumale P, Costa DFG, Gibson MI, Wang YE, Newman JV, McCauley TG. Programmable mRNA therapeutics for controlled epigenomic modulation of single and multiplexed gene expression in diverse diseases. Nat Commun 2025; 16:2517. [PMID: 40082450 PMCID: PMC11906599 DOI: 10.1038/s41467-025-57920-5] [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: 11/15/2023] [Accepted: 03/07/2025] [Indexed: 03/16/2025] Open
Abstract
Pathogenic gene dysregulation can be attributed to chromatin state change that pre-transcriptionally regulates expression. Recent breakthroughs elucidating the rules governing this DNA control layer, an epigenetic code, unlock a modality in precision medicine to target gene dysregulation across myriad diseases. Here we present a modular platform to design programmable mRNA therapeutics, Epigenomic Controllers (EC), that control gene expression through directed epigenetic change. By leveraging natural mechanisms, ECs tune expression levels of one or multiple genes with durable effect of weeks-to-months in female mice following a single dose. We design and characterize ECs to multiple target genes and identify an EC that effectively inhibits the cancer- and inflammatory-disorder-associated multi-gene cluster CXCL1-8. With precision targeting of NF-kB signaling and identification of homologous murine surrogates, ECs significantly reduce neutrophil migration in vivo during acute lung inflammation in female mice. A platform approach to EC design for epigenomic modulation expands treatment frontiers for diverse gene targets, including those considered "undruggable."
Collapse
Affiliation(s)
| | | | | | - Justin Chen
- Omega Therapeutics, Inc., Cambridge, MA, USA
| | | | | | | | | | - Adam Katz
- Omega Therapeutics, Inc., Cambridge, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Lu C, Li Y, Cummings JR, Banskota S. Delivery of genome editors with engineered virus-like particles. Methods Enzymol 2025; 712:475-516. [PMID: 40121085 DOI: 10.1016/bs.mie.2025.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Genome editing technologies have revolutionized biomedical sciences and biotechnology. However, their delivery in vivo remains one of the major obstacles for clinical translation. Here, we introduce various emerging genome editing systems and review different delivery systems have been developed to realize the promise of in vivo gene editing therapies. In particular, we focus on virus-like particles (VLPs), an emerging delivery platform and provide in depth analysis on recent advancements to improve VLPs delivery potential and highlight opportunities for future improvements. To this end, we also provide detail workflows for engineered VLP (eVLP) selection, production, and purification, along with methods for characterization and validation.
Collapse
Affiliation(s)
- Christopher Lu
- Department of Biomedical Engineering, Boston University, Boston, MA, United States; Biological Design Center, Boston University, Boston, MA, United States
| | - Yuanhang Li
- Biological Design Center, Boston University, Boston, MA, United States; Department of Mechanical Engineering, Boston University, Boston, MA, United States
| | - Jacob Ryan Cummings
- Department of Biomedical Engineering, Boston University, Boston, MA, United States; Biological Design Center, Boston University, Boston, MA, United States
| | - Samagya Banskota
- Department of Biomedical Engineering, Boston University, Boston, MA, United States; Biological Design Center, Boston University, Boston, MA, United States.
| |
Collapse
|
18
|
Mishra P, Shakespear S, Carpenter SCD, Hamsa S, Vigi S, Anith KN, Dash PK, Bogdanove AJ, Rai R. PthXo2B Orthologue Tal7 of Xanthomonas oryzae pv. oryzae Strain IX-221 Acts as a Major Virulence Factor in Indica Rice Without Activating a Clade III SWEET Gene. MOLECULAR PLANT PATHOLOGY 2025; 26:e70067. [PMID: 40019023 PMCID: PMC11868982 DOI: 10.1111/mpp.70067] [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: 10/02/2024] [Revised: 01/08/2025] [Accepted: 02/03/2025] [Indexed: 03/01/2025]
Abstract
In rice bacterial blight, Xanthomonas oryzae pv. oryzae deploys transcription activator-like effectors (TALEs) that upregulate host susceptibility genes. Thirty-four amino acid repeats in TALEs each specify a base in the DNA target, via a repeat-variable diresidue (RVD; positions 12 and 13). Some aberrant-length repeats can disengage to accommodate single base deletions. Clade III SWEET genes SWEET11, -13 and -14 are major susceptibility targets of different TALEs. xa13 is a SWEET11 allele lacking the TALE binding site and thus confers resistance to some strains. It has been deployed widely in India. We report that an xa13-breaking Indian isolate, IX-221, harbours one SWEET14- and two SWEET13-activating TALEs, with one or two disengageable repeats. One, Tal7, orthologous to PthXo2B of Philippines strain PXO61 but with minor, non-RVD sequence differences, like PthXo2B upregulates SWEET13 in a japonica variety and no clade III SWEET in an indica, yet unlike PthXo2B renders both varieties susceptible. A designer TALE with distinct, minor differences also failed to render the indica susceptible. The results suggest that Tal7 activates an alternative susceptibility gene and that non-RVD polymorphism can affect TALE targeting. Moreover, IX-221 provides evidence that the deployment of xa13 in India resulted in strains super-equipped with TALEs that break it.
Collapse
Affiliation(s)
- Prashant Mishra
- Plant Pathogen Interaction, ICAR‐National Institute for Plant BiotechnologyPusaNew DelhiIndia
| | - S. Shakespear
- Plant Pathogen Interaction, ICAR‐National Institute for Plant BiotechnologyPusaNew DelhiIndia
| | - Sara C. D. Carpenter
- Plant Pathology and Plant‐Microbe Biology Section, School of Integrative Plant ScienceCornell UniversityIthacaNew YorkUSA
| | - S. Hamsa
- Plant Pathogen Interaction, ICAR‐National Institute for Plant BiotechnologyPusaNew DelhiIndia
| | - S. Vigi
- Department of Microbiology, College of AgricultureKerala Agricultural UniversityThiruvanathpuramKeralaIndia
| | - K. N. Anith
- Department of Microbiology, College of AgricultureKerala Agricultural UniversityThiruvanathpuramKeralaIndia
| | - Prasanta K. Dash
- Plant Pathogen Interaction, ICAR‐National Institute for Plant BiotechnologyPusaNew DelhiIndia
| | - Adam J. Bogdanove
- Plant Pathology and Plant‐Microbe Biology Section, School of Integrative Plant ScienceCornell UniversityIthacaNew YorkUSA
| | - Rhitu Rai
- Plant Pathogen Interaction, ICAR‐National Institute for Plant BiotechnologyPusaNew DelhiIndia
| |
Collapse
|
19
|
Ahmadikhah A, Zarabizadeh H, Nayeri S, Abbasi MS. Advancements in genome editing tools for genetic studies and crop improvement. FRONTIERS IN PLANT SCIENCE 2025; 15:1370675. [PMID: 39963359 PMCID: PMC11830681 DOI: 10.3389/fpls.2024.1370675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 12/31/2024] [Indexed: 02/20/2025]
Abstract
The rapid increase in global population poses a significant challenge to food security, compounded by the adverse effects of climate change, which limit crop productivity through both biotic and abiotic stressors. Despite decades of progress in plant breeding and genetic engineering, the development of new crop varieties with desirable agronomic traits remains a time-consuming process. Traditional breeding methods often fall short of addressing the urgent need for improved crop varieties. Genome editing technologies, which enable precise modifications at specific genomic loci, have emerged as powerful tools for enhancing crop traits. These technologies, including RNA interference, Meganucleases, ZFNs, TALENs, and CRISPR/Cas systems, allow for the targeted insertion, deletion, or alteration of DNA fragments, facilitating improvements in traits such as herbicide and insect resistance, nutritional quality, and stress tolerance. Among these, CRISPR/Cas9 stands out for its simplicity, efficiency, and ability to reduce off-target effects, making it a valuable tool in both agricultural biotechnology and plant functional genomics. This review examines the functional mechanisms and applications of various genome editing technologies for crop improvement, highlighting their advantages and limitations. It also explores the ethical considerations associated with genome editing in agriculture and discusses the potential of these technologies to contribute to sustainable food production in the face of growing global challenges.
Collapse
Affiliation(s)
- Asadollah Ahmadikhah
- Department of Cellular and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | | | | | | |
Collapse
|
20
|
Mormile BW, Yan Y, Bauer T, Wang L, Rivero RC, Carpenter SCD, Danmaigona Clement C, Cox KL, Zhang L, Ma X, Wheeler TA, Dever JK, He P, Bogdanove AJ, Shan L. Activation of three targets by a TAL effector confers susceptibility to bacterial blight of cotton. Nat Commun 2025; 16:644. [PMID: 39809734 PMCID: PMC11733179 DOI: 10.1038/s41467-025-55926-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 01/04/2025] [Indexed: 01/16/2025] Open
Abstract
Bacterial transcription activator-like effectors (TALEs) promote pathogenicity by activating host susceptibility (S) genes. To understand the pathogenicity and host adaptation of Xanthomonas citri pv. malvacearum (Xcm), we assemble the genome and the TALE repertoire of three recent Xcm Texas isolates. A newly evolved TALE, Tal7b, activates GhSWEET14a and GhSWEET14b, different from GhSWEET10 targeted by a TALE in an early Xcm isolate. Activation of GhSWEET14a and GhSWEET14b results in water-soaked lesions. Transcriptome profiling coupled with TALE-binding element prediction identify a pectin lyase gene as an additional Tal7b target, quantitatively contributing to Xcm virulence alongside GhSWEET14a/b. CRISPR-Cas9 gene editing supports the function of GhSWEETs in cotton bacterial blight and the promise of disrupting the TALE-binding site in S genes for disease management. Collectively, our findings elucidate the rapid evolution of TALEs in Xanthomonas field isolates and highlight the virulence mechanism wherein TALEs induce multiple S genes to promote pathogenicity.
