1
|
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] [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
|
2
|
Pepe S, Guerra F, Russo M, Duardo RC, Capranico G. Genomic context influences translesion synthesis DNA polymerase-dependent mechanisms of micronuclei induction by G-quadruplexes. Cell Rep 2025; 44:115706. [PMID: 40349342 DOI: 10.1016/j.celrep.2025.115706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 03/11/2025] [Accepted: 04/25/2025] [Indexed: 05/14/2025] Open
Abstract
Guanine quadruplexes (G4s) are non-canonical DNA structures that can trigger micronuclei (MNi). Mechanisms of micronuclei formation by G4s are not fully understood. Here, we show that G4 stabilization can trigger cell-cycle-phase-specific mechanisms of replication fork stalling and DNA synthesis restart dependent on translesion synthesis (TLS) DNA polymerases (Pols). Fork stalling is caused by G-loops and high transcription during early S only. Moreover, while induction of micronuclei is dependent on DNA Pol η throughout S phase, primase and DNA-directed polymerase (PrimPol) is required in late S only. DNA breakage is not an immediate response to stabilized G4s but rather a consequence of persistent G4-mediated replication stress. Thus, different modes of fork stalling and restart, based on genomic context and TLS Pols, avoid immediate DNA breakage at stalled forks but at the expense of a risk of later mitotic chromosomal instability. The insights can lead to the development of more effective therapies for cancer and neurological diseases.
Collapse
Affiliation(s)
- Simona Pepe
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Federico Guerra
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Marco Russo
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy; Preclinical & Translational Research in Oncology (PRO), IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Renée C Duardo
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Giovanni Capranico
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy; Preclinical & Translational Research in Oncology (PRO), IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.
| |
Collapse
|
3
|
Zhang H, Li Y, Shah SB, Li S, Li Q, Oaks J, Lv T, Shi LZ, Wang H, Wang D, Wu X. ATM priming and end resection-coupled phosphorylation of MRE11 is important for fork protection and replication restart. Proc Natl Acad Sci U S A 2025; 122:e2422720122. [PMID: 40249789 PMCID: PMC12037065 DOI: 10.1073/pnas.2422720122] [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: 11/05/2024] [Accepted: 02/25/2025] [Indexed: 04/20/2025] Open
Abstract
The MRE11/RAD50/NBS1 (MRN) complex plays multiple roles in the maintenance of genome stability. MRN is associated with replication forks to preserve fork integrity and is also required for end resection at double-strand breaks (DSBs) to facilitate homologous recombination (HR). The critical need for proper control of the MRE11 nuclease activity is highlighted by the extensive nascent strand DNA degradation driven by MRE11 in BRCA-deficient cells, leading to genome instability and increased sensitivity to chemotherapeutics. In this study, we identified a tightly controlled mechanism, elicited by sequential phosphorylation of MRE11 by ATM and ATR to regulate MRE11 nuclease activities through its DNA binding. Specifically, at DSBs, MRE11 phosphorylation by ATM at the C-terminal S676/S678 primes it for subsequent phosphorylation by ATR, whose activation is triggered by end resection which requires the MRE11 nuclease activity. This ATR-mediated phosphorylation in turn induces MRE11 dissociation from DNA, providing a feedback mechanism to regulate the extent of end resection. At stalled replication forks, however, without ATM priming, MRN is stably associated with forks despite ATR activation. Furthermore, the ATR phosphorylation-defective MRE11 mutants are retained at single-ended DSBs formed by fork reversal upon replication stress, leading to extensive degradation of nascent DNA strands. Importantly, this end resection-coupled MRE11 phosphorylation elicits another critical layer of fork protection of nascent DNA in addition to BRCA2, ensuring proper end resection that is sufficient for replication restart at reversed forks while maintaining fork stability.
Collapse
Affiliation(s)
- Huimin Zhang
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA92037
| | - Youhang Li
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA92037
- College of Life Science, Capital Normal University, Beijing100037, China
| | - Sameer Bikram Shah
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA92037
| | - Shibo Li
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA92037
| | - Qingrong Li
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, CA92093
| | - Joshua Oaks
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA92037
| | - Tinghong Lv
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA92037
| | - Linda Z. Shi
- The Institute of Engineering in Medicine, University of California, San Diego, CA92093
| | - Hailong Wang
- College of Life Science, Capital Normal University, Beijing100037, China
| | - Dong Wang
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, CA92093
| | - Xiaohua Wu
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA92037
| |
Collapse
|
4
|
Wu W, Wu W, Xie X, Li J, Gao Y, Xie L, Zhong C, Xiao J, Cai M, Yin D, Hu K. DNMT1 is required for efficient DSB repair and maintenance of replication fork stability, and its loss reverses resistance to PARP inhibitors in cancer cells. Oncogene 2025:10.1038/s41388-025-03409-w. [PMID: 40234721 DOI: 10.1038/s41388-025-03409-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 04/02/2025] [Accepted: 04/07/2025] [Indexed: 04/17/2025]
Abstract
Cancer cells with breast cancer susceptibility gene (BRCA) mutations inevitably acquire resistance to PARP inhibitors (PARPi), and new strategies to maximize the efficacy of PARPi are urgently needed for the treatment of patients with BRCA1/2-mutant cancers. Here, we provide evidence that DNMT1 plays essential roles in DNA repair and the maintenance of replication fork stability by associating with the RPA complex and the SFPQ/NONO/FUS complex. DNMT1 depletion impairs RPA1 recruitment to stalled replication forks and inhibits DNA‒RNA hybrid (R-loop) resolution as well as the retention of RPA1 and SFPQ/NONO/FUS complexes at double-stranded DNA breaks (DSBs). Moreover, PARP1 activity is required for DNMT1 retention at DSB sites by modulating its protein stability, which is tightly and dynamically regulated by PARP1-mediated PARylation and PARG- and NUDT16-mediated dePARylation. DNMT1 PARylation further recruits the E3 ubiquitin ligase CHFR to enhance its ubiquitination and target it for proteasome-dependent degradation. Notably, DNMT1 is also required for irradiation (IR)-mediated and PARPi-induced activation of the G2 arrest checkpoint. The combination of DNMT1i with PARPi significantly attenuates PARPi-induced ATR-Chk1 signaling and enhances the degradation of the stalled replication fork mediated by PARPi, resulting in increased chromosomal aberrations and cell death in BRCA-proficient and BRCA-deficient cancer cells. Therefore, our findings provide novel insights into the mechanism by which DNMT1 inhibitors (DNMT1i) reverse PARPi resistance and indicate that targeting the PARP-DNMT1 pathway is a promising strategy for cancer therapy.
Collapse
Affiliation(s)
- Wenjing Wu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Department of Breast Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Weijun Wu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiaojuan Xie
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jing Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Department of Gynecologic Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yuan Gao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Limin Xie
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Caixia Zhong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jianhong Xiao
- Department of Hematology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Manbo Cai
- Department of Oncology Radiotherapy, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China.
| | - Dong Yin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Kaishun Hu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
5
|
Nikulenkov F, Carbain B, Biswas R, Havel S, Prochazkova J, Sisakova A, Zacpalova M, Chavdarova M, Marini V, Vsiansky V, Weisova V, Slavikova K, Biradar D, Khirsariya P, Vitek M, Sedlak D, Bartunek P, Daniel L, Brezovsky J, Damborsky J, Paruch K, Krejci L. Discovery of new inhibitors of nuclease MRE11. Eur J Med Chem 2025; 285:117226. [PMID: 39793442 DOI: 10.1016/j.ejmech.2024.117226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 12/10/2024] [Accepted: 12/27/2024] [Indexed: 01/13/2025]
Abstract
MRE11 nuclease is a central player in signaling and processing DNA damage, and in resolving stalled replication forks. Here, we describe the identification and characterization of new MRE11 inhibitors MU147 and MU1409. Both compounds inhibit MRE11 nuclease more specifically and effectively than the relatively weak state-of-the-art inhibitor mirin. They also abrogate double-strand break repair mechanisms that rely on MRE11 nuclease activity, without impairing ATM activation. Inhibition of MRE11 also impairs nascent strand degradation of stalled replication forks and selectively affects BRCA2-deficient cells. Herein, we illustrate that our newly discovered compounds MU147 and MU1409 can be used as chemical probes to further explore the biological role of MRE11 and support the potential clinical relevance of pharmacological inhibition of this nuclease.
Collapse
Affiliation(s)
- Fedor Nikulenkov
- Department of Biology, Faculty of Medicine, Masaryk University, 62500, Brno, Czech Republic
| | - Benoit Carbain
- Department of Biology, Faculty of Medicine, Masaryk University, 62500, Brno, Czech Republic; Department of Chemistry, Faculty of Science, Masaryk University, 62500, Brno, Czech Republic
| | - Raktim Biswas
- Department of Biology, Faculty of Medicine, Masaryk University, 62500, Brno, Czech Republic
| | - Stepan Havel
- International Clinical Research Center, St. Anne's University Hospital in Brno, 62500, Brno, Czech Republic; Department of Chemistry, Faculty of Science, Masaryk University, 62500, Brno, Czech Republic
| | - Jana Prochazkova
- Department of Biology, Faculty of Medicine, Masaryk University, 62500, Brno, Czech Republic
| | - Alexandra Sisakova
- Department of Biology, Faculty of Medicine, Masaryk University, 62500, Brno, Czech Republic
| | - Magdalena Zacpalova
- Department of Biology, Faculty of Medicine, Masaryk University, 62500, Brno, Czech Republic
| | - Melita Chavdarova
- Department of Biology, Faculty of Medicine, Masaryk University, 62500, Brno, Czech Republic
| | - Victoria Marini
- Department of Biology, Faculty of Medicine, Masaryk University, 62500, Brno, Czech Republic
| | - Vit Vsiansky
- Department of Biology, Faculty of Medicine, Masaryk University, 62500, Brno, Czech Republic
| | - Veronika Weisova
- Department of Biology, Faculty of Medicine, Masaryk University, 62500, Brno, Czech Republic
| | - Kristina Slavikova
- Department of Biology, Faculty of Medicine, Masaryk University, 62500, Brno, Czech Republic
| | - Dhanraj Biradar
- International Clinical Research Center, St. Anne's University Hospital in Brno, 62500, Brno, Czech Republic; Department of Chemistry, Faculty of Science, Masaryk University, 62500, Brno, Czech Republic
| | - Prashant Khirsariya
- International Clinical Research Center, St. Anne's University Hospital in Brno, 62500, Brno, Czech Republic; Department of Chemistry, Faculty of Science, Masaryk University, 62500, Brno, Czech Republic
| | - Marco Vitek
- Department of Chemistry, Faculty of Science, Masaryk University, 62500, Brno, Czech Republic
| | - David Sedlak
- CZ-OPENSCREEN, Institute of Molecular Genetics of the ASCR, v.v.i., Prague 4, Czech Republic
| | - Petr Bartunek
- CZ-OPENSCREEN, Institute of Molecular Genetics of the ASCR, v.v.i., Prague 4, Czech Republic
| | - Lukas Daniel
- International Clinical Research Center, St. Anne's University Hospital in Brno, 62500, Brno, Czech Republic; Loschmidt Laboratories, Department of Experimental Biology and RECETOX, Faculty of Science, Masaryk University, 62500, Brno, Czech Republic
| | - Jan Brezovsky
- International Clinical Research Center, St. Anne's University Hospital in Brno, 62500, Brno, Czech Republic; Loschmidt Laboratories, Department of Experimental Biology and RECETOX, Faculty of Science, Masaryk University, 62500, Brno, Czech Republic
| | - Jiri Damborsky
- International Clinical Research Center, St. Anne's University Hospital in Brno, 62500, Brno, Czech Republic; Loschmidt Laboratories, Department of Experimental Biology and RECETOX, Faculty of Science, Masaryk University, 62500, Brno, Czech Republic
| | - Kamil Paruch
- International Clinical Research Center, St. Anne's University Hospital in Brno, 62500, Brno, Czech Republic; Department of Chemistry, Faculty of Science, Masaryk University, 62500, Brno, Czech Republic.
| | - Lumir Krejci
- Department of Biology, Faculty of Medicine, Masaryk University, 62500, Brno, Czech Republic; NCBR, Faculty of Science, Masaryk University, 62500, Brno, Czech Republic.
| |
Collapse
|
6
|
Lu M, Wu J, Gao Q, Jin R, An C, Ma T. To cleave or not and how? The DNA exonucleases and endonucleases in immunity. Genes Dis 2025; 12:101219. [PMID: 39759116 PMCID: PMC11697192 DOI: 10.1016/j.gendis.2024.101219] [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: 07/19/2023] [Revised: 11/02/2023] [Accepted: 12/29/2023] [Indexed: 01/07/2025] Open
Abstract
DNA exonucleases and endonucleases are key executors of the genome during many physiological processes. They generate double-stranded DNA by cleaving damaged endogenous or exogenous DNA, triggering the activation of the innate immune pathways such as cGAS-STING-IFN, and enabling the body to produce anti-viral or anti-tumor immune responses. This is of great significance for maintaining the stability of the genome and improving the therapeutic efficacy of tumors. In addition, genomic instability caused by exonuclease mutations contributes to the development of various autoimmune diseases. This review summarizes the DNA exonucleases and endonucleases which have critical functions in immunity and associated diseases.
Collapse
Affiliation(s)
- Mingjun Lu
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| | - Jinghong Wu
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| | - Qing Gao
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| | - Renjing Jin
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| | - Changming An
- Department of Head and Neck Surgery, Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China
| | - Teng Ma
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| |
Collapse
|
7
|
Buckley-Benbow L, Agnarelli A, Bellelli R. 'Where is my gap': mechanisms underpinning PARP inhibitor sensitivity in cancer. Biochem Soc Trans 2025; 53:BST20241633. [PMID: 39927794 DOI: 10.1042/bst20241633] [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: 12/08/2024] [Revised: 01/20/2025] [Accepted: 01/23/2025] [Indexed: 02/11/2025]
Abstract
The introduction of poly-ADP ribose polymerase (PARP) inhibitors (PARPi) has completely changed the treatment landscape of breast cancer susceptibility 1-2 (BRCA1-BRCA2)-mutant cancers and generated a new avenue of research in the fields of DNA damage response and cancer therapy. Despite this, primary and secondary resistances to PARPi have become a challenge in the clinic, and novel therapies are urgently needed to address this problem. After two decades of research, a unifying model explaining sensitivity of cancer cells to PARPi is still missing. Here, we review the current knowledge in the field and the increasing evidence pointing to a crucial role for replicative gaps in mediating sensitization to PARPi in BRCA-mutant and 'wild-type' cancer cells. Finally, we discuss the challenges to be addressed to further improve the utilization of PARPi and tackle the emergence of resistance in the clinical context.
