1
|
Towa S, Okada S, Ito T. Catalytically inactive Cas9 attenuates DNA end resection: A potential application for region-restricted random mutagenesis. iScience 2025; 28:112702. [PMID: 40520111 PMCID: PMC12164011 DOI: 10.1016/j.isci.2025.112702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 03/25/2025] [Accepted: 05/16/2025] [Indexed: 06/18/2025] Open
Abstract
Gene duplication followed by sequence diversification is a key driver of innovation in genome evolution. To mimic this process in genome engineering, a method for region-restricted mutagenesis is needed to selectively mutate one copy of a duplicated gene. Notably, regions flanking a double-strand break (DSB) become hypersensitive to mutagens due to end resection, which converts them into single-stranded DNA (ssDNA). Blocking end resection could, therefore, confine hypermutation to a limited region. To achieve this, we investigated a catalytically inactive variant of Streptococcus pyogenes Cas9 (dSpCas9) and demonstrated its ability to attenuate end resection in the budding yeast Saccharomyces cerevisiae using ssDNA-specific quantitative PCR, live-cell imaging, and Southern blot analysis. By leveraging the bisulfite sensitivity of ssDNA, we further validated the concept of DSB-coupled, dSpCas9-mediated region-restricted mutagenesis. We anticipate that dSpCas9-mediated modulation of end resection at induced DSB sites will have valuable applications in both genome engineering and mechanistic studies.
Collapse
Affiliation(s)
- Suchin Towa
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Satoshi Okada
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Takashi Ito
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
2
|
Gilglioni EH, Bansal M, St-Pierre-Wijckmans W, Talamantes S, Kasarinaite A, Hay DC, Gurzov EN. Therapeutic potential of stem cell-derived somatic cells to treat metabolic dysfunction-associated steatotic liver disease and diabetes. Obes Rev 2025; 26:e13899. [PMID: 39861937 DOI: 10.1111/obr.13899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 10/22/2024] [Accepted: 12/04/2024] [Indexed: 01/27/2025]
Abstract
Developments in basic stem cell biology have paved the way for technology translation in human medicine. An exciting prospective use of stem cells is the ex vivo generation of hepatic and pancreatic endocrine cells for biomedical applications. This includes creating novel models 'in a dish' and developing therapeutic strategies for complex diseases, such as metabolic dysfunction-associated steatotic liver disease (MASLD) and diabetes. In this review, we explore recent advances in the generation of stem cell-derived hepatocyte-like cells and insulin-producing β-like cells. We cover the different differentiation strategies, new discoveries, and the caveats that still exist regarding their routine use. Finally, we discuss the challenges and limitations of stem cell-derived therapies as a clinical strategy to manage metabolic diseases in humans.
Collapse
Affiliation(s)
- Eduardo H Gilglioni
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, Brussels, Belgium
| | - Mayank Bansal
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, Brussels, Belgium
| | | | - Stephanie Talamantes
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, Brussels, Belgium
| | - Alvile Kasarinaite
- Institute for Regeneration and Repair, Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - David C Hay
- Institute for Regeneration and Repair, Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Esteban N Gurzov
- Signal Transduction and Metabolism Laboratory, Université libre de Bruxelles, Brussels, Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| |
Collapse
|
3
|
Xiong J, Deng C, Fu Y, Tang J, Xie J, Chen Y. Prognostic and Potential Therapeutic Roles of PRKDC Expression in Lung Cancer. Mol Biotechnol 2025; 67:2455-2466. [PMID: 39044064 DOI: 10.1007/s12033-024-01209-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 05/06/2024] [Indexed: 07/25/2024]
Abstract
PRKDC is a key factor involved in the ligation step of the non-homologous end joining pathway. Its dysfunction has proven to be a biomarker for radiosensitivity of cancer cells. However, the prognostic value of PRKDC and its underlying mechanisms have not been clarified yet. In this study, we found that PRKDC overexpressed in lung adenocarcinoma (LUAD) and is significantly related to unfavorable survival, while downregulation of PRKDC is link to inflamed tumor immune signature. Our further in vitro results also showed a potent antitumor efficacy of PRKDC inhibitors alone or combined with cisplatin in human lung cancer cells. This study demonstrated that PRKDC is a potential prognostic biomarker, immunotherapy target, and promising combination candidate for chemotherapy for lung cancer, and highlighted the potential of PRKDC-targeted inhibitors for the treatment of lung cancer.
Collapse
Affiliation(s)
- Jiani Xiong
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
- Cancer Bio-immunotherapy Center, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
| | - Cuimin Deng
- Department of Pharmacy, QuanZhou Women's and Children's Hospital, Quanzhou, Fujian Province, People's Republic of China
| | - YunRong Fu
- Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Jingji Tang
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
- Cancer Bio-immunotherapy Center, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China
| | - Jieming Xie
- Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China.
| | - Yu Chen
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China.
- Cancer Bio-immunotherapy Center, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China.
| |
Collapse
|
4
|
Xu Z, Xie H, Song L, Huang Y, Huang J. BRCA1 and BRCA2 in DNA damage and replication stress response: Insights into their functions, mechanisms, and implications for cancer treatment. DNA Repair (Amst) 2025; 150:103847. [PMID: 40373656 DOI: 10.1016/j.dnarep.2025.103847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 05/04/2025] [Accepted: 05/08/2025] [Indexed: 05/17/2025]
Abstract
Genomic stability is a cornerstone of cellular survival and proliferation. To counter the constant threat posed by endogenous and exogenous DNA-damaging agents, cells rely on a network of intricate mechanisms to safeguard DNA integrity and ensure accurate replication. Among these, the BRCA1 and BRCA2 tumor suppressor proteins play pivotal roles. While traditionally recognized for their involvement in homologous recombination repair and cell cycle checkpoints, emerging evidence highlights their essential functions in protecting stalled replication forks during replication stress. Mutations in BRCA1 or BRCA2 disrupt these critical functions, leading to compromised genome stability and an increased susceptibility to various cancers, particularly breast and ovarian cancers. This review provides a comprehensive analysis of the multifaceted roles of BRCA1 and BRCA2, focusing on their contributions to DNA damage responses and replication stress management. By elucidating the molecular pathways through which BRCA1 and BRCA2 operate, we aim to provide insights into their pivotal roles in maintaining genomic integrity and their implications for cancer treatment.
Collapse
Affiliation(s)
- Ziqi Xu
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Haihua Xie
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Lizhi Song
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yuhua Huang
- Department of Urology, the First Affiliated Hospital of Soochow University, Suzhou 215000, China.
| | - Jun Huang
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; The MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China; Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing 321000, China.
| |
Collapse
|
5
|
Wu F, Li N, Xiao Y, Palanki R, Yamagata H, Mitchell MJ, Han X. Lipid Nanoparticles for Delivery of CRISPR Gene Editing Components. SMALL METHODS 2025:e2401632. [PMID: 40434188 DOI: 10.1002/smtd.202401632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 05/05/2025] [Indexed: 05/29/2025]
Abstract
Gene editing has emerged as a promising therapeutic option for treating genetic diseases. However, a central challenge in the field is the safe and efficient delivery of these large editing tools, especially in vivo. Lipid nanoparticles (LNPs) are attractive nonviral vectors due to their low immunogenicity and high delivery efficiency. To maximize editing efficiency, LNPs should efficiently protect gene editing components against multiple biological barriers and release them into the cytoplasm of target cells. In this review, the widely used CRISPR gene editing systems are first overviewed. Then, each component of LNPs, as well as their effects on delivery, are systematically discussed. Following this, the current LNP engineering strategies to achieve non-liver targeting are summarized. Finally, preclinical and clinical applications of LNPs for in vivo genome editing are highlighted, and perspectives for the future development of LNPs are provided.
Collapse
Affiliation(s)
- Fan Wu
- Key Laboratory of RNA Innovation, Science and Engineering, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Nei Li
- Key Laboratory of RNA Innovation, Science and Engineering, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yudian Xiao
- Key Laboratory of RNA Innovation, Science and Engineering, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Rohan Palanki
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Hannah Yamagata
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Xuexiang Han
- Key Laboratory of RNA Innovation, Science and Engineering, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| |
Collapse
|
6
|
Marconato N, Tommasi OD, Paladin D, Boscarino D, Spagnol G, Saccardi C, Maggino T, Tozzi R, Noventa M, Marchetti M. Unraveling Homologous Recombination Deficiency in Ovarian Cancer: A Review of Currently Available Testing Platforms. Cancers (Basel) 2025; 17:1771. [PMID: 40507252 PMCID: PMC12153926 DOI: 10.3390/cancers17111771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2025] [Revised: 05/22/2025] [Accepted: 05/23/2025] [Indexed: 06/16/2025] Open
Abstract
Homologous recombination deficiency (HRD) is a key biomarker associated with increased sensitivity to PARP inhibitors (PARPi) in advanced epithelial ovarian cancer. Accurate identification of HRD status is essential for selecting patients most likely to benefit from these therapies. Current diagnostic approaches combine sequencing to detect mutations in homologous recombination repair genes-particularly BRCA1 and BRCA2-with genome-wide analysis of structural genomic alterations indicative of HRD. This review briefly outlines the biological basis of HRD and its clinical significance and then focuses on currently available assays for HRD assessment. We compare their molecular strategies, including the use of targeted gene panels and genomic instability metrics such as loss of heterozygosity, telomeric allelic imbalance, and large-scale state transitions. The review also highlights the strengths and limitations of each platform and discusses their role in guiding clinical decision-making. Challenges related to dynamic tumor evolution and the interpretation of HRD status in recurrent disease settings are also addressed.
Collapse
Affiliation(s)
- Nicola Marconato
- Department of Women and Children’s Health, University of Padua, 35128 Padua, Italy;
| | - Orazio De Tommasi
- Unit of Gynecology and Obstetrics, Department of Women and Children’s Health, University of Padua, 35128 Padua, Italy
| | - Dino Paladin
- AB ANALITICA S.r.l., Via Svizzera, 16, 35127 Padova, Italy; (D.P.); (D.B.)
| | - Diego Boscarino
- AB ANALITICA S.r.l., Via Svizzera, 16, 35127 Padova, Italy; (D.P.); (D.B.)
| | - Giulia Spagnol
- Unit of Gynecology and Obstetrics, Department of Women and Children’s Health, University of Padua, 35128 Padua, Italy
| | - Carlo Saccardi
- Unit of Gynecology and Obstetrics, Department of Women and Children’s Health, University of Padua, 35128 Padua, Italy
| | - Tiziano Maggino
- Unit of Gynecology and Obstetrics, Department of Women and Children’s Health, University of Padua, 35128 Padua, Italy
| | - Roberto Tozzi
- Unit of Gynecology and Obstetrics, Department of Women and Children’s Health, University of Padua, 35128 Padua, Italy
| | - Marco Noventa
- Unit of Gynecology and Obstetrics, Department of Women and Children’s Health, University of Padua, 35128 Padua, Italy
| | - Matteo Marchetti
- Unit of Gynecology and Obstetrics, Department of Women and Children’s Health, University of Padua, 35128 Padua, Italy
| |
Collapse
|
7
|
Ho CH, Tsai CY, Chang CC, Hu CJ, Huang CY, Lu YC, Lin PH, Lin CH, Lin HI, OuYang CH, Hsu CJ, Liu TC, Chen YT, Chan YH, Cheng YF, Wu CC. Low Efficiency of Homology-Independent Targeted Integration for CRISPR/Cas9 Correction in the Vicinity of the SLC26A4 c.919-2A>G Variant. Int J Mol Sci 2025; 26:4980. [PMID: 40507794 PMCID: PMC12155412 DOI: 10.3390/ijms26114980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 05/15/2025] [Accepted: 05/17/2025] [Indexed: 06/16/2025] Open
Abstract
Recessive variants of SLC26A4 are a common cause of hereditary hearing impairment and are responsible for non-syndromic enlarged vestibular aqueducts and Pendred syndrome. Patients with bi-allelic SLC26A4 variants often suffer from fluctuating hearing loss and recurrent vertigo, ultimately leading to severe to profound hearing impairment. However, there are currently no satisfactory prevention or treatment options for this condition. The CRISPR/Cas9 genome-editing technique is a well-known tool for correcting point mutations or manipulating genes and shows potential therapeutic applications for hereditary disorders. In this study, we used the homology-independent targeted integration (HITI) strategy to correct the SLC26A4 c.919-2A>G variant, the most common SLC26A4 variant in the Han Chinese population. Next-generation sequencing was performed to evaluate the editing efficiency of the HITI strategy. The results showed that only 0.15% of the reads successfully exhibited HITI integration, indicating that the c.919-2 region may not be a suitable region for HITI selection. This suggests that other site selection or insertion strategies may be needed to improve the efficiency of correcting the SLC26A4 c.919-2A>G variant. This experience may serve as a valuable reference for other researchers considering CRISPR target design in this region.