Collapse
Affiliation(s)
- Brendan W Mormile
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Plant Pathology and Microbiology, Texas A&M University, College Station, TX, 77843, USA
| | - Yan Yan
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Taran Bauer
- Plant Pathology and Plant-Microbe Biology Section, School of Integrative Plant Science, Cornell University, Ithaca, NY, 14853, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Li Wang
- Plant Pathology and Plant-Microbe Biology Section, School of Integrative Plant Science, Cornell University, Ithaca, NY, 14853, USA
| | - Rachel C Rivero
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Sara C D Carpenter
- Plant Pathology and Plant-Microbe Biology Section, School of Integrative Plant Science, Cornell University, Ithaca, NY, 14853, USA
| | - Catherine Danmaigona Clement
- Department of Plant Pathology and Microbiology, Texas A&M University, College Station, TX, 77843, USA
- Bayer Research and Development Services LLC, 800 N. Lindbergh Blvd., St. Louis, MO, 63167, USA
| | - Kevin L Cox
- Department of Plant Pathology and Microbiology, Texas A&M University, College Station, TX, 77843, USA
- Department of Biology, Washington University, St. Louis, MO, 63130, USA
| | - Lin Zhang
- Department of Plant Pathology and Microbiology, Texas A&M University, College Station, TX, 77843, USA
| | - Xiyu Ma
- Department of Plant Pathology and Microbiology, Texas A&M University, College Station, TX, 77843, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, 02115, USA
| | | | - Jane K Dever
- Texas A&M AgriLife Research, Lubbock, TX, 79403, USA
- Pee Dee Research and Education Center, 2200 Pocket Road, Florence, SC, 29506, USA
| | - Ping He
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Adam J Bogdanove
- Plant Pathology and Plant-Microbe Biology Section, School of Integrative Plant Science, Cornell University, Ithaca, NY, 14853, USA.
| | - Libo Shan
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Plant Pathology and Microbiology, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
21
|
Gaudin C, Preveaux A, Aubineau N, Le Goff D, Jacques MA, Chen NWG. A dTALE approach demonstrates that induction of common bean OVATE Family Protein 7 promotes resistance to common bacterial blight. JOURNAL OF EXPERIMENTAL BOTANY 2025; 76:607-620. [PMID: 39437252 DOI: 10.1093/jxb/erae433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 10/21/2024] [Indexed: 10/25/2024]
Abstract
Common bacterial blight (CBB) is a devastating seed-transmitted disease of common bean (Phaseolus vulgaris L.), caused by Xanthomonas phaseoli pv. phaseoli and Xanthomonas citri pv. fuscans. The genes responsible for CBB resistance are largely unknown. Moreover, the lack of a reproducible and universal transformation protocol limits the study of genetic traits in common bean. We produced X. phaseoli pv. phaseoli strains expressing artificially designed transcription-activator like effectors (dTALEs) to target 14 candidate genes for resistance to CBB based on previous transcriptomic data. In planta assays in a susceptible common bean genotype showed that induction of PvOFP7, PvAP2-ERF71, or PvExpansinA17 expression by dTALEs resulted in CBB symptom reduction. After PvOFP7 induction, in planta bacterial growth was reduced at early colonization stages, and RNA-seq analysis revealed up-regulation of cell wall formation and primary metabolism, together with major down-regulation of heat shock proteins. Our results demonstrated that PvOFP7 contributes to CBB resistance, and underlined the usefulness of dTALEs for functional validation of genes whose induction impacts Xanthomonas-plant interactions.
Collapse
Affiliation(s)
- Charlotte Gaudin
- Université Angers, Institut Agro, INRAE, IRHS, SFR QUASAV, F-49000 Angers, France
| | - Anne Preveaux
- Université Angers, Institut Agro, INRAE, IRHS, SFR QUASAV, F-49000 Angers, France
| | - Nathan Aubineau
- Université Angers, Institut Agro, INRAE, IRHS, SFR QUASAV, F-49000 Angers, France
| | - Damien Le Goff
- Université Angers, Institut Agro, INRAE, IRHS, SFR QUASAV, F-49000 Angers, France
| | - Marie-Agnès Jacques
- Université Angers, Institut Agro, INRAE, IRHS, SFR QUASAV, F-49000 Angers, France
| | - Nicolas W G Chen
- Université Angers, Institut Agro, INRAE, IRHS, SFR QUASAV, F-49000 Angers, France
| |
Collapse
|
22
|
Garcia DA, Pierre AF, Quirino L, Acharya G, Vasudevan A, Pei Y, Chung E, Chang JYH, Lee S, Endow M, Kuakini K, Bresnahan M, Chumpitaz M, Rajappan K, Parker S, Chivukula P, Boehme SA, Diaz-Trelles R. Lipid nanoparticle delivery of TALEN mRNA targeting LPA causes gene disruption and plasma lipoprotein(a) reduction in transgenic mice. Mol Ther 2025; 33:90-103. [PMID: 39563033 PMCID: PMC11764125 DOI: 10.1016/j.ymthe.2024.11.020] [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: 02/26/2024] [Revised: 08/11/2024] [Accepted: 11/15/2024] [Indexed: 11/21/2024] Open
Abstract
Lipoprotein(a), or Lp(a), is encoded by the LPA gene and is a causal genetic risk factor for cardiovascular disease. Individuals with high Lp(a) are at risk for cardiovascular morbidity and are refractory to standard lipid-lowering agents. Lp(a)-lowering therapies currently in clinical development require repetitive dosing, while a gene editing approach presents an opportunity for a single-dose treatment. In this study, mRNAs encoding transcription activator-like effector nucleases (TALENs) were designed to target human LPA for gene disruption and permanent Lp(a) reduction. TALEN mRNAs were screened in vitro and found to cause on-target gene editing and target protein reduction with minimal off-target editing. TALEN mRNAs were then encapsulated with LUNAR, a proprietary lipid nanoparticle (LNP), and administered to transgenic mice that expressed a human LPA transgene. A single dose of TALEN mRNA-LNPs reduced plasma Lp(a) levels in mice by over 80%, which was sustained for at least 5 weeks. Moreover, both standard and long-read next-generation sequencing confirmed the presence of gene-inactivating deletions at LPA transgene loci. Overall, this study serves as a proof-of-concept for using TALEN-mediated gene editing to disrupt LPA in vivo, paving the way for the development of a feasible gene editing therapy for patients with high Lp(a).
Collapse
Affiliation(s)
- Daniel A Garcia
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA.
| | - Abigail F Pierre
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| | - Linda Quirino
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| | - Grishma Acharya
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| | - Aishwarya Vasudevan
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| | - Yihua Pei
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| | - Emily Chung
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| | - Jason Y H Chang
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| | - Samuel Lee
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| | - Michael Endow
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| | - Kristen Kuakini
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| | - Michael Bresnahan
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| | - Maria Chumpitaz
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| | - Kumar Rajappan
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| | - Suezanne Parker
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| | - Pad Chivukula
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| | - Stefen A Boehme
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA.
| | - Ramon Diaz-Trelles
- Arcturus Therapeutics, Inc., 10628 Science Center Drive, Suite 250, San Diego, CA 92121, USA
| |
Collapse
|
23
|
Jacobs R, Singh P, Smith T, Arbuthnot P, Maepa MB. Prospects of viral vector-mediated delivery of sequences encoding anti-HBV designer endonucleases. Gene Ther 2025; 32:8-15. [PMID: 35606493 DOI: 10.1038/s41434-022-00342-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 11/09/2022]
Abstract
Available treatment for chronic hepatitis B virus (HBV) infection offers modest functional curative efficacy. The viral replicative intermediate comprising covalently closed circular DNA (cccDNA) is responsible for persistent chronic HBV infection. Hence, current efforts have focused on developing therapies that disable cccDNA. Employing gene editing tools has emerged as an attractive strategy, with the end goal of establishing permanently inactivated cccDNA. Although anti-HBV designer nucleases are effective in vivo, none has yet progressed to clinical trial. Lack of safe and efficient delivery systems remains the limiting factor. Several vectors may be used to deliver anti-HBV gene editor-encoding sequences, with viral vectors being at the forefront. Despite the challenges associated with packaging large gene editor-encoding sequences into viral vectors, advancement in the field is overcoming such limitations. Translation of viral vector-mediated gene editing against HBV to clinical application is within reach. This review discusses the prospects of delivering HBV targeted designer nucleases using viral vectors.
Collapse
Affiliation(s)
- Ridhwaanah Jacobs
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Prashika Singh
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Tiffany Smith
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Patrick Arbuthnot
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Mohube Betty Maepa
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| |
Collapse
|
24
|
Engel-Pizcueta C, Hevia CF, Voltes A, Livet J, Pujades C. Her9 controls the stemness properties of hindbrain boundary cells. Development 2025; 152:dev203164. [PMID: 39628452 PMCID: PMC11829766 DOI: 10.1242/dev.203164] [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: 06/18/2024] [Accepted: 11/26/2024] [Indexed: 01/03/2025]
Abstract
The different spatiotemporal distribution of progenitor and neurogenic capacities permits that brain regions engage asynchronously in neurogenesis. In the hindbrain, rhombomere progenitor cells contribute to neurons during the first neurogenic phase, whereas boundary cells participate later. To analyze what maintains boundary cells as non-neurogenic progenitors, we addressed the role of Her9, a zebrafish Hes1-related protein. her9 expression is temporarily sustained in boundary cells independently of Notch at early embryonic stages, while they are non-neurogenic progenitors. Complementary functional approaches show that Her9 inhibits the onset of Notch signaling and the neurogenic program, keeping boundary cells as progenitors. Multicolor clonal analysis combined with genetic perturbations reveal that Her9 expands boundary progenitors by promoting symmetric proliferative and preventing neurogenic cell divisions. Her9 also regulates the proliferation of boundary cells by inhibiting the cell cycle arrest gene cdkn1ca and interplaying with Cyclin D1. Moreover, her9 is enriched in hindbrain radial glial cells at late embryonic stages independently of Notch. Together these data demonstrate that Her9 maintains the stemness properties of hindbrain boundary progenitors and late radial glial cells, ensuring the different temporal distribution of neurogenic capacities within the hindbrain.