Collapse
Affiliation(s)
- Lauryn Buckley-Benbow
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, Barbican, London EC1M 6BQ, U.K
| | - Alessandro Agnarelli
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, Barbican, London EC1M 6BQ, U.K
| | - Roberto Bellelli
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, Barbican, London EC1M 6BQ, U.K
| |
Collapse
|
8
|
Turner JL, Moore G, McCraw TJ, Mason JM. Overexpression of the NEK8 kinase inhibits homologous recombination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.07.637121. [PMID: 39975112 PMCID: PMC11839122 DOI: 10.1101/2025.02.07.637121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Homologous recombination maintains genome stability by repairing double strand breaks and protecting replication fork stability. Defects in homologous recombination results in cancer predisposition but can be exploited due to increased sensitivity to certain chemotherapeutics such as PARP inhibitors. The NEK8 kinase has roles in the replication response and homologous recombination. NEK8 is overexpressed in breast cancer, but the impact of NEK8 overexpression on homologous recombination has not been determined. Here, we demonstrate NEK8 overexpression inhibits RAD51 focus formation resulting in a defect in homologous recombination and degradation of stalled replication forks. Importantly, NEK8 overexpression sensitizes cells to the PARP inhibitor, Olaparib. Together, our results suggest NEK8 overexpressing tumors may be recombination-deficient and respond to chemotherapeutics that target defects in recombination such as Olaparib.
Collapse
Affiliation(s)
| | - Georgia Moore
- Department of Genetics and Biochemistry, Clemson University
| | | | | |
Collapse
|
9
|
Song L, Xie H, Fan H, Zhang Y, Cheng Z, Chen J, Guo Y, Zhang S, Zhou X, Li Z, Liao H, Han J, Huang J, Zhou J, Fang D, Liu T. Dynamic control of RNA-DNA hybrid formation orchestrates DNA2 activation at stalled forks by RNAPII and DDX39A. Mol Cell 2025; 85:506-522.e7. [PMID: 39706186 DOI: 10.1016/j.molcel.2024.11.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 10/17/2024] [Accepted: 11/25/2024] [Indexed: 12/23/2024]
Abstract
Stalled replication forks, susceptible to nucleolytic threats, necessitate protective mechanisms involving pivotal factors such as the tumor suppressors BRCA1 and BRCA2. Here, we demonstrate that, upon replication stress, RNA polymerase II (RNAPII) is recruited to stalled forks, actively promoting the transient formation of RNA-DNA hybrids. These hybrids act as safeguards, preventing premature engagement by the DNA2 nuclease and uncontrolled DNA2-mediated degradation of nascent DNA. Furthermore, we provide evidence that DExD box polypeptide 39A (DDX39A), serving as an RNA-DNA resolver, unwinds these structures and facilitates regulated DNA2 access to stalled forks. This orchestrated process enables controlled DNA2-dependent stalled fork processing and restart. Finally, we reveal that loss of DDX39A enhances stalled fork protection in BRCA1/2-deficient cells, consequently conferring chemoresistance. Our results suggest that the dynamic regulation of RNA-DNA hybrid formation at stalled forks by RNAPII and DDX39A precisely governs the timing of DNA2 activation, contributing to stalled fork protection, processing, and restart, ultimately promoting genome stability.
Collapse
Affiliation(s)
- Lizhi Song
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310058 Hangzhou, China; Department of Cell Biology, Zhejiang University School of Medicine, 310058 Hangzhou, China; MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, 310058 Hangzhou, China
| | - Haihua Xie
- Department of Cell Biology, Zhejiang University School of Medicine, 310058 Hangzhou, China
| | - Haonan Fan
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, 310058 Hangzhou, China
| | - Yanjun Zhang
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, 322000 Yiwu, China
| | - Zixiu Cheng
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, 310058 Hangzhou, China
| | - Junliang Chen
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, 321000 Shaoxing, China
| | - Yuzun Guo
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, 321000 Shaoxing, China
| | - Shudi Zhang
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, 310058 Hangzhou, China
| | - Xinyu Zhou
- Department of Cell Biology, Zhejiang University School of Medicine, 310058 Hangzhou, China
| | - Zhaoshuang Li
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, 310058 Hangzhou, China
| | - Haoxiang Liao
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, 310058 Hangzhou, China
| | - Jinhua Han
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, 310030 Hangzhou, China
| | - Jun Huang
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310058 Hangzhou, China; MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, 310058 Hangzhou, China; Center for Life Sciences, Shaoxing Institute, Zhejiang University, 321000 Shaoxing, China
| | - Jianwei Zhou
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310058 Hangzhou, China
| | - Dong Fang
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, 310058 Hangzhou, China
| | - Ting Liu
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310058 Hangzhou, China; Department of Cell Biology, Zhejiang University School of Medicine, 310058 Hangzhou, China.
| |
Collapse
|
10
|
Yano K, Kato M, Endo S, Igarashi T, Wada R, Kohno T, Zimmermann A, Dahmen H, Zenke FT, Shiotani B. PARP inhibition-associated heterochromatin confers increased DNA replication stress and vulnerability to ATR inhibition in SMARCA4-deficient cells. Cell Death Discov 2025; 11:31. [PMID: 39875375 PMCID: PMC11775187 DOI: 10.1038/s41420-025-02306-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/18/2024] [Accepted: 01/16/2025] [Indexed: 01/30/2025] Open
Abstract
DNA replication stress (RS), a prevalent feature of various malignancies, arises from both genetic mutations and genotoxic exposure. Elevated RS levels increase the vulnerability of cancer cells to ataxia telangiectasia and Rad3-related kinase inhibitors (ATRis). Here, we screened for DNA damage response inhibitors that enhance ATRi-induced cytotoxicity using SWI/SNF complex-deficient cells and identified a potent synergy between ATRi and poly(ADP-ribose) polymerase inhibitor (PARPi), particularly in SMARCA4-deficient cells. PARP inhibition triggers chromatin changes, namely elevated histone H3 at lysine 9 di-methylation (H3K9me2), a hallmark of facultative heterochromatin, increasing dependence on ATR activity for replication fork progression and cell survival. Interestingly, SMARCA4 deficient cells, intrinsically vulnerable to replication stress, exhibited exacerbated DNA damage upon combined ATRi and PARPi treatment in a Mre11- and Mus81-mediated manner. In vivo, combined treatment with intermittent ATRi and continuous PARPi showed greater inhibition of tumor growth than ATRi alone in SMARCA4-deficient lung adenocarcinoma xenograft models. These findings demonstrate that PARPi-induced heterochromatin amplifies RS and ATRi susceptibility, providing a potential rationale for therapeutic strategies targeting SMARCA4-deficient tumors.
Collapse
Affiliation(s)
- Kimiyoshi Yano
- Laboratory of Genome Stress Signaling, National Cancer Center Research Institute, Chuo-ku, Tokyo, 104-0045, Japan
- Department of Cellular and Molecular Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima-city, Hiroshima, 734-8553, Japan
| | - Megumi Kato
- Laboratory of Genome Stress Signaling, National Cancer Center Research Institute, Chuo-ku, Tokyo, 104-0045, Japan
- Department of NCC Cancer Science, Health Sciences and Biomedical Engineering, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Syoju Endo
- Laboratory of Genome Stress Signaling, National Cancer Center Research Institute, Chuo-ku, Tokyo, 104-0045, Japan
- Department of NCC Cancer Science, Division of Integrative Molecular Biomedicine, Biomedical Sciences and Engineering, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Taichi Igarashi
- Laboratory of Genome Stress Signaling, National Cancer Center Research Institute, Chuo-ku, Tokyo, 104-0045, Japan
- Department of Biosciences, School of Science, Kitasato University, Minami-ku, Sagamihara-city, Kanagawa, 252-0373, Japan
| | - Ryoga Wada
- Laboratory of Genome Stress Signaling, National Cancer Center Research Institute, Chuo-ku, Tokyo, 104-0045, Japan
- Department of Pharmacology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Takashi Kohno
- Division of Genome Biology, National Cancer Center Research Institute, Chuo-ku, Tokyo, 104-0045, Japan
| | - Astrid Zimmermann
- Research Unit Oncology, The healthcare business of Merck KGaA, Frankfurter Str. 250, 64293, Darmstadt, Germany
| | - Heike Dahmen
- Research Unit Oncology, The healthcare business of Merck KGaA, Frankfurter Str. 250, 64293, Darmstadt, Germany
| | - Frank T Zenke
- Research Unit Oncology, The healthcare business of Merck KGaA, Frankfurter Str. 250, 64293, Darmstadt, Germany
| | - Bunsyo Shiotani
- Laboratory of Genome Stress Signaling, National Cancer Center Research Institute, Chuo-ku, Tokyo, 104-0045, Japan.
- Department of Genome Stress Signaling, Institute of Medical Science, Tokyo Medical University, Shinjuku-ku, Tokyo, 160-0023, Japan.
| |
Collapse
|
11
|
Xie H, Song L, Mao G, Han J, Pu J, Wu Z, Chen J, Zhou J, Huang J, Fang D, Liu T. Synergistic protection of nascent DNA at stalled forks by MSANTD4 and BRCA1/2-RAD51. Nat Chem Biol 2025:10.1038/s41589-024-01833-9. [PMID: 39809895 DOI: 10.1038/s41589-024-01833-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 12/19/2024] [Indexed: 01/16/2025]
Abstract
The regressed arms of reversed replication forks exhibit structural similarities to one-ended double-stranded breaks and need to be protected against uncontrolled nucleolytic degradation. Here, we identify MSANTD4 (Myb/SANT-like DNA-binding domain-containing protein 4), a functionally uncharacterized protein that uniquely counters the replication protein A (RPA)-Bloom (BLM)/Werner syndrome helicase (WRN)-DNA replication helicase/nuclease 2 (DNA2) complex to safeguard reversed replication forks from detrimental degradation, independently of the breast cancer susceptibility proteins (BRCA1/2)-DNA repair protein RAD51 pathway. MSANTD4 specifically interacts with the junctions between single-stranded DNA (ssDNA) and double-stranded DNA (dsDNA) in DNA substrates harboring a 3' overhang, which resemble the structural features of regressed arms processed by WRN-DNA2. This DNA-binding capability allows MSANTD4 to accumulate at reversed forks, strategically antagonizing the RPA-BLM/WRN-DNA2 complex by impeding its access to the ssDNA-dsDNA junction of the regressed arms. Loss of MSANTD4 exacerbates genome instability induced by replication stress in BRCA1/2-deficient cells. Our findings unveil a collaborative defense mechanism orchestrated by MSANTD4 and BRCA1/2-RAD51, effectively counteracting nucleolytic attacks on the regressed arms and synergistically preserving the integrity of reversed forks.
Collapse
Affiliation(s)
- Haihua Xie
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lizhi Song
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, China
| | - Genxiang Mao
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinhua Han
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiali Pu
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhibing Wu
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Chen
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianwei Zhou
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Huang
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China.
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, China.
| | - Dong Fang
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China.
| | - Ting Liu
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China.
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
12
|
Chiou LF, Jayaprakash D, Droby GN, Zhang X, Yang Y, Mills CA, Webb TS, Barker NK, Wu D, Herring LE, Bowser J, Vaziri C. The RING Finger E3 Ligase RNF25 Protects DNA Replication Forks Independently of its Canonical Roles in Ubiquitin Signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.09.632184. [PMID: 39829812 PMCID: PMC11741350 DOI: 10.1101/2025.01.09.632184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The DNA damage response (DDR) mechanisms that allow cells to tolerate DNA replication stress are critically important for genome stability and cell viability. Using an unbiased genetic screen we identify a role for the RING finger E3 ubiquitin ligase RNF25 in promoting DNA replication stress tolerance. In response to DNA replication stress, RNF25-deficient cells generate aberrantly high levels of single-stranded DNA (ssDNA), accumulate in S-phase and show reduced mitotic entry. Using single-molecule DNA fiber analysis, we show that RNF25 protects reversed DNA replication forks generated by the fork remodeler HLTF from nucleolytic degradation by MRE11 and CtIP. Mechanistically, RNF25 interacts with the replication fork protection factor REV7 and recruits REV7 to nascent DNA after replication stress. The role of RNF25 in protecting replication forks is fully separable from its canonical functions in ubiquitin conjugation. This work reveals the RNF25-REV7 signaling axis as an important protective mechanism in cells experiencing replication stress.
Collapse
Affiliation(s)
- Lilly F. Chiou
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Deepika Jayaprakash
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Oral and Craniofacial Biomedicine Program, Adams School of Dentistry, University of North Carolina at Chapel Hill, NC 27599, USA
- Present address: Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37237, USA
| | - Gaith N. Droby
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Xingyuan Zhang
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Present address: Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - Yang Yang
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Present address: In Vivo Neurobiology Group, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709, USA
| | - C. Allie Mills
- UNC Metabolomics & Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Thomas S. Webb
- UNC Metabolomics & Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Natalie K. Barker
- UNC Metabolomics & Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Di Wu
- Division of Oral and Craniofacial Health Science, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Laura E. Herring
- UNC Metabolomics & Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jessica Bowser
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Cyrus Vaziri
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lead Contact
| |
Collapse
|
13
|
Vega-Letter AM, García-Guerrero C, Yantén-Fuentes L, Pradenas C, Herrera-Luna Y, Lara-Barba E, Bustamante-Barrientos FA, Rojas M, Araya MJ, Jeraldo N, Aros C, Troncoso F, Poblete D, Court A, Ortloff A, Barraza J, Velarde F, Farkas C, Carril C, Luque-Campos N, Almarza G, Barahona M, Matas J, Cereceda L, Lorca R, Toledo J, Oyarce K, Vernal R, Caicedo A, Del Campo A, Hidalgo Y, Elizondo-Vega R, Djouad F, Khoury M, Figueroa FE, Luz-Crawford P. Safety and efficacy of mesenchymal stromal cells mitochondria transplantation as a cell-free therapy for osteoarthritis. J Transl Med 2025; 23:26. [PMID: 39773289 PMCID: PMC11706173 DOI: 10.1186/s12967-024-05945-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
OBJECTIVE The inflammatory responses from synovial fibroblasts and macrophages and the mitochondrial dysfunction in chondrocytes lead to oxidative stress, disrupt extracellular matrix (ECM) homeostasis, and accelerate the deterioration process of articular cartilage in osteoarthritis (OA). In recent years, it has been proposed that mesenchymal stromal cells (MSC) transfer their functional mitochondria to damaged cells in response to cellular stress, becoming one of the mechanisms underpinning their therapeutic effects. Therefore, we hypothesize that a novel cell-free treatment for OA could involve direct mitochondria transplantation, restoring both cellular and mitochondrial homeostasis. METHODS Mitochondria were isolated from Umbilical Cord (UC)-MSC (Mito-MSC) and characterized based on their morphology, phenotype, functions, and their ability to be internalized by different articular cells. Furthermore, the transcriptional changes following mitochondrial uptake by chondrocytes were evaluated using an Affymetrix analysis, Lastly, the dose dependence therapeutic efficacy, biodistribution and immunogenicity of Mito-MSC were assessed in vivo, through an intra-articular injection in male C57BL6 mice in a collagenase-induced OA (CIOA) model. RESULTS Our findings demonstrate the functional integrity of Mito-MSC and their ability to be efficiently transferred into chondrocytes, synovial macrophages, and synovial fibroblasts. Moreover, the transcriptomic analysis showed the upregulation of genes involved in stress such as DNA reparative machinery and inflammatory antiviral responses. Finally, Mito-MSC transplantation yielded significant reductions in joint mineralization, a hallmark of OA progression, as well as improvements in OA-related histological signs, with the lower dose exhibiting better therapeutic efficacy. Furthermore, Mito-MSC was detected within the knee joint for up to 24 h post-injection without eliciting an inflammatory response in CIOA mice. CONCLUSION Collectively, our results reveal that mitochondria derived from MSC are transferred to key articular cells and are retained in the joint without generating an inflammatory immune response mitigating articular cartilage degradation in OA, probably through a restorative effect triggered by the stress antiviral response within OA chondrocytes.