Collapse
Affiliation(s)
- Chang-Han Ho
- Department of Otolaryngology-Head and Neck Surgery, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Cheng-Yu Tsai
- Department of Otolaryngology-Head and Neck Surgery, National Taiwan University Hospital, Taipei 100225, Taiwan
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei 100233, Taiwan
| | - Chi-Chieh Chang
- Department of Otolaryngology-Head and Neck Surgery, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Chin-Ju Hu
- Department of Otolaryngology-Head and Neck Surgery, National Taiwan University Hospital, Taipei 100225, Taiwan
- Program in Speech and Hearing Biosciences and Technology, Harvard Medical School, Boston, MA 02115, USA
| | - Cheng-Yen Huang
- Gene Knockout/in Cell Line Modeling Core, Human Disease Modeling Center, First Core Laboratory, Branch Office of Research and Development, National Taiwan University College of Medicine, Taipei 100233, Taiwan
| | - Ying-Chang Lu
- Department of Otolaryngology-Head and Neck Surgery, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Pei-Hsuan Lin
- Department of Otolaryngology-Head and Neck Surgery, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Chin-Hsien Lin
- Department of Neurology, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Han-I Lin
- Department of Neurology, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Chih-Hsin OuYang
- Department of Neurology, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Chuan-Jen Hsu
- Department of Otolaryngology-Head and Neck Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung 427, Taiwan
| | - Tien-Chen Liu
- Department of Otolaryngology-Head and Neck Surgery, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - You-Tzung Chen
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei 100233, Taiwan
| | - Yen-Hui Chan
- Department of Otolaryngology-Head and Neck Surgery, National Taiwan University Hospital, Taipei 100225, Taiwan
- Department of Otolaryngology-Head and Neck Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung 427, Taiwan
| | - Yen-Fu Cheng
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Department of Otolaryngology-Head and Neck Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- School of Medicine, National Yang-Ming Chiao-Tung University, Taipei 112304, Taiwan
- Department of Speech Language Pathology and Audiology, National Taipei University of Nursing Health Sciences, Taipei 112303, Taiwan
| | - Chen-Chi Wu
- Department of Otolaryngology-Head and Neck Surgery, National Taiwan University Hospital, Taipei 100225, Taiwan
- Department of Medical Research, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu 302058, Taiwan
| |
Collapse
|
8
|
Gallala M. Application of CRISPR/Cas gene editing for infectious disease control in poultry. Open Life Sci 2025; 20:20251095. [PMID: 40417002 PMCID: PMC12103187 DOI: 10.1515/biol-2025-1095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 02/11/2025] [Accepted: 03/11/2025] [Indexed: 05/27/2025] Open
Abstract
The poultry industry faces multifaceted challenges, including escalating demand for poultry products, climate change impacting feed availability, emergence of novel avian pathogens, and antimicrobial resistance. Traditional disease control measures are costly and not always effective, prompting the need for complementary methods. Gene editing (GE, also called genome editing) technologies, particularly CRISPR/Cas9, offer promising solutions. This article summarizes recent advancements in utilizing CRISPR/Cas GE to enhance infectious disease control in poultry. It begins with an overview of modern GE techniques, highlighting CRISPR/Cas9's advantages over other methods. The potential applications of CRISPR/Cas in poultry infectious disease prevention and control are explored, including the engineering of innovative vaccines, the generation of disease-resilient birds, and in vivo pathogen targeting. Additionally, insights are provided regarding regulatory frameworks and future perspectives in this rapidly evolving field.
Collapse
Affiliation(s)
- Mahdi Gallala
- Animal Resources Department, Ministry of Municipality, Doha, State of Qatar
| |
Collapse
|
9
|
Tenjo-Castaño F, Rout SS, Dey S, Montoya G. Unlocking the potential of CRISPR-associated transposons: from structural to functional insights. Trends Genet 2025:S0168-9525(25)00080-0. [PMID: 40393858 DOI: 10.1016/j.tig.2025.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/14/2025] [Accepted: 04/14/2025] [Indexed: 05/22/2025]
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR)-associated transposons (CASTs) are emerging genome-editing tools that enable RNA-guided DNA integration without inducing double-strand breaks (DSBs). Unlike CRISPR-associated (Cas) nucleases, CASTs use transposon machinery to insert large DNA segments with high precision, potentially reducing off-target effects and bypassing DNA damage responses. CASTs are categorized into classes 1 and 2, each employing distinct mechanisms for DNA targeting and integration. Recent structural insights have elucidated how CASTs recognize target sites, recruit transposases, and mediate insertion. These advances position CASTs as promising tools for genome engineering in bacteria and possibly in mammalian cells. Key challenges remain in enhancing efficiency and specificity, particularly for therapeutic use. Ongoing research aims to evolve CAST systems for precise, large-scale genome editing in human cells.
Collapse
Affiliation(s)
- Francisco Tenjo-Castaño
- Structural Molecular Biology Group, Novo Nordisk Foundation Centre for Protein Research, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences University of Copenhagen, Blegdamsvej 3B, Copenhagen 2200, Denmark
| | - Sweta Suman Rout
- Structural Molecular Biology Group, Novo Nordisk Foundation Centre for Protein Research, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences University of Copenhagen, Blegdamsvej 3B, Copenhagen 2200, Denmark
| | - Sanjay Dey
- Structural Molecular Biology Group, Novo Nordisk Foundation Centre for Protein Research, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences University of Copenhagen, Blegdamsvej 3B, Copenhagen 2200, Denmark
| | - Guillermo Montoya
- Structural Molecular Biology Group, Novo Nordisk Foundation Centre for Protein Research, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences University of Copenhagen, Blegdamsvej 3B, Copenhagen 2200, Denmark.
| |
Collapse
|
10
|
Zhang L, Wang E, Wu L, Zhang J, You S, Su R, Qi W. Rational Design of UvsX Recombinase Variants for Enhanced Performance in Recombinase Polymerase Amplification Applications. Biochemistry 2025; 64:2025-2038. [PMID: 40261914 DOI: 10.1021/acs.biochem.5c00098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Homologous recombination is a vital biological process for DNA repair, genomic stability, and genetic diversity, driven by the RecA/Rad51 recombinase family. However, as a T4 bacteriophage recombinase homologous to RecA/Rad51, UvsX has limited in vitro performance during recombinase polymerase amplification (RPA) due to ATP utilization and DNA affinity. In this study, UvsX was rationally engineered to enhance these properties through homology modeling, virtual saturation mutations, and consensus mutation strategies. Targeted mutagenesis produced UvsX variants (E198N, E198R, E198K, and K35G) with a 16 ± 4% to 39 ± 6% improvement in RPA activity, while the double mutant K35G/E198R showed an increase of up to 43 ± 4%. Structural analysis revealed that the K35G/E198R mutation enlarged ATP-binding pockets and increased the positive surface potential of DNA-binding sites, resulting in a 12 ± 4% improvement in ATP utilization and more ADP and less AMP generated, a 10 ± 2% enhancement in DNA interaction compared to the wild-type, and better inhibitor tolerance. These findings establish a foundation for the rational optimization of recombinases in nucleic acid amplification and promote their potential for industrial RPA applications.
Collapse
Affiliation(s)
- Lin Zhang
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P. R. China
| | - Enjie Wang
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P. R. China
| | - Lvping Wu
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P. R. China
| | - Jiaxing Zhang
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P. R. China
- State Key Laboratory of Chemical Engineering, Tianjin University, Tianjin 300350, P. R. China
| | - Shengping You
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P. R. China
- Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Rongxin Su
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P. R. China
- State Key Laboratory of Chemical Engineering, Tianjin University, Tianjin 300350, P. R. China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072, P. R. China
- Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Wei Qi
- Chemical Engineering Research Center, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, P. R. China
- State Key Laboratory of Chemical Engineering, Tianjin University, Tianjin 300350, P. R. China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072, P. R. China
- Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin 300072, P. R. China
| |
Collapse
|
11
|
Straume T, Mora AM, Brown JB, Bansal I, Rabin BM, Braby LA, Wyrobek AJ. Non-DNA radiosensitive targets that initiate persistent behavioral deficits in rats exposed to space radiation. LIFE SCIENCES IN SPACE RESEARCH 2025; 45:44-60. [PMID: 40280642 DOI: 10.1016/j.lssr.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 12/16/2024] [Accepted: 12/20/2024] [Indexed: 04/29/2025]
Abstract
Predicting future CNS risks for astronauts during deep-space missions will rely substantially on ground-based rodent data with space-relevant ions and behaviors. For rats, the accumulated evidence indicates that less densely ionizing radiation, such as 4He and 12C ions, induce behavior deficits at lower doses than densely ionizing ions, such as 48Ti and 56Fe. However, this observation conflicts with standard somatic radiobiology, in which densely ionizing ions are generally more effective than less densely ionizing ions, and where the DNA/nucleus is the accepted target for radiation-induced tumorigenesis, cytogenetic aberrations, genetic mutations, and reproductive cell death. To gain deeper insight into the subcellular nature of the radiation targets for behavior risks, we compared the effects of dose, fluence, and linear energy transfer (LET) of 4He and 56Fe particles using existing datasets for four distinct behavioral outcomes in rats: elevated plus maze (EPM-anxiety), novel object recognition (NOR-memory), operant responding (OR-response to environmental stimuli), and attentional set-shifting (ATSET-cognitive flexibility). We confirmed that less densely ionizing particles (except protons) showed ∼100-fold lower threshold doses than densely ionizing particles for behavioral deficits (0.1-1 cGy for 4He vs. 15-100 cGy for 56Fe). However, when analyzed by fluence the behavioral responses converged, indicating that 4He and 56Fe were equally effective on a per-track basis. When analyzed by LET, there were ∼100-fold differences in the LET for maximum effectiveness for behavioral deficits and DNA endpoints (∼1 vs ∼100 keV/μm, respectively). These unique features of radiation-induced behavioral deficits (high sensitivity to particles in the 1-keV/μm range, insensitivity to protons in the 0.2 keV/μm range, and isofluence dependence for particles with LET>1 keV/μm) provide evidence in support of a new hypothesis of sub-micron sized radiosensitive targets for behavioral effects consistent with the thickness of plasma membranes and/or small subcellular structures, smaller than a whole synapse. Like our behavior findings, mouse immature oocyte killing which is known to have a plasma membrane target was also better explained by fluence, rather than dose. In contrast, fluence analyses for DNA/nuclear endpoints in somatic cells (e.g., tumor induction, chromosome aberrations) showed opposite results, suggesting that behavior targets are not DNA. Our findings raise questions regarding the identity of subcellular targets and the multi-cellular functional unit for behavior risks, low-dose susceptibility, and generalizability from rat to other species and astronauts.
Collapse
Affiliation(s)
- Tore Straume
- Lawrence Berkeley National Laboratory, University of California, 1 Cyclotron Road, Berkeley, CA 94720, USA; NASA Ames Research Center (retired affiliation), Moffett Field, CA 94035, USA.
| | - Ana M Mora
- School of Public Health, University of California, Berkeley, CA 94720, USA
| | - James B Brown
- Lawrence Berkeley National Laboratory, University of California, 1 Cyclotron Road, Berkeley, CA 94720, USA
| | - Ishan Bansal
- Lawrence Berkeley National Laboratory, University of California, 1 Cyclotron Road, Berkeley, CA 94720, USA
| | | | | | - Andrew J Wyrobek
- Lawrence Berkeley National Laboratory, University of California, 1 Cyclotron Road, Berkeley, CA 94720, USA.
| |
Collapse
|
12
|
Huang J, Ding K, Chen J, Fan J, Huang L, Qiu S, Wang L, Du X, Wang C, Pan H, Yuan Z, Liu H, Song H. Comparison of CRISPR-Cas9, CRISPR-Cas12f1, and CRISPR-Cas3 in eradicating resistance genes KPC-2 and IMP-4. Microbiol Spectr 2025:e0257224. [PMID: 40293254 DOI: 10.1128/spectrum.02572-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 02/22/2025] [Indexed: 04/30/2025] Open
Abstract
Bacterial plasmid encoding antibiotic resistance could be eradicated by various CRISPR systems, such as CRISPR-Cas9, Cas12f1, and Cas3. However, the efficacy of these gene editing tools against bacterial resistance has not been systematically assessed and compared. This study eliminates carbapenem resistance genes KPC-2 and IMP-4 via CRISPR-Cas9, Cas12f1, and Cas3 systems, respectively. The eradication efficiency of the three CRISPR systems was evaluated. First, the target sites for the three CRISPR systems were designed within the regions 542-576 bp of the KPC-2 gene and 213-248 bp of the IMP-4 gene, respectively. The recombinant CRISPR plasmids were transformed into Escherichia coli carrying KPC-2 or IMP-4-encoding plasmid. Colony PCR of transformants showed that KPC-2 and IMP-4 were eradicated by the three different CRISPR systems, and the elimination efficacy was both 100.00%. The drug sensitivity test results showed that the resistant E. coli strain was resensitized to ampicillin. In addition, the three CRISPR plasmids could block the horizontal transfer of drug-resistant plasmids, with a blocking rate as high as 99%. Importantly, a qPCR assay was performed to analyze the copy number changes of drug-resistant plasmids in E. coli cells. The results indicated that CRISPR-Cas3 showed higher eradication efficiency than CRISPR-Cas9 and Cas12f1 systems. IMPORTANCE With the continuous development and application of CRISPR-based resistance removal technologies, CRISPR-Cas9, Cas12f1, and Cas3 have gradually come into focus. However, it remains uncertain which system exhibits more potent efficacy in the removal of bacterial resistance. This study verifies that CRISPR-Cas9, Cas12f1, and Cas3 can eradicate the carbapenem-resistant genes KPC-2 and IMP-4 and restore the sensitivity of drug-resistant model bacteria to antibiotics. Among the three CRISPR systems, the CRISPR-Cas3 system showed the highest eradication efficiency. Although each system has its advantages and characteristics, our results provide guidance on the selection of the CRISPR system from the perspective of resistance gene removal efficiency, contributing to the further application of CRISPR-based bacterial resistance removal technologies.