Collapse
Affiliation(s)
- Carolyn Engel-Pizcueta
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - Covadonga F. Hevia
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - Adrià Voltes
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - Jean Livet
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Cristina Pujades
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| |
Collapse
|
25
|
Bilal M, Geng J, Chen L, García-Caparros P, Hu T. Genome editing for grass improvement and future agriculture. HORTICULTURE RESEARCH 2025; 12:uhae293. [PMID: 39906167 PMCID: PMC11789526 DOI: 10.1093/hr/uhae293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 10/06/2024] [Indexed: 02/06/2025]
Abstract
Grasses, including turf and forage, cover most of the earth's surface; predominantly important for land, water, livestock feed, soil, and water conservation, as well as carbon sequestration. Improved production and quality of grasses by modern molecular breeding is gaining more research attention. Recent advances in genome-editing technologies are helping to revolutionize plant breeding and also offering smart and efficient acceleration on grass improvement. Here, we reviewed all recent researches using (CRISPR)/CRISPR-associated protein (Cas)-mediated genome editing tools to enhance the growth and quality of forage and turf grasses. Furthermore, we highlighted emerging approaches aimed at advancing grass breeding program. We assessed the CRISPR-Cas effectiveness, discussed the challenges associated with its application, and explored future perspectives primarily focusing on turf and forage grasses. Despite the promising potential of genome editing in grasses, its current efficiency remains limited due to several bottlenecks, such as the absence of comprehensive reference genomes, the lack of efficient gene delivery tools, unavailability of suitable vector and delivery for grass species, high polyploidization, and multiple homoeoalleles, etc. Despite these challenges, the CRISPR-Cas system holds great potential to fully harness its benefits in grass breeding and genetics, aiming to improve and sustain the quantity and quality of turf and forage grasses.
Collapse
Affiliation(s)
- Muhammad Bilal
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou 730020, China
| | - Jie Geng
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou 730020, China
| | - Lin Chen
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Pedro García-Caparros
- Agronomy Department of Superior School Engineering, University of Almería, Almeria, Spain
| | - Tao Hu
- State Key Laboratory of Herbage Improvement and Grassland Agro-ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou 730020, China
| |
Collapse
|
26
|
Nanasato Y, Kawabe H, Ueno S, Konagaya KI, Endo M, Taniguchi T. Improvement of genome editing efficiency by Cas9 codon optimization in Japanese cedar ( Cryptomeria japonica D. Don). PLANT BIOTECHNOLOGY (TOKYO, JAPAN) 2024; 41:335-344. [PMID: 40083578 PMCID: PMC11897717 DOI: 10.5511/plantbiotechnology.24.0709a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 07/09/2024] [Indexed: 03/16/2025]
Abstract
Japanese cedar or sugi (Cryptomeria japonica D. Don) is among the most important plantation conifers in Japan, occupying 12% of the total land area in the country. We have successfully established a CRISPR/Cas9-based genome editing system in C. japonica. However, in practical use, we encountered problems of low efficiency when generating biallelic mutations, i.e., target gene knockouts. As part of our efforts to improve efficiency, we codon-optimized the Cas9 gene, evaluated by the genome editing efficiency of CjChl I, a gene encoding a chlorophyll biosynthesis enzyme. As a result, our codon-optimized SpCas9, named CjSpCas9, performed the highest genome editing efficiency of two targets (t4, t1+t2). Specifically, the biallelic disruption efficiency of the CjChl I with CjSpCas9 was 1.8-fold higher than that of the SpCas9 gene optimized for Arabidopsis thaliana (AtSpCas9) and 2.0-fold higher than that of the SpCas9 gene optimized for Orysa sativa (OsSpCas9) for t4, respectively. For t1+t2, the efficiency was 4.9-fold higher than that of AtSpCas9 and 1.4-fold higher than that of OsSpCas9, respectively. Our western blotting analysis proved that the Cas9 protein accumulation increased upon codon frequency optimization. We concluded that the observed efficiency improvement was due to the increased Cas9 protein quantity. The efficient genome editing system we report here would accelerate molecular breeding in conifers.
Collapse
Affiliation(s)
- Yoshihiko Nanasato
- Forest Bio-Research Center, Forestry and Forest Products Research Institute (FFPRI), Forest Research and Management Organization (FRMO), 3809-1 Ishi, Juo, Hitachi, Ibaraki 319-1301, Japan
| | - Harunori Kawabe
- Forest Bio-Research Center, Forestry and Forest Products Research Institute (FFPRI), Forest Research and Management Organization (FRMO), 3809-1 Ishi, Juo, Hitachi, Ibaraki 319-1301, Japan
| | - Saneyoshi Ueno
- Department of Forest Molecular Genetics and Biotechnology, FFPRI, FRMO, 1 Matsunosato, Tsukuba, Ibaraki 305-8687, Japan
| | - Ken-ichi Konagaya
- Forest Bio-Research Center, Forestry and Forest Products Research Institute (FFPRI), Forest Research and Management Organization (FRMO), 3809-1 Ishi, Juo, Hitachi, Ibaraki 319-1301, Japan
| | - Masaki Endo
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, 1-2 Owashi, Tsukuba, Ibaraki 305-8634, Japan
| | - Toru Taniguchi
- Forest Bio-Research Center, Forestry and Forest Products Research Institute (FFPRI), Forest Research and Management Organization (FRMO), 3809-1 Ishi, Juo, Hitachi, Ibaraki 319-1301, Japan
| |
Collapse
|
27
|
Defossez PA. Chromatin and transcription in Nucleic Acids Research: the first 50 years. Nucleic Acids Res 2024; 52:13485-13489. [PMID: 39607690 DOI: 10.1093/nar/gkae1151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Affiliation(s)
- Pierre-Antoine Defossez
- Université Paris Cité, CNRS, Epigenetics and Cell Fate, 35 Rue H. Brion, F-75013 Paris, France
| |
Collapse
|
28
|
Zhang L, Zhou J. Zebrafish: A smart tool for heart disease research. JOURNAL OF FISH BIOLOGY 2024; 105:1487-1500. [PMID: 37824489 DOI: 10.1111/jfb.15585] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 09/07/2023] [Accepted: 10/09/2023] [Indexed: 10/14/2023]
Abstract
The increasing prevalence of heart disease poses a significant threat to human survival and safety. However, the current treatments available for heart disease are quite limited. Therefore, it is important to utilize suitable animal models that can accurately simulate the physiological characteristics of heart disease. This would help improve our understanding of this disease and aid in the development of new treatment methods and drugs. Zebrafish heart not only exhibits similarities to mammalian hearts, but they also share ~70% of homologous genes with humans. Utilizing zebrafish as an alternative to expensive and time-consuming mammalian models offers numerous advantages. Zebrafish models can be easily established and maintained, and compound screening and genetic methods allow for the development of various economical and easily controlled zebrafish and zebrafish embryonic heart disease models in a short period of time. Consequently, zebrafish have become a powerful tool for exploring the pathological mechanisms of heart disease and identifying new effective genes. In this review, we summarize recent studies on different zebrafish models of heart disease. We also describe the techniques and protocols used to develop zebrafish models of myocardial infarction, heart failure, and congenital heart disease, including surgical procedures, forward and reverse genetics, and drug and combination screening. This review aims to promote the utilization of zebrafish models in investigating diverse pathological mechanisms of heart disease, enhancing our knowledge and comprehension of heart disease, and offering novel insights and objectives for exploring the prevention and treatment of heart disease.
Collapse
Affiliation(s)
- Lantian Zhang
- Education Branch, Chongqing Publishing Group, Chongqing, China
| | - Jinrun Zhou
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Qingdao, China
| |
Collapse
|
29
|
Yu S, Ye X, Dai X, Dai X, Wang X, Zheng H, Zhong B. A strategy for improving the mechanical properties of silk fibers through the combination of genetic manipulation and zinc ion crosslinking. Int J Biol Macromol 2024; 282:137075. [PMID: 39488310 DOI: 10.1016/j.ijbiomac.2024.137075] [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: 06/28/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024]
Abstract
Silk fiber is generally considered an excellent biological material due to its good biocompatibility, morphological plasticity and biodegradability. Previously, the construction of silkworm silk gland bioreactors based on the piggyBac transposon has been optimized. However, the inserted exogenous genes have problems such as position uncertainty, and expression is not strictly controlled. Here, we applied transcription activator-like effector nuclease (TALEN)-mediated homology-directed repair (HDR) to precisely insert histidine-rich cuticular protein (CP) into silkworm Sericin1 (Ser1) gene. The Ser1-CP fusion protein was successfully secreted into cocoon shell. Subsequently, based on the metal coordination ability of the histidine imidazole group, we crosslinked cocoon with metal ions in vitro. In this strategy, the mechanical properties of the fused silk fibers with crosslinked Zn2+ improved, and the maximum breaking stress of the crosslinked Zn2+-fused silk fibers was 23.5 % greater than that of the wild-type fibers. Analysis of the secondary structure of the silk protein showed that the fused silk fibers crosslinked with Zn2+ had more β-sheet structures. This study pioneered a method of improving the mechanical properties of silk fibers by crosslinking metal ions with fused exogenous proteins and expanded the application value of silk gland bioreactors in the development of novel biomaterials.