Collapse
Affiliation(s)
- Ana Maria Vega-Letter
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Cynthia García-Guerrero
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Liliana Yantén-Fuentes
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Carolina Pradenas
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Yeimi Herrera-Luna
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Eliana Lara-Barba
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Felipe A Bustamante-Barrientos
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Masyelly Rojas
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - María Jesús Araya
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Nicole Jeraldo
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
- Red de Equipamiento Científico Avanzado-REDECA, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Constanza Aros
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Francisca Troncoso
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Daniela Poblete
- Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Angela Court
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
- Cells for Cells and Regenero the Chilean Consortium for Regenerative Medicine, Santiago, Chile
| | - Alexander Ortloff
- Departamento de Ciencias Veterinarias y Salud Pública, Facultad de Recursos Naturales, Universidad Católica de Temuco, Temuco, Chile
| | - Jose Barraza
- Departamento de Ciencias Veterinarias y Salud Pública, Facultad de Recursos Naturales, Universidad Católica de Temuco, Temuco, Chile
| | - Francesca Velarde
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Carlos Farkas
- Laboratorio de Investigación en Ciencias Biomédicas, Departamento de Ciencias Básicas y Morfología, Facultad de Medicina, Universidad Católica de la Santísima Concepción, Concepción, Chile
| | - Claudio Carril
- Laboratorio de Neuroinmunología, Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Concepción, Chile
| | - Noymar Luque-Campos
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Gonzalo Almarza
- Laboratorio de Fisiología y Bioenergetica Celular, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Maximiliano Barahona
- Departamento de Ortopedia y Traumatología, Hospital Clinico Universidad de Chile, Independencia, Chile
| | - Jose Matas
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
| | - Lucas Cereceda
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Rocío Lorca
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Jorge Toledo
- Red de Equipamiento Científico Avanzado-REDECA, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Karina Oyarce
- Laboratorio de Neuroinmunología, Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Concepción, Chile
| | - Rolando Vernal
- Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Andrés Caicedo
- Universidad San Francisco de Quito USFQ, Colegio de Ciencias de la Salud e Instituto de Investigaciones en Biomedicina iBioMed, Escuela de Medicina, Quito, Ecuador-Mito-Act Research Consortium, Quito, Ecuador
| | - Andrea Del Campo
- Laboratorio de Fisiología y Bioenergetica Celular, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Yessia Hidalgo
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
- Cells for Cells and Regenero the Chilean Consortium for Regenerative Medicine, Santiago, Chile
| | - Roberto Elizondo-Vega
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Farida Djouad
- IRMB, Université de Montpellier, INSERM, Montpellier, France
- CHU Montpellier, Montpellier, France
| | - Maroun Khoury
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile.
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile.
- Cells for Cells and Regenero the Chilean Consortium for Regenerative Medicine, Santiago, Chile.
| | - Fernando E Figueroa
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile.
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile.
- Cells for Cells and Regenero the Chilean Consortium for Regenerative Medicine, Santiago, Chile.
| | - Patricia Luz-Crawford
- Center of Interventional Medicine for Precision and Advanced Cellular Therapy, IMPACT, Santiago, Chile.
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile.
| |
Collapse
|
14
|
Raina VB, Greene EC. Single-Molecule Visualization of BLM-DNA2-Mediated DNA End Resection Using DNA Curtains. Methods Mol Biol 2025; 2881:195-211. [PMID: 39704945 PMCID: PMC12013495 DOI: 10.1007/978-1-0716-4280-1_10] [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] [Indexed: 12/21/2024]
Abstract
Homologous recombination (HR) is the principal pathway undertaken by a cell for the error-free repair of DNA double-strand breaks that are frequently encountered by the cell. HR can be initiated at the sites of DNA double-strand breaks by generating long stretches of single-stranded 3' DNA overhang through a process called DNA end resection. In one DNA end resection pathway, this is achieved via the concerted effort of specialized machinery involving the RecQ family helicase BLM, the helicase/endonuclease DNA2, and a single-strand DNA binding protein complex RPA. BLM unwinds the DNA at the sites of DNA damage. The DNA unwound by BLM is cleaved in a 5' to 3' direction by DNA2 leaving behind a long single-stranded 3' DNA tail which is rapidly coated with RPA. This long single-stranded DNA provides the loading platform for recombinase proteins to form nucleoprotein filaments which initiate the homology search to undergo HR-based repair. Most of the insights into these processes have been gained using ensemble biochemical methods. However, these approaches have proven to be challenging for dissecting complex molecular mechanisms, especially because of the dynamic nature of these processes. Experiments involving single-molecule techniques have enabled us to counter these issues by allowing us to gather information on intermediates that are heterogeneous and transient in nature. We have developed a single-molecule technique called DNA curtains where we can study hundreds of protein-nucleic acid interaction events in real time. Here, we describe a method to prepare a single-tethered double-stranded DNA curtain to visualize, in combination with total internal reflection microscopy (TIRFM), the BLM- and DNA2-mediated DNA end resection in real time.
Collapse
Affiliation(s)
- Vivek B Raina
- Department of Biochemistry & Molecular Biophysics, Columbia University, New York, NY, USA
| | - Eric C Greene
- Department of Biochemistry & Molecular Biophysics, Columbia University, New York, NY, USA.
| |
Collapse
|
15
|
Duzanic FD, Penengo L. The interferon response at the intersection of genome integrity and innate immunity. DNA Repair (Amst) 2025; 145:103786. [PMID: 39577202 DOI: 10.1016/j.dnarep.2024.103786] [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: 07/15/2024] [Revised: 10/24/2024] [Accepted: 11/08/2024] [Indexed: 11/24/2024]
Abstract
In recent years, numerous reports indicated that, besides pathogen infections, DNA replication stress and defective DNA repair can trigger the innate immune response by introducing a state of viral mimicry, due to cytosolic accumulation of the self-nucleic acid species, which culminates in the activation of type I interferon (IFN) pathway. In turn, IFN upregulates a variety of factors mutually implicated in immune- and genome-related mechanisms, shedding light on the unprecedented causality between genome stability and innate immunity. Intriguingly, in addition to being induced by replication stress, IFN-regulated factors can also promote it, pinpointing IFN signaling as both a consequence and a cause of replication stress. Here, we provide an overview of the factors and molecular mechanisms implicated in the evolutionary conserved crosstalk between genome maintenance and innate immunity, highlighting the role of the IFN-stimulated gene 15 (ISG15), which appears to be at the hub of this intersection. Moreover, we discuss the potential significance and clinical implications of the immune-mediated modulation of DNA replication and repair upon pathogen infection and in human diseases such as cancer and autoinflammatory syndromes. Finally, we discuss the relevant open questions and future directions.
Collapse
Affiliation(s)
- Filip D Duzanic
- University of Zurich, Institute of Molecular Cancer Research, Zurich 8057, Switzerland
| | - Lorenza Penengo
- University of Zurich, Institute of Molecular Cancer Research, Zurich 8057, Switzerland.
| |
Collapse
|
16
|
Mashayekhi F, Ganje C, Caron MC, Heyza JR, Gao Y, Zeinali E, Fanta M, Li L, Ali J, Mersaoui SY, Schmidt JC, Godbout R, Masson JY, Weinfeld M, Ismail IH. PNKP safeguards stalled replication forks from nuclease-dependent degradation during replication stress. Cell Rep 2024; 43:115066. [PMID: 39671289 DOI: 10.1016/j.celrep.2024.115066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 09/04/2024] [Accepted: 11/22/2024] [Indexed: 12/15/2024] Open
Abstract
Uncontrolled degradation and collapse of stalled replication forks (RFs) are primary sources of genomic instability, yet the molecular mechanisms for protecting forks from degradation/collapse remain to be fully elaborated. Here, we show that polynucleotide kinase-phosphatase (PNKP) localizes at stalled forks and protects stalled forks from excessive degradation. The loss of PNKP results in nucleolytic degradation of nascent DNA at stalled RFs. This mechanism is different from the BRCA2-dependent fork protection pathway, which protects stalled forks from excessive MRE11-dependent nucleolytic degradation. Our research shows that hydroxyurea treatment leads to increased misincorporation of ribonucleotides in DNA, which in turn traps TOP1 on stalled RFs. We have also found that reducing the levels of TOP1 or TDP1 in cells can reverse the degradation of nascent DNA observed in PNKP-deficient cells. In summary, our data suggest that PNKP plays a role in maintaining the stability of stalled RFs.
Collapse
Affiliation(s)
- Fatemeh Mashayekhi
- Department of Oncology, Faculty of Medicine & Dentistry, University of Alberta, 11560 University Avenue, Edmonton, AB T6G 1Z2, Canada
| | - Cassandra Ganje
- Department of Oncology, Faculty of Medicine & Dentistry, University of Alberta, 11560 University Avenue, Edmonton, AB T6G 1Z2, Canada
| | - Marie-Christine Caron
- Department of Molecular Biology, Medical Biochemistry and Pathology, Laval University, 9 McMahon, Québec City, QC G1R 3S3, Canada
| | - Joshua R Heyza
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Yuandi Gao
- Department of Molecular Biology, Medical Biochemistry and Pathology, Laval University, 9 McMahon, Québec City, QC G1R 3S3, Canada
| | - Elham Zeinali
- Department of Oncology, Faculty of Medicine & Dentistry, University of Alberta, 11560 University Avenue, Edmonton, AB T6G 1Z2, Canada
| | - Mesfin Fanta
- Department of Oncology, Faculty of Medicine & Dentistry, University of Alberta, 11560 University Avenue, Edmonton, AB T6G 1Z2, Canada
| | - Lei Li
- Department of Oncology, Faculty of Medicine & Dentistry, University of Alberta, 11560 University Avenue, Edmonton, AB T6G 1Z2, Canada
| | - Jana Ali
- Department of Oncology, Faculty of Medicine & Dentistry, University of Alberta, 11560 University Avenue, Edmonton, AB T6G 1Z2, Canada
| | - Sofiane Yacine Mersaoui
- Department of Molecular Biology, Medical Biochemistry and Pathology, Laval University, 9 McMahon, Québec City, QC G1R 3S3, Canada
| | - Jens C Schmidt
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Roseline Godbout
- Department of Oncology, Faculty of Medicine & Dentistry, University of Alberta, 11560 University Avenue, Edmonton, AB T6G 1Z2, Canada
| | - Jean-Yves Masson
- Department of Molecular Biology, Medical Biochemistry and Pathology, Laval University, 9 McMahon, Québec City, QC G1R 3S3, Canada
| | - Michael Weinfeld
- Department of Oncology, Faculty of Medicine & Dentistry, University of Alberta, 11560 University Avenue, Edmonton, AB T6G 1Z2, Canada
| | - Ismail Hassan Ismail
- Department of Oncology, Faculty of Medicine & Dentistry, University of Alberta, 11560 University Avenue, Edmonton, AB T6G 1Z2, Canada; Biophysics Department, Faculty of Science, Cairo University, Giza 12613, Egypt.
| |
Collapse
|
17
|
Ghorai A, Singh B, Dutt S. Biphasic DNA damage and non-canonical replication stress response govern radiation-induced senescence in glioblastoma. J Cell Sci 2024; 137:jcs261844. [PMID: 39568404 DOI: 10.1242/jcs.261844] [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: 11/28/2023] [Accepted: 10/28/2024] [Indexed: 11/22/2024] Open
Abstract
Therapy-induced senescence (TIS) in glioblastoma (GBM) residual disease and escape from TIS account for resistance and recurrence, but the mechanism of TIS manifestation remains obscure. Here, we demonstrate that replication stress (RS) is critical for the induction of TIS in residual cells by employing an in vitro GBM therapy-resistance cellular model. Interestingly, we found a 'biphasic' mode of DNA damage after radiation treatment and reveal that the second phase of DNA damage arises majorly in the S phase of residual cells due to RS. Mechanistically, we show that persistent phosphorylated ATR is a safeguard for radiation resilience, whereas the other canonical RS molecules remain unaltered during the second phase of DNA damage. Importantly, RS preceded the induction of senescence, and ATR inhibition resulted in TIS reduction, leading to apoptosis. Moreover, ATR inhibition sensitized PARP-1 inhibitor-induced enhanced TIS-mediated resistance, leading to cell death. Our study demonstrates the crucial role of RS in TIS induction and maintenance in GBM residual cells, and targeting ATR alone or in combination with a PARP-1 inhibitor will be an effective strategy to eliminate TIS for better treatment outcomes.
Collapse
Affiliation(s)
- Atanu Ghorai
- Shilpee Dutt Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai 410210, India
| | - Bhawna Singh
- Shilpee Dutt Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai 410210, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400085, India
| | - Shilpee Dutt
- Shilpee Dutt Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai 410210, India
- Shilpee Dutt Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi 110067, India
| |
Collapse
|
18
|
Pale LM, Khatib JB, Nusawardhana A, Straka J, Nicolae CM, Moldovan GL. CRISPR knockout genome-wide screens identify the HELQ-RAD52 axis in regulating the repair of cisplatin-induced single-stranded DNA gaps. Nucleic Acids Res 2024; 52:13832-13848. [PMID: 39530221 DOI: 10.1093/nar/gkae998] [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: 04/22/2024] [Revised: 10/09/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
Treatment with genotoxic agents, such as platinum compounds, is still the mainstay therapeutical approach for the majority of cancers. Our understanding of the mechanisms of action of these drugs is, however, imperfect and continuously evolving. Recent advances highlighted single-stranded DNA (ssDNA) gap accumulation as a potential determinant underlying cisplatin chemosensitivity, at least in some genetic backgrounds, such as BRCA mutations. Cisplatin-induced ssDNA gaps form upon restart of DNA synthesis downstream of cisplatin-induced lesions through repriming catalyzed by the PRIMPOL enzyme. Here, we show that PRIMPOL overexpression in otherwise wild-type cells results in accumulation of cisplatin-induced ssDNA gaps without sensitizing cells to cisplatin, suggesting that ssDNA gap accumulation does not confer cisplatin sensitivity in BRCA-proficient cells. To understand how ssDNA gaps may cause cellular sensitivity, we employed CRISPR-mediated genome-wide genetic screening to identify factors which enable the cytotoxicity of cisplatin-induced ssDNA gaps. We found that the helicase HELQ specifically suppresses cisplatin sensitivity in PRIMPOL-overexpressing cells, and this is associated with reduced ssDNA accumulation. We moreover identify RAD52 as a mediator of this pathway. RAD52 promotes ssDNA gap accumulation through a BRCA-mediated mechanism. Our work identified the HELQ-RAD52-BRCA axis as a regulator of ssDNA gap processing and cisplatin sensitization.