Collapse
Affiliation(s)
- Jun Huang
- Department of Infectious Disease Prevention and Control, Chinese People's Liberation Army Center for Disease Control and Prevention, Beijing, China
- Department of Human Anatomy and Histology, School of Basic Medicine, Capital Medical University, Beijing, China
| | - Kanghui Ding
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Jiahui Chen
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, China
| | - Jiao Fan
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, China
| | - Luyao Huang
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, China
| | - Shaofu Qiu
- Department of Infectious Disease Prevention and Control, Chinese People's Liberation Army Center for Disease Control and Prevention, Beijing, China
| | - Ligui Wang
- Department of Infectious Disease Prevention and Control, Chinese People's Liberation Army Center for Disease Control and Prevention, Beijing, China
| | - Xinying Du
- Department of Infectious Disease Prevention and Control, Chinese People's Liberation Army Center for Disease Control and Prevention, Beijing, China
| | - Chao Wang
- Department of Infectious Disease Prevention and Control, Chinese People's Liberation Army Center for Disease Control and Prevention, Beijing, China
| | - Haifeng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Zhengquan Yuan
- Department of Infectious Disease Prevention and Control, Chinese People's Liberation Army Center for Disease Control and Prevention, Beijing, China
| | - Hongbo Liu
- Department of Infectious Disease Prevention and Control, Chinese People's Liberation Army Center for Disease Control and Prevention, Beijing, China
| | - Hongbin Song
- Department of Infectious Disease Prevention and Control, Chinese People's Liberation Army Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
13
|
Xiong Q, Zhu C, Yin X, Zhu L. CRISPR/Cas and Argonaute-based biosensors for nucleic acid detection. Talanta 2025; 294:128210. [PMID: 40280080 DOI: 10.1016/j.talanta.2025.128210] [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: 03/06/2025] [Revised: 04/20/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Nowadays, nucleic acid detection technology has been applied to disease diagnosis, prevention, food safety, environmental testing and many other aspects. However, traditional methods still have shortcomings. Therefore, there is an urgent need for a simple, rapid, sensitive, and specific new method to supersede traditional nucleic acid detection technology. CRISPR/Cas(Clustered Regularly Interspaced Short Palindromic Repeats/CRISPR-associated) system and Argonaute (Ago) system play an important role in microbial immune defense. Their targeting specificity, programmability and special trans-cleavage activity make it possible to develop some new platforms for nucleic acid detection in combination with a variety of biosensors. We introduce the origins of these two systems and the biosensors developed based on CRISPR/Cas system and Ago system, respectively, especially the prospects for the future development of Cascade Amplification biosensors. This review is expected to provide useful guidance for researchers in related fields and provide inspiration for the development of Cascade Amplification biosensors in the future.
Collapse
Affiliation(s)
- Qiangyuan Xiong
- Anhui Institute of Optics and Fine Mechanics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, PR China; University of Science and Technology of China, Hefei, 230026, PR China
| | - Cancan Zhu
- Anhui Institute of Optics and Fine Mechanics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, PR China.
| | - Xueer Yin
- Anhui Institute of Optics and Fine Mechanics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, PR China; University of Science and Technology of China, Hefei, 230026, PR China
| | - Ling Zhu
- Anhui Institute of Optics and Fine Mechanics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, PR China; University of Science and Technology of China, Hefei, 230026, PR China.
| |
Collapse
|
14
|
Yang L, Wei W, Yuan X, Guo E, Peng P, Wang J, Sun W. Targeting DNA Damage Repair to Enhance Antitumor Immunity in Radiotherapy: Mechanisms and Opportunities. Int J Mol Sci 2025; 26:3743. [PMID: 40332379 PMCID: PMC12027993 DOI: 10.3390/ijms26083743] [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: 03/08/2025] [Revised: 04/07/2025] [Accepted: 04/13/2025] [Indexed: 05/08/2025] Open
Abstract
Radiotherapy is a standard cancer treatment that involves the induction of DNA damage. DNA damage repair (DDR) pathways maintain genomic integrity and make tumors resistant to radiotherapy and certain chemotherapies. In turn, DDR dysfunction results in cumulative DNA damage, leading to increased sensitivity for antitumor treatment. Moreover, radiotherapy has been shown to trigger antitumor immunity. Currently, immunotherapy has become a new and widely used standard strategy for treating a broad spectrum of tumor types. Notably, recent studies have demonstrated that DDR pathways play important roles in driving the response to immunotherapy. Herein, we review and discuss how DDR affects antitumor immunity induced by radiotherapy. Furthermore, we summarize the development of strategies for combining DDR inhibitors with radiotherapy and/or immunotherapy to enhance their efficacy against cancers.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wei Sun
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (L.Y.); (W.W.); (X.Y.); (E.G.); (P.P.); (J.W.)
| |
Collapse
|
15
|
Haque US, Yokota T. Gene Editing for Duchenne Muscular Dystrophy: From Experimental Models to Emerging Therapies. Degener Neurol Neuromuscul Dis 2025; 15:17-40. [PMID: 40241992 PMCID: PMC12002074 DOI: 10.2147/dnnd.s495536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025] Open
Abstract
The CRISPR system has emerged as a ground-breaking gene-editing tool, offering promising therapeutic potential for Duchenne muscular dystrophy (DMD), a severe genetic disorder affecting approximately 1 in 5000 male births globally. DMD is caused by mutations in the dystrophin gene, which encodes a critical membrane-associated protein essential for maintaining muscle structure, function and repair. Patients with DMD experience progressive muscle degeneration, loss of ambulation, respiratory insufficiency, and cardiac failure, with most succumbing to the disease by their third decade of life. Despite the well-characterized genetic basis of DMD, curative treatments- such as exon skipping therapies, micro-dystrophin, and steroids- remain elusive. Recent preclinical studies have demonstrated the promise of CRISPR-based approaches in restoring dystrophin expression across various models, including human cells, murine systems, and large animal models. These advancements highlight the potential of gene editing to fundamentally alter the trajectory of the disease. However, significant challenges persist, including immunogenicity, off-target effects, and limited editing efficiency, which hinder clinical translation. This review provides a comprehensive analysis of the latest developments in CRISPR-based therapeutic strategies for DMD. It emphasizes the need for further innovation in gene-editing technologies, delivery systems, and rigorous safety evaluations to overcome current barriers and harness the full potential of CRISPR/Cas as a durable and effective treatment for DMD.
Collapse
Affiliation(s)
- Umme Sabrina Haque
- Department of Neuroscience, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
| | - Toshifumi Yokota
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
- The Friends of Garrett Cumming Research & Muscular Dystrophy Canada HM Toupin Neurological Science Research Chair, Edmonton, AB, T6G 2H7, Canada
| |
Collapse
|
16
|
Menon AV, Song B, Chao L, Sriram D, Chansky P, Bakshi I, Ulianova J, Li W. Unraveling the future of genomics: CRISPR, single-cell omics, and the applications in cancer and immunology. Front Genome Ed 2025; 7:1565387. [PMID: 40292231 PMCID: PMC12021818 DOI: 10.3389/fgeed.2025.1565387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 03/26/2025] [Indexed: 04/30/2025] Open
Abstract
The CRISPR system has transformed many research areas, including cancer and immunology, by providing a simple yet effective genome editing system. Its simplicity has facilitated large-scale experiments to assess gene functionality across diverse biological contexts, generating extensive datasets that boosted the development of computational methods and machine learning/artificial intelligence applications. Integrating CRISPR with single-cell technologies has further advanced our understanding of genome function and its role in many biological processes, providing unprecedented insights into human biology and disease mechanisms. This powerful combination has accelerated AI-driven analyses, enhancing disease diagnostics, risk prediction, and therapeutic innovations. This review provides a comprehensive overview of CRISPR-based genome editing systems, highlighting their advancements, current progress, challenges, and future opportunities, especially in cancer and immunology.
Collapse
Affiliation(s)
- A. Vipin Menon
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, DC, United States
- Department of Genomics and Precision Medicine, George Washington University, Washington, DC, DC, United States
| | - Bicna Song
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, DC, United States
- Department of Genomics and Precision Medicine, George Washington University, Washington, DC, DC, United States
| | - Lumen Chao
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, DC, United States
- Department of Genomics and Precision Medicine, George Washington University, Washington, DC, DC, United States
| | - Diksha Sriram
- The George Washington University, Washington, DC, DC, United States
| | - Pamela Chansky
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, DC, United States
- Integrated Biomedical Sciences (IBS) Program, The George Washington University, Washington, DC, DC, United States
| | - Ishnoor Bakshi
- The George Washington University, Washington, DC, DC, United States
| | - Jane Ulianova
- Integrated Biomedical Sciences (IBS) Program, The George Washington University, Washington, DC, DC, United States
| | - Wei Li
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, DC, United States
- Department of Genomics and Precision Medicine, George Washington University, Washington, DC, DC, United States
| |
Collapse
|
17
|
Canny MD, Beikzadeh M, Kaur N, Pendse R, Latham MP. Dynamic conformations of the P. furiosus MR-DNA complex link Mre11 nuclease activity to DNA-stimulated Rad50 ATP hydrolysis. Commun Biol 2025; 8:548. [PMID: 40175510 PMCID: PMC11965522 DOI: 10.1038/s42003-025-08003-0] [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] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 03/26/2025] [Indexed: 04/04/2025] Open
Abstract
The MRE11-RAD50-NBS1/Xrs2 (MRN/X) protein complex has essential roles in the repair of damaged DNA. The current understanding of the conformational landscape of the core MR complex comes from various structural studies. However, given the heterogeneous nature of these structures, we suspect that several conformational states may still be unaccounted for. Here, we use methyl-based NMR experiments on P. furiosus MR to determine an ensemble of distinct conformations of MR bound to DNA, consistent with the highly dynamic nature of the MR-DNA complex. Interrogation of these structures via in vitro activity assays on MR mutants reveal an unexpected, striking correlation between the nuclease activity of Mre11 and the magnitude of DNA-stimulated ATP hydrolysis by Rad50. Together, the structures and activity data support a model for MR activity where DNA-stimulated ATP hydrolysis unlocks Rad50 to provide access to the Mre11 active sites and further demonstrate how a heterogeneous ensemble of conformations can be used to coordinate various functions to direct biological outcomes. By elucidating the dynamic conformations of the DNA-bound MR complex, this work lays the foundation for future studies aimed at further characterizing this landscape and dissecting its role in the molecular mechanism of DNA repair and genome stability.
Collapse
Affiliation(s)
- Marella D Canny
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Mahtab Beikzadeh
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX, USA
| | - Navneet Kaur
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX, USA
- Doherty Institute of Infection and Immunity, The University of Melbourne, Melbourne, Australia
| | - Rohan Pendse
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX, USA
| | - Michael P Latham
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA.
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX, USA.
| |
Collapse
|
18
|
Zhang Z, Samsa WE, Gong Z. NBS1 dePARylation by NUDT16 is critical for DNA double-strand break repair. Mol Cell Biochem 2025; 480:2595-2609. [PMID: 39438373 DOI: 10.1007/s11010-024-05140-8] [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: 05/28/2024] [Accepted: 10/12/2024] [Indexed: 10/25/2024]
Abstract
NBS1, a protein linked to the autosomal recessive disorder Nijmegen breakage syndrome, plays an essential role in the DNA damage response and DNA repair. Despite its importance, the mechanisms regulating NBS1 and the impact of this regulation on DNA repair processes remain obscure. In this study, we discovered a new post-translational modification of NBS1, ADP-ribosylation. This modification can be removed by the NUDT16 hydrolase. The loss of NUDT16 results in a reduction of NBS1 protein levels due to NBS1 PARylation-dependent ubiquitination and degradation, which is mediated by the PAR-binding E3 ubiquitin ligase, RNF146. Importantly, ADP-ribosylation of NBS1 is crucial for its localization at DSBs and its involvement in homologous recombination (HR) repair. Additionally, the NUDT16-NBS1 interaction is regulated in response to DNA damage, providing further rationale for NBS1 regulation by NUDT16 hydrolase. In summary, our study unveils the critical role of NUDT16 in governing both the stability of NBS1 and recruitment of NBS1 to DNA double-strand breaks, providing novel insights into the regulation of NBS1 in the HR repair pathway.
Collapse
Affiliation(s)
- Zhen Zhang
- Department of Cancer Biology, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - William E Samsa
- Department of Cancer Biology, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Zihua Gong
- Department of Cancer Biology, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH, 44195, USA.
| |
Collapse
|
19
|
Yuan H, Song C, Xu H, Sun Y, Anthon C, Bolund L, Lin L, Benabdellah K, Lee C, Hou Y, Gorodkin J, Luo Y. An Overview and Comparative Analysis of CRISPR-SpCas9 gRNA Activity Prediction Tools. CRISPR J 2025; 8:89-104. [PMID: 40151952 DOI: 10.1089/crispr.2024.0058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025] Open
Abstract
Design of guide RNA (gRNA) with high efficiency and specificity is vital for successful application of the CRISPR gene editing technology. Although many machine learning (ML) and deep learning (DL)-based tools have been developed to predict gRNA activities, a systematic and unbiased evaluation of their predictive performance is still needed. Here, we provide a brief overview of in silico tools for CRISPR design and assess the CRISPR datasets and statistical metrics used for evaluating model performance. We benchmark seven ML and DL-based CRISPR-Cas9 editing efficiency prediction tools across nine CRISPR datasets covering six cell types and three species. The DL models CRISPRon and DeepHF outperform the other models exhibiting greater accuracy and higher Spearman correlation coefficient across multiple datasets. We compile all CRISPR datasets and in silico prediction tools into a GuideNet resource web portal, aiming to facilitate and streamline the sharing of CRISPR datasets. Furthermore, we summarize features affecting CRISPR gene editing activity, providing important insights into model performance and the further development of more accurate CRISPR prediction models.
Collapse
Affiliation(s)
- Hao Yuan
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- Lars Bolund Institute of Regenerative Medicine, Qingdao-Europe Advanced Institute for Life Sciences, BGI Research, Qingdao, China
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Chunping Song
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Huixin Xu
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Ying Sun
- Center for non-coding RNA in Technology and Health, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Christian Anthon
- Center for non-coding RNA in Technology and Health, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Lars Bolund
- Lars Bolund Institute of Regenerative Medicine, Qingdao-Europe Advanced Institute for Life Sciences, BGI Research, Qingdao, China
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Lin Lin
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Karim Benabdellah
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), Granada, Spain
| | - Ciaran Lee
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Yong Hou
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Jan Gorodkin
- Center for non-coding RNA in Technology and Health, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Yonglun Luo
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
20
|
Li M, Lin Y, Cheng Q, Wei T. Prime Editing: A Revolutionary Technology for Precise Treatment of Genetic Disorders. Cell Prolif 2025; 58:e13808. [PMID: 40014809 PMCID: PMC11969253 DOI: 10.1111/cpr.13808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/06/2024] [Accepted: 01/03/2025] [Indexed: 03/01/2025] Open
Abstract
Genetic diseases have long posed significant challenges, with limited breakthroughs in treatment. Recent advances in gene editing technologies offer new possibilities in gene therapy for the treatment of inherited disorders. However, traditional gene editing methods have limitations that hinder their potential for clinical use, such as limited editing capabilities and the production of unintended byproducts. To overcome these limitations, prime editing (PE) has been developed as a powerful tool for precise and efficient genome modification. In this review, we provide an overview of the latest advancements in PE and its potential applications in the treatment of inherited disorders. Furthermore, we examine the current delivery vehicles employed for delivering PE systems in vitro and in vivo, and analyze their respective benefits and limitations. Ultimately, we discuss the challenges that need to be addressed to fully unlock the potential of PE for the remission or cure of genetic diseases.