Collapse
Affiliation(s)
- Shihua Yu
- College of Animal Sciences, Zhejiang University, 310058 Hangzhou, China; Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, 310058 Hangzhou, China
| | - Xiaogang Ye
- College of Animal Sciences, Zhejiang University, 310058 Hangzhou, China; Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, 310058 Hangzhou, China
| | - Xiaoyan Dai
- Suposik Bioscience Technologies Ltd., 314031 Jiaxing, China
| | - Xiangping Dai
- College of Animal Sciences, Zhejiang University, 310058 Hangzhou, China; Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, 310058 Hangzhou, China
| | - Xinqiu Wang
- College of Animal Sciences, Zhejiang University, 310058 Hangzhou, China; Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, 310058 Hangzhou, China
| | - Huoqing Zheng
- College of Animal Sciences, Zhejiang University, 310058 Hangzhou, China; Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, 310058 Hangzhou, China
| | - Boxiong Zhong
- College of Animal Sciences, Zhejiang University, 310058 Hangzhou, China; Suposik Bioscience Technologies Ltd., 314031 Jiaxing, China; Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, 310058 Hangzhou, China.
| |
Collapse
|
30
|
Castillo SR, Simone BW, Clark KJ, Devaux P, Ekker SC. Unconstrained Precision Mitochondrial Genome Editing with αDdCBEs. Hum Gene Ther 2024; 35:798-813. [PMID: 39212664 PMCID: PMC11511777 DOI: 10.1089/hum.2024.073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
DddA-derived cytosine base editors (DdCBEs) enable the targeted introduction of C•G-to-T•A conversions in mitochondrial DNA (mtDNA). DdCBEs work in pairs, with each arm composed of a transcription activator-like effector (TALE), a split double-stranded DNA deaminase half, and a uracil glycosylase inhibitor. This pioneering technology has helped improve our understanding of cellular processes involving mtDNA and has paved the way for the development of models and therapies for genetic disorders caused by pathogenic mtDNA variants. Nonetheless, given the intrinsic properties of TALE proteins, several target sites in human mtDNA are predicted to remain out of reach to DdCBEs and other TALE-based technologies. Specifically, due to the conventional requirement for a thymine immediately upstream of the TALE target sequences (i.e., the 5'-T constraint), over 150 loci in the human mitochondrial genome are presumed to be inaccessible to DdCBEs. Previous attempts at circumventing this requirement, either by developing monomeric DdCBEs or utilizing DNA-binding domains alternative to TALEs, have resulted in suboptimal specificity profiles with reduced therapeutic potential. Here, aiming to challenge and elucidate the relevance of the 5'-T constraint in the context of DdCBE-mediated mtDNA editing, and to expand the range of motifs that are editable by this technology, we generated DdCBEs containing TALE proteins engineered to recognize all 5' bases. These modified DdCBEs are herein referred to as αDdCBEs. Notably, 5'-T-noncompliant canonical DdCBEs efficiently edited mtDNA at diverse loci. However, they were frequently outperformed by αDdCBEs, which exhibited significant improvements in activity and specificity, regardless of the most 5' bases of their TALE binding sites. Furthermore, we showed that αDdCBEs are compatible with the enhanced DddAtox variants DddA6 and DddA11, and we validated TALE shifting with αDdCBEs as an effective approach to optimize base editing outcomes. Overall, αDdCBEs enable efficient, specific, and unconstrained mitochondrial base editing.
Collapse
Affiliation(s)
- Santiago R. Castillo
- Virology and Gene Therapy Graduate Program, Mayo Clinic, Rochester, Minnesota, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Brandon W. Simone
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Karl J. Clark
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Patricia Devaux
- Virology and Gene Therapy Graduate Program, Mayo Clinic, Rochester, Minnesota, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Stephen C. Ekker
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Pediatrics and Department of Molecular Biosciences, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
31
|
Hu X, Zhang X, Sun W, Liu C, Deng P, Cao Y, Zhang C, Xu N, Zhang T, Zhang Y, Liu JJ, Wang H. Systematic discovery of DNA-binding tandem repeat proteins. Nucleic Acids Res 2024; 52:10464-10489. [PMID: 39189466 PMCID: PMC11417379 DOI: 10.1093/nar/gkae710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/30/2024] [Accepted: 08/07/2024] [Indexed: 08/28/2024] Open
Abstract
Tandem repeat proteins (TRPs) are widely distributed and bind to a wide variety of ligands. DNA-binding TRPs such as zinc finger (ZNF) and transcription activator-like effector (TALE) play important roles in biology and biotechnology. In this study, we first conducted an extensive analysis of TRPs in public databases, and found that the enormous diversity of TRPs is largely unexplored. We then focused our efforts on identifying novel TRPs possessing DNA-binding capabilities. We established a protein language model for DNA-binding protein prediction (PLM-DBPPred), and predicted a large number of DNA-binding TRPs. A subset was then selected for experimental screening, leading to the identification of 11 novel DNA-binding TRPs, with six showing sequence specificity. Notably, members of the STAR (Short TALE-like Repeat proteins) family can be programmed to target specific 9 bp DNA sequences with high affinity. Leveraging this property, we generated artificial transcription factors using reprogrammed STAR proteins and achieved targeted activation of endogenous gene sets. Furthermore, the members of novel families such as MOON (Marine Organism-Originated DNA binding protein) and pTERF (prokaryotic mTERF-like protein) exhibit unique features and distinct DNA-binding characteristics, revealing interesting biological clues. Our study expands the diversity of DNA-binding TRPs, and demonstrates that a systematic approach greatly enhances the discovery of new biological insights and tools.
Collapse
Affiliation(s)
- Xiaoxuan Hu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Xuechun Zhang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Wen Sun
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Chunhong Liu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Pujuan Deng
- State Key Laboratory of Membrane Biology, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yuanwei Cao
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Chenze Zhang
- National Key Laboratory of Efficacy and Mechanism on Chinese Medicine for Metabolic Diseases, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Ning Xu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Tongtong Zhang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Yong E Zhang
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jun-Jie Gogo Liu
- State Key Laboratory of Membrane Biology, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Haoyi Wang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| |
Collapse
|
32
|
Guo Z, Su Y, Huang WK, Yao XS, Hong Y, Gordin A, Nguyen HN, Wen Z, Ringeling FR, Chen G, Li S, Lu L, Xia M, Zheng W, Sawa A, Chen G, Christian KM, Song H, Ming GL. GABAergic neuron dysregulation in a human neurodevelopmental model for major psychiatric disorders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614564. [PMID: 39372772 PMCID: PMC11451812 DOI: 10.1101/2024.09.23.614564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
"GABA dysfunction" is a major hypothesis for the biological basis of schizophrenia with indirect supporting evidence from human post-mortem brain and genetic studies. Patient-derived induced pluripotent stem cells (iPSCs) have emerged as a valuable platform for modeling psychiatric disorders, and previous modeling has revealed glutamatergic synapse deficits. Whether GABAergic synapse properties are affected in patient-derived human neurons and how this impacts neuronal network activity remain poorly understood. Here we optimized a protocol to differentiate iPSCs into highly enriched ganglionic eminence-like neural progenitors and GABAergic neurons. Using a collection of iPSCs derived from patients of psychiatric disorders carrying a Disrupted-in-Schizophrenia 1 ( DISC1 ) mutation and their unaffected family member, together with respective isogenic lines, we identified mutation-dependent deficits in GABAergic synapse formation and function, a phenotype similar to that of mutant glutamatergic neurons. However, mutant glutamatergic and GABAergic neurons contribute differentially to neuronal network excitability and synchrony deficits. Finally, we showed that GABAergic synaptic transmission is also defective in neurons derived from several idiopathic schizophrenia patient iPSCs. Transcriptome analysis further showed some shared gene expression dysregulation, which is more prominent in DISC1 mutant neurons. Together, our study supports a functional GABAergic synaptic deficit in major psychiatric disorders.
Collapse
|
33
|
Rodriguez-Villamil P, Beaton BP, Krisher RL. Gene editing in livestock: innovations and applications. Anim Reprod 2024; 21:e20240054. [PMID: 39372257 PMCID: PMC11452096 DOI: 10.1590/1984-3143-ar2024-0054] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/05/2024] [Indexed: 10/08/2024] Open
Abstract
Gene editing technologies have revolutionized the field of livestock breeding, offering unprecedented opportunities to enhance animal welfare, productivity, and sustainability. This paper provides a comprehensive review of recent innovations and applications of gene editing in livestock, exploring the diverse applications of gene editing in livestock breeding, as well as the regulatory and ethical considerations, and the current challenges and prospects of the technology in the industry. Overall, this review underscores the transformative potential of gene editing in livestock breeding and its pivotal role in shaping the future of agriculture and biomedicine.
Collapse
|
34
|
Ng BW, Kaukonen MK, McClements ME, Shamsnajafabadi H, MacLaren RE, Cehajic-Kapetanovic J. Genetic therapies and potential therapeutic applications of CRISPR activators in the eye. Prog Retin Eye Res 2024; 102:101289. [PMID: 39127142 DOI: 10.1016/j.preteyeres.2024.101289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
Conventional gene therapy involving supplementation only treats loss-of-function diseases and is limited by viral packaging sizes, precluding therapy of large genes. The discovery of CRISPR/Cas has led to a paradigm shift in the field of genetic therapy, with the promise of precise gene editing, thus broadening the range of diseases that can be treated. The initial uses of CRISPR/Cas have focused mainly on gene editing or silencing of abnormal variants via utilising Cas endonuclease to trigger the target cell endogenous non-homologous end joining. Subsequently, the technology has evolved to modify the Cas enzyme and even its guide RNA, leading to more efficient editing tools in the form of base and prime editing. Further advancements of this CRISPR/Cas technology itself have expanded its functional repertoire from targeted editing to programmable transactivation, shifting the therapeutic focus to precise endogenous gene activation or upregulation with the potential for epigenetic modifications. In vivo experiments using this platform have demonstrated the potential of CRISPR-activators (CRISPRa) to treat various loss-of-function diseases, as well as in regenerative medicine, highlighting their versatility to overcome limitations associated with conventional strategies. This review summarises the molecular mechanisms of CRISPRa platforms, the current applications of this technology in vivo, and discusses potential solutions to translational hurdles for this therapy, with a focus on ophthalmic diseases.
Collapse
Affiliation(s)
- Benjamin Wj Ng
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Maria K Kaukonen
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK; Department of Medical and Clinical Genetics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Michelle E McClements
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Hoda Shamsnajafabadi
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Robert E MacLaren
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Jasmina Cehajic-Kapetanovic
- Oxford Eye Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK.
| |
Collapse
|
35
|
Roeschlin RA, Azad SM, Grove RP, Chuan A, García L, Niñoles R, Uviedo F, Villalobos L, Massimino ME, Marano MR, Boch J, Gadea J. Designer TALEs enable discovery of cell death-inducer genes. PLANT PHYSIOLOGY 2024; 195:2985-2996. [PMID: 38723194 PMCID: PMC11288752 DOI: 10.1093/plphys/kiae230] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 03/26/2024] [Indexed: 08/02/2024]
Abstract
Transcription activator-like effectors (TALEs) in plant-pathogenic Xanthomonas bacteria activate expression of plant genes and support infection or cause a resistance response. PthA4AT is a TALE with a particularly short DNA-binding domain harboring only 7.5 repeats which triggers cell death in Nicotiana benthamiana; however, the genetic basis for this remains unknown. To identify possible target genes of PthA4AT that mediate cell death in N. benthamiana, we exploited the modularity of TALEs to stepwise enhance their specificity and reduce potential target sites. Substitutions of individual repeats suggested that PthA4AT-dependent cell death is sequence specific. Stepwise addition of repeats to the C-terminal or N-terminal end of the repeat region narrowed the sequence requirements in promoters of target genes. Transcriptome profiling and in silico target prediction allowed the isolation of two cell death inducer genes, which encode a patatin-like protein and a bifunctional monodehydroascorbate reductase/carbonic anhydrase protein. These two proteins are not linked to known TALE-dependent resistance genes. Our results show that the aberrant expression of different endogenous plant genes can cause a cell death reaction, which supports the hypothesis that TALE-dependent executor resistance genes can originate from various plant processes. Our strategy further demonstrates the use of TALEs to scan genomes for genes triggering cell death and other relevant phenotypes.