Collapse
Affiliation(s)
- Lindsey M Pale
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Jude B Khatib
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Alexandra Nusawardhana
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Joshua Straka
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - Claudia M Nicolae
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | - George-Lucian Moldovan
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| |
Collapse
|
19
|
Straka J, Khatib JB, Pale L, Nicolae CM, Moldovan GL. CAF-1 promotes efficient PrimPol recruitment to nascent DNA for single-stranded DNA gap formation. Nucleic Acids Res 2024; 52:13865-13880. [PMID: 39558157 DOI: 10.1093/nar/gkae1068] [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: 09/18/2024] [Accepted: 10/22/2024] [Indexed: 11/20/2024] Open
Abstract
Suppression of single-stranded DNA (ssDNA) gap accumulation at replication forks has emerged as a potential determinant of chemosensitivity in homologous recombination (HR)-deficient tumors, as ssDNA gaps are transformed into cytotoxic double-stranded DNA breaks. We have previously shown that the histone chaperone CAF-1's nucleosome deposition function is vital to preventing degradation of stalled replication forks correlating with HR-deficient cells' response to genotoxic drugs. Here we report that the CAF-1-ASF1 pathway promotes ssDNA gap accumulation at replication forks in both wild-type and breast cancer (BRCA)-deficient backgrounds. We show that this is independent of CAF-1's nucleosome deposition function but instead may rely on its proper localization to replication forks. Moreover, we show that the efficient localization to nascent DNA of PrimPol, the enzyme responsible for repriming upon replication stress, is dependent on CAF-1. As PrimPol has been shown to be responsible for generating ssDNA gaps as a byproduct of its repriming function, CAF-1's role in its recruitment could directly impact ssDNA gap formation. We also show that chemoresistance observed in HR-deficient cells when CAF-1 or ASF1A are lost correlates with suppression of ssDNA gaps rather than protection of stalled replication forks. Overall, this work identifies an unexpected role of CAF-1 in regulating PrimPol recruitment and ssDNA gap generation.
Collapse
Affiliation(s)
- Joshua Straka
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Jude B Khatib
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Lindsey Pale
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Claudia M Nicolae
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - George-Lucian Moldovan
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
20
|
Nelligan A, Dungrawala H. SNF2L suppresses nascent DNA gap formation to promote DNA synthesis. Nucleic Acids Res 2024; 52:13003-13018. [PMID: 39413208 PMCID: PMC11602140 DOI: 10.1093/nar/gkae903] [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: 05/16/2024] [Revised: 09/26/2024] [Accepted: 10/01/2024] [Indexed: 10/18/2024] Open
Abstract
Nucleosome remodelers at replication forks function in the assembly and maturation of chromatin post DNA synthesis. The ISWI chromatin remodeler SNF2L (or SMARCA1) travels with replication forks but its contribution to DNA replication remains largely unknown. We find that fork elongation is curtailed when SNF2L is absent. SNF2L deficiency elevates replication stress and causes fork collapse due to remodeling activities by fork reversal enzymes. Mechanistically, SNF2L regulates nucleosome assembly to suppress post-replicative ssDNA gap accumulation. Gap induction is not dependent on fork remodeling and PRIMPOL. Instead, gap synthesis is driven by MRE11 and EXO1 indicating susceptibility of nascent DNA to nucleolytic cleavage and resection when SNF2L is removed. Additionally, nucleosome remodeling by SNF2L protects nascent chromatin from MNase digestion and gap induction highlighting a critical role of SNF2L in chromatin assembly post DNA synthesis to maintain unperturbed replication.
Collapse
Affiliation(s)
- Anthony Nelligan
- Department of Molecular Biosciences, University of South Florida, Tampa, FL 33620, USA
| | - Huzefa Dungrawala
- Department of Molecular Biosciences, University of South Florida, Tampa, FL 33620, USA
| |
Collapse
|
21
|
Uhrig M, Sharma N, Maxwell P, Gomez J, Selemenakis P, Mazin A, Wiese C. Disparate requirements for RAD54L in replication fork reversal. Nucleic Acids Res 2024; 52:12390-12404. [PMID: 39315725 PMCID: PMC11551752 DOI: 10.1093/nar/gkae828] [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: 05/15/2024] [Revised: 09/05/2024] [Accepted: 09/13/2024] [Indexed: 09/25/2024] Open
Abstract
RAD54L is a DNA motor protein with multiple roles in homologous recombination DNA repair. In vitro, RAD54L was shown to also catalyze the reversal and restoration of model replication forks. In cells, however, little is known about how RAD54L may regulate the dynamics of DNA replication. Here, we show that RAD54L restrains the progression of replication forks and functions as a fork remodeler in human cancer cell lines and non-transformed cells. Analogous to HLTF, SMARCAL1 and FBH1, and consistent with a role in fork reversal, RAD54L decelerates fork progression in response to replication stress and suppresses the formation of replication-associated ssDNA gaps. Interestingly, loss of RAD54L prevents nascent strand DNA degradation in both BRCA1/2- and 53BP1-deficient cells, suggesting that RAD54L functions in both pathways of RAD51-mediated replication fork reversal. In the HLTF/SMARCAL1 pathway, RAD54L is critical, but its ability to catalyze branch migration is dispensable, indicative of its function downstream of HLTF/SMARCAL1. Conversely, in the FBH1 pathway, branch migration activity of RAD54L is essential, and FBH1 engagement is dependent on its concerted action with RAD54L. Collectively, our results reveal disparate requirements for RAD54L in two distinct RAD51-mediated fork reversal pathways, positing its potential as a future therapeutic target.
Collapse
Affiliation(s)
- Mollie E Uhrig
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Neelam Sharma
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Petey Maxwell
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Jordi Gomez
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Platon Selemenakis
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Alexander V Mazin
- Department of Biochemistry and Structural Biology, UT Health San Antonio, San Antonio, TX 78229, USA
| | - Claudia Wiese
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
22
|
Elfar G, Aning O, Ngai T, Yeo P, Chan J, Sim S, Goh L, Yuan J, Phua C, Yeo J, Mak S, Goh B, Chow PH, Tam W, Ho Y, Cheok C. p53-dependent crosstalk between DNA replication integrity and redox metabolism mediated through a NRF2-PARP1 axis. Nucleic Acids Res 2024; 52:12351-12377. [PMID: 39315696 PMCID: PMC11551750 DOI: 10.1093/nar/gkae811] [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: 02/01/2024] [Revised: 08/24/2024] [Accepted: 09/10/2024] [Indexed: 09/25/2024] Open
Abstract
Mechanisms underlying p53-mediated protection of the replicating genome remain elusive, despite the quintessential role of p53 in maintaining genomic stability. Here, we uncover an unexpected function of p53 in curbing replication stress by limiting PARP1 activity and preventing the unscheduled degradation of deprotected stalled forks. We searched for p53-dependent factors and elucidated RRM2B as a prime factor. Deficiency in p53/RRM2B results in the activation of an NRF2 antioxidant transcriptional program, with a concomitant elevation in basal PARylation in cells. Dissecting the consequences of p53/RRM2B loss revealed a crosstalk between redox metabolism and genome integrity that is negotiated through a hitherto undescribed NRF2-PARP1 axis, and pinpoint G6PD as a primary oxidative stress-induced NRF2 target and activator of basal PARylation. This study elucidates how loss of p53 could be destabilizing for the replicating genome and, importantly, describes an unanticipated crosstalk between redox metabolism, PARP1 and p53 tumor suppressor pathway that is broadly relevant in cancers and can be leveraged therapeutically.
Collapse
Affiliation(s)
- Gamal Ahmed Elfar
- NUS Department of Pathology, National University of Singapore, Yong Loo Lin School of Medicine, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Obed Aning
- NUS Department of Pathology, National University of Singapore, Yong Loo Lin School of Medicine, Singapore
| | - Tsz Wai Ngai
- NUS Department of Pathology, National University of Singapore, Yong Loo Lin School of Medicine, Singapore
| | - Pearlyn Yeo
- NUS Department of Pathology, National University of Singapore, Yong Loo Lin School of Medicine, Singapore
| | - Joel Wai Kit Chan
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Shang Hong Sim
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Leonard Goh
- NUS Department of Pathology, National University of Singapore, Yong Loo Lin School of Medicine, Singapore
| | - Ju Yuan
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Cheryl Zi Jin Phua
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Joanna Zhen Zhen Yeo
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Shi Ya Mak
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Brian Kim Poh Goh
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore and National Cancer Centre Singapore, Singapore
| | - Pierce Kah-Hoe Chow
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore and National Cancer Centre Singapore, Singapore
- Surgery Academic ClinicalProgramme, Duke-NUS Medical School, National University of Singapore, Singapore
| | - Wai Leong Tam
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University Singapore, Singapore
| | - Ying Swan Ho
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Chit Fang Cheok
- NUS Department of Pathology, National University of Singapore, Yong Loo Lin School of Medicine, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University Singapore, Singapore
| |
Collapse
|
23
|
Osia B, Merkell A, Lopezcolorado FW, Ping X, Stark JM. RAD52 and ERCC6L/PICH have a compensatory relationship for genome stability in mitosis. PLoS Genet 2024; 20:e1011479. [PMID: 39561207 PMCID: PMC11614213 DOI: 10.1371/journal.pgen.1011479] [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: 04/25/2024] [Revised: 12/03/2024] [Accepted: 10/31/2024] [Indexed: 11/21/2024] Open
Abstract
Mammalian RAD52 is a DNA repair factor with strand annealing and recombination mediator activities that appear important in both interphase and mitotic cells. Nonetheless, RAD52 is dispensable for cell viability. To query RAD52 synthetic lethal relationships, we performed genome-wide CRISPR knock-out screens and identified hundreds of candidate synthetic lethal interactions. We then performed secondary screening and identified genes for which depletion causes reduced viability and elevated genome instability (increased 53BP1 nuclear foci) in RAD52-deficient cells. One such factor was ERCC6L, which marks DNA bridges during anaphase, and hence is important for genome stability in mitosis. Thus, we investigated the functional interrelationship between RAD52 and ERCC6L. We found that RAD52 deficiency increases ERCC6L-coated anaphase ultrafine bridges, and that ERCC6L depletion causes elevated RAD52 foci in prometaphase and interphase cells. These effects were enhanced with replication stress (i.e. hydroxyurea) and topoisomerase IIα inhibition (ICRF-193), where post-treatment effect timings were consistent with defects in addressing stress in mitosis. Altogether, we suggest that RAD52 and ERCC6L co-compensate to protect genome stability in mitosis.
Collapse
Affiliation(s)
- Beth Osia
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute of the City of Hope, Duarte, California, United States of America
| | - Arianna Merkell
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute of the City of Hope, Duarte, California, United States of America
| | - Felicia Wednesday Lopezcolorado
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute of the City of Hope, Duarte, California, United States of America
| | - Xiaoli Ping
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute of the City of Hope, Duarte, California, United States of America
| | - Jeremy M. Stark
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute of the City of Hope, Duarte, California, United States of America
| |
Collapse
|
24
|
Ahmad T, Kawasumi R, Hirota K. RAD18- and BRCA1-dependent pathways promote cellular tolerance to the nucleoside analog ganciclovir. Genes Cells 2024; 29:935-950. [PMID: 39169841 PMCID: PMC11555630 DOI: 10.1111/gtc.13155] [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: 07/16/2024] [Revised: 08/09/2024] [Accepted: 08/10/2024] [Indexed: 08/23/2024]
Abstract
Ganciclovir (GCV) is a clinically important drug as it is used to treat viral infections. GCV is incorporated into the DNA during replication, where it interferes with subsequent replication on GCV-incorporated templates. However, the effects of GCV on the host genome and the mechanisms underlying cellular tolerance to GCV remain unclear. In this study, we explored these mechanisms using a collection of mutant DT40 cells. We identified RAD17/-, BRCA1-/-, and RAD18-/- cells as highly GCV-sensitive. RAD17, a component of the alternative checkpoint-clamp loader RAD17-RFC, was required for the activation of the intra-S checkpoint following GCV treatment. BRCA1, a critical factor for promoting homologous recombination (HR), was required for suppressing DNA double-strand breaks (DSBs). Moreover, RAD18, an E3-ligase involved in DNA repair, was critical in suppressing the aberrant ligation of broken chromosomes caused by GCV. We found that BRCA1 suppresses DSBs through HR-mediated repair and template switching (TS)-mediated damage bypass. Moreover, the strong GCV sensitivity of BRCA1-/- cells was rescued by the loss of 53BP1, despite the only partial restoration in the sister chromatid exchange events which are hallmarks of HR. These results indicate that BRCA1 promotes cellular tolerance to GCV through two mechanisms, TS and HR-mediated repair.
Collapse
Affiliation(s)
- Tasnim Ahmad
- Department of Chemistry, Graduate School of ScienceTokyo Metropolitan UniversityHachioji‐shiTokyoJapan
| | - Ryotaro Kawasumi
- Department of Chemistry, Graduate School of ScienceTokyo Metropolitan UniversityHachioji‐shiTokyoJapan
| | - Kouji Hirota
- Department of Chemistry, Graduate School of ScienceTokyo Metropolitan UniversityHachioji‐shiTokyoJapan
| |
Collapse
|
25
|
Akita M, Girvan P, Spirek M, Novacek J, Rueda D, Prokop Z, Krejci L. Mechanism of BCDX2-mediated RAD51 nucleation on short ssDNA stretches and fork DNA. Nucleic Acids Res 2024; 52:11738-11752. [PMID: 39268578 PMCID: PMC11514458 DOI: 10.1093/nar/gkae770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 08/20/2024] [Accepted: 08/26/2024] [Indexed: 09/17/2024] Open
Abstract
Homologous recombination (HR) factors are crucial for DSB repair and processing stalled replication forks. RAD51 paralogs, including RAD51B, RAD51C, RAD51D, XRCC2 and XRCC3, have emerged as essential tumour suppressors, forming two subcomplexes, BCDX2 and CX3. Mutations in these genes are associated with cancer susceptibility and Fanconi anaemia, yet their biochemical activities remain unclear. This study reveals a linear arrangement of BCDX2 subunits compared to the RAD51 ring. BCDX2 shows a strong affinity towards single-stranded DNA (ssDNA) via unique binding mechanism compared to RAD51, and a contribution of DX2 subunits in binding branched DNA substrates. We demonstrate that BCDX2 facilitates RAD51 loading on ssDNA by suppressing the cooperative requirement of RAD51 binding to DNA and stabilizing the filament. Notably, BCDX2 also promotes RAD51 loading on short ssDNA and reversed replication fork substrates. Moreover, while mutants defective in ssDNA binding retain the ability to bind branched DNA substrates, they still facilitate RAD51 loading onto reversed replication forks. Our study provides mechanistic insights into how the BCDX2 complex stimulates the formation of BRCA2-independent RAD51 filaments on short stretches of ssDNA present at ssDNA gaps or stalled replication forks, highlighting its role in genome maintenance and DNA repair.