Collapse
Affiliation(s)
- Mengyao Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of ZoologyChinese Academy of SciencesBeijingChina
- Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yi Lin
- Department of Biomedical Engineering, College of Future TechnologyPeking UniversityBeijingChina
| | - Qiang Cheng
- Department of Biomedical Engineering, College of Future TechnologyPeking UniversityBeijingChina
- Beijing Advanced Center of RNA BiologyPeking UniversityBeijingChina
| | - Tuo Wei
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of ZoologyChinese Academy of SciencesBeijingChina
- Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| |
Collapse
|
21
|
Carnie CJ, Jackson SP, Stingele J. Transcription-coupled repair of DNA-protein crosslinks. Trends Cell Biol 2025; 35:316-329. [PMID: 39617652 DOI: 10.1016/j.tcb.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/30/2024] [Accepted: 11/06/2024] [Indexed: 04/06/2025]
Abstract
DNA-protein crosslinks (DPCs) are highly toxic DNA lesions that are relevant to multiple human diseases. They are caused by various endogenous and environmental agents, and from the actions of enzymes such as topoisomerases. DPCs impede DNA polymerases, triggering replication-coupled DPC repair. Until recently the consequences of DPC blockade of RNA polymerases remained unclear. New methodologies for studying DPC repair have enabled the discovery of a transcription-coupled (TC) DPC repair pathway. Briefly, RNA polymerase II (RNAPII) stalling initiates TC-DPC repair, leading to sequential engagement of Cockayne syndrome (CS) proteins CSB and CSA, and to proteasomal degradation of the DPC. Deficient TC-DPC repair caused by loss of CSA or CSB function may help to explain the complex clinical presentation of CS patients.
Collapse
Affiliation(s)
| | - Stephen P Jackson
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.
| | - Julian Stingele
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
22
|
Gedeonová D, Bianchi C, Štembírek J, Hrdinka M, Chyra Z, Buchtová M, Hurník P, Blažek T, Režnarová J. BRCA1 and BRCA2 as prognostic markers in oral squamous cell carcinoma: a minireview. Front Oncol 2025; 15:1528822. [PMID: 40224184 PMCID: PMC11986421 DOI: 10.3389/fonc.2025.1528822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/26/2025] [Indexed: 04/15/2025] Open
Abstract
Oral squamous cell carcinoma (OSCC), a subset of head and neck cancers, primarily originates in the epithelial tissues of the oral cavity. Despite advancements in treatment, the mortality rate for OSCC remains around 50%, underscoring the urgent need for improved prognostic markers. This review explores the role of the BRCA1 and BRCA2 genes-traditionally associated with breast and ovarian cancers-in the context of OSCC. We discuss the molecular pathways involving BRCA genes, their potential as diagnostics and prognostic biomarkers, and their implications for personalized treatment strategies, including addressing chemotherapy resistance. Furthermore, this review emphasizes the significance of genome stability in cancer progression and examines both current and emerging methodologies for detecting BRCA mutations in OSCC patients. Despite limited prevalence of BRCA mutations in OSCC compared to other cancers, their role in DNA repair and therapeutic response underscores their potential as clinical biomarkers. However, standardized, multicenter studies are still needed to validate their utility in OSCC management. A better understanding of the role of BRCA genes in OSCC could pave the way for more effective therapeutic approaches and improved patient outcomes.
Collapse
Affiliation(s)
- Dominika Gedeonová
- Department of Oral and Maxillofacial Surgery, University Hospital Ostrava, Ostrava, Czechia
- Department of Craniofacial Surgery, Faculty of Medicine, University of Ostrava, Ostrava, Czechia
| | - Claretta Bianchi
- Department of Oral and Maxillofacial Surgery, University Hospital Ostrava, Ostrava, Czechia
- Health Research Centre, Faculty of Medicine, University of Ostrava, Ostrava, Czechia
| | - Jan Štembírek
- Department of Oral and Maxillofacial Surgery, University Hospital Ostrava, Ostrava, Czechia
- Department of Craniofacial Surgery, Faculty of Medicine, University of Ostrava, Ostrava, Czechia
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
| | - Matouš Hrdinka
- Department of Oral and Maxillofacial Surgery, University Hospital Ostrava, Ostrava, Czechia
- Health Research Centre, Faculty of Medicine, University of Ostrava, Ostrava, Czechia
| | - Zuzana Chyra
- Department of Hematooncology, University Hospital Ostrava, Ostrava, Czechia
- Department of Hematology, Faculty of Medicine, University of Ostrava, Ostrava, Czechia
| | - Marcela Buchtová
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Pavel Hurník
- Institute of Molecular and Clinical Pathology and Medical Genetics, University Hospital Ostrava, Ostrava, Czechia
- Institute of Molecular and Clinical Pathology and Medical Genetics, Faculty of Medicine, University of Ostrava, Ostrava, Czechia
| | - Tomáš Blažek
- Clinic of Oncology, University Hospital Ostrava, Ostrava, Czechia
| | - Jana Režnarová
- Department of Oral and Maxillofacial Surgery, University Hospital Ostrava, Ostrava, Czechia
- Health Research Centre, Faculty of Medicine, University of Ostrava, Ostrava, Czechia
| |
Collapse
|
23
|
Vats A, Laimins L. How human papillomavirus (HPV) targets DNA repair pathways for viral replication: from guardian to accomplice. Microbiol Mol Biol Rev 2025; 89:e0015323. [PMID: 39868790 PMCID: PMC11948491 DOI: 10.1128/mmbr.00153-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2025] Open
Abstract
SUMMARYHuman papillomaviruses (HPVs) are small DNA viruses that are responsible for significant disease burdens worldwide, including cancers of the cervix, anogenital tract, and oropharynx. HPVs infect stratified epithelia at a variety of body locations and link their productive life cycles to the differentiation of the host cell. These viruses have evolved sophisticated mechanisms to exploit cellular pathways, such as DNA damage repair (DDR), to regulate their life cycles. HPVs activate key DDR pathways such as ATM, ATR, and FA, which are critical for maintaining genomic integrity but are often dysregulated in cancers. Importantly, these DDR pathways are essential for HPV replication in undifferentiated cells and amplification upon differentiation. The ability to modulate these DDR pathways not only enables HPV persistence but also contributes to cellular transformation. In this review, we discuss the recent advances in understanding the mechanisms by which HPV manipulates the host DDR pathways and how these depend upon enhanced topoisomerase activity and R-loop formation. Furthermore, the strategies to manipulate DDR pathways utilized by high-risk HPVs are compared with those used by other DNA viruses that exhibit similarities and distinct differences.
Collapse
Affiliation(s)
- Arushi Vats
- Department of Microbiology-Immunology, Northwestern University, Chicago, Illinois, USA
| | - Laimonis Laimins
- Department of Microbiology-Immunology, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
24
|
Rijal S, Mainali A, Acharya S, Bhattarai HK. Evolutionary history of the DNA repair protein, Ku, in eukaryotes and prokaryotes. PLoS One 2025; 20:e0308593. [PMID: 40131966 PMCID: PMC11936186 DOI: 10.1371/journal.pone.0308593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 01/21/2025] [Indexed: 03/27/2025] Open
Abstract
Ku is essential in non-homologous end-joining (NHEJ) across prokaryotes and eukaryotes, primarily in double-stranded breaks (DSBs) repair. It often presents as a multi-domain protein in eukaryotes, unlike their prokaryotic single-domain homologs. We systematically searched for Ku proteins across different domains of life. To elucidate the evolutionary history of the Ku protein, we constructed a maximum likelihood phylogenetic tree using Ku protein sequences from 100 representative eukaryotic, prokaryotic, and viral species. The resulting tree revealed a common node for eukaryotic Ku proteins, while viral and prokaryotic species clustered into a distinct clade. Our phylogenetic analysis reveals that the common ancestry of Ku70 and Ku80 likely resulted from a gene duplication event in the ancestral eukaryote. This inference is supported by BLASTp results, which indicate a close resemblance between archaeal Ku and eukaryotic Ku, particularly Ku70. The presence of both Ku protein paralogs in the Discoba group further supports the hypothesis that the gene duplication occurred early in eukaryotic evolution. It is plausible that archaea, which may have acted as intermediaries for Ku transfer, subsequently lost the Ku protein. Nonetheless, the extensive horizontal transfer of Ku among prokaryotes and its relatively higher prevalence in bacteria complicates our understanding of how Ku protein was inherited by early-branching eukaryotes.
Collapse
Affiliation(s)
- Sadikshya Rijal
- Department of Biotechnology, Kathmandu University, Dhulikhel, Nepal.
| | - Ashmita Mainali
- Department of Biotechnology, Kathmandu University, Dhulikhel, Nepal.
| | - Sandesh Acharya
- Department of Biotechnology, Kathmandu University, Dhulikhel, Nepal.
| | | |
Collapse
|
25
|
Teng Y, Arbogast K, Junge H, Chen Z. Protocol for generating splice isoform-specific mouse mutants using CRISPR-Cas9 and a minigene splicing reporter. STAR Protoc 2025; 6:103543. [PMID: 39756031 PMCID: PMC11758566 DOI: 10.1016/j.xpro.2024.103543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/04/2024] [Accepted: 12/09/2024] [Indexed: 01/07/2025] Open
Abstract
Here, we present a protocol to alter the production of alternatively spliced mRNA variants, without affecting the overall gene expression, through CRISPR-Cas9-engineered genomic mutations in mice. We describe steps for designing guide RNA to direct Cas9 endonuclease to consensus splice sites, producing transgenic mice through pronuclear injection, and screening for desired mutations in cultured mammalian cells using a minigene splicing reporter. Splice isoform-specific mouse mutants provide valuable tools for genetic analyses beyond loss-of-function and transgenic alleles. For complete details on the use and execution of this protocol, please refer to Dailey-Krempel et al.1 and Johnson et al.2.
Collapse
Affiliation(s)
- Yudong Teng
- The Genetically Engineered Murine Models Core, Department of Immunology & Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kelsey Arbogast
- Colorado Center for Personalized Medicine Biobank, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Harald Junge
- Department of Ophthalmology and Visual Neuroscience, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Zhe Chen
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN 55455, USA.
| |
Collapse
|
26
|
Harris H, Kittur J. Unlocking the potential of CRISPR-Cas9 for cystic fibrosis: A systematic literature review. Gene 2025; 942:149257. [PMID: 39832688 DOI: 10.1016/j.gene.2025.149257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 01/13/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
CRISPR-Cas9 technology has revolutionized genetic engineering, offering precise and efficient genome editing capabilities. This review explores the application of CRISPR-Cas9 for cystic fibrosis (CF), particularly targeting mutations in the CFTR gene. CF is a multiorgan disease primarily affecting the lungs, gastrointestinal system (e.g., CF-related diabetes (CFRD), CF-associated liver disease (CFLD)), bones (CF-bone disease), and the reproductive system. CF, a genetic disorder characterized by defective ion transport leading to thick mucus accumulation, is often caused by mutations like ΔF508 in the CFTR gene. This review employs a systematic methodology, incorporating an extensive literature search across multiple academic databases, including PubMed, Web of Science, and ScienceDirect, to identify 40 high-quality studies focused on CRISPR-Cas9 applications for CFTR gene editing. The data collection process involved predefined inclusion criteria targeting experimental approaches, gene-editing outcomes, delivery methods, and verification techniques. Data analysis synthesized findings on editing efficiency, off-target effects, and delivery system optimization to present a comprehensive overview of the field. The review highlights the historical development of CRISPR-Cas9, its mechanism, and its transformative role in genetic engineering and medicine. A detailed examination of CRISPR-Cas9's application in CFTR gene correction emphasizes the potential for therapeutic interventions while addressing challenges such as off-target effects, delivery efficiency, and ethical considerations. Future directions include optimizing delivery systems, integrating advanced editing tools like prime and base editing, and expanding personalized medicine approaches to improve treatment outcomes. By systematically analyzing the current landscape, this review provides a foundation for advancing CRISPR-Cas9 technologies for cystic fibrosis treatment and related disorders.
Collapse
Affiliation(s)
- Hudson Harris
- Department of Biomedical Engineering, Gallogly College of Engineering, University of Oklahoma Norman OK USA.
| | - Javeed Kittur
- Department of Biomedical Engineering, Gallogly College of Engineering, University of Oklahoma Norman OK USA
| |
Collapse
|
27
|
Liu D, Cao D, Han R. Recent advances in therapeutic gene-editing technologies. Mol Ther 2025:S1525-0016(25)00200-X. [PMID: 40119516 DOI: 10.1016/j.ymthe.2025.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/26/2025] [Accepted: 03/17/2025] [Indexed: 03/24/2025] Open
Abstract
The advent of gene-editing technologies, particularly CRISPR-based systems, has revolutionized the landscape of biomedical research and gene therapy. Ongoing research in gene editing has led to the rapid iteration of CRISPR technologies, such as base and prime editors, enabling precise nucleotide changes without the need for generating harmful double-strand breaks (DSBs). Furthermore, innovations such as CRISPR fusion systems with DNA recombinases, DNA polymerases, and DNA ligases have expanded the size limitations for edited sequences, opening new avenues for therapeutic development. Beyond the CRISPR system, mobile genetic elements (MGEs) and epigenetic editors are emerging as efficient alternatives for precise large insertions or stable gene manipulation in mammalian cells. These advances collectively set the stage for next-generation gene therapy development. This review highlights recent developments of genetic and epigenetic editing tools and explores preclinical innovations poised to advance the field.