Collapse
Affiliation(s)
- Roxana A Roeschlin
- Instituto de Biología Molecular y Celular de Rosario (IBR)-Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Ocampo y Esmeralda S/n, S2002LRK, Rosario, Argentina
| | - Sepideh M Azad
- Instituto de Biología Molecular y celular de Plantas (IBMCP), Universidad Politécnica de Valencia-CSIC, Ingeniero Fausto Elio S/N., 46022, Valencia, España
| | - René P Grove
- Institute of Plant Genetics, Leibniz Universität Hannover, 30419 Hannover, Germany
| | - Ana Chuan
- Instituto de Biología Molecular y celular de Plantas (IBMCP), Universidad Politécnica de Valencia-CSIC, Ingeniero Fausto Elio S/N., 46022, Valencia, España
| | - Lucila García
- Instituto de Biología Molecular y Celular de Rosario (IBR)-Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Ocampo y Esmeralda S/n, S2002LRK, Rosario, Argentina
- Área Virología, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario (UNR), Suipacha 590, S2002LRK, Rosario, Argentina
| | - Regina Niñoles
- Instituto de Biología Molecular y celular de Plantas (IBMCP), Universidad Politécnica de Valencia-CSIC, Ingeniero Fausto Elio S/N., 46022, Valencia, España
| | - Facundo Uviedo
- Instituto de Biología Molecular y Celular de Rosario (IBR)-Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Ocampo y Esmeralda S/n, S2002LRK, Rosario, Argentina
| | - Liara Villalobos
- Instituto de Biología Molecular y Celular de Rosario (IBR)-Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Ocampo y Esmeralda S/n, S2002LRK, Rosario, Argentina
| | - Maria E Massimino
- Instituto de Biología Molecular y celular de Plantas (IBMCP), Universidad Politécnica de Valencia-CSIC, Ingeniero Fausto Elio S/N., 46022, Valencia, España
| | - María R Marano
- Instituto de Biología Molecular y Celular de Rosario (IBR)-Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Ocampo y Esmeralda S/n, S2002LRK, Rosario, Argentina
- Área Virología, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario (UNR), Suipacha 590, S2002LRK, Rosario, Argentina
| | - Jens Boch
- Institute of Plant Genetics, Leibniz Universität Hannover, 30419 Hannover, Germany
| | - José Gadea
- Instituto de Biología Molecular y celular de Plantas (IBMCP), Universidad Politécnica de Valencia-CSIC, Ingeniero Fausto Elio S/N., 46022, Valencia, España
| |
Collapse
|
36
|
Teles D, Fine BM. Using induced pluripotent stem cells for drug discovery in arrhythmias. Expert Opin Drug Discov 2024; 19:827-840. [PMID: 38825838 PMCID: PMC11227103 DOI: 10.1080/17460441.2024.2360420] [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: 03/18/2024] [Accepted: 05/23/2024] [Indexed: 06/04/2024]
Abstract
INTRODUCTION Arrhythmias are disturbances in the normal rhythm of the heart and account for significant cardiovascular morbidity and mortality worldwide. Historically, preclinical research has been anchored in animal models, though physiological differences between these models and humans have limited their clinical translation. The discovery of human induced pluripotent stem cells (iPSC) and subsequent differentiation into cardiomyocyte has led to the development of new in vitro models of arrhythmias with the hope of a new pathway for both exploration of pathogenic variants and novel therapeutic discovery. AREAS COVERED The authors describe the latest two-dimensional in vitro models of arrhythmias, several examples of the use of these models in drug development, and the role of gene editing when modeling diseases. They conclude by discussing the use of three-dimensional models in the study of arrythmias and the integration of computational technologies and machine learning with experimental technologies. EXPERT OPINION Human iPSC-derived cardiomyocytes models have significant potential to augment disease modeling, drug discovery, and toxicity studies in preclinical development. While there is initial success with modeling arrhythmias, the field is still in its nascency and requires advances in maturation, cellular diversity, and readouts to emulate arrhythmias more accurately.
Collapse
Affiliation(s)
- Diogo Teles
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Barry M. Fine
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
37
|
Ebrahimi V, Hashemi A. CRISPR-based gene editing in plants: Focus on reagents and their delivery tools. BIOIMPACTS : BI 2024; 15:30019. [PMID: 39963563 PMCID: PMC11830140 DOI: 10.34172/bi.30019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 01/16/2024] [Accepted: 02/06/2024] [Indexed: 02/20/2025]
Abstract
Introduction CRISPR-Cas9 technology has revolutionized plant genome editing, providing precise and efficient methods for genetic modification. This study focuses on the advancements and delivery of CRISPR-Cas9 in plant gene editing. Methods A comprehensive search in scientific databases, including PubMed, ScienceDirect, and Google Scholar, was conducted to gather information on CRISPR-Cas9 gene editing and its delivery in precise gene modification in plants. Results The evolving landscape of CRISPR nucleases has led to the development of innovative technologies, enhancing plant research. However, successful editing is contingent on efficient delivery of genome engineering reagents. CRISPR-based gene editing in plants utilizes diverse delivery methods: Agrobacterium-mediated transformation for bacterial transfer, biolistic transformation for physical gene insertion, electroporation for direct gene entry, expression of developmental regulators for gene expression modulation, and tobacco rattle virus as a viral vector, each offering distinct advantages for precise and efficient genetic modification in plants. Conclusion CRISPR-Cas9 gene editing stands as a pivotal advancement in plant genetics, offering precise gene manipulation with applications in agriculture and biotechnology. The continuous refinement of reagent delivery tools reinforces CRISPR-Cas9's transformative role in plant genome editing, with significant implications for broader scientific applications.
Collapse
Affiliation(s)
- Vida Ebrahimi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atieh Hashemi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
38
|
Villiger L, Joung J, Koblan L, Weissman J, Abudayyeh OO, Gootenberg JS. CRISPR technologies for genome, epigenome and transcriptome editing. Nat Rev Mol Cell Biol 2024; 25:464-487. [PMID: 38308006 DOI: 10.1038/s41580-023-00697-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2023] [Indexed: 02/04/2024]
Abstract
Our ability to edit genomes lags behind our capacity to sequence them, but the growing understanding of CRISPR biology and its application to genome, epigenome and transcriptome engineering is narrowing this gap. In this Review, we discuss recent developments of various CRISPR-based systems that can transiently or permanently modify the genome and the transcriptome. The discovery of further CRISPR enzymes and systems through functional metagenomics has meaningfully broadened the applicability of CRISPR-based editing. Engineered Cas variants offer diverse capabilities such as base editing, prime editing, gene insertion and gene regulation, thereby providing a panoply of tools for the scientific community. We highlight the strengths and weaknesses of current CRISPR tools, considering their efficiency, precision, specificity, reliance on cellular DNA repair mechanisms and their applications in both fundamental biology and therapeutics. Finally, we discuss ongoing clinical trials that illustrate the potential impact of CRISPR systems on human health.
Collapse
Affiliation(s)
- Lukas Villiger
- McGovern Institute for Brain Research, Massachusetts Institute of Technology Cambridge, Cambridge, MA, USA
| | - Julia Joung
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Luke Koblan
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jonathan Weissman
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Omar O Abudayyeh
- McGovern Institute for Brain Research, Massachusetts Institute of Technology Cambridge, Cambridge, MA, USA.
| | - Jonathan S Gootenberg
- McGovern Institute for Brain Research, Massachusetts Institute of Technology Cambridge, Cambridge, MA, USA.
| |
Collapse
|
39
|
Yu S, Zheng H, Ye X, Dai X, Wang X, Zhao S, Dai X, Zhong B. TALEN-mediated homologous-recombination-based fibroin light chain in-fusion expression system in Bombyx mori. Front Bioeng Biotechnol 2024; 12:1399629. [PMID: 38832132 PMCID: PMC11144906 DOI: 10.3389/fbioe.2024.1399629] [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: 03/12/2024] [Accepted: 04/25/2024] [Indexed: 06/05/2024] Open
Abstract
Silkworm was the first domesticated insect and has important economic value. It has also become an ideal model organism with applications in genetic and expression studies. In recent years, the use of transgenic strategies has made the silkworm silk gland an attractive bioreactor for the production of recombinant proteins, in particular, piggyBac-mediated transgenes. However, owing to differences in regulatory elements such as promoters, the expression levels of exogenous proteins have not reached expectations. Here, we used targeted gene editing to achieve site-specific integration of exogenous genes on genomic DNA and established the fibroin light chain (FibL) in-fusion expression system by TALEN-mediated homology-directed recombination. First, the histidine-rich cuticular protein (CP) was successfully site-directed inserted into the native FibL, and the FibL-CP fusion gene was correctly transcribed and expressed in the posterior silk gland under the control of the endogenous FibL promoter, with a protein expression level comparable with that of the native FibL protein. Moreover, we showed based on molecular docking that the fusion of FibL with cuticular protein may have a negative effect on disulfide bond formation between the C-terminal domain of fibroin heavy chain (FibH) and FibL-CP, resulting in abnormal spinning and cocoon in homozygotes, indicating a significant role of FibL in silk protein formation and secretion. Our results demonstrate the feasibility of using the FibL fusion system to express exogenous proteins in silkworm. We expect that this bioreactor system will be used to produce more proteins of interest, expanding the application value of the silk gland bioreactor.