Collapse
Affiliation(s)
- Masaki Akita
- Department of Biology and National Centre for Biomolecular Research, Masaryk University, Brno, Czech Republic
| | - Paul Girvan
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
| | - Mario Spirek
- Department of Biology and National Centre for Biomolecular Research, Masaryk University, Brno, Czech Republic
| | - Jiri Novacek
- Cryo-Electron Microscopy and Tomography Core Facility, Central European Institute of Technology, Brno, Czech Republic
| | - David Rueda
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
- Single Molecule Imaging Group, MRC-London Institute of Medical Sciences, London, UK
| | - Zbynek Prokop
- Loschmidt Laboratories, Department of Experimental Biology and RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St Anne's University Hospital, Brno, Czech Republic
| | - Lumir Krejci
- Department of Biology and National Centre for Biomolecular Research, Masaryk University, Brno, Czech Republic
| |
Collapse
|
26
|
MacGilvary N, Cantor SB. Positioning loss of PARP1 activity as the central toxic event in BRCA-deficient cancer. DNA Repair (Amst) 2024; 144:103775. [PMID: 39461277 DOI: 10.1016/j.dnarep.2024.103775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/05/2024] [Accepted: 10/13/2024] [Indexed: 10/29/2024]
Abstract
The mechanisms by which poly(ADP-ribose) polymerase 1 (PARP1) inhibitors (PARPi)s inflict replication stress and/or DNA damage are potentially numerous. PARPi toxicity could derive from loss of its catalytic activity and/or its physical trapping of PARP1 onto DNA that perturbs not only PARP1 function in DNA repair and DNA replication, but also obstructs compensating pathways. The combined disruption of PARP1 with either of the hereditary breast and ovarian cancer genes, BRCA1 or BRCA2 (BRCA), results in synthetic lethality. This has driven the development of PARP inhibitors as therapies for BRCA-mutant cancers. In this review, we focus on recent findings that highlight loss of PARP1 catalytic activity, rather than PARPi-induced allosteric trapping, as central to PARPi efficacy in BRCA deficient cells. However, we also review findings that PARP-trapping is an effective strategy in other genetic deficiencies. Together, we conclude that the mechanism-of-action of PARP inhibitors is not unilateral; with loss of activity or enhanced trapping differentially killing depending on the genetic context. Therefore, effectively targeting cancer cells requires an intricate understanding of their key underlying vulnerabilities.
Collapse
Affiliation(s)
- Nathan MacGilvary
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Sharon B Cantor
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
27
|
Colicino-Murbach E, Hathaway C, Dungrawala H. Replication fork stalling in late S-phase elicits nascent strand degradation by DNA mismatch repair. Nucleic Acids Res 2024; 52:10999-11013. [PMID: 39180395 PMCID: PMC11472054 DOI: 10.1093/nar/gkae721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 07/03/2024] [Accepted: 08/06/2024] [Indexed: 08/26/2024] Open
Abstract
Eukaryotic chromosomal replication occurs in a segmented, temporal manner wherein open euchromatin and compact heterochromatin replicate during early and late S-phase respectively. Using single molecule DNA fiber analyses coupled with cell synchronization, we find that newly synthesized strands remain stable at perturbed forks in early S-phase. Unexpectedly, stalled forks are susceptible to nucleolytic digestion during late replication resulting in defective fork restart. This inherent vulnerability to nascent strand degradation is dependent on fork reversal enzymes and resection nucleases MRE11, DNA2 and EXO1. Inducing chromatin compaction elicits digestion of nascent DNA in response to fork stalling due to reduced association of RAD51 with nascent DNA. Furthermore, RAD51 occupancy at stalled forks in late S-phase is diminished indicating that densely packed chromatin limits RAD51 accessibility to mediate replication fork protection. Genetic analyses reveal that susceptibility of late replicating forks to nascent DNA digestion is dependent on EXO1 via DNA mismatch repair (MMR) and that the BRCA2-mediated replication fork protection blocks MMR from degrading nascent DNA. Overall, our findings illustrate differential regulation of fork protection between early and late replication and demonstrate nascent strand degradation as a critical determinant of heterochromatin instability in response to replication stress.
Collapse
Affiliation(s)
| | - Caitlin Hathaway
- Department of Molecular Biosciences, University of South Florida, Tampa, FL, USA
| | - Huzefa Dungrawala
- Department of Molecular Biosciences, University of South Florida, Tampa, FL, USA
| |
Collapse
|
28
|
Yu T, Lok BH. PARP inhibitor resistance mechanisms and PARP inhibitor derived imaging probes. Expert Rev Anticancer Ther 2024; 24:989-1008. [PMID: 39199000 DOI: 10.1080/14737140.2024.2398494] [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: 07/15/2024] [Revised: 08/19/2024] [Accepted: 08/27/2024] [Indexed: 09/01/2024]
Abstract
INTRODUCTION Poly(ADP-ribose) polymerase 1 (PARP1) inhibition has become a major target in anticancer therapy. While PARP inhibitors (PARPi) are approved for homologous recombination (HR) deficient cancers, therapeutic resistance is a challenge and PARPi are now being investigated in cancers lacking HR deficiencies. This creates a need to develop molecular and imaging biomarkers of PARPi response to improve patient selection and circumvent therapeutic resistance. AREAS COVERED PubMed and clinicaltrials.gov were queried for studies on PARPi resistance and imaging. This review summarizes established and emerging resistance mechanisms to PARPi, and the current state of imaging and theragnostic probes for PARPi, including fluorescently labeled and radiolabeled probes. EXPERT OPINION While progress has been made in understanding PARPi therapeutic resistance, clinical evidence remains lacking and relatively little is known regarding PARPi response outside of HR deficiencies. Continued research will clarify the importance of known biomarkers and resistance mechanisms in patient cohorts and the broader utility of PARPi. Progress has also been made in PARPi imaging, particularly with radiolabeled probes, and both imaging and theragnostic probes have now reached clinical validation. Reducing abdominal background signal from probe clearance will broaden their applicability, and improvements to molecular synthesis and radiation delivery will increase their utility.
Collapse
Affiliation(s)
- Tony Yu
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Benjamin H Lok
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Department of Radiation Oncology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
29
|
Petta TB, Carlson J. Exploring Molecular Drivers of PARPi Resistance in BRCA1-Deficient Ovarian Cancer: The Role of LY6E and Immunomodulation. Int J Mol Sci 2024; 25:10427. [PMID: 39408764 PMCID: PMC11477368 DOI: 10.3390/ijms251910427] [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: 08/01/2024] [Revised: 09/04/2024] [Accepted: 09/10/2024] [Indexed: 10/20/2024] Open
Abstract
Approximately 50% of patients diagnosed with ovarian cancer harbor tumors with mutations in BRCA1, BRCA2, or other genes involved in homologous recombination repair (HR). The presence of homologous recombination deficiency (HRD) is an approved biomarker for poly-ADP-ribose polymerase inhibitors (PARPis) as a maintenance treatment following a positive response to initial platinum-based chemotherapy. Despite this treatment option, the development of resistance to PARPis is common among recurrent disease patients, leading to a poor prognosis. In this study, we conducted a comprehensive analysis using publicly available datasets to elucidate the molecular mechanisms driving PARPi resistance in BRCA1-deficient ovarian cancer. Our findings reveal a central role for the interferon (IFN) pathway in mediating resistance in the context of BRCA1 deficiency. Through integrative bioinformatics approaches, we identified LY6E, an interferon-stimulated gene, as a key mediator of PARPi resistance, with its expression linked to an immunosuppressive tumor microenvironment (TME) encouraging tumor progression and invasion. LY6E amplification correlates with poor prognosis and increased expression of immune-related gene signatures, which is predictive of immunotherapy response. Interestingly, LY6E expression upon PARPi treatment resistance was found to be dependent on BRCA1 status. Gene expression analysis in the Orien/cBioPortal database revealed an association between LY6E and genes involved in DNA repair, such as Rad21 and PUF60, emphasizing the interplay between DNA repair pathways and immune modulation. Moreover, PUF60, Rad21, and LY6E are located on chromosome 8q24, a locus often amplified and associated with the progression of ovarian cancer. Overall, our study provides novel insights into the molecular determinants of PARPi resistance and highlights LY6E as a promising prognostic biomarker in the management of HRD ovarian cancer. Future studies are needed to fully elucidate the molecular mechanisms underlying the role of LY6E in PARPi resistance.
Collapse
Affiliation(s)
- Tirzah Braz Petta
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
- Centro de Biociencias, Departmento de Biologia Celular e Genetica, Universidade Federal do Rio Grande do Norte, Natal CEP 59078-970, Brazil
| | - Joseph Carlson
- Department of Pathology, City of Hope, Duarte, CA 91010, USA;
| |
Collapse
|
30
|
Rao S, Andrs M, Shukla K, Isik E, König C, Schneider S, Bauer M, Rosano V, Prokes J, Müller A, Janscak P. Senataxin RNA/DNA helicase promotes replication restart at co-transcriptional R-loops to prevent MUS81-dependent fork degradation. Nucleic Acids Res 2024; 52:10355-10369. [PMID: 39119900 PMCID: PMC11417401 DOI: 10.1093/nar/gkae673] [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: 02/19/2024] [Revised: 07/12/2024] [Accepted: 07/23/2024] [Indexed: 08/10/2024] Open
Abstract
Replication forks stalled at co-transcriptional R-loops can be restarted by a mechanism involving fork cleavage-religation cycles mediated by MUS81 endonuclease and DNA ligase IV (LIG4), which presumably relieve the topological barrier generated by the transcription-replication conflict (TRC) and facilitate ELL-dependent reactivation of transcription. Here, we report that the restart of R-loop-stalled replication forks via the MUS81-LIG4-ELL pathway requires senataxin (SETX), a helicase that can unwind RNA:DNA hybrids. We found that SETX promotes replication fork progression by preventing R-loop accumulation during S-phase. Interestingly, loss of SETX helicase activity leads to nascent DNA degradation upon induction of R-loop-mediated fork stalling by hydroxyurea. This fork degradation phenotype is independent of replication fork reversal and results from DNA2-mediated resection of MUS81-cleaved replication forks that accumulate due to defective replication restart. Finally, we demonstrate that SETX acts in a common pathway with the DEAD-box helicase DDX17 to suppress R-loop-mediated replication stress in human cells. A possible cooperation between these RNA/DNA helicases in R-loop unwinding at TRC sites is discussed.
Collapse
Affiliation(s)
- Satyajeet Rao
- Institute of Molecular Cancer Research, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Martin Andrs
- Institute of Molecular Cancer Research, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Kaustubh Shukla
- Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic
| | - Esin Isik
- Institute of Molecular Cancer Research, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Christiane König
- Institute of Molecular Cancer Research, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Stefan Schneider
- Institute of Molecular Cancer Research, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Michael Bauer
- Institute of Molecular Cancer Research, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Vinicio Rosano
- Institute of Molecular Cancer Research, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Jiri Prokes
- Institute of Molecular Cancer Research, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Anne Müller
- Institute of Molecular Cancer Research, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Pavel Janscak
- Institute of Molecular Cancer Research, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
- Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic
| |
Collapse
|
31
|
Adolph MB, Cortez D. Mechanisms and regulation of replication fork reversal. DNA Repair (Amst) 2024; 141:103731. [PMID: 39089193 PMCID: PMC11877614 DOI: 10.1016/j.dnarep.2024.103731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 08/03/2024]
Abstract
DNA replication is remarkably accurate with estimates of only a handful of mutations per human genome per cell division cycle. Replication stress caused by DNA lesions, transcription-replication conflicts, and other obstacles to the replication machinery must be efficiently overcome in ways that minimize errors and maximize completion of DNA synthesis. Replication fork reversal is one mechanism that helps cells tolerate replication stress. This process involves reannealing of parental template DNA strands and generation of a nascent-nascent DNA duplex. While fork reversal may be beneficial by facilitating DNA repair or template switching, it must be confined to the appropriate contexts to preserve genome stability. Many enzymes have been implicated in this process including ATP-dependent DNA translocases like SMARCAL1, ZRANB3, HLTF, and the helicase FBH1. In addition, the RAD51 recombinase is required. Many additional factors and regulatory activities also act to ensure reversal is beneficial instead of yielding undesirable outcomes. Finally, reversed forks must also be stabilized and often need to be restarted to complete DNA synthesis. Disruption or deregulation of fork reversal causes a variety of human diseases. In this review we will describe the latest models for reversal and key mechanisms of regulation.
Collapse
Affiliation(s)
- Madison B Adolph
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, United States
| | - David Cortez
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, United States.
| |
Collapse
|
32
|
Schreuder A, Wendel TJ, Dorresteijn CGV, Noordermeer SM. (Single-stranded DNA) gaps in understanding BRCAness. Trends Genet 2024; 40:757-771. [PMID: 38789375 DOI: 10.1016/j.tig.2024.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024]
Abstract
The tumour-suppressive roles of BRCA1 and 2 have been attributed to three seemingly distinct functions - homologous recombination, replication fork protection, and single-stranded (ss)DNA gap suppression - and their relative importance is under debate. In this review, we examine the origin and resolution of ssDNA gaps and discuss the recent advances in understanding the role of BRCA1/2 in gap suppression. There are ample data showing that gap accumulation in BRCA1/2-deficient cells is linked to genomic instability and chemosensitivity. However, it remains unclear whether there is a causative role and the function of BRCA1/2 in gap suppression cannot unambiguously be dissected from their other functions. We therefore conclude that the three functions of BRCA1 and 2 are closely intertwined and not mutually exclusive.
Collapse
Affiliation(s)
- Anne Schreuder
- Leiden University Medical Center, Department of Human Genetics, Leiden, The Netherlands; Oncode Institute, Utrecht, The Netherlands
| | - Tiemen J Wendel
- Leiden University Medical Center, Department of Human Genetics, Leiden, The Netherlands; Oncode Institute, Utrecht, The Netherlands
| | - Carlo G V Dorresteijn
- Leiden University Medical Center, Department of Human Genetics, Leiden, The Netherlands
| | - Sylvie M Noordermeer
- Leiden University Medical Center, Department of Human Genetics, Leiden, The Netherlands; Oncode Institute, Utrecht, The Netherlands.
| |
Collapse
|
33
|
Dixit S, Nagraj T, Bhattacharya D, Saxena S, Sahoo S, Chittela RK, Somyajit K, Nagaraju G. RTEL1 helicase counteracts RAD51-mediated homologous recombination and fork reversal to safeguard replicating genomes. Cell Rep 2024; 43:114594. [PMID: 39116203 DOI: 10.1016/j.celrep.2024.114594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 05/23/2024] [Accepted: 07/22/2024] [Indexed: 08/10/2024] Open
Abstract
Homologous recombination (HR) plays an essential role in the repair of DNA double-strand breaks (DSBs), replication stress responses, and genome maintenance. However, unregulated HR during replication can impair genome duplication and compromise genome stability. The mechanisms underlying HR regulation during DNA replication are obscure. Here, we find that RTEL1 helicase, RAD51, and RAD51 paralogs are enriched at stalled replication sites. The absence of RTEL1 leads to an increase in the RAD51-mediated HR and fork reversal during replication and affects genome-wide replication, which can be rescued by co-depleting RAD51 and RAD51 paralogs. Interestingly, co-depletion of fork remodelers such as SMARCAL1/ZRANB3/HLTF/FBH1 and expression of HR-defective RAD51 mutants also rescues replication defects in RTEL1-deficient cells. The anti-recombinase function of RTEL1 during replication depends on its interaction with PCNA and helicase activity. Together, our data identify the role of RTEL1 helicase in restricting RAD51-mediated fork reversal and HR activity to facilitate error-free genome duplication.