Collapse
Affiliation(s)
- Dongqi Liu
- Department of Pediatrics, Department of Molecular and Medical Genetics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Di Cao
- Department of Pediatrics, Department of Molecular and Medical Genetics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Renzhi Han
- Department of Pediatrics, Department of Molecular and Medical Genetics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
28
|
Russell M, Trofimov A, Bradley P, Matsen IV F. Statistical analysis of repertoire data demonstrates the influence of microhomology in V(D)J recombination. Nucleic Acids Res 2025; 53:gkaf250. [PMID: 40173015 PMCID: PMC11963759 DOI: 10.1093/nar/gkaf250] [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: 11/14/2024] [Revised: 02/11/2025] [Accepted: 03/24/2025] [Indexed: 04/04/2025] Open
Abstract
V(D)J recombination generates the diverse B and T cell receptors essential for recognizing a wide array of antigens. This diversity arises from the combinatorial assembly of V(D)J genes and the junctional deletion and insertion of nucleotides. While previous in vitro studies have shown that microhomology-short stretches of sequence homology between gene ends-can bias the recombination process, the extent of microhomology's impact in vivo, particularly in humans, remains unknown. In this paper, we assess how germline-encoded microhomology influences trimming and ligation during V(D)J recombination using statistical inference on previously published high-throughput TCRα repertoire sequencing data. We find that microhomology increases both trimming and ligation probabilities, making it an important predictor of recombination outcomes. These effects are consistent across other receptor loci and sequence types. Further, we demonstrate that accounting for germline microhomology effects significantly alters sequence annotation probabilities and rankings, highlighting its practical importance for accurately inferring the V(D)J recombination events that generated an observed sequence. Together, these results enhance our understanding of how germline-encoded microhomologous nucleotides shape the human V(D)J recombination process.
Collapse
Affiliation(s)
- Magdalena L Russell
- Computational Biology Program, Fred Hutchinson Cancer Center, Seattle, WA 98109, United States
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, United States
| | - Assya Trofimov
- Computational Biology Program, Fred Hutchinson Cancer Center, Seattle, WA 98109, United States
- Department of Physics, University of Washington, Seattle, WA 98195, United States
| | - Philip Bradley
- Computational Biology Program, Fred Hutchinson Cancer Center, Seattle, WA 98109, United States
- Institute for Protein Design, Department of Biochemistry, University of Washington, Seattle, WA 98195, United States
| | - Frederick A Matsen IV
- Computational Biology Program, Fred Hutchinson Cancer Center, Seattle, WA 98109, United States
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, United States
- Department of Statistics, University of Washington, Seattle, WA 98195, United States
- Howard Hughes Medical Institute, Seattle, WA 98195, United States
| |
Collapse
|
29
|
Zhang T, Zhao W, Wirth C, Díaz-Gay M, Yin J, Cecati M, Marchegiani F, Hoang PH, Leduc C, Baine MK, Travis WD, Sholl LM, Joubert P, Sang J, McElderry JP, Klein A, Khandekar A, Hartman C, Rosenbaum J, Colón-Matos FJ, Miraftab M, Saha M, Lee OW, Jones KM, Caporaso NE, Wong MP, Leung KC, Agnes Hsiung C, Chen CY, Edell ES, Martínez Santamaría J, Schabath MB, Yendamuri SS, Manczuk M, Lissowska J, Świątkowska B, Mukeria A, Shangina O, Zaridze D, Holcatova I, Mates D, Milosavljevic S, Savic M, Bossé Y, Gould Rothberg BE, Christiani DC, Gaborieau V, Brennan P, Liu G, Hofman P, Homer R, Yang SR, Pesatori AC, Consonni D, Yang L, Zhu B, Shi J, Brown K, Rothman N, Chanock SJ, Alexandrov LB, Choi J, Cardelli M, Lan Q, Nowak MA, Wedge DC, Landi MT. Deciphering lung adenocarcinoma evolution and the role of LINE-1 retrotransposition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.643063. [PMID: 40161734 PMCID: PMC11952568 DOI: 10.1101/2025.03.14.643063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Understanding lung cancer evolution can identify tools for intercepting its growth. In a landscape analysis of 1024 lung adenocarcinomas (LUAD) with deep whole-genome sequencing integrated with multiomic data, we identified 542 LUAD that displayed diverse clonal architecture. In this group, we observed an interplay between mobile elements, endogenous and exogenous mutational processes, distinct driver genes, and epidemiological features. Our results revealed divergent evolutionary trajectories based on tobacco smoking exposure, ancestry, and sex. LUAD from smokers showed an abundance of tobacco-related C:G>A:T driver mutations in KRAS plus short subclonal diversification. LUAD in never smokers showed early occurrence of copy number alterations and EGFR mutations associated with SBS5 and SBS40a mutational signatures. Tumors harboring EGFR mutations exhibited long latency, particularly in females of European-ancestry (EU_N). In EU_N, EGFR mutations preceded the occurrence of other driver genes, including TP53 and RBM10. Tumors from Asian never smokers showed a short clonal evolution and presented with heterogeneous repetitive patterns for the inferred mutational order. Importantly, we found that the mutational signature ID2 is a marker of a previously unrecognized mechanism for LUAD evolution. Tumors with ID2 showed short latency and high L1 retrotransposon activity linked to L1 promoter demethylation. These tumors exhibited an aggressive phenotype, characterized by increased genomic instability, elevated hypoxia scores, low burden of neoantigens, propensity to develop metastasis, and poor overall survival. Reactivated L1 retrotransposition-induced mutagenesis can contribute to the origin of the mutational signature ID2, including through the regulation of the transcriptional factor ZNF695, a member of the KZFP family. The complex nature of LUAD evolution creates both challenges and opportunities for screening and treatment plans.
Collapse
Affiliation(s)
- Tongwu Zhang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Wei Zhao
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Christopher Wirth
- Manchester Cancer Research Centre, The University of Manchester, Manchester, UK
| | - Marcos Díaz-Gay
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
- Digital Genomics Group, Structural Biology Program, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Jinhu Yin
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Monia Cecati
- Advanced Technology Center for Aging Research, IRCCS INRCA, Ancona, Italy
| | | | - Phuc H Hoang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Charles Leduc
- Department of Pathology, Centre Hospitalier de l'Université de Montréal, Montreal, Canada
| | - Marina K Baine
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - William D Travis
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lynette M Sholl
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Philippe Joubert
- Institut universitaire de cardiologie et de pneumologie de Québec, Laval University, Quebec City, Canada
| | - Jian Sang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - John P McElderry
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Alyssa Klein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Azhar Khandekar
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Caleb Hartman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | | | - Frank J Colón-Matos
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Mona Miraftab
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Monjoy Saha
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Olivia W Lee
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Kristine M Jones
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
- Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Neil E Caporaso
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
- Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Maria Pik Wong
- Queen Mary Hospital, The University of Hong Kong, Hong Kong, China
| | - Kin Chung Leung
- Department of Pathology, The University of Hong Kong, Hong Kong, China
| | - Chao Agnes Hsiung
- Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Taiwan
| | - Chih-Yi Chen
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Surgery, Division of Thoracic Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Eric S Edell
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Matthew B Schabath
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Sai S Yendamuri
- Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Marta Manczuk
- Department of Cancer Epidemiology and Primary Prevention, Maria Skłodowska-Curie National Research Institute of Oncology, Warshaw, Poland
| | - Jolanta Lissowska
- Department of Cancer Epidemiology and Primary Prevention, Maria Skłodowska-Curie National Research Institute of Oncology, Warshaw, Poland
| | - Beata Świątkowska
- Department of Environmental Epidemiology, Nofer Institute of Occupational Medicine, Łódź, Poland
| | - Anush Mukeria
- Department of Clinical Epidemiology, N.N. Blokhin National Medical Research Centre of Oncology, Moscow, Russia
| | - Oxana Shangina
- Department of Clinical Epidemiology, N.N. Blokhin National Medical Research Centre of Oncology, Moscow, Russia
| | - David Zaridze
- Department of Clinical Epidemiology, N.N. Blokhin National Medical Research Centre of Oncology, Moscow, Russia
| | - Ivana Holcatova
- Institute of Hygiene and Epidemiology, 1st Faculty of Medicine, Charles University, Prague, Czech Republic
- Department of Oncology, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Dana Mates
- Department of Occupational Health and Toxicology, National Center for Environmental Risk Monitoring, National Institute of Public Health, Bucharest, Romania
| | - Sasa Milosavljevic
- International Organisation for Cancer Prevention and Research (IOCPR), Belgrade, Serbia
| | - Milan Savic
- Department of Thoracic Surgery, Clinical Center of Serbia, Belgrade, Serbia
| | - Yohan Bossé
- Institut universitaire de cardiologie et de pneumologie de Québec, Laval University, Quebec City, Canada
| | - Bonnie E Gould Rothberg
- Sylvester Comprehensive Cancer Center, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - David C Christiani
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Valerie Gaborieau
- Genomic Epidemiology Branch, International Agency for Research on Cancer (IARC/WHO), Lyon, France
| | - Paul Brennan
- Genomic Epidemiology Branch, International Agency for Research on Cancer (IARC/WHO), Lyon, France
| | - Geoffrey Liu
- Princess Margaret Cancer Center, University of Toronto, Toronto, Ontario, Canada
| | - Paul Hofman
- IHU RespirERA, Biobank-BB-0033-0025, Côte d'Azur University, Nice, France
| | - Robert Homer
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Soo-Ryum Yang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Angela C Pesatori
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Dario Consonni
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Lixing Yang
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA
- Department of Human Genetics, The University of Chicago, Chicago, IL, USA
- The University of Chicago Medicine Comprehensive Cancer Center, The University of Chicago, Chicago, IL, USA
| | - Bin Zhu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Jianxin Shi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Kevin Brown
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Nathaniel Rothman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Ludmil B Alexandrov
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
- Sanford Stem Cell Institute, University of California San Diego, La Jolla, CA, USA
| | - Jiyeon Choi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Maurizio Cardelli
- Advanced Technology Center for Aging Research, IRCCS INRCA, Ancona, Italy
| | - Qing Lan
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Martin A Nowak
- Department of Mathematics, Harvard University, Cambridge, MA, USA
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - David C Wedge
- Manchester Cancer Research Centre, The University of Manchester, Manchester, UK
- Manchester NIHR Biomedical Research Centre, Manchester, UK
| | - Maria Teresa Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
30
|
Solomon AD, Gouttia OG, Wang L, Zhu S, Wang F, Li Y, Paydar M, Bessho T, Kwok BH, Peng A. γ-tubulin mediates DNA double-strand break repair. J Cell Sci 2025; 138:jcs262255. [PMID: 40135584 PMCID: PMC12050090 DOI: 10.1242/jcs.262255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 12/20/2024] [Indexed: 03/27/2025] Open
Abstract
Double-strand breaks (DSBs) in DNA pose a critical threat to genomic integrity, potentially leading to the onset and progression of various diseases, including cancer. Cellular responses to such lesions entail sophisticated repair mechanisms primarily mediated by non-homologous end joining (NHEJ) and homologous recombination (HR). Interestingly, the efficient recruitment of repair proteins and completion of DSB repair likely involve complex, inter-organelle communication and coordination of cellular components. In this study, we report a role of γ-tubulin in DSB repair. γ-tubulin is a major microtubule nucleation factor governing microtubule dynamics. We show that γ-tubulin is recruited to the site of DNA damage and is required for efficient DSB repair via both NHEJ and HR. Suppression of γ-tubulin impedes DNA repair and exacerbates DNA damage accumulation. Furthermore, γ-tubulin mediates the mobilization and formation of DNA damage foci, which serve as repair centers, thereby facilitating the recruitment of HR and NHEJ repair proteins on damaged chromatin. Finally, pharmacological inhibition of γ-tubulin enhances the cytotoxic effect of DNA-damaging agents, consistent with the DNA repair function of γ-tubulin, and underscoring the potential of its therapeutic intervention in cancer therapy.
Collapse
Affiliation(s)
- Abhishikt David Solomon
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Odjo G. Gouttia
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ling Wang
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Songli Zhu
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE 68583, USA
| | - Feifei Wang
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE 68583, USA
| | - Yanqui Li
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE 68583, USA
| | - Mohammadjavad Paydar
- Institute for Research in Immunology and Cancer (IRIC), Département de médecine, Université de Montréal, Montréal H3C 3J7, Canada
| | - Tadayoshi Bessho
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Benjamin H. Kwok
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE 68583, USA
| | - Aimin Peng
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
31
|
Gong P, Guo Z, Wang S, Gao S, Cao Q. Histone Phosphorylation in DNA Damage Response. Int J Mol Sci 2025; 26:2405. [PMID: 40141048 PMCID: PMC11941871 DOI: 10.3390/ijms26062405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/01/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
The DNA damage response (DDR) is crucial for maintaining genomic stability and preventing the accumulation of mutations that can lead to various diseases, including cancer. The DDR is a complex cellular regulatory network that involves DNA damage sensing, signal transduction, repair, and cell cycle arrest. Modifications in histone phosphorylation play important roles in these processes, facilitating DNA repair factor recruitment, damage signal transduction, chromatin remodeling, and cell cycle regulation. The precise regulation of histone phosphorylation is critical for the effective repair of DNA damage, genomic integrity maintenance, and the prevention of diseases such as cancer, where DNA repair mechanisms are often compromised. Thus, understanding histone phosphorylation in the DDR provides insights into DDR mechanisms and offers potential therapeutic targets for diseases associated with genomic instability, including cancers.