Collapse
Affiliation(s)
- Shihua Yu
- College of Animal Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Hangzhou, China
| | - Huoqing Zheng
- College of Animal Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Hangzhou, China
| | - Xiaogang Ye
- College of Animal Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Hangzhou, China
| | - Xiangping Dai
- College of Animal Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Hangzhou, China
| | - Xinqiu Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Hangzhou, China
| | - Shuo Zhao
- Department of Laboratory Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zheng Zhou, China
| | - Xiaoyan Dai
- Suposik Bioscience Technologies Ltd., Jiaxing, China
| | - Boxiong Zhong
- College of Animal Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Hangzhou, China
| |
Collapse
|
40
|
Morooka N, Gui N, Ando K, Sako K, Fukumoto M, Hasegawa U, Hußmann M, Schulte-Merker S, Mochizuki N, Nakajima H. Angpt1 binding to Tie1 regulates the signaling required for lymphatic vessel development in zebrafish. Development 2024; 151:dev202269. [PMID: 38742432 DOI: 10.1242/dev.202269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 04/26/2024] [Indexed: 05/16/2024]
Abstract
Development of the vascular system is regulated by multiple signaling pathways mediated by receptor tyrosine kinases. Among them, angiopoietin (Ang)/Tie signaling regulates lymphatic and blood vessel development in mammals. Of the two Tie receptors, Tie2 is well known as a key mediator of Ang/Tie signaling, but, unexpectedly, recent studies have revealed that the Tie2 locus has been lost in many vertebrate species, whereas the Tie1 gene is more commonly present. However, Tie1-driven signaling pathways, including ligands and cellular functions, are not well understood. Here, we performed comprehensive mutant analyses of angiopoietins and Tie receptors in zebrafish and found that only angpt1 and tie1 mutants show defects in trunk lymphatic vessel development. Among zebrafish angiopoietins, only Angpt1 binds to Tie1 as a ligand. We indirectly monitored Ang1/Tie1 signaling and detected Tie1 activation in sprouting endothelial cells, where Tie1 inhibits nuclear import of EGFP-Foxo1a. Angpt1/Tie1 signaling functions in endothelial cell migration and proliferation, and in lymphatic specification during early lymphangiogenesis, at least in part by modulating Vegfc/Vegfr3 signaling. Thus, we show that Angpt1/Tie1 signaling constitutes an essential signaling pathway for lymphatic development in zebrafish.
Collapse
Affiliation(s)
- Nanami Morooka
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan
- Department of Medical Physiology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| | - Ning Gui
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan
| | - Koji Ando
- Department of Cardiac Regeneration Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan
| | - Keisuke Sako
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan
| | - Moe Fukumoto
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan
| | - Urara Hasegawa
- Department of Materials Science and Engineering, The Pennsylvania State University, Steidle Building, University Park, Pennsylvania 16802, United States
| | - Melina Hußmann
- Institute of Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WU Münster, 48149 Münster, Germany
| | - Stefan Schulte-Merker
- Institute of Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WU Münster, 48149 Münster, Germany
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan
| | - Hiroyuki Nakajima
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan
| |
Collapse
|
41
|
Arimura SI, Nakazato I. Genome Editing of Plant Mitochondrial and Chloroplast Genomes. PLANT & CELL PHYSIOLOGY 2024; 65:477-483. [PMID: 38113380 PMCID: PMC11094758 DOI: 10.1093/pcp/pcad162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/30/2023] [Accepted: 12/16/2023] [Indexed: 12/21/2023]
Abstract
Plastids (including chloroplasts) and mitochondria are remnants of endosymbiotic bacteria, yet they maintain their own genomes, which encode vital components for photosynthesis and respiration, respectively. Organellar genomes have distinctive features, such as being present as multicopies, being mostly inherited maternally, having characteristic genomic structures and undergoing frequent homologous recombination. To date, it has proven to be challenging to modify these genomes. For example, while CRISPR/Cas9 is a widely used system for editing nuclear genes, it has not yet been successfully applied to organellar genomes. Recently, however, precise gene-editing technologies have been successfully applied to organellar genomes. Protein-based enzymes, especially transcription activator-like effector nucleases (TALENs) and artificial enzymes utilizing DNA-binding domains of TALENs (TALEs), have been successfully used to modify these genomes by harnessing organellar-targeting signals. This short review introduces and discusses the use of targeted nucleases and base editors in organellar genomes, their effects and their potential applications in plant science and breeding.
Collapse
Affiliation(s)
- Shin-ichi Arimura
- Laboratory of Plant Molecular Genetics, Graduate School of Agricultural and Life Science, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo, 113-8657 Japan
| | - Issei Nakazato
- Laboratory of Plant Molecular Genetics, Graduate School of Agricultural and Life Science, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo, 113-8657 Japan
| |
Collapse
|
42
|
Castillo SR, Simone BW, Clark KJ, Devaux P, Ekker SC. Unconstrained Precision Mitochondrial Genome Editing with αDdCBEs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.13.593977. [PMID: 38798498 PMCID: PMC11118498 DOI: 10.1101/2024.05.13.593977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
DddA-derived cytosine base editors (DdCBEs) enable the targeted introduction of C•G-to-T•A conversions in mitochondrial DNA (mtDNA). DdCBEs are often deployed as pairs, with each arm comprised of a transcription activator-like effector (TALE), a split double-stranded DNA deaminase half, and a uracil glycosylase inhibitor. This pioneering technology has helped improve our understanding of cellular processes involving mtDNA and has paved the way for the development of models and therapies for genetic disorders caused by pathogenic mtDNA variants. Nonetheless, given the intrinsic properties of TALE proteins, several target sites in human mtDNA remain out of reach to DdCBEs and other TALE-based technologies. Specifically, due to the conventional requirement for a thymine immediately upstream of the TALE target sequences (i.e., the 5'-T constraint), over 150 loci in the human mitochondrial genome are presumed to be inaccessible to DdCBEs. Previous attempts at circumventing this constraint, either by developing monomeric DdCBEs or utilizing DNA-binding domains alternative to TALEs, have resulted in suboptimal specificity profiles with reduced therapeutic potential. Here, aiming to challenge and elucidate the relevance of the 5'-T constraint in the context of DdCBE-mediated mtDNA editing, and to expand the range of motifs that are editable by this technology, we generated αDdCBEs that contain modified TALE proteins engineered to recognize all 5' bases. Notably, 5'-T-noncompliant, canonical DdCBEs efficiently edited mtDNA at diverse loci. However, DdCBEs were frequently outperformed by αDdCBEs, which consistently displayed significant improvements in activity and specificity, regardless of the 5'-most bases of their TALE binding sites. Furthermore, we showed that αDdCBEs are compatible with DddA tox and its derivatives DddA6, and DddA11, and we validated TALE shifting with αDdCBEs as an effective approach to optimize base editing outcomes at a single target site. Overall, αDdCBEs enable efficient, specific, and unconstrained mitochondrial base editing.
Collapse
|
43
|
Wu M, Hau PM, Li L, Tsang CM, Yang Y, Taghbalout A, Chung GTY, Hui SY, Tang WC, Jillette N, Zhu JJ, Lee HHY, Kong EL, Chan MSA, Chan JYK, Ma BBY, Chen MR, Lee C, To KF, Cheng AW, Lo KW. Synthetic BZLF1-targeted transcriptional activator for efficient lytic induction therapy against EBV-associated epithelial cancers. Nat Commun 2024; 15:3729. [PMID: 38702330 PMCID: PMC11068728 DOI: 10.1038/s41467-024-48031-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 04/17/2024] [Indexed: 05/06/2024] Open
Abstract
The unique virus-cell interaction in Epstein-Barr virus (EBV)-associated malignancies implies targeting the viral latent-lytic switch is a promising therapeutic strategy. However, the lack of specific and efficient therapeutic agents to induce lytic cycle in these cancers is a major challenge facing clinical implementation. We develop a synthetic transcriptional activator that specifically activates endogenous BZLF1 and efficiently induces lytic reactivation in EBV-positive cancer cells. A lipid nanoparticle encapsulating nucleoside-modified mRNA which encodes a BZLF1-specific transcriptional activator (mTZ3-LNP) is synthesized for EBV-targeted therapy. Compared with conventional chemical inducers, mTZ3-LNP more efficiently activates EBV lytic gene expression in EBV-associated epithelial cancers. Here we show the potency and safety of treatment with mTZ3-LNP to suppress tumor growth in EBV-positive cancer models. The combination of mTZ3-LNP and ganciclovir yields highly selective cytotoxic effects of mRNA-based lytic induction therapy against EBV-positive tumor cells, indicating the potential of mRNA nanomedicine in the treatment of EBV-associated epithelial cancers.
Collapse
Affiliation(s)
- Man Wu
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Translational Oncology, Sir YK Pao Centre for Cancer, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Pok Man Hau
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Linxian Li
- Ming Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Shatin, Hong Kong SAR, China
- Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong SAR, China
| | - Chi Man Tsang
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Translational Oncology, Sir YK Pao Centre for Cancer, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yike Yang
- Ming Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Shatin, Hong Kong SAR, China
- College of Chemistry and Green Catalysis Center, Zhengzhou University, Zhengzhou, China
| | - Aziz Taghbalout
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Grace Tin-Yun Chung
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Shin Yee Hui
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wing Chung Tang
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | | | - Jacqueline Jufen Zhu
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85281, USA
| | - Horace Hok Yeung Lee
- Ming Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Shatin, Hong Kong SAR, China
| | - Ee Ling Kong
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Melissa Sue Ann Chan
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jason Ying Kuen Chan
- Department of Otorhinolaryngology, Head and Neck Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Brigette Buig Yue Ma
- Department of Clinical Oncology, State Key Laboratory of Translational Oncology, Charlie Lee Precision Immuno-oncology program, Sir Y.K. Pao Centre for Cancer, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Mei-Ru Chen
- Graduate Institute and Department of Microbiology, College of Medicine, National Taiwan University, Taipei, 100233, Taiwan
| | - Charles Lee
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Ka Fai To
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Translational Oncology, Sir YK Pao Centre for Cancer, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Albert Wu Cheng
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA.