Collapse
Affiliation(s)
- Suruchi Dixit
- Department of Biochemistry Indian Institute of Science, Bangalore 560012, India
| | - Tarun Nagraj
- Department of Biochemistry Indian Institute of Science, Bangalore 560012, India
| | | | - Sneha Saxena
- Department of Biochemistry Indian Institute of Science, Bangalore 560012, India
| | - Satyaranjan Sahoo
- Department of Biochemistry Indian Institute of Science, Bangalore 560012, India
| | - Rajani Kant Chittela
- Applied Genomics Section, Bioscience Group, Bhabha Atomic Research Centre, Mumbai 400085, India
| | - Kumar Somyajit
- Department of Biochemistry Indian Institute of Science, Bangalore 560012, India; Functional Genomics & Metabolism Research Unit, Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, Odense M 5230, Denmark.
| | - Ganesh Nagaraju
- Department of Biochemistry Indian Institute of Science, Bangalore 560012, India.
| |
Collapse
|
34
|
Cybulla E, Wallace S, Meroni A, Jackson J, Agashe S, Tennakoon M, Limbu M, Quinet A, Lomonosova E, Noia H, Tirman S, Wood M, Lemacon D, Fuh K, Zou L, Vindigni A. A RAD18-UBC13-PALB2-RNF168 axis mediates replication fork recovery in BRCA1-deficient cancer cells. Nucleic Acids Res 2024; 52:8861-8879. [PMID: 38943334 PMCID: PMC11347138 DOI: 10.1093/nar/gkae563] [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: 02/07/2024] [Revised: 05/24/2024] [Accepted: 06/21/2024] [Indexed: 07/01/2024] Open
Abstract
BRCA1/2 proteins function in genome stability by promoting repair of double-stranded DNA breaks through homologous recombination and by protecting stalled replication forks from nucleolytic degradation. In BRCA1/2-deficient cancer cells, extensively degraded replication forks can be rescued through distinct fork recovery mechanisms that also promote cell survival. Here, we identified a novel pathway mediated by the E3 ubiquitin ligase RAD18, the E2-conjugating enzyme UBC13, the recombination factor PALB2, the E3 ubiquitin ligase RNF168 and PCNA ubiquitination that promotes fork recovery in BRCA1- but not BRCA2-deficient cells. We show that this pathway does not promote fork recovery by preventing replication fork reversal and degradation in BRCA1-deficient cells. We propose a mechanism whereby the RAD18-UBC13-PALB2-RNF168 axis facilitates resumption of DNA synthesis by promoting re-annealing of the complementary single-stranded template strands of the extensively degraded forks, thereby allowing re-establishment of a functional replication fork. We also provide preliminary evidence for the potential clinical relevance of this novel fork recovery pathway in BRCA1-mutated cancers, as RAD18 is over-expressed in BRCA1-deficient cancers, and RAD18 loss compromises cell viability in BRCA1-deficient cancer cells.
Collapse
Affiliation(s)
- Emily Cybulla
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Sierra Wallace
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Alice Meroni
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Jessica Jackson
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Sumedha Agashe
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Mithila Tennakoon
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Mangsi Limbu
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Annabel Quinet
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Elena Lomonosova
- Department of Obstetrics and Gynecology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Hollie Noia
- Department of Obstetrics and Gynecology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Stephanie Tirman
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Matthew Wood
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Delphine Lemacon
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Katherine Fuh
- Division of Gynecologic Oncology, Department of Ob/Gyn and Reproductive Sciences, University of California San Francisco, San Francisco, CA 94143, USA
| | - Lee Zou
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27708, USA
| | - Alessandro Vindigni
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| |
Collapse
|
35
|
Di Biagi L, Marozzi G, Malacaria E, Honda M, Aiello FA, Valenzisi P, Spies M, Franchitto A, Pichierri P. RAD52 prevents accumulation of Polα -dependent replication gaps at perturbed replication forks in human cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.12.536536. [PMID: 37090680 PMCID: PMC10120653 DOI: 10.1101/2023.04.12.536536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Replication gaps can arise as a consequence of perturbed DNA replication and their accumulation might undermine the stability of the genome. Loss of RAD52, a protein involved in the regulation of fork reversal, promotes accumulation of parental ssDNA gaps during replication perturbation. Here, we demonstrate that this is due to the engagement of Polα downstream of the extensive degradation of perturbed replication forks after their reversal, and is not dependent on PrimPol. Polα is hyper-recruited at parental ssDNA in the absence of RAD52, and this recruitment is dependent on fork reversal enzymes and RAD51. Of note, we report that the interaction between Polα and RAD51 is stimulated by RAD52 inhibition, and Polα -dependent gap accumulation requires nucleation of RAD51 suggesting that it occurs downstream strand invasion. Altogether, our data indicate that RAD51- Polα -dependent repriming is essential to promote fork restart and limit DNA damage accumulation when RAD52 function is disabled.
Collapse
Affiliation(s)
- Ludovica Di Biagi
- Mechanisms, Biomarkers and Models Section, Genome Stability Group, Department of Environment and Health, Istituto Superiore di Sanità - Viale Regina Elena 299, 00161 Rome (Italy)
| | - Giorgia Marozzi
- Mechanisms, Biomarkers and Models Section, Genome Stability Group, Department of Environment and Health, Istituto Superiore di Sanità - Viale Regina Elena 299, 00161 Rome (Italy)
| | - Eva Malacaria
- Mechanisms, Biomarkers and Models Section, Genome Stability Group, Department of Environment and Health, Istituto Superiore di Sanità - Viale Regina Elena 299, 00161 Rome (Italy)
| | - Masayoshi Honda
- Department of Biochemistry and Molecular Biology, University of Iowa Carver College of Medicine, 51 Newton Road, Iowa City, IA 52242 (USA)
| | - Francesca Antonella Aiello
- Mechanisms, Biomarkers and Models Section, Genome Stability Group, Department of Environment and Health, Istituto Superiore di Sanità - Viale Regina Elena 299, 00161 Rome (Italy)
| | - Pasquale Valenzisi
- Mechanisms, Biomarkers and Models Section, Genome Stability Group, Department of Environment and Health, Istituto Superiore di Sanità - Viale Regina Elena 299, 00161 Rome (Italy)
| | - Maria Spies
- Department of Biochemistry and Molecular Biology, University of Iowa Carver College of Medicine, 51 Newton Road, Iowa City, IA 52242 (USA)
| | - Annapaola Franchitto
- Mechanisms, Biomarkers and Models Section, Genome Stability Group, Department of Environment and Health, Istituto Superiore di Sanità - Viale Regina Elena 299, 00161 Rome (Italy)
| | - Pietro Pichierri
- Mechanisms, Biomarkers and Models Section, Genome Stability Group, Department of Environment and Health, Istituto Superiore di Sanità - Viale Regina Elena 299, 00161 Rome (Italy)
- Istituto Nazionale Biostrutture e Biosistemi - Roma Area Research - Via delle Medaglie d’Oro 305, 00136 Rome (Italy)
| |
Collapse
|
36
|
Chiang HC, Qi L, Mitra P, Huang Y, Hu Y, Li R. R-loop functions in Brca1-associated mammary tumorigenesis. Proc Natl Acad Sci U S A 2024; 121:e2403600121. [PMID: 39116124 PMCID: PMC11331088 DOI: 10.1073/pnas.2403600121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 07/11/2024] [Indexed: 08/10/2024] Open
Abstract
Deleterious accumulation of R-loops, a DNA-RNA hybrid structure, contributes to genome instability. They are associated with BRCA1 mutation-related breast cancer, an estrogen receptor α negative (ERα-) tumor type originating from luminal progenitor cells. However, a presumed causality of R-loops in tumorigenesis has not been established in vivo. Here, we overexpress mouse Rnaseh1 (Rh1-OE) in vivo to remove accumulated R-loops in Brca1-deficient mouse mammary epithelium (BKO). R-loop removal exacerbates DNA replication stress in proliferating BKO mammary epithelial cells, with little effect on homology-directed repair of double-strand breaks following ionizing radiation. Compared to their BKO counterparts, BKO-Rh1-OE mammary glands contain fewer luminal progenitor cells but more mature luminal cells. Despite a similar incidence of spontaneous mammary tumors in BKO and BKO-Rh1-OE mice, a significant percentage of BKO-Rh1-OE tumors express ERα and progesterone receptor. Our results suggest that rather than directly elevating the overall tumor incidence, R-loops influence the mammary tumor subtype by shaping the cell of origin for Brca1 tumors.
Collapse
Affiliation(s)
- Huai-Chin Chiang
- Department of Biochemistry and Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, DC20037
| | - Leilei Qi
- Department of Anatomy and Cell Biology, School of Medicine & Health Sciences, The George Washington University, Washington, DC20037
| | - Payal Mitra
- Department of Biochemistry and Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, DC20037
| | - Yimeng Huang
- Department of Biochemistry and Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, DC20037
| | - Yanfen Hu
- Department of Anatomy and Cell Biology, School of Medicine & Health Sciences, The George Washington University, Washington, DC20037
| | - Rong Li
- Department of Biochemistry and Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, DC20037
| |
Collapse
|
37
|
Li X, Zou L. BRCAness, DNA gaps, and gain and loss of PARP inhibitor-induced synthetic lethality. J Clin Invest 2024; 134:e181062. [PMID: 39007266 PMCID: PMC11245158 DOI: 10.1172/jci181062] [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: 07/16/2024] Open
Abstract
Mutations in the tumor-suppressor genes BRCA1 and BRCA2 resulting in BRCA1/2 deficiency are frequently identified in breast, ovarian, prostate, pancreatic, and other cancers. Poly(ADP-ribose) polymerase (PARP) inhibitors (PARPis) selectively kill BRCA1/2-deficient cancer cells by inducing synthetic lethality, providing an effective biomarker-guided strategy for targeted cancer therapy. However, a substantial fraction of cancer patients carrying BRCA1/2 mutations do not respond to PARPis, and most patients develop resistance to PARPis over time, highlighting a major obstacle to PARPi therapy in the clinic. Recent studies have revealed that changes of specific functional defects of BRCA1/2-deficient cells, particularly their defects in suppressing and protecting single-stranded DNA gaps, contribute to the gain or loss of PARPi-induced synthetic lethality. These findings not only shed light on the mechanism of action of PARPis, but also lead to revised models that explain how PARPis selectively kill BRCA-deficient cancer cells. Furthermore, new mechanistic principles of PARPi sensitivity and resistance have emerged from these studies, generating potentially useful guidelines for predicting the PARPi response and design therapies for overcoming PARPi resistance. In this Review, we will discuss these recent studies and put them in context with the classic views of PARPi-induced synthetic lethality, aiming to stimulate the development of new therapeutic strategies to overcome PARPi resistance and improve PARPi therapy.
Collapse
|
38
|
Khodaverdian V, Sano T, Maggs LR, Tomarchio G, Dias A, Tran M, Clairmont C, McVey M. REV1 coordinates a multi-faceted tolerance response to DNA alkylation damage and prevents chromosome shattering in Drosophila melanogaster. PLoS Genet 2024; 20:e1011181. [PMID: 39074150 PMCID: PMC11309488 DOI: 10.1371/journal.pgen.1011181] [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: 02/09/2024] [Revised: 08/08/2024] [Accepted: 07/09/2024] [Indexed: 07/31/2024] Open
Abstract
When replication forks encounter damaged DNA, cells utilize damage tolerance mechanisms to allow replication to proceed. These include translesion synthesis at the fork, postreplication gap filling, and template switching via fork reversal or homologous recombination. The extent to which these different damage tolerance mechanisms are utilized depends on cell, tissue, and developmental context-specific cues, the last two of which are poorly understood. To address this gap, we have investigated damage tolerance responses in Drosophila melanogaster. We report that tolerance of DNA alkylation damage in rapidly dividing larval tissues depends heavily on translesion synthesis. Furthermore, we show that the REV1 protein plays a multi-faceted role in damage tolerance in Drosophila. Larvae lacking REV1 are hypersensitive to methyl methanesulfonate (MMS) and have highly elevated levels of γ-H2Av (Drosophila γ-H2AX) foci and chromosome aberrations in MMS-treated tissues. Loss of the REV1 C-terminal domain (CTD), which recruits multiple translesion polymerases to damage sites, sensitizes flies to MMS. In the absence of the REV1 CTD, DNA polymerases eta and zeta become critical for MMS tolerance. In addition, flies lacking REV3, the catalytic subunit of polymerase zeta, require the deoxycytidyl transferase activity of REV1 to tolerate MMS. Together, our results demonstrate that Drosophila prioritize the use of multiple translesion polymerases to tolerate alkylation damage and highlight the critical role of REV1 in the coordination of this response to prevent genome instability.
Collapse
Affiliation(s)
- Varandt Khodaverdian
- Department of Biology, Tufts University, Medford, Massachusetts, United States of America
| | - Tokio Sano
- Department of Biology, Tufts University, Medford, Massachusetts, United States of America
| | - Lara R. Maggs
- Department of Biology, Tufts University, Medford, Massachusetts, United States of America
| | - Gina Tomarchio
- Department of Biology, Tufts University, Medford, Massachusetts, United States of America
| | - Ana Dias
- Department of Biology, Tufts University, Medford, Massachusetts, United States of America
| | - Mai Tran
- Department of Biology, Tufts University, Medford, Massachusetts, United States of America
| | - Connor Clairmont
- Department of Biology, Tufts University, Medford, Massachusetts, United States of America
| | - Mitch McVey
- Department of Biology, Tufts University, Medford, Massachusetts, United States of America
| |
Collapse
|
39
|
Técher H, Gopaul D, Heuzé J, Bouzalmad N, Leray B, Vernet A, Mettling C, Moreaux J, Pasero P, Lin YL. MRE11 and TREX1 control senescence by coordinating replication stress and interferon signaling. Nat Commun 2024; 15:5423. [PMID: 38926338 PMCID: PMC11208572 DOI: 10.1038/s41467-024-49740-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Oncogene-induced senescence (OIS) arrests cell proliferation in response to replication stress (RS) induced by oncogenes. OIS depends on the DNA damage response (DDR), but also on the cGAS-STING pathway, which detects cytosolic DNA and induces type I interferons (IFNs). Whether and how RS and IFN responses cooperate to promote OIS remains unknown. Here, we show that the induction of OIS by the H-RASV12 oncogene in immortalized human fibroblasts depends on the MRE11 nuclease. Indeed, treatment with the MRE11 inhibitor Mirin prevented RS, micronuclei formation and IFN response induced by RASV12. Overexpression of the cytosolic nuclease TREX1 also prevented OIS. Conversely, overexpression of a dominant negative mutant of TREX1 or treatment with IFN-β was sufficient to induce RS and DNA damage, independent of RASV12 induction. These data suggest that the IFN response acts as a positive feedback loop to amplify DDR in OIS through a process regulated by MRE11 and TREX1.