Collapse
Affiliation(s)
- Ping Gong
- Hunan Institute of Microbiology, Changsha 410009, China; (Z.G.); (S.W.); (S.G.)
| | - Zhaohui Guo
- Hunan Institute of Microbiology, Changsha 410009, China; (Z.G.); (S.W.); (S.G.)
| | - Shengping Wang
- Hunan Institute of Microbiology, Changsha 410009, China; (Z.G.); (S.W.); (S.G.)
| | - Shufeng Gao
- Hunan Institute of Microbiology, Changsha 410009, China; (Z.G.); (S.W.); (S.G.)
| | - Qinhong Cao
- College of Biological Sciences, China Agricultural University, No.2 Yuan-Ming-Yuan West Road, Beijing 100193, China
| |
Collapse
|
32
|
Bhat A, Bhan S, Kabiraj A, Pandita RK, Ramos KS, Nandi S, Sopori S, Sarkar PS, Dhar A, Pandita S, Kumar R, Das C, Tainer JA, Pandita TK. A predictive chromatin architecture nexus regulates transcription and DNA damage repair. J Biol Chem 2025; 301:108300. [PMID: 39947477 PMCID: PMC11931391 DOI: 10.1016/j.jbc.2025.108300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 12/16/2024] [Accepted: 01/16/2025] [Indexed: 03/28/2025] Open
Abstract
Genomes are blueprints of life essential for an organism's survival, propagation, and evolutionary adaptation. Eukaryotic genomes comprise of DNA, core histones, and several other nonhistone proteins, packaged into chromatin in the tiny confines of nucleus. Chromatin structural organization restricts transcription factors to access DNA, permitting binding only after specific chromatin remodeling events. The fundamental processes in living cells, including transcription, replication, repair, and recombination, are thus regulated by chromatin structure through ATP-dependent remodeling, histone variant incorporation, and various covalent histone modifications including phosphorylation, acetylation, and ubiquitination. These modifications, particularly involving histone variant H2AX, furthermore play crucial roles in DNA damage responses by enabling repair protein's access to damaged DNA. Chromatin also stabilizes the genome by regulating DNA repair mechanisms while suppressing damage from endogenous and exogenous sources. Environmental factors such as ionizing radiations induce DNA damage, and if repair is compromised, can lead to chromosomal abnormalities and gene amplifications as observed in several tumor types. Consequently, chromatin architecture controls the genome fidelity and activity: it orchestrates correct gene expression, genomic integrity, DNA repair, transcription, replication, and recombination. This review considers connecting chromatin organization to functional outcomes impacting transcription, DNA repair and genomic integrity as an emerging grand challenge for predictive molecular cell biology.
Collapse
Affiliation(s)
- Audesh Bhat
- Centre for Molecular Biology, Central University of Jammu, Jammu and Kashmir, India.
| | - Sonali Bhan
- Centre for Molecular Biology, Central University of Jammu, Jammu and Kashmir, India
| | - Aindrila Kabiraj
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India; Homi Bhabha National Institute, BARC Training School Complex, Mumbai, Maharashtra, India
| | - Raj K Pandita
- Center for Genomics and Precision Medicine, Texas A&M College of Medicine, Houston, Texas, USA
| | - Keneth S Ramos
- Center for Genomics and Precision Medicine, Texas A&M College of Medicine, Houston, Texas, USA
| | - Sandhik Nandi
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India; Homi Bhabha National Institute, BARC Training School Complex, Mumbai, Maharashtra, India
| | - Shreya Sopori
- Centre for Molecular Biology, Central University of Jammu, Jammu and Kashmir, India
| | - Parthas S Sarkar
- Department of Neurobiology and Neurology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Arti Dhar
- Department of Pharmacy, Birla Institute of Technology and Sciences Pilani, Hyderabad Campus, Telangana, India
| | | | - Rakesh Kumar
- Department of Biotechnology, Shri Mata Vaishnav Devi University, Katra, India
| | - Chandrima Das
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India; Homi Bhabha National Institute, BARC Training School Complex, Mumbai, Maharashtra, India.
| | - John A Tainer
- Department of Molecular & Cellular Oncology and Department of Cancer Biology, UT MD Anderson Cancer Center, Houston, Texas, USA
| | - Tej K Pandita
- Center for Genomics and Precision Medicine, Texas A&M College of Medicine, Houston, Texas, USA.
| |
Collapse
|
33
|
Chung OW, Yao S, Yang F, Wang L, Cerda-Smith C, Hutchinson HM, Wood KC, Su W, Khasraw M, Zou L, Ramsden DA, Zhang ZZZ. BRCA1-A and LIG4 complexes mediate ecDNA biogenesis and cancer drug resistance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.18.638901. [PMID: 40027615 PMCID: PMC11870461 DOI: 10.1101/2025.02.18.638901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Extrachromosomal circular DNA (ecDNA) are commonly produced within the nucleus to drive genome dynamics and heterogeneity, enabling cancer cell evolution and adaptation. However, the mechanisms underlying ecDNA biogenesis remain poorly understood. Here using genome-wide CRISPR screening in human cells, we identified the BRCA1-A and the LIG4 complexes mediate ecDNA production. Following DNA fragmentation, the upstream BRCA1-A complex protects DNA ends from excessive resection, promoting end-joining for circularization. Conversely, the MRN complex, which mediates end resection and thus antagonizes the BRCA1-A complex, suppresses ecDNA formation. Downstream, LIG4 conservatively catalyzes ecDNA production in Drosophila and mammals, with patient tumor ecDNA harboring junctions marked by LIG4 activity. Notably, disrupting LIG4 or BRCA1-A in cancer cells impairs ecDNA-mediated adaptation, hindering resistance to both chemotherapy and targeted therapies. Together, our study reveals the roles of the LIG4 and BRCA1-A complexes in ecDNA biogenesis, and uncovers new therapeutic targets to block ecDNA-mediated adaptation for cancer treatment.
Collapse
Affiliation(s)
- Oliver W. Chung
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, USA
- These authors contributed equally
| | - Shun Yao
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, USA
- These authors contributed equally
| | - Fu Yang
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, USA
- Present address: Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- These authors contributed equally
| | - Ling Wang
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, USA
| | - Christian Cerda-Smith
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, USA
| | - Haley M. Hutchinson
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, USA
| | - Kris C. Wood
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, USA
| | - Weijia Su
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, USA
| | - Mustafa Khasraw
- The Preston Robert Tisch Brain Tumor Center at Duke, Department of Neurosurgery, Duke University Medical Center, Durham, USA
| | - Lee Zou
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, USA
| | - Dale A. Ramsden
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - ZZ Zhao Zhang
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, USA
- Lead contact
| |
Collapse
|
34
|
Tang H, Gao J, Wang H, Sun M, Zhang S, Song C, Li Q. Characterization of the genome editing with miniature DNA nucleases TnpB and IscB in Escherichia coli strains. Commun Biol 2025; 8:261. [PMID: 39972101 PMCID: PMC11840021 DOI: 10.1038/s42003-025-07521-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 01/11/2025] [Indexed: 02/21/2025] Open
Abstract
DNA endonucleases TnpB and IscB are emerging candidates for combating drug-resistant bacteria, particularly Escherichia coli, due to their specificity in targeting DNA and smaller size. However, the genome-editing of TnpB/IscB in E. coli remains unclear. This study characterized the genome editing of TnpB/IscB in different E. coli strains. First, the toxicity and cleavage results indicated TnpB was effective only in MG1655, whereas IscB and enIscB demonstrated functionality in ATCC9637/BL21(DE3). Subsequently, a genome-editing tool was established in MG1655 by using TnpB (as a thermophilic programmable endonuclease), achieving up to 100% editing efficiency, while IscB/enIscB achieved editing in ATCC9637/BL21(DE3). Additionally, the editing plasmids were successfully cured. Finally, the mechanism underlying the escape of E. coli during TnpB/IscB editing was elucidated. Overall, this study successfully applied TnpB/IscB/enIscB to genome editing in E. coli, which will expand the genetic manipulation toolbox in E. coli and facilitate the development of the antimicrobial drugs.
Collapse
Affiliation(s)
- Hongjie Tang
- College of Life Sciences, Sichuan Normal University, Chengdu, 610101, China
| | - Jie Gao
- College of Life Sciences, Sichuan Normal University, Chengdu, 610101, China
| | - Hengyi Wang
- College of Life Sciences, Sichuan Normal University, Chengdu, 610101, China
| | - Mingjun Sun
- College of Life Sciences, Sichuan Normal University, Chengdu, 610101, China
| | - Suyi Zhang
- Luzhou Laojiao Co. Ltd, Luzhou, 646000, China
- National Engineering Research Center of Solid-State Brewing, Luzhou, 646000, China
| | - Chuan Song
- Luzhou Laojiao Co. Ltd, Luzhou, 646000, China
- National Engineering Research Center of Solid-State Brewing, Luzhou, 646000, China
| | - Qi Li
- College of Life Sciences, Sichuan Normal University, Chengdu, 610101, China.
| |
Collapse
|
35
|
Wang T, Wang Z. Targeting the "Undruggable": Small-Molecule Inhibitors of Proliferating Cell Nuclear Antigen (PCNA) in the Spotlight in Cancer Therapy. J Med Chem 2025; 68:2058-2088. [PMID: 39904718 DOI: 10.1021/acs.jmedchem.4c00526] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
PCNA plays multiple roles in cancer development, including cell proliferation regulation, DNA repair, replication, and serving as a widely used biomarker and therapeutic target. Despite its significant role in oncology, PCNA has historically been considered "undruggable" due to the absence of known endogenous small molecule modulators and identifiable ligand binding sites. Unlike other protein-protein interfaces, PCNA lacks explicit binding grooves, featuring a relatively small and shallow surface pocket, which hinders the discovery of traditional small molecule targets. Recent breakthroughs have introduced promising PCNA-targeting candidates, with ATX-101 and AOH1996 entering phase I clinical trials for cancer therapy, garnering academic and industry interest. These achievements provide new evidence for PCNA as a drug target. This article provides insight and perspective on the application of small-molecule PCNA inhibitors in cancer treatment, covering PCNA function, its relationship with cancer, structural modification of small molecule inhibitors, and discovery strategies.
Collapse
Affiliation(s)
- Tiantian Wang
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, P. R. China
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, P. R. China
| | - Zengtao Wang
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, P. R. China
- Jiangxi Provincial Key Laboratory of TCM Female Reproductive Health and Related Diseases Research and Transformation, Jiangxi University of Chinese Medicine, Nanchang 330004, P. R. China
| |
Collapse
|
36
|
Tripathi D, Pyla M, Dutta AK, Matsika S. Impact of solvation on the electronic resonances in uracil. Phys Chem Chem Phys 2025; 27:3588-3601. [PMID: 39903129 DOI: 10.1039/d4cp04333b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Interactions of low-energy electrons with the DNA and RNA nucleobases are known to form metastable states, known as electronic resonances. In this work, we study electron attachment to solvated uracil, an RNA nucleobase, using the orbital stabilization method at the Equation of Motion-Coupled Cluster for Electron Affinities with Singles and Doubles (EOM-EA-CCSD) level of theory with the Effective Fragment Potential (EFP) solvation method. We benchmarked the approach using multireference methods, as well as by comparing EFP and full quantum calculations. The impact of solvation on the first one particle (1p) shape resonance, formed by electron attachment to the π* LUMO orbital, as well as the first two particle one hole (2p1h) resonance, formed by electron attachment to neutral uracil's π-π* excited state, was investigated. We used molecular dynamics simulations for solvent configurations and applied charge stabilization technique-based biased sampling to procure configurations adequate to cover the entire range of the electron attachment energy distribution. The electron attachment energy in solution is found to be distributed over a wide range of energies, between 4.6 eV to 6.8 eV for the 2p1h resonance, and between -0.1 eV to 2 eV for the 1p resonance. The solvent effects were similar for the two resonances, indicating that the exact electron density of the state is not as important as the solvent configurations. Multireference calculations extended the findings showing that solvation effects are similar for the lowest four resonances, further indicating that the specific solute electron density is not as important, but rather the water configurations play the most important role in solvation effects. Finally, by comparing bulk solvation to clusters of uracil with a few water molecules around it, we find that the impact of microsolvation is very different from that of bulk solvation.
Collapse
|
37
|
Shokoohi M, Sedaghatshoar S, Arian H, Mokarami M, Habibi F, Bamarinejad F. Genetic advancements in breast cancer treatment: a review. Discov Oncol 2025; 16:127. [PMID: 39918655 PMCID: PMC11805739 DOI: 10.1007/s12672-025-01884-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/03/2025] [Indexed: 02/09/2025] Open
Abstract
Breast cancer (BC) remains a leading cause of cancer-related deaths among women globally, highlighting the urgent need for more effective and targeted therapies. Traditional treatments, including surgery, chemotherapy, and radiation, face limitations such as drug resistance, metastasis, and severe side effects. Recent advancements in gene therapy, particularly CRISPR/Cas9 technology and Oncolytic Virotherapy (OVT), are transforming the BC treatment landscape. CRISPR/Cas9 enables precise gene editing to correct mutations in oncogenes like HER2 and MYC, directly addressing tumor growth and immune evasion. Simultaneously, OVT leverages genetically engineered viruses to selectively destroy cancer cells and stimulate robust antitumor immune responses. Despite their potential, gene therapies face challenges, including off-target effects, delivery issues, and ethical concerns. Innovations in delivery systems, combination strategies, and integrating gene therapy with existing treatments offer promising solutions to overcome these barriers. Personalized medicine, guided by genomic profiling, further enhances treatment precision by identifying patient-specific mutations, such as BRCA1 and BRCA2, allowing for more tailored and effective interventions. As research progresses, the constructive interaction between gene therapy, immunotherapy, and traditional approaches is paving the way for groundbreaking advancements in BC care. Continued collaboration between researchers and clinicians is essential to translate these innovations into clinical practice, ultimately improving BC patients' survival rates and quality of life.
Collapse
Affiliation(s)
- Marzieh Shokoohi
- Department of Life Sciences Engineering, Faculty of New Sciences & Technologies, University of Tehran, Tehran, Iran.