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85281, USA.
- Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT, 06030, USA.
- Institute for Systems Genomics, University of Connecticut Health Center, Farmington, CT, 06030, USA.
- The Jackson Laboratory Cancer Center, Bar Harbor, ME, 04609, USA.
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Key Laboratory of Organ Regeneration and Reconstruction, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Kwok-Wai Lo
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China.
- State Key Laboratory of Translational Oncology, Sir YK Pao Centre for Cancer, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
44
|
Barriga FM, Lowe SW. Engineering megabase-sized genomic deletions with MACHETE (Molecular Alteration of Chromosomes with Engineered Tandem Elements). Nat Protoc 2024; 19:1381-1399. [PMID: 38326496 PMCID: PMC11642290 DOI: 10.1038/s41596-024-00953-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 11/21/2023] [Indexed: 02/09/2024]
Abstract
The elimination of large genomic regions has been enabled by the advent of site-specific nucleases. However, as the intended deletions get larger, the efficiency of successful engineering decreases to a point where it is not feasible to retrieve edited cells due to the rarity of on-target events. To address this issue, we developed a system called molecular alteration of chromosomes with engineered tandem elements (MACHETE). MACHETE is a CRISPR-Cas9-based system involving two stages: the initial insertion of a bicistronic positive/negative selection cassette to the locus of interest. This is followed by the introduction of single-guide RNAs flanking the knockin cassette to engineer the intended deletion, where only cells that have lost the locus survive the negative selection. In contrast to other approaches optimizing the activity of sequence-specific nucleases, MACHETE selects for the deletion event itself, thus greatly enriching for cells with the engineered alteration. The procedure routinely takes 4-6 weeks from design to selection of polyclonal populations bearing the deletion of interest. We have successfully deployed MACHETE to engineer deletions of up to 45 Mb, as well as the rapid creation of allelic series to map the relevant activities within a locus. This protocol details the design and step-by-step procedure to engineer megabase-sized deletions in cells of interest, with potential application for cancer genetics, transcriptional regulation, genome architecture and beyond.
Collapse
Affiliation(s)
- Francisco M Barriga
- Systems Oncology Program, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain.
| | - Scott W Lowe
- Cancer Biology and Genetics Program and Howard Hughes Medical Institute, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
45
|
Kieda J, Shakeri A, Landau S, Wang EY, Zhao Y, Lai BF, Okhovatian S, Wang Y, Jiang R, Radisic M. Advances in cardiac tissue engineering and heart-on-a-chip. J Biomed Mater Res A 2024; 112:492-511. [PMID: 37909362 PMCID: PMC11213712 DOI: 10.1002/jbm.a.37633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/26/2023] [Accepted: 10/13/2023] [Indexed: 11/03/2023]
Abstract
Recent advances in both cardiac tissue engineering and hearts-on-a-chip are grounded in new biomaterial development as well as the employment of innovative fabrication techniques that enable precise control of the mechanical, electrical, and structural properties of the cardiac tissues being modelled. The elongated structure of cardiomyocytes requires tuning of substrate properties and application of biophysical stimuli to drive its mature phenotype. Landmark advances have already been achieved with induced pluripotent stem cell-derived cardiac patches that advanced to human testing. Heart-on-a-chip platforms are now commonly used by a number of pharmaceutical and biotechnology companies. Here, we provide an overview of cardiac physiology in order to better define the requirements for functional tissue recapitulation. We then discuss the biomaterials most commonly used in both cardiac tissue engineering and heart-on-a-chip, followed by the discussion of recent representative studies in both fields. We outline significant challenges common to both fields, specifically: scalable tissue fabrication and platform standardization, improving cellular fidelity through effective tissue vascularization, achieving adult tissue maturation, and ultimately developing cryopreservation protocols so that the tissues are available off the shelf.
Collapse
Affiliation(s)
- Jennifer Kieda
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Amid Shakeri
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Shira Landau
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Erika Yan Wang
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Yimu Zhao
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Benjamin Fook Lai
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Sargol Okhovatian
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Ying Wang
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Richard Jiang
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Milica Radisic
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
46
|
Portolés I, Ribera J, Fernandez-Galán E, Lecue E, Casals G, Melgar-Lesmes P, Fernández-Varo G, Boix L, Sanduzzi M, Aishwarya V, Reig M, Jiménez W, Morales-Ruiz M. Identification of Dhx15 as a Major Regulator of Liver Development, Regeneration, and Tumor Growth in Zebrafish and Mice. Int J Mol Sci 2024; 25:3716. [PMID: 38612527 PMCID: PMC11011938 DOI: 10.3390/ijms25073716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/15/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
RNA helicase DHX15 plays a significant role in vasculature development and lung metastasis in vertebrates. In addition, several studies have demonstrated the overexpression of DHX15 in the context of hepatocellular carcinoma. Therefore, we hypothesized that this helicase may play a significant role in liver regeneration, physiology, and pathology. Dhx15 gene deficiency was generated by CRISPR/Cas9 in zebrafish and by TALEN-RNA in mice. AUM Antisense-Oligonucleotides were used to silence Dhx15 in wild-type mice. The hepatocellular carcinoma tumor induction model was generated by subcutaneous injection of Hepa 1-6 cells. Homozygous Dhx15 gene deficiency was lethal in zebrafish and mouse embryos. Dhx15 gene deficiency impaired liver organogenesis in zebrafish embryos and liver regeneration after partial hepatectomy in mice. Also, heterozygous mice presented decreased number and size of liver metastasis after Hepa 1-6 cells injection compared to wild-type mice. Dhx15 gene silencing with AUM Antisense-Oligonucleotides in wild-type mice resulted in 80% reduced expression in the liver and a significant reduction in other major organs. In addition, Dhx15 gene silencing significantly hindered primary tumor growth in the hepatocellular carcinoma experimental model. Regarding the potential use of DHX15 as a diagnostic marker for liver disease, patients with hepatocellular carcinoma showed increased levels of DHX15 in blood samples compared with subjects without hepatic affectation. In conclusion, Dhx15 is a key regulator of liver physiology and organogenesis, is increased in the blood of cirrhotic and hepatocellular carcinoma patients, and plays a key role in controlling hepatocellular carcinoma tumor growth and expansion in experimental models.
Collapse
Affiliation(s)
- Irene Portolés
- Biochemistry and Molecular Genetics Department-CDB, Hospital Clínic of Barcelona, Fundació de Recerca Clínic Barcelona-Institut d’Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), 170 Villarroel St. Barcelona, 08036 Barcelona, Spain; (I.P.); (J.R.); (E.F.-G.); (E.L.); (G.C.); (P.M.-L.); (G.F.-V.); (W.J.)
| | - Jordi Ribera
- Biochemistry and Molecular Genetics Department-CDB, Hospital Clínic of Barcelona, Fundació de Recerca Clínic Barcelona-Institut d’Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), 170 Villarroel St. Barcelona, 08036 Barcelona, Spain; (I.P.); (J.R.); (E.F.-G.); (E.L.); (G.C.); (P.M.-L.); (G.F.-V.); (W.J.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28222 Madrid, Spain; (L.B.); (M.S.); (M.R.)
| | - Esther Fernandez-Galán
- Biochemistry and Molecular Genetics Department-CDB, Hospital Clínic of Barcelona, Fundació de Recerca Clínic Barcelona-Institut d’Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), 170 Villarroel St. Barcelona, 08036 Barcelona, Spain; (I.P.); (J.R.); (E.F.-G.); (E.L.); (G.C.); (P.M.-L.); (G.F.-V.); (W.J.)
| | - Elena Lecue
- Biochemistry and Molecular Genetics Department-CDB, Hospital Clínic of Barcelona, Fundació de Recerca Clínic Barcelona-Institut d’Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), 170 Villarroel St. Barcelona, 08036 Barcelona, Spain; (I.P.); (J.R.); (E.F.-G.); (E.L.); (G.C.); (P.M.-L.); (G.F.-V.); (W.J.)
| | - Gregori Casals
- Biochemistry and Molecular Genetics Department-CDB, Hospital Clínic of Barcelona, Fundació de Recerca Clínic Barcelona-Institut d’Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), 170 Villarroel St. Barcelona, 08036 Barcelona, Spain; (I.P.); (J.R.); (E.F.-G.); (E.L.); (G.C.); (P.M.-L.); (G.F.-V.); (W.J.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28222 Madrid, Spain; (L.B.); (M.S.); (M.R.)
- Commission for the Biochemical Evaluation of the Hepatic Disease-SEQCML, 08036 Barcelona, Spain
| | - Pedro Melgar-Lesmes
- Biochemistry and Molecular Genetics Department-CDB, Hospital Clínic of Barcelona, Fundació de Recerca Clínic Barcelona-Institut d’Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), 170 Villarroel St. Barcelona, 08036 Barcelona, Spain; (I.P.); (J.R.); (E.F.-G.); (E.L.); (G.C.); (P.M.-L.); (G.F.-V.); (W.J.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28222 Madrid, Spain; (L.B.); (M.S.); (M.R.)
- Biomedicine Department, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
| | - Guillermo Fernández-Varo
- Biochemistry and Molecular Genetics Department-CDB, Hospital Clínic of Barcelona, Fundació de Recerca Clínic Barcelona-Institut d’Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), 170 Villarroel St. Barcelona, 08036 Barcelona, Spain; (I.P.); (J.R.); (E.F.-G.); (E.L.); (G.C.); (P.M.-L.); (G.F.-V.); (W.J.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28222 Madrid, Spain; (L.B.); (M.S.); (M.R.)
| | - Loreto Boix
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28222 Madrid, Spain; (L.B.); (M.S.); (M.R.)
- Barcelona Clinic Liver Cancer Group, Liver Unit, Hospital Clinic, University of Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Marco Sanduzzi
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28222 Madrid, Spain; (L.B.); (M.S.); (M.R.)
- Barcelona Clinic Liver Cancer Group, Liver Unit, Hospital Clinic, University of Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Veenu Aishwarya
- AUM LifeTech, Inc., 3675 Market Street, Suite 200, Philadelphia, PA 19104, USA;
| | - Maria Reig
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28222 Madrid, Spain; (L.B.); (M.S.); (M.R.)