Collapse
Affiliation(s)
- Hervé Técher
- Institut de Génétique Humaine, University of Montpellier, CNRS, Equipe Labellisée Ligue contre le Cancer, Montpellier, France
- Institute for Research on Cancer and Aging of Nice (IRCAN), Université Côte d'Azur, CNRS UMR7284 - INSERM U1081, Nice, France
| | - Diyavarshini Gopaul
- Institut de Génétique Humaine, University of Montpellier, CNRS, Equipe Labellisée Ligue contre le Cancer, Montpellier, France
- Biotech Research and Innovation Centre, University of Copenhagen, 2200 N, Copenhagen, Denmark
| | - Jonathan Heuzé
- Institut de Génétique Humaine, University of Montpellier, CNRS, Equipe Labellisée Ligue contre le Cancer, Montpellier, France
| | - Nail Bouzalmad
- Institut de Génétique Humaine, University of Montpellier, CNRS, Equipe Labellisée Ligue contre le Cancer, Montpellier, France
| | - Baptiste Leray
- Institut de Génétique Humaine, University of Montpellier, CNRS, Equipe Labellisée Ligue contre le Cancer, Montpellier, France
| | - Audrey Vernet
- Institut de Génétique Humaine, University of Montpellier, CNRS, Equipe Labellisée Ligue contre le Cancer, Montpellier, France
| | - Clément Mettling
- Institut de Génétique Humaine, University of Montpellier, CNRS, Montpellier, France
| | - Jérôme Moreaux
- Institut de Génétique Humaine, University of Montpellier, CNRS, Montpellier, France
- Department of Biological Hematology, CHU Montpellier, Montpellier, France
- University of Montpellier, UFR Medicine, Montpellier, France
| | - Philippe Pasero
- Institut de Génétique Humaine, University of Montpellier, CNRS, Equipe Labellisée Ligue contre le Cancer, Montpellier, France.
| | - Yea-Lih Lin
- Institut de Génétique Humaine, University of Montpellier, CNRS, Equipe Labellisée Ligue contre le Cancer, Montpellier, France.
| |
Collapse
|
40
|
Matsuzaki K, Shinohara A, Shinohara M. Human AAA+ ATPase FIGNL1 suppresses RAD51-mediated ultra-fine bridge formation. Nucleic Acids Res 2024; 52:5774-5791. [PMID: 38597669 PMCID: PMC11162793 DOI: 10.1093/nar/gkae263] [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: 08/31/2023] [Revised: 03/09/2024] [Accepted: 03/29/2024] [Indexed: 04/11/2024] Open
Abstract
RAD51 filament is crucial for the homology-dependent repair of DNA double-strand breaks and stalled DNA replication fork protection. Positive and negative regulators control RAD51 filament assembly and disassembly. RAD51 is vital for genome integrity but excessive accumulation of RAD51 on chromatin causes genome instability and growth defects. However, the detailed mechanism underlying RAD51 disassembly by negative regulators and the physiological consequence of abnormal RAD51 persistence remain largely unknown. Here, we report the role of the human AAA+ ATPase FIGNL1 in suppressing a novel type of RAD51-mediated genome instability. FIGNL1 knockout human cells were defective in RAD51 dissociation after replication fork restart and accumulated ultra-fine chromosome bridges (UFBs), whose formation depends on RAD51 rather than replication fork stalling. FIGNL1 suppresses homologous recombination intermediate-like UFBs generated between sister chromatids at genomic loci with repeated sequences such as telomeres and centromeres. These data suggest that RAD51 persistence per se induces the formation of unresolved linkage between sister chromatids resulting in catastrophic genome instability. FIGNL1 facilitates post-replicative disassembly of RAD51 filament to suppress abnormal recombination intermediates and UFBs. These findings implicate FIGNL1 as a key factor required for active RAD51 removal after processing of stalled replication forks, which is essential to maintain genome stability.
Collapse
Affiliation(s)
- Kenichiro Matsuzaki
- Department of Advanced Bioscience, Graduate School of Agriculture, Kindai University, Nara City, Nara 631-8505, Japan
| | - Akira Shinohara
- Laboratory of Genome and Chromosome Functions, Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Miki Shinohara
- Department of Advanced Bioscience, Graduate School of Agriculture, Kindai University, Nara City, Nara 631-8505, Japan
- Agricultural Technology and Innovation Research Institute, Kindai University, Nara City, Nara 631-8505, Japan
| |
Collapse
|
41
|
Goehring L, Keegan S, Lahiri S, Xia W, Kong M, Jimenez-Sainz J, Gupta D, Drapkin R, Jensen RB, Smith DJ, Rothenberg E, Fenyö D, Huang TT. Dormant origin firing promotes head-on transcription-replication conflicts at transcription termination sites in response to BRCA2 deficiency. Nat Commun 2024; 15:4716. [PMID: 38830843 PMCID: PMC11148086 DOI: 10.1038/s41467-024-48286-1] [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: 08/19/2023] [Accepted: 04/24/2024] [Indexed: 06/05/2024] Open
Abstract
BRCA2 is a tumor suppressor protein responsible for safeguarding the cellular genome from replication stress and genotoxicity, but the specific mechanism(s) by which this is achieved to prevent early oncogenesis remains unclear. Here, we provide evidence that BRCA2 acts as a critical suppressor of head-on transcription-replication conflicts (HO-TRCs). Using Okazaki-fragment sequencing (Ok-seq) and computational analysis, we identified origins (dormant origins) that are activated near the transcription termination sites (TTS) of highly expressed, long genes in response to replication stress. Dormant origins are a source for HO-TRCs, and drug treatments that inhibit dormant origin firing led to a reduction in HO-TRCs, R-loop formation, and DNA damage. Using super-resolution microscopy, we showed that HO-TRC events track with elongating RNA polymerase II, but not with transcription initiation. Importantly, RNase H2 is recruited to sites of HO-TRCs in a BRCA2-dependent manner to help alleviate toxic R-loops associated with HO-TRCs. Collectively, our results provide a mechanistic basis for how BRCA2 shields against genomic instability by preventing HO-TRCs through both direct and indirect means occurring at predetermined genomic sites based on the pre-cancer transcriptome.
Collapse
Affiliation(s)
- Liana Goehring
- Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| | - Sarah Keegan
- Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
- Institute for Systems Genetics, New York University School of Medicine, New York University School of Medicine, New York, NY, USA
| | - Sudipta Lahiri
- Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
- Department of Therapeutic Radiology, Yale University, New Haven, CT, USA
| | - Wenxin Xia
- Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| | - Michael Kong
- Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| | | | - Dipika Gupta
- Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| | - Ronny Drapkin
- Penn Ovarian Cancer Research Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Basser Center for BRCA, Abramson Cancer Center, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Ryan B Jensen
- Department of Therapeutic Radiology, Yale University, New Haven, CT, USA
| | - Duncan J Smith
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Eli Rothenberg
- Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| | - David Fenyö
- Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
- Institute for Systems Genetics, New York University School of Medicine, New York University School of Medicine, New York, NY, USA
| | - Tony T Huang
- Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
42
|
Teresa BGD, Ayala-Zambrano C, González-Suárez M, Molina B, Torres L, Rodríguez A, Frías S. Reversion from basal histone H4 hypoacetylation at the replication fork increases DNA damage in FANCA deficient cells. PLoS One 2024; 19:e0298032. [PMID: 38820384 PMCID: PMC11142588 DOI: 10.1371/journal.pone.0298032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 01/16/2024] [Indexed: 06/02/2024] Open
Abstract
The FA/BRCA pathway safeguards DNA replication by repairing interstrand crosslinks (ICL) and maintaining replication fork stability. Chromatin structure, which is in part regulated by histones posttranslational modifications (PTMs), has a role in maintaining genomic integrity through stabilization of the DNA replication fork and promotion of DNA repair. An appropriate balance of PTMs, especially acetylation of histones H4 in nascent chromatin, is required to preserve a stable DNA replication fork. To evaluate the acetylation status of histone H4 at the replication fork of FANCA deficient cells, we compared histone acetylation status at the DNA replication fork of isogenic FANCA deficient and FANCA proficient cell lines by using accelerated native immunoprecipitation of nascent DNA (aniPOND) and in situ protein interactions in the replication fork (SIRF) assays. We found basal hypoacetylation of multiple residues of histone H4 in FA replication forks, together with increased levels of Histone Deacetylase 1 (HDAC1). Interestingly, high-dose short-term treatment with mitomycin C (MMC) had no effect over H4 acetylation abundance at the replication fork. However, chemical inhibition of histone deacetylases (HDAC) with Suberoylanilide hydroxamic acid (SAHA) induced acetylation of the FANCA deficient DNA replication forks to levels comparable to their isogenic control counterparts. This forced permanence of acetylation impacted FA cells homeostasis by inducing DNA damage and promoting G2 cell cycle arrest. Altogether, this caused reduced RAD51 foci formation and increased markers of replication stress, including phospho-RPA-S33. Hypoacetylation of the FANCA deficient replication fork, is part of the cellular phenotype, the perturbation of this feature by agents that prevent deacetylation, such as SAHA, have a deleterious effect over the delicate equilibrium they have reached to perdure despite a defective FA/BRCA pathway.
Collapse
Affiliation(s)
- Benilde García-de Teresa
- Laboratorio de Citogenética, Instituto Nacional de Pediatría, Mexico City, Ciudad de México, Mexico
- Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Ciudad de México, Mexico
| | - Cecilia Ayala-Zambrano
- Laboratorio de Citogenética, Instituto Nacional de Pediatría, Mexico City, Ciudad de México, Mexico
- Doctorado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City, Ciudad de México, Mexico
| | - Mirna González-Suárez
- Laboratorio de Citogenética, Instituto Nacional de Pediatría, Mexico City, Ciudad de México, Mexico
| | - Bertha Molina
- Laboratorio de Citogenética, Instituto Nacional de Pediatría, Mexico City, Ciudad de México, Mexico
| | - Leda Torres
- Laboratorio de Citogenética, Instituto Nacional de Pediatría, Mexico City, Ciudad de México, Mexico
| | - Alfredo Rodríguez
- Laboratorio de Falla Medular y Carcinogénesis, Unidad de Genética de la Nutrición, Instituto Nacional de Pediatría, Mexico City, Ciudad de México, Mexico
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Mexico City, Ciudad de México, Mexico
| | - Sara Frías
- Laboratorio de Citogenética, Instituto Nacional de Pediatría, Mexico City, Ciudad de México, Mexico
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Mexico City, Ciudad de México, Mexico
| |
Collapse
|
43
|
Kong L, Cheng C, Cheruiyot A, Yuan J, Yang Y, Hwang S, Foust D, Tsao N, Wilkerson E, Mosammaparast N, Major MB, Piston DW, Li S, You Z. TCAF1 promotes TRPV2-mediated Ca 2+ release in response to cytosolic DNA to protect stressed replication forks. Nat Commun 2024; 15:4609. [PMID: 38816425 PMCID: PMC11139906 DOI: 10.1038/s41467-024-48988-6] [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: 09/19/2023] [Accepted: 05/21/2024] [Indexed: 06/01/2024] Open
Abstract
The protection of the replication fork structure under stress conditions is essential for genome maintenance and cancer prevention. A key signaling pathway for fork protection involves TRPV2-mediated Ca2+ release from the ER, which is triggered after the generation of cytosolic DNA and the activation of cGAS/STING. This results in CaMKK2/AMPK activation and subsequent Exo1 phosphorylation, which prevent aberrant fork processing, thereby ensuring genome stability. However, it remains poorly understood how the TRPV2 channel is activated by the presence of cytosolic DNA. Here, through a genome-wide CRISPR-based screen, we identify TRPM8 channel-associated factor 1 (TCAF1) as a key factor promoting TRPV2-mediated Ca2+ release under replication stress or other conditions that activate cGAS/STING. Mechanistically, TCAF1 assists Ca2+ release by facilitating the dissociation of STING from TRPV2, thereby relieving TRPV2 repression. Consistent with this function, TCAF1 is required for fork protection, chromosomal stability, and cell survival after replication stress.
Collapse
Affiliation(s)
- Lingzhen Kong
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Chen Cheng
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Abigael Cheruiyot
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jiayi Yuan
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Yichan Yang
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Sydney Hwang
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Daniel Foust
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Ning Tsao
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, 63110, USA
| | - Emily Wilkerson
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Nima Mosammaparast
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, 63110, USA
| | - Michael B Major
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - David W Piston
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Shan Li
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Zhejiang Provincial Key Laboratory of Pancreatic Disease in the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310029, China.
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310029, China.
| | - Zhongsheng You
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
44
|
Rivard RS, Chang YC, Ragland RL, Thu YM, Kassab M, Mandal RS, Van Riper SK, Kulej K, Higgins L, Markowski TM, Shang D, Hedberg J, Erber L, Garcia B, Chen Y, Bielinsky AK, Brown EJ. Improved detection of DNA replication fork-associated proteins. Cell Rep 2024; 43:114178. [PMID: 38703364 PMCID: PMC12034227 DOI: 10.1016/j.celrep.2024.114178] [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: 07/12/2022] [Revised: 03/06/2024] [Accepted: 04/16/2024] [Indexed: 05/06/2024] Open
Abstract
Innovative methods to retrieve proteins associated with actively replicating DNA have provided a glimpse into the molecular dynamics of replication fork stalling. We report that a combination of density-based replisome enrichment by isolating proteins on nascent DNA (iPOND2) and label-free quantitative mass spectrometry (iPOND2-DRIPPER) substantially increases both replication factor yields and the dynamic range of protein quantification. Replication protein abundance in retrieved nascent DNA is elevated up to 300-fold over post-replicative controls, and recruitment of replication stress factors upon fork stalling is observed at similar levels. The increased sensitivity of iPOND2-DRIPPER permits direct measurement of ubiquitination events without intervening retrieval of diglycine tryptic fragments of ubiquitin. Using this approach, we find that stalled replisomes stimulate the recruitment of a diverse cohort of DNA repair factors, including those associated with poly-K63-ubiquitination. Finally, we uncover the temporally controlled association of stalled replisomes with nuclear pore complex components and nuclear cytoskeleton networks.