- Amino Techno Gene Virtual Private Laboratory, Tehran, Iran.
| | - Sadaf Sedaghatshoar
- Kent School of Social Work and Family Science, University of Louisville, Louisville, KY, USA
| | - Homaira Arian
- Pharmaceutical Biotechnology Department, Pharmacy Faculty, Anadolu University, Eskishehir, Turkey.
| | - Milad Mokarami
- Student Research Committee, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Fatemeh Habibi
- Department of Speech Therapy, School of Rehabilitation, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Bamarinejad
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
38
|
Chen Y, Zhu H, Guo F, Wang L, Zhang W, Liu R, Zhang X, Dai X. Vitrification affects the post-implantation development of mouse embryos by inducing DNA damage and epigenetic modifications. Clin Epigenetics 2025; 17:20. [PMID: 39920865 PMCID: PMC11806629 DOI: 10.1186/s13148-025-01826-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 01/27/2025] [Indexed: 02/09/2025] Open
Abstract
Vitrification is widely used in assisted reproductive technology (ART) for female infertility, but the long-term effect on the embryo of vitrification has not been comprehensively studied. The study aimed to investigate the effect of vitrification on long-term development of mouse embryos. The warmed embryos which were frozen at 8-cell stage were cultured in vitro until the blastocyst stage and were transferred into recipients. Immunofluorescence staining was performed to evaluate the reactive oxygen species (ROS) level, mitochondrial function, cell apoptosis, DNA damage and histone epigenetic modification in blastocysts. Transmission electron microscopy (TEM) analysis was performed to exam the mitochondrial ultrastructure in blastocysts. The related gene expression and transcriptome profiles were investigated by RT-qPCR and RNA-seq, respectively. Blastocyst and implantation frequencies were not significantly affected by vitrification. However, vitrification significantly reduced blastocyst cell number and the live pup frequency. Vitrification induced ROS accumulation, DNA damage, and apoptosis in mouse blastocysts. The homologous recombination (NHEJ) is the major DNA repair pathway for vitrified embryos. Vitrification elevated H3K4me2/3, H4K12ac, and H4K16ac levels and reduced m6A modification in blastocysts. Moreover, vitrification significantly altered transcriptome profiles of mice placentas and brains at embryonic day 18.5 (E18.5). Thus, vitrification exhibited a long-term effect on mouse embryo viability by increasing ROS levels, DNA damage, altering the epigenetic modifications and transcriptome profiles.
Collapse
Affiliation(s)
- Yurong Chen
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital of Jilin University, Changchun, China
- Reproductive Medicine Center, Xiamen University Affiliated Chenggong Hospital, Xiamen, Fujian, China
| | - Haibo Zhu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital of Jilin University, Changchun, China
- Center of Reproductive Medicine and Center of Prenatal Diagnosis, First Hospital of Jilin University, Changchun, China
| | - Fucheng Guo
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital of Jilin University, Changchun, China
| | - Luyao Wang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital of Jilin University, Changchun, China
| | - Wenli Zhang
- Comprehensive Testing and Analytical Center of North China University of Science and Technology, Tangshan, Hebei, China
| | - Ruizhi Liu
- Center of Reproductive Medicine and Center of Prenatal Diagnosis, First Hospital of Jilin University, Changchun, China
| | - Xiaoling Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital of Jilin University, Changchun, China.
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital of Jilin University, Changchun, China.
| | - Xiangpeng Dai
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital of Jilin University, Changchun, China.
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
39
|
Borphukan B, Khatun M, Fartyal D, James D, Reddy MK. A Gemini Virus-Derived Autonomously Replicating System for HDR-Mediated Genome Editing of the EPSP Synthase Gene in Indica Rice. PLANTS (BASEL, SWITZERLAND) 2025; 14:477. [PMID: 39943039 PMCID: PMC11821261 DOI: 10.3390/plants14030477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/22/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025]
Abstract
CRISPR/Cas9-mediated homology-directed repair (HDR) is a powerful tool for precise genome editing in plants, but its efficiency remains low, particularly for targeted amino acid substitutions or gene knock-ins. Successful HDR requires the simultaneous presence of Cas9, guide RNA, and a repair template (RT) in the same cell nucleus. Among these, the timely availability of the RT at the double-strand break (DSB) site is a critical bottleneck. To address this, we developed a sequential transformation strategy incorporating a deconstructed wheat dwarf virus (dWDV)-based autonomously replicating delivery system, effectively simplifying the process into a two-component system. Using this approach, we successfully achieved the targeted editing of the OsEPSPS gene in rice with a 10 percent HDR efficiency, generating three lines (TIPS1, TIPS2, and TIPS3) with amino acid substitutions (T172I and P177S) in the native EPSPS protein. The modifications were confirmed through Sanger sequencing and restriction digestion assays, and the edited lines showed no yield penalties compared to wild-type plants. This study demonstrates the utility of viral replicons in delivering gene-editing tools for precise genome modification, offering a promising approach for efficient HDR in crop improvement programs.
Collapse
Affiliation(s)
- Bhabesh Borphukan
- Department of Crop and Soil Sciences, Washington State University, Pullman, WA 99163, USA
- Crop Improvement Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India;
| | - Muslima Khatun
- Molecular Biotechnology Division, National Institute of Biotechnology, Dhaka 1349, Bangladesh;
| | - Dhirendra Fartyal
- Plant Nutrition Division, INRES, University of Bonn, 53113 Bonn, Germany;
| | - Donald James
- Department of Biotechnology, Forest Genetics and Biotechnology Division, Kerala Forest Research Institute, Thrissur 680653, India;
| | - Malireddy K. Reddy
- Crop Improvement Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India;
| |
Collapse
|
40
|
Karaaslan BG, Demirkale ZH, Turan I, Aydemir S, Meric Z, Taskin Z, Kilinc OC, Burtecene N, Topcu B, Yucel E, Aydogmus C, Cokugras H, Kiykim A. Evaluation of T-cell repertoire by flow cytometric analysis in primary immunodeficiencies with DNA repair defects. Scand J Immunol 2025; 101:e70003. [PMID: 39967281 PMCID: PMC11836546 DOI: 10.1111/sji.70003] [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: 07/05/2024] [Revised: 01/07/2025] [Accepted: 01/13/2025] [Indexed: 02/20/2025]
Abstract
The group of patients with DNA-repair-defects increases susceptibility to infections due to impaired repertoire diversity. In this context, we aimed to investigate the TCRvβ-repertoire by flow cytometric analysis and its correlation with clinical entities in a group of IEI patients with DNA repair defects. Peripheral lymphocyte subset and TCRvβ-repertoire analyses were performed by flow cytometric analysis. The aim was to explore the changing TCR-Vβ-repertoire that can predict some clinical entities by investigating the repertoire using flow-cytometric-analysis-based TCR-Vβ and its interaction with clinical entities in a group of IEI patients with DNA repair defects. TCR-repertoire of the patients with DNA-repair-defects and healthy controls was analysed with flow-cytometer. The potential of flow-cytometric analysis of the TCR repertoire as a practical and easily accessible clinical prediction method was investigated. Thirty-nine-IEI patients with DNA-repair-defects and 15 age-matched healthy-controls were included in this study. Peripheral lymphocyte subset and TCR-Vβ repertoire analyses were performed by flow cytometry. Compared to the control group, 9 out of 24 clones (37.5%) exhibited a statistically significant reduction, while only 3 clones showed a statistically significant increase (p < 0.05). Preferential use of vβ-genes was associated with some clinical entities. Lower TCR-vβ-9 and TCR-vβ23, higher TCR-vβ7.2 were found in the patients with pneumonia (n = 13) (p = 0.018, p = 0.044 p = 0.032). AT patients with pneumonia had lower TCR-vβ-9 clone than patients without pneumonia (p = 0.008). Skewed proliferation of most TCR-vβ clones was seen DNA-repair-defects, especially AT. In addition, this study showed that preferential use of TCR-vβ genes could be predictive for some clinical entities.
Collapse
Affiliation(s)
- Betul Gemici Karaaslan
- Cerrahpasa School of Medicine, Department of Pediatric Immunology and AllergyIstanbul University‐CerrahpasaIstanbulTürkiye
| | - Zeynep Hizli Demirkale
- Istanbul Medical Faculty, Department of Pediatric Immunology and AllergyIstanbul UniversityIstanbulTürkiye
| | - Isilay Turan
- Department of Pediatric Immunology and AllergyBasaksehir Cam and Sakura City HospitalIstanbulTürkiye
| | - Sezin Aydemir
- Cerrahpasa School of Medicine, Department of Pediatric Immunology and AllergyIstanbul University‐CerrahpasaIstanbulTürkiye
| | - Zeynep Meric
- Cerrahpasa School of Medicine, Department of Pediatric Immunology and AllergyIstanbul University‐CerrahpasaIstanbulTürkiye
| | - Zuleyha Taskin
- Cerrahpasa School of MedicineIstanbul University‐CerrahpasaIstanbulTürkiye
| | - Ozgur Can Kilinc
- Cerrahpasa School of MedicineIstanbul University‐CerrahpasaIstanbulTürkiye
| | - Nihan Burtecene
- Cerrahpasa School of Medicine, Department of Pediatric Immunology and AllergyIstanbul University‐CerrahpasaIstanbulTürkiye
| | - Birol Topcu
- Department of BiostatisticsTekirdag Namik Kemal UniversityTekirdagTürkiye
| | - Esra Yucel
- Istanbul Medical Faculty, Department of Pediatric Immunology and AllergyIstanbul UniversityIstanbulTürkiye
| | - Cigdem Aydogmus
- Department of Pediatric Immunology and AllergyBasaksehir Cam and Sakura City HospitalIstanbulTürkiye
| | - Haluk Cokugras
- Cerrahpasa School of Medicine, Department of Pediatric Immunology and AllergyIstanbul University‐CerrahpasaIstanbulTürkiye
| | - Ayca Kiykim
- Cerrahpasa School of Medicine, Department of Pediatric Immunology and AllergyIstanbul University‐CerrahpasaIstanbulTürkiye
| |
Collapse
|
41
|
Karami Fath M, Najafiyan B, Morovatshoar R, Khorsandi M, Dashtizadeh A, Kiani A, Farzam F, Kazemi KS, Nabi Afjadi M. Potential promising of synthetic lethality in cancer research and treatment. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:1403-1431. [PMID: 39305329 DOI: 10.1007/s00210-024-03444-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 09/08/2024] [Indexed: 02/14/2025]
Abstract
Cancer is a complex disease driven by multiple genetic changes, including mutations in oncogenes, tumor suppressor genes, DNA repair genes, and genes involved in cancer metabolism. Synthetic lethality (SL) is a promising approach in cancer research and treatment, where the simultaneous dysfunction of specific genes or pathways causes cell death. By targeting vulnerabilities created by these dysfunctions, SL therapies selectively kill cancer cells while sparing normal cells. SL therapies, such as PARP inhibitors, WEE1 inhibitors, ATR and ATM inhibitors, and DNA-PK inhibitors, offer a distinct approach to cancer treatment compared to conventional targeted therapies. Instead of directly inhibiting specific molecules or pathways, SL therapies exploit genetic or molecular vulnerabilities in cancer cells to induce selective cell death, offering benefits such as targeted therapy, enhanced treatment efficacy, and minimized harm to healthy tissues. SL therapies can be personalized based on each patient's unique genetic profile and combined with other treatment modalities to potentially achieve synergistic effects. They also broaden the effectiveness of treatment across different cancer types, potentially overcoming drug resistance and improving patient outcomes. This review offers an overview of the current understanding of SL mechanisms, advancements, and challenges, as well as the preclinical and clinical development of SL. It also discusses new directions and opportunities for utilizing SL in targeted therapy for anticancer treatment.
Collapse
Affiliation(s)
- Mohsen Karami Fath
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Behnam Najafiyan
- Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Morovatshoar
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mahdieh Khorsandi
- Department of Biotechnology, Faculty of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Arash Kiani
- Student Research Committee, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Farnoosh Farzam
- Department of Biochemistry, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran
| | - Kimia Sadat Kazemi
- Faculty of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohsen Nabi Afjadi
- Department of Biochemistry, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
42
|
Kumari N, Kaur E, Raghavan SC, Sengupta S. Regulation of pathway choice in DNA repair after double-strand breaks. Curr Opin Pharmacol 2025; 80:102496. [PMID: 39724838 DOI: 10.1016/j.coph.2024.102496] [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: 01/27/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024]
Abstract
DNA damage signaling is a highly coordinated cellular process which is required for the removal of DNA lesions. Amongst the different types of DNA damage, double-strand breaks (DSBs) are the most harmful type of lesion that attenuates cellular proliferation. DSBs are repaired by two major pathways-homologous recombination (HR), and non-homologous end-joining (NHEJ) and in some cases by microhomology-mediated end-joining (MMEJ). Preference of the pathway depends on multiple parameters including site of the DNA damage, the cell cycle phase and topology of the DNA lesion. Deregulated repair response contributes to genomic instability resulting in a plethora of diseases including cancer. This review discusses the different molecular players of HR, NHEJ, and MMEJ pathways that control the switch among the different DSB repair pathways. We also highlight the various functions of chromatin modifications in modulating repair response and how deregulated DNA damage repair response may promote oncogenic transformation.