- Barcelona Clinic Liver Cancer Group, Liver Unit, Hospital Clinic, University of Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Wladimiro Jiménez
- Biochemistry and Molecular Genetics Department-CDB, Hospital Clínic of Barcelona, Fundació de Recerca Clínic Barcelona-Institut d’Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), 170 Villarroel St. Barcelona, 08036 Barcelona, Spain; (I.P.); (J.R.); (E.F.-G.); (E.L.); (G.C.); (P.M.-L.); (G.F.-V.); (W.J.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28222 Madrid, Spain; (L.B.); (M.S.); (M.R.)
- Biomedicine Department, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
| | - Manuel Morales-Ruiz
- Biochemistry and Molecular Genetics Department-CDB, Hospital Clínic of Barcelona, Fundació de Recerca Clínic Barcelona-Institut d’Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), 170 Villarroel St. Barcelona, 08036 Barcelona, Spain; (I.P.); (J.R.); (E.F.-G.); (E.L.); (G.C.); (P.M.-L.); (G.F.-V.); (W.J.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28222 Madrid, Spain; (L.B.); (M.S.); (M.R.)
- Commission for the Biochemical Evaluation of the Hepatic Disease-SEQCML, 08036 Barcelona, Spain
- Biomedicine Department, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
| |
Collapse
|
47
|
Mabesoone MF, Leopold-Messer S, Minas HA, Chepkirui C, Chawengrum P, Reiter S, Meoded RA, Wolf S, Genz F, Magnus N, Piechulla B, Walker AS, Piel J. Evolution-guided engineering of trans-acyltransferase polyketide synthases. Science 2024; 383:1312-1317. [PMID: 38513027 PMCID: PMC11260071 DOI: 10.1126/science.adj7621] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 02/13/2024] [Indexed: 03/23/2024]
Abstract
Bacterial multimodular polyketide synthases (PKSs) are giant enzymes that generate a wide range of therapeutically important but synthetically challenging natural products. Diversification of polyketide structures can be achieved by engineering these enzymes. However, notwithstanding successes made with textbook cis-acyltransferase (cis-AT) PKSs, tailoring such large assembly lines remains challenging. Unlike textbook PKSs, trans-AT PKSs feature an extraordinary diversity of PKS modules and commonly evolve to form hybrid PKSs. In this study, we analyzed amino acid coevolution to identify a common module site that yields functional PKSs. We used this site to insert and delete diverse PKS parts and create 22 engineered trans-AT PKSs from various pathways and in two bacterial producers. The high success rates of our engineering approach highlight the broader applicability to generate complex designer polyketides.
Collapse
Affiliation(s)
- Mathijs F.J. Mabesoone
- Institute of Microbiology, Eidgenössische Technische Hochschule (ETH) Zürich, Vladimir-Prelog-Weg 4, 8093 Zürich, Switzerland
| | - Stefan Leopold-Messer
- Institute of Microbiology, Eidgenössische Technische Hochschule (ETH) Zürich, Vladimir-Prelog-Weg 4, 8093 Zürich, Switzerland
| | - Hannah A. Minas
- Institute of Microbiology, Eidgenössische Technische Hochschule (ETH) Zürich, Vladimir-Prelog-Weg 4, 8093 Zürich, Switzerland
| | - Clara Chepkirui
- Institute of Microbiology, Eidgenössische Technische Hochschule (ETH) Zürich, Vladimir-Prelog-Weg 4, 8093 Zürich, Switzerland
| | - Pornsuda Chawengrum
- Institute of Microbiology, Eidgenössische Technische Hochschule (ETH) Zürich, Vladimir-Prelog-Weg 4, 8093 Zürich, Switzerland
- Chemical Biology Program, Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok 10210, Thailand
| | - Silke Reiter
- Institute of Microbiology, Eidgenössische Technische Hochschule (ETH) Zürich, Vladimir-Prelog-Weg 4, 8093 Zürich, Switzerland
| | - Roy A. Meoded
- Institute of Microbiology, Eidgenössische Technische Hochschule (ETH) Zürich, Vladimir-Prelog-Weg 4, 8093 Zürich, Switzerland
| | - Sarah Wolf
- Institute of Microbiology, Eidgenössische Technische Hochschule (ETH) Zürich, Vladimir-Prelog-Weg 4, 8093 Zürich, Switzerland
| | - Ferdinand Genz
- Institute of Microbiology, Eidgenössische Technische Hochschule (ETH) Zürich, Vladimir-Prelog-Weg 4, 8093 Zürich, Switzerland
| | - Nancy Magnus
- Institute for Biological Sciences, University of Rostock, Albert-Einstein-Straße 3, 18059 Rostock, Germany
| | - Birgit Piechulla
- Institute for Biological Sciences, University of Rostock, Albert-Einstein-Straße 3, 18059 Rostock, Germany
| | - Allison S. Walker
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 240 Longwood Avenue, Boston, Massachusetts 02115, United States
- Department of Chemistry, Vanderbilt University, 1234 Stevenson Center Lane, Nashville, Tennessee 37240, United States
- Department of Biological Sciences, Vanderbilt University, 465 21st Avenue S, Nashville, Tennesee 37232, United States
| | - Jörn Piel
- Institute of Microbiology, Eidgenössische Technische Hochschule (ETH) Zürich, Vladimir-Prelog-Weg 4, 8093 Zürich, Switzerland
- Lead contact
| |
Collapse
|
48
|
Zheng Y, Li Y, Zhou K, Li T, VanDusen NJ, Hua Y. Precise genome-editing in human diseases: mechanisms, strategies and applications. Signal Transduct Target Ther 2024; 9:47. [PMID: 38409199 PMCID: PMC10897424 DOI: 10.1038/s41392-024-01750-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 02/28/2024] Open
Abstract
Precise genome-editing platforms are versatile tools for generating specific, site-directed DNA insertions, deletions, and substitutions. The continuous enhancement of these tools has led to a revolution in the life sciences, which promises to deliver novel therapies for genetic disease. Precise genome-editing can be traced back to the 1950s with the discovery of DNA's double-helix and, after 70 years of development, has evolved from crude in vitro applications to a wide range of sophisticated capabilities, including in vivo applications. Nonetheless, precise genome-editing faces constraints such as modest efficiency, delivery challenges, and off-target effects. In this review, we explore precise genome-editing, with a focus on introduction of the landmark events in its history, various platforms, delivery systems, and applications. First, we discuss the landmark events in the history of precise genome-editing. Second, we describe the current state of precise genome-editing strategies and explain how these techniques offer unprecedented precision and versatility for modifying the human genome. Third, we introduce the current delivery systems used to deploy precise genome-editing components through DNA, RNA, and RNPs. Finally, we summarize the current applications of precise genome-editing in labeling endogenous genes, screening genetic variants, molecular recording, generating disease models, and gene therapy, including ex vivo therapy and in vivo therapy, and discuss potential future advances.
Collapse
Affiliation(s)
- Yanjiang Zheng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Kaiyu Zhou
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Tiange Li
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Nathan J VanDusen
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - Yimin Hua
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
49
|
Larriba E, Yaroshko O, Pérez-Pérez JM. Recent Advances in Tomato Gene Editing. Int J Mol Sci 2024; 25:2606. [PMID: 38473859 PMCID: PMC10932025 DOI: 10.3390/ijms25052606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/19/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
The use of gene-editing tools, such as zinc finger nucleases, TALEN, and CRISPR/Cas, allows for the modification of physiological, morphological, and other characteristics in a wide range of crops to mitigate the negative effects of stress caused by anthropogenic climate change or biotic stresses. Importantly, these tools have the potential to improve crop resilience and increase yields in response to challenging environmental conditions. This review provides an overview of gene-editing techniques used in plants, focusing on the cultivated tomatoes. Several dozen genes that have been successfully edited with the CRISPR/Cas system were selected for inclusion to illustrate the possibilities of this technology in improving fruit yield and quality, tolerance to pathogens, or responses to drought and soil salinity, among other factors. Examples are also given of how the domestication of wild species can be accelerated using CRISPR/Cas to generate new crops that are better adapted to the new climatic situation or suited to use in indoor agriculture.
Collapse
Affiliation(s)
- Eduardo Larriba
- Instituto de Bioingeniería, Universidad Miguel Hernández, 03202 Elche, Spain;
| | | | | |
Collapse
|
50
|
Aksoy YA, Cole AJ, Deng W, Hesselson D. Zebrafish CCNF and FUS Mediate Stress-Specific Motor Responses. Cells 2024; 13:372. [PMID: 38474336 PMCID: PMC10930717 DOI: 10.3390/cells13050372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/09/2024] [Accepted: 02/10/2024] [Indexed: 03/14/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease characterized by the degeneration of motor neurons. Mutations in the cyclin F (CCNF) and fused in sarcoma (FUS) genes have been associated with ALS pathology. In this study, we aimed to investigate the functional role of CCNF and FUS in ALS by using genome editing techniques to generate zebrafish models with genetic disruptions in these genes. Sequence comparisons showed significant homology between human and zebrafish CCNF and FUS proteins. We used CRISPR/Cas9 and TALEN-mediated genome editing to generate targeted disruptions in the zebrafish ccnf and fus genes. Ccnf-deficient zebrafish exhibited abnormal motor neuron development and axonal outgrowth, whereas Fus-deficient zebrafish did not exhibit developmental abnormalities or axonopathies in primary motor neurons. However, Fus-deficient zebrafish displayed motor impairments in response to oxidative and endoplasmic reticulum stress. The Ccnf-deficient zebrafish were only sensitized to endoplasmic reticulum stress, indicating that ALS genes have overlapping as well as unique cellular functions. These zebrafish models provide valuable platforms for studying the functional consequences of CCNF and FUS mutations in ALS pathogenesis. Furthermore, these zebrafish models expand the drug screening toolkit used to evaluate possible ALS treatments.
Collapse
Affiliation(s)
- Yagiz Alp Aksoy
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia;
- Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
- Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia
| | - Alexander J Cole
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia;
| | - Wei Deng
- School of Biomedical Engineering, University of Technology Sydney, Ultimo, NSW 2007, Australia;
| | - Daniel Hesselson
- Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia;
| |
Collapse
|