Collapse
Affiliation(s)
- Rebecca S Rivard
- Department of Cancer Biology and the Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ya-Chu Chang
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Ryan L Ragland
- Department of Cancer Biology and the Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yee-Mon Thu
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Muzaffer Kassab
- Department of Cancer Biology and the Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rahul Shubhra Mandal
- Department of Cancer Biology and the Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan K Van Riper
- University of Minnesota Informatics Institute, University of Minnesota, Minneapolis, MN, USA
| | - Katarzyna Kulej
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - LeeAnn Higgins
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Todd M Markowski
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - David Shang
- Department of Cancer Biology and the Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jack Hedberg
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Luke Erber
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Benjamin Garcia
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yue Chen
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Anja-Katrin Bielinsky
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA.
| | - Eric J Brown
- Department of Cancer Biology and the Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
45
|
Dibitetto D, Liptay M, Vivalda F, Dogan H, Gogola E, González Fernández M, Duarte A, Schmid JA, Decollogny M, Francica P, Przetocka S, Durant ST, Forment JV, Klebic I, Siffert M, de Bruijn R, Kousholt AN, Marti NA, Dettwiler M, Sørensen CS, Tille JC, Undurraga M, Labidi-Galy I, Lopes M, Sartori AA, Jonkers J, Rottenberg S. H2AX promotes replication fork degradation and chemosensitivity in BRCA-deficient tumours. Nat Commun 2024; 15:4430. [PMID: 38789420 PMCID: PMC11126719 DOI: 10.1038/s41467-024-48715-1] [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: 03/20/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Histone H2AX plays a key role in DNA damage signalling in the surrounding regions of DNA double-strand breaks (DSBs). In response to DNA damage, H2AX becomes phosphorylated on serine residue 139 (known as γH2AX), resulting in the recruitment of the DNA repair effectors 53BP1 and BRCA1. Here, by studying resistance to poly(ADP-ribose) polymerase (PARP) inhibitors in BRCA1/2-deficient mammary tumours, we identify a function for γH2AX in orchestrating drug-induced replication fork degradation. Mechanistically, γH2AX-driven replication fork degradation is elicited by suppressing CtIP-mediated fork protection. As a result, H2AX loss restores replication fork stability and increases chemoresistance in BRCA1/2-deficient tumour cells without restoring homology-directed DNA repair, as highlighted by the lack of DNA damage-induced RAD51 foci. Furthermore, in the attempt to discover acquired genetic vulnerabilities, we find that ATM but not ATR inhibition overcomes PARP inhibitor (PARPi) resistance in H2AX-deficient tumours by interfering with CtIP-mediated fork protection. In summary, our results demonstrate a role for H2AX in replication fork biology in BRCA-deficient tumours and establish a function of H2AX separable from its classical role in DNA damage signalling and DSB repair.
Collapse
Affiliation(s)
- Diego Dibitetto
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012, Bern, Switzerland.
- Cancer Therapy Resistance Cluster and Bern Center for Precision Medicine, Department for Biomedical Research, University of Bern, 3012, Bern, Switzerland.
- Department of Experimental Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via Mario Negri 2, 20156, Milan, Italy.
| | - Martin Liptay
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012, Bern, Switzerland
- Cancer Therapy Resistance Cluster and Bern Center for Precision Medicine, Department for Biomedical Research, University of Bern, 3012, Bern, Switzerland
| | - Francesca Vivalda
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Hülya Dogan
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012, Bern, Switzerland
- Cancer Therapy Resistance Cluster and Bern Center for Precision Medicine, Department for Biomedical Research, University of Bern, 3012, Bern, Switzerland
| | - Ewa Gogola
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066CX, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Martín González Fernández
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012, Bern, Switzerland
- Cancer Therapy Resistance Cluster and Bern Center for Precision Medicine, Department for Biomedical Research, University of Bern, 3012, Bern, Switzerland
| | - Alexandra Duarte
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066CX, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Jonas A Schmid
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Morgane Decollogny
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012, Bern, Switzerland
- Cancer Therapy Resistance Cluster and Bern Center for Precision Medicine, Department for Biomedical Research, University of Bern, 3012, Bern, Switzerland
| | - Paola Francica
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012, Bern, Switzerland
- Cancer Therapy Resistance Cluster and Bern Center for Precision Medicine, Department for Biomedical Research, University of Bern, 3012, Bern, Switzerland
| | - Sara Przetocka
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Stephen T Durant
- DDR Biology, Bioscience, Oncology R&D, AstraZeneca, Cambridge, CB4 0WG, UK
| | - Josep V Forment
- DDR Biology, Bioscience, Oncology R&D, AstraZeneca, Cambridge, CB4 0WG, UK
| | - Ismar Klebic
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012, Bern, Switzerland
| | - Myriam Siffert
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012, Bern, Switzerland
| | - Roebi de Bruijn
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066CX, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Arne N Kousholt
- Oncode Institute, Amsterdam, The Netherlands
- Biotech Research and Innovation Centre, University of Copenhagen, 2200 N, Copenhagen, Denmark
| | - Nicole A Marti
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012, Bern, Switzerland
- Cancer Therapy Resistance Cluster and Bern Center for Precision Medicine, Department for Biomedical Research, University of Bern, 3012, Bern, Switzerland
| | - Martina Dettwiler
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012, Bern, Switzerland
| | - Claus S Sørensen
- Biotech Research and Innovation Centre, University of Copenhagen, 2200 N, Copenhagen, Denmark
| | - Jean-Christophe Tille
- Division of Clinical Pathology, Department of Diagnostics, Hôpitaux Universitaires de Genève, Geneva, Switzerland
| | - Manuela Undurraga
- Division of Gynecology, Department of Pediatrics and Gynecology, Hôpitaux Universitaires de Genève, Geneva, Switzerland
| | - Intidhar Labidi-Galy
- Faculty of Medicine, Department of Medicine and Center of Translational Research in Onco-Hematology, University of Geneva, Swiss Cancer Center Leman, Geneva, Switzerland
- Department of Oncology, Hôpitaux Universitaires de Genève, 4, Rue Gabrielle Perret-Gentil, Geneva, 1205, Switzerland
| | - Massimo Lopes
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Alessandro A Sartori
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Jos Jonkers
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066CX, Amsterdam, The Netherlands.
- Oncode Institute, Amsterdam, The Netherlands.
| | - Sven Rottenberg
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, 3012, Bern, Switzerland.
- Cancer Therapy Resistance Cluster and Bern Center for Precision Medicine, Department for Biomedical Research, University of Bern, 3012, Bern, Switzerland.
- Division of Molecular Pathology, The Netherlands Cancer Institute, 1066CX, Amsterdam, The Netherlands.
| |
Collapse
|
46
|
Batenburg NL, Sowa DJ, Walker JR, Andres SN, Zhu XD. CSB and SMARCAL1 compete for RPA32 at stalled forks and differentially control the fate of stalled forks in BRCA2-deficient cells. Nucleic Acids Res 2024; 52:5067-5087. [PMID: 38416570 PMCID: PMC11109976 DOI: 10.1093/nar/gkae154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 02/14/2024] [Accepted: 02/19/2024] [Indexed: 03/01/2024] Open
Abstract
CSB (Cockayne syndrome group B) and SMARCAL1 (SWI/SNF-related, matrix-associated, actin-dependent, regulator of chromatin, subfamily A-like 1) are DNA translocases that belong to the SNF2 helicase family. They both are enriched at stalled replication forks. While SMARCAL1 is recruited by RPA32 to stalled forks, little is known about whether RPA32 also regulates CSB's association with stalled forks. Here, we report that CSB directly interacts with RPA, at least in part via a RPA32C-interacting motif within the N-terminal region of CSB. Modeling of the CSB-RPA32C interaction suggests that CSB binds the RPA32C surface previously shown to be important for binding of UNG2 and SMARCAL1. We show that this interaction is necessary for promoting fork slowing and fork degradation in BRCA2-deficient cells but dispensable for mediating restart of stalled forks. CSB competes with SMARCAL1 for RPA32 at stalled forks and acts non-redundantly with SMARCAL1 to restrain fork progression in response to mild replication stress. In contrast to CSB stimulated restart of stalled forks, SMARCAL1 inhibits restart of stalled forks in BRCA2-deficient cells, likely by suppressing BIR-mediated repair of collapsed forks. Loss of CSB leads to re-sensitization of SMARCAL1-depleted BRCA2-deficient cells to chemodrugs, underscoring a role of CSB in targeted cancer therapy.
Collapse
Affiliation(s)
- Nicole L Batenburg
- Department of Biology, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Dana J Sowa
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - John R Walker
- Department of Biology, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Sara N Andres
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| | - Xu-Dong Zhu
- Department of Biology, McMaster University, Hamilton, Ontario L8S 4K1, Canada
| |
Collapse
|
47
|
Herr LM, Schaffer ED, Fuchs KF, Datta A, Brosh RM. Replication stress as a driver of cellular senescence and aging. Commun Biol 2024; 7:616. [PMID: 38777831 PMCID: PMC11111458 DOI: 10.1038/s42003-024-06263-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 04/29/2024] [Indexed: 05/25/2024] Open
Abstract
Replication stress refers to slowing or stalling of replication fork progression during DNA synthesis that disrupts faithful copying of the genome. While long considered a nexus for DNA damage, the role of replication stress in aging is under-appreciated. The consequential role of replication stress in promotion of organismal aging phenotypes is evidenced by an extensive list of hereditary accelerated aging disorders marked by molecular defects in factors that promote replication fork progression and operate uniquely in the replication stress response. Additionally, recent studies have revealed cellular pathways and phenotypes elicited by replication stress that align with designated hallmarks of aging. Here we review recent advances demonstrating the role of replication stress as an ultimate driver of cellular senescence and aging. We discuss clinical implications of the intriguing links between cellular senescence and aging including application of senotherapeutic approaches in the context of replication stress.
Collapse
Affiliation(s)
- Lauren M Herr
- Helicases and Genomic Integrity Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Ethan D Schaffer
- Helicases and Genomic Integrity Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Kathleen F Fuchs
- Helicases and Genomic Integrity Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Arindam Datta
- Helicases and Genomic Integrity Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA.
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Robert M Brosh
- Helicases and Genomic Integrity Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA.
| |
Collapse
|
48
|
Uhrig ME, Sharma N, Maxwell P, Selemenakis P, Mazin AV, Wiese C. Disparate requirements for RAD54L in replication fork reversal. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.26.550704. [PMID: 37546955 PMCID: PMC10402051 DOI: 10.1101/2023.07.26.550704] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
RAD54L is a DNA motor protein with multiple roles in homologous recombination DNA repair (HR). In vitro , RAD54L was shown to also catalyze the reversal and restoration of model replication forks. In cells, however, little is known about how RAD54L may regulate the dynamics of DNA replication. Here, we show that RAD54L restrains the progression of replication forks and functions as a fork remodeler in human cells. Analogous to HLTF, SMARCAL1, and FBH1, and consistent with a role in fork reversal, RAD54L decelerates fork progression in response to replication stress and suppresses the formation of replication-associated ssDNA gaps. Interestingly, loss of RAD54L prevents nascent strand DNA degradation in both BRCA1/2- and 53BP1-deficient cells, suggesting that RAD54L functions in both pathways of RAD51-mediated replication fork reversal. In the HLTF/SMARCAL1 pathway, RAD54L is critical, but its ability to catalyze branch migration is dispensable, indicative of its function downstream of HLTF/SMARCAL1. Conversely, in the FBH1 pathway, branch migration activity of RAD54L is essential, and FBH1 engagement is dependent on its concerted action with RAD54L. Collectively, our results reveal disparate requirements for RAD54L in two distinct RAD51-mediated fork reversal pathways, positing its potential as a future therapeutic target.
Collapse
|
49
|
Ubieto-Capella P, Ximénez-Embún P, Giménez-Llorente D, Losada A, Muñoz J, Méndez J. A rewiring of DNA replication mediated by MRE11 exonuclease underlies primed-to-naive cell de-differentiation. Cell Rep 2024; 43:114024. [PMID: 38581679 DOI: 10.1016/j.celrep.2024.114024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 02/01/2024] [Accepted: 03/15/2024] [Indexed: 04/08/2024] Open
Abstract
Mouse embryonic stem cells (mESCs) in the primed pluripotency state, which resembles the post-implantation epiblast, can be de-differentiated in culture to a naive state that resembles the pre-implantation inner cell mass. We report that primed-to-naive mESC transition entails a significant slowdown of DNA replication forks and the compensatory activation of dormant origins. Using isolation of proteins on nascent DNA coupled to mass spectrometry, we identify key changes in replisome composition that are responsible for these effects. Naive mESC forks are enriched in MRE11 nuclease and other DNA repair proteins. MRE11 is recruited to newly synthesized DNA in response to transcription-replication conflicts, and its inhibition or genetic downregulation in naive mESCs is sufficient to restore the fork rate of primed cells. Transcriptomic analyses indicate that MRE11 exonuclease activity is required for the complete primed-to-naive mESC transition, demonstrating a direct link between DNA replication dynamics and the mESC de-differentiation process.
Collapse
Affiliation(s)
- Patricia Ubieto-Capella
- DNA Replication Group, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
| | - Pilar Ximénez-Embún
- Proteomics Unit-ProteoRed-ISCIII, Biotechnology Programme, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
| | - Daniel Giménez-Llorente
- Chromosome Dynamics Group, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
| | - Ana Losada
- Chromosome Dynamics Group, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
| | - Javier Muñoz
- Proteomics Unit-ProteoRed-ISCIII, Biotechnology Programme, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
| | - Juan Méndez
- DNA Replication Group, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain.
| |
Collapse
|
50
|
Tian T, Chen J, Zhao H, Li Y, Xia F, Huang J, Han J, Liu T. UFL1 triggers replication fork degradation by MRE11 in BRCA1/2-deficient cells. Nat Chem Biol 2024:10.1038/s41589-024-01611-7. [PMID: 38649452 DOI: 10.1038/s41589-024-01611-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 03/26/2024] [Indexed: 04/25/2024]
Abstract
The stabilization of stalled forks has emerged as a crucial mechanism driving resistance to poly(ADP-ribose) polymerase (PARP) inhibitors in BRCA1/2-deficient tumors. Here, we identify UFL1, a UFM1-specific E3 ligase, as a pivotal regulator of fork stability and the response to PARP inhibitors in BRCA1/2-deficient cells. On replication stress, UFL1 localizes to stalled forks and catalyzes the UFMylation of PTIP, a component of the MLL3/4 methyltransferase complex, specifically at lysine 148. This modification facilitates the assembly of the PTIP-MLL3/4 complex, resulting in the enrichment of H3K4me1 and H3K4me3 at stalled forks and subsequent recruitment of the MRE11 nuclease. Consequently, loss of UFL1, disruption of PTIP UFMylation or overexpression of the UFM1 protease UFSP2 protects nascent DNA strands from extensive degradation and confers resistance to PARP inhibitors in BRCA1/2-deficient cells. These findings provide mechanistic insights into the processes underlying fork instability in BRCA1/2-deficient cells and offer potential therapeutic avenues for the treatment of BRCA1/2-deficient tumors.
Collapse
Affiliation(s)
- Tian Tian
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, China
- The MOE Key Laboratory of Biosystems Homeostasis and Protection and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Junliang Chen
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, China
| | - Huacun Zhao
- Department of Cell Biology, and Department of General Surgery of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yulin Li
- The MOE Key Laboratory of Biosystems Homeostasis and Protection and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Feiyu Xia
- Department of Cell Biology, and Department of General Surgery of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Huang
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, China
- The MOE Key Laboratory of Biosystems Homeostasis and Protection and Zhejiang Provincial Key Laboratory of Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Jinhua Han
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ting Liu
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Department of Cell Biology, and Department of General Surgery of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|