Collapse
Affiliation(s)
- Nitu Kumari
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | - Ekjot Kaur
- Biotechnology Research and Innovation Council - National Institute of Immunology (BRIC-NII), Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Sathees C Raghavan
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India.
| | - Sagar Sengupta
- Biotechnology Research and Innovation Council - National Institute of Immunology (BRIC-NII), Aruna Asaf Ali Marg, New Delhi 110067, India; Biotechnology Research and Innovation Council - National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani 741251, India.
| |
Collapse
|
43
|
Zhu Y, Li A, Maji S, Lee BJ, Korn SM, Gertie JA, Dorrity TJ, Wang J, Wang KJ, Pelletier A, Moakley DF, Kelly RD, Holmes AB, Rabadan R, Edgell DR, Schild Poulter C, Modesti M, Steckelberg AL, Hendrickson EA, Chung H, Zhang C, Zha S. Ku suppresses RNA-mediated innate immune responses in human cells to accommodate primate-specific Alu expansion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.31.633084. [PMID: 39975384 PMCID: PMC11838425 DOI: 10.1101/2025.01.31.633084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Ku70 and Ku80 form Ku, a ring-shaped protein that initiates the non-homologous end-joining (NHEJ) DNA repair pathway. 1 Specifically, Ku binds to double-stranded DNA (dsDNA) ends and recruits other NHEJ factors ( e.g. , DNA-PKcs and LIG4). While Ku binds to double-stranded RNA (dsRNA) 2 and traps mutated-DNA-PKcs on ribosomal RNA in vivo, 3,4 the physiological significance of Ku-dsRNA interactions in otherwise wild-type cells remains elusive. Intriguingly, while dispensable for murine development, 5,6 Ku is essential in human cells. 7 Despite similar genome sizes, human cells express ∼100-fold more Ku than mouse cells, implying functions beyond NHEJ, possibly through a dose-sensitive interaction with dsRNA, which is ∼100 times weaker than with dsDNA. 2,8 While investigating the essentiality of Ku in human cells, we found that depletion of Ku - unlike LIG4 - induces profound interferon (IFN) and NF-kB responses reliant on the dsRNA-sensor MDA5/RIG-I and adaptor MAVS. Prolonged Ku-degradation also activates other dsRNA-sensors, e.g. PKR that suppresses protein translation, and OAS/RNaseL that cleaves rRNAs and eventually induces growth arrest and cell death. MAVS, RIG-I, or MDA5 knockouts suppressed IFN signaling and, together with PKR knockouts, partially rescued Ku-depleted human cells. Ku-irCLIP analyses revealed that Ku binds to diverse dsRNA, predominantly stem-loops in primate-specific Alu elements 9 at anti-sense orientation in introns and 3'-UTRs. Ku expression rose sharply in higher primates tightly correlating with Alu-expansion (r = 0.94/0.95). Together, our study identified a vital role of Ku in accommodating Alu-expansion in primates by mitigating a dsRNA-induced innate immune response, explaining the rise of Ku levels and its essentiality in human cells.
Collapse
|
44
|
Shahi A, Kidane D. Decoding mitochondrial DNA damage and repair associated with H. pylori infection. Front Cell Infect Microbiol 2025; 14:1529441. [PMID: 39906209 PMCID: PMC11790445 DOI: 10.3389/fcimb.2024.1529441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 12/19/2024] [Indexed: 02/06/2025] Open
Abstract
Mitochondrial genomic stability is critical to prevent various human inflammatory diseases. Bacterial infection significantly increases oxidative stress, driving mitochondrial genomic instability and initiating inflammatory human disease. Oxidative DNA base damage is predominantly repaired by base excision repair (BER) in the nucleus (nBER) as well as in the mitochondria (mtBER). In this review, we summarize the molecular mechanisms of spontaneous and H. pylori infection-associated oxidative mtDNA damage, mtDNA replication stress, and its impact on innate immune signaling. Additionally, we discuss how mutations located on mitochondria targeting sequence (MTS) of BER genes may contribute to mtDNA genome instability and innate immune signaling activation. Overall, the review summarizes evidence to understand the dynamics of mitochondria genome and the impact of mtBER in innate immune response during H. pylori-associated pathological outcomes.
Collapse
Affiliation(s)
| | - Dawit Kidane
- Department of Physiology and Biophysics, College of Medicine, Howard University, Washington, DC, United States
| |
Collapse
|
45
|
Collins RL, Talkowski ME. Diversity and consequences of structural variation in the human genome. Nat Rev Genet 2025:10.1038/s41576-024-00808-9. [PMID: 39838028 DOI: 10.1038/s41576-024-00808-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2024] [Indexed: 01/23/2025]
Abstract
The biomedical community is increasingly invested in capturing all genetic variants across human genomes, interpreting their functional consequences and translating these findings to the clinic. A crucial component of this endeavour is the discovery and characterization of structural variants (SVs), which are ubiquitous in the human population, heterogeneous in their mutational processes, key substrates for evolution and adaptation, and profound drivers of human disease. The recent emergence of new technologies and the remarkable scale of sequence-based population studies have begun to crystalize our understanding of SVs as a mutational class and their widespread influence across phenotypes. In this Review, we summarize recent discoveries and new insights into SVs in the human genome in terms of their mutational patterns, population genetics, functional consequences, and impact on human traits and disease. We conclude by outlining three frontiers to be explored by the field over the next decade.
Collapse
Affiliation(s)
- Ryan L Collins
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Michael E Talkowski
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
46
|
Carvalho JE, Burtin M, Detournay O, Amiel AR, Röttinger E. Optimized husbandry and targeted gene-editing for the cnidarian Nematostella vectensis. Development 2025; 152:dev204387. [PMID: 39776154 DOI: 10.1242/dev.204387] [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: 09/12/2024] [Accepted: 11/23/2024] [Indexed: 01/11/2025]
Abstract
Optimized laboratory conditions for research models are crucial for the success of scientific projects. This includes controlling the entire life cycle, having access to all developmental stages and maintaining stable physiological conditions. Reducing the life cycle of a research model can also enhance the access to biological material and speed up genetic tool development. Thus, we optimized the rearing conditions for the sea anemone Nematostella vectensis, a cnidarian research model, to study embryonic and post-metamorphic processes, such as regeneration. We adopted a semi-automated aquaculture system for N. vectensis and developed a dietary protocol optimized for the different life stages. Thereby, we increased spawning efficiencies, juvenile growth and survival rates, and considerably reduced the overall life cycle down to 2 months. To further improve the obtention of CRISPR-Cas9 mutants, we optimized the design of sgRNAs leading to full knockout animals in F0 polyps using a single sgRNA. Finally, we show that NHEJ-mediated transgene insertion is possible in N. vectensis. In summary, our study provides additional resources for the scientific community that uses or plans to use N. vectensis as a research model.
Collapse
Affiliation(s)
- João E Carvalho
- Université Côte d'Azur, CNRS, INSERM, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, 06107France
- Université Côte d'Azur, Federative Research Institute - Marine Resources (IFR MARRES), 28 Avenue de Valrose, Nice, 06103France
| | - Maxence Burtin
- Université Côte d'Azur, CNRS, INSERM, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, 06107France
- Université Côte d'Azur, Federative Research Institute - Marine Resources (IFR MARRES), 28 Avenue de Valrose, Nice, 06103France
| | | | - Aldine R Amiel
- Université Côte d'Azur, CNRS, INSERM, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, 06107France
- Université Côte d'Azur, Federative Research Institute - Marine Resources (IFR MARRES), 28 Avenue de Valrose, Nice, 06103France
| | - Eric Röttinger
- Université Côte d'Azur, CNRS, INSERM, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, 06107France
- Université Côte d'Azur, Federative Research Institute - Marine Resources (IFR MARRES), 28 Avenue de Valrose, Nice, 06103France
| |
Collapse
|
47
|
Medina-Suárez D, Han L, O’Reilly S, Liu J, Wei C, Brenière M, Goff N, Chen C, Modesti M, Meek K, Harrington B, Yu K. Lig3-dependent rescue of mouse viability and DNA double-strand break repair by catalytically inactive Lig4. Nucleic Acids Res 2025; 53:gkae1216. [PMID: 39673806 PMCID: PMC11754673 DOI: 10.1093/nar/gkae1216] [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: 02/12/2024] [Revised: 11/16/2024] [Accepted: 11/22/2024] [Indexed: 12/16/2024] Open
Abstract
Recent studies have revealed a structural role for DNA ligase 4 (Lig4) in the maintenance of a repair complex during non-homologous end joining (NHEJ) of DNA double-strand breaks. In cultured cell lines, catalytically inactive Lig4 can partially alleviate the severe DNA repair phenotypes observed in cells lacking Lig4. To study the structural role of Lig4 in vivo, a mouse strain harboring a point mutation to Lig4's catalytic site was generated. In contrast to the ablation of Lig4, catalytically inactive Lig4 mice are born alive. These mice display marked growth retardation and have clear deficits in lymphocyte development. We considered that the milder phenotype results from inactive Lig4 help to recruit another ligase to the repair complex. We next generated a mouse strain deficient for nuclear Lig3. Nuclear Lig3-deficient mice are moderately smaller and have elevated incidences of cerebral ventricle dilation but otherwise appear normal. Strikingly, in experiments crossing these two strains, mice lacking nuclear Lig3 and expressing inactive Lig4 were not obtained. Timed mating revealed that fetuses harboring both mutations underwent resorption, establishing an embryonic lethal genetic interaction. These data suggest that Lig3 is recruited to NHEJ complexes to facilitate end joining in the presence (but not activity) of Lig4.
Collapse
Affiliation(s)
- David Medina-Suárez
- Department of Microbiology, Genetics and Immunology, Michigan State University, 567 Wilson Rd., East Lansing, MI 48824, USA
| | - Li Han
- Department of Microbiology, Genetics and Immunology, Michigan State University, 567 Wilson Rd., East Lansing, MI 48824, USA
| | - Sandra O’Reilly
- Research Technology Support Facility, and Department of Physiology, Michigan State University, 567 Wilson Rd., East Lansing, MI 48824, USA
| | - Jiali Liu
- Department of Animal Science, Michigan State University, 3018 Interdisciplinary Science and Technology Building, 766 Service Rd, East Lansing, MI 48824, USA
| | - Chao Wei
- Department of Animal Science, Michigan State University, 3018 Interdisciplinary Science and Technology Building, 766 Service Rd, East Lansing, MI 48824, USA
| | - Manon Brenière
- Cancer Research Center of Marseille, Department of Genome Integrity, CNRS UMR7258, Inserm U1068, Institut Paoli-Calmettes, Aix Marseille Univ, 27 Boulevard Leï Roure CS30059, 13273 Marseille Cedex 09, Marseille, France
| | - Noah J Goff
- Department of Microbiology, Genetics and Immunology, Michigan State University, 567 Wilson Rd., East Lansing, MI 48824, USA
- Department of Pathobiology & Diagnostic Investigation, Michigan State University, 567 Wilson Rd., East Lansing, MI 48824, USA
| | - Chen Chen
- Department of Animal Science, Michigan State University, 3018 Interdisciplinary Science and Technology Building, 766 Service Rd, East Lansing, MI 48824, USA
| | - Mauro Modesti
- Cancer Research Center of Marseille, Department of Genome Integrity, CNRS UMR7258, Inserm U1068, Institut Paoli-Calmettes, Aix Marseille Univ, 27 Boulevard Leï Roure CS30059, 13273 Marseille Cedex 09, Marseille, France
| | - Katheryn Meek
- Department of Microbiology, Genetics and Immunology, Michigan State University, 567 Wilson Rd., East Lansing, MI 48824, USA
- Department of Pathobiology & Diagnostic Investigation, Michigan State University, 567 Wilson Rd., East Lansing, MI 48824, USA
| | - Bonnie Harrington
- Department of Pathobiology & Diagnostic Investigation, Michigan State University, 567 Wilson Rd., East Lansing, MI 48824, USA
| | - Kefei Yu
- Department of Microbiology, Genetics and Immunology, Michigan State University, 567 Wilson Rd., East Lansing, MI 48824, USA
| |
Collapse
|
48
|
Lee C, Oh JS. Novel BRCA1-PLK1-CIP2A axis orchestrates homologous recombination-mediated DNA repair to maintain chromosome integrity during oocyte meiosis. Nucleic Acids Res 2025; 53:gkae1207. [PMID: 39657788 PMCID: PMC11754672 DOI: 10.1093/nar/gkae1207] [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: 06/12/2024] [Revised: 11/18/2024] [Accepted: 11/21/2024] [Indexed: 12/12/2024] Open
Abstract
Double-strand breaks (DSBs) are a formidable threat to genome integrity, potentially leading to cancer and various genetic diseases. The prolonged lifespan of mammalian oocytes increases their susceptibility to DNA damage over time. While somatic cells suppress DSB repair during mitosis, oocytes exhibit a remarkable capacity to repair DSBs during meiotic maturation. However, the precise mechanisms underlying DSB repair in oocytes remain poorly understood. Here, we describe the pivotal role of the BRCA1-PLK1-CIP2A axis in safeguarding genomic integrity during meiotic maturation in oocytes. We found that inhibition of homologous recombination (HR) severely impaired chromosome integrity by generating chromosome fragments during meiotic maturation. Notably, HR inhibition impaired the recruitment of CIP2A to damaged chromosomes, and the depletion of CIP2A led to chromosome fragmentation following DSB induction. Moreover, BRCA1 depletion impaired chromosomal recruitment of CIP2A, but not vice versa. Importantly, the impaired chromosomal recruitment of CIP2A could be rescued by PLK1 inhibition. Consequently, our findings not only underscore the importance of the chromosomal recruitment of CIP2A in preventing chromosome fragmentation, but also demonstrate the regulatory role of the BRCA1-PLK1-CIP2A axis in this process during oocyte meiotic maturation.
Collapse
Affiliation(s)
- Crystal Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Suwon 16419, South Korea
| | - Jeong Su Oh
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Suwon 16419, South Korea
| |
Collapse
|
49
|
Scully R, Glodzik D, Menghi F, Liu ET, Zhang CZ. Mechanisms of tandem duplication in the cancer genome. DNA Repair (Amst) 2025; 145:103802. [PMID: 39742573 PMCID: PMC11843477 DOI: 10.1016/j.dnarep.2024.103802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/17/2024] [Accepted: 12/17/2024] [Indexed: 01/03/2025]
Abstract
Tandem duplications (TD) are among the most frequent type of structural variant (SV) in the cancer genome. They are characterized by a single breakpoint junction that defines the boundaries and the size of the duplicated segment. Cancer-associated TDs often increase oncogene copy number or disrupt tumor suppressor gene function, and thus have important roles in tumor evolution. TDs in cancer genomes fall into three classes, defined by the size of duplications, and are associated with distinct genetic drivers. In this review, we survey key features of cancer-related TDs and consider possible underlying mechanisms in relation to stressed DNA replication and the 3D organization of the S phase genome.
Collapse
Affiliation(s)
- Ralph Scully
- Department of Medicine, Division of Hematology-Oncology and Cancer Research Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA.
| | - Dominik Glodzik
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Francesca Menghi
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Edison T Liu
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Cheng-Zhong Zhang
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Pathology, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
50
|
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
|