1
|
Merigliano C, Ryu T, Cibulka J, Rawal CC, See CD, Mitra A, Reynolds TW, Butova NL, Caridi CP, Li X, Wang J, Deng C, Chenoweth DM, Sung P, Capelson M, Krejčí L, Chiolo I. Off-pore Nup98 condensates mobilize heterochromatic breaks and exclude Rad51. Mol Cell 2025; 85:2355-2373.e11. [PMID: 40480227 DOI: 10.1016/j.molcel.2025.05.012] [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: 05/16/2024] [Revised: 03/17/2025] [Accepted: 05/08/2025] [Indexed: 06/22/2025]
Abstract
Phase separation forms membraneless compartments, including heterochromatin "domains" and repair foci. Pericentromeric heterochromatin mostly comprises repeated sequences prone to aberrant recombination. In Drosophila cells, "safe" homologous recombination (HR) repair of these sequences requires their relocalization to the nuclear periphery before Rad51 recruitment and strand invasion. How this mobilization initiates is unknown, and the contribution of phase separation is unclear. Here, we show that Nup98 nucleoporin is recruited to repair sites before relocalization by Sec13 or Nup88, and downstream of the Smc5/6 complex and heterochromatin protein 1 (HP1). Remarkably, Nup98 condensates are immiscible with HP1 condensates, and they are required and sufficient to mobilize repair sites and exclude Rad51, thus preventing aberrant recombination while promoting HR repair. Disrupting this pathway results in heterochromatin repair defects and widespread chromosome rearrangements, revealing an "off-pore" role for nucleoporins and phase separation in nuclear dynamics and genome integrity in a multicellular eukaryote.
Collapse
Affiliation(s)
- Chiara Merigliano
- Department of Biological Sciences, Molecular and Computational Biology Section, University of Southern California, Los Angeles, CA 90089, USA
| | - Taehyun Ryu
- Department of Biological Sciences, Molecular and Computational Biology Section, University of Southern California, Los Angeles, CA 90089, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Jakub Cibulka
- Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5/A7, Brno 62500, Czech Republic
| | - Chetan C Rawal
- Department of Biological Sciences, Molecular and Computational Biology Section, University of Southern California, Los Angeles, CA 90089, USA
| | - Colby D See
- Department of Biological Sciences, Molecular and Computational Biology Section, University of Southern California, Los Angeles, CA 90089, USA
| | - Anik Mitra
- Department of Biological Sciences, Molecular and Computational Biology Section, University of Southern California, Los Angeles, CA 90089, USA
| | - Trevor W Reynolds
- Department of Biological Sciences, Molecular and Computational Biology Section, University of Southern California, Los Angeles, CA 90089, USA
| | - Nadejda L Butova
- Department of Biological Sciences, Molecular and Computational Biology Section, University of Southern California, Los Angeles, CA 90089, USA
| | - Christopher P Caridi
- Department of Biological Sciences, Molecular and Computational Biology Section, University of Southern California, Los Angeles, CA 90089, USA
| | - Xiao Li
- Department of Biological Sciences, Molecular and Computational Biology Section, University of Southern California, Los Angeles, CA 90089, USA; Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Jeff Wang
- Department of Biochemistry and Structural Biology and Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Changfeng Deng
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David M Chenoweth
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Patrick Sung
- Department of Biochemistry and Structural Biology and Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Maya Capelson
- Department of Biology, San Diego State University, San Diego, CA 92182, USA
| | - Lumír Krejčí
- Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5/A7, Brno 62500, Czech Republic; National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5/A4, 62500 Brno, Czech Republic
| | - Irene Chiolo
- Department of Biological Sciences, Molecular and Computational Biology Section, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
2
|
Longo MA, Ahmed SM, Chen Y, Tsai CL, Namjoshi S, Shen R, Ahmed Z, Wang X, Perera RL, Arvai A, Lee M, Kong LR, Engl W, Ng WS, Zhao ZW, Venkitaraman AR, Tainer JA, Schlacher K. BRCA2 C-terminal clamp restructures RAD51 dimers to bind B-DNA for replication fork stability. Mol Cell 2025; 85:2080-2096.e6. [PMID: 40441151 PMCID: PMC12186269 DOI: 10.1016/j.molcel.2025.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 04/04/2025] [Accepted: 05/06/2025] [Indexed: 06/29/2025]
Abstract
Tumor suppressor protein breast cancer susceptibility protein 2 (BRCA2) acts with RAD51 in replication fork protection (FP) and homology-directed DNA-break repair (HDR). Critical for cancer etiology and therapy resistance, the BRCA2 C terminus was thought to stabilize recombinogenic RAD51 after the assembly of ATP-extended RAD51 filaments on single-stranded DNA (ssDNA). Here, the detailed crystal structure of the human BRCA2 C-terminal interaction domain (TR2 interface [TR2i]) complexed with ATP-bound RAD51 prior to DNA binding instead reveals TR2i unexpectedly induces a unique ATP-RAD51 dimer conformation that accommodates nucleation onto double-stranded B-DNA unsuited for HDR initiation. Structural, biochemical, and molecular results with interface-guided mutations uncover TR2i's FP mechanism. Proline-driven secondary structure stabilizes residue triads and spans the RAD51 dimer, engaging pivotal interactions of RAD51 M210 and BRCA2 S3291/P3292, the cyclin-dependent kinase (CDK) phosphorylation site that toggles between FP during S phase and HDR in G2. TR2i evidently acts as an allosteric clamp, switching RAD51 from ssDNA to double-stranded and B-DNA binding, enforcing FP over HDR, challenging the current BRCA2-RAD51 dogma.
Collapse
Affiliation(s)
- Michael A Longo
- Department of Molecular & Cellular Oncology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Syed Moiz Ahmed
- The Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Yue Chen
- Department of Cancer Biology, UT MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Chi-Lin Tsai
- Department of Molecular & Cellular Oncology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sarita Namjoshi
- Department of Molecular & Cellular Oncology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Runze Shen
- Department of Molecular & Cellular Oncology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zamal Ahmed
- Department of Molecular & Cellular Oncology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiaoyan Wang
- Department of Molecular & Cellular Oncology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Andy Arvai
- The Department of Integrative Structural & Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Miyoung Lee
- Medical Research Council Cancer Unit, University of Cambridge, Hills Road, Cambridge CB2 0XZ, UK
| | - Li Ren Kong
- The Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; Medical Research Council Cancer Unit, University of Cambridge, Hills Road, Cambridge CB2 0XZ, UK
| | - Wilfried Engl
- Department of Chemistry and Centre for BioImaging Sciences, National University of Singapore, Singapore 117543, Singapore
| | - Woei Shyuan Ng
- Department of Chemistry and Centre for BioImaging Sciences, National University of Singapore, Singapore 117543, Singapore
| | - Ziqing Winston Zhao
- Department of Chemistry and Centre for BioImaging Sciences, National University of Singapore, Singapore 117543, Singapore; Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Ashok R Venkitaraman
- The Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; Medical Research Council Cancer Unit, University of Cambridge, Hills Road, Cambridge CB2 0XZ, UK; Institute of Molecular & Cell Biology, Agency for Science, Technology and Research (A∗STAR), Singapore 138673, Singapore.
| | - John A Tainer
- Department of Molecular & Cellular Oncology, UT MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Cancer Biology, UT MD Anderson Cancer Center, Houston, TX 77054, USA.
| | - Katharina Schlacher
- Department of Cancer Biology, UT MD Anderson Cancer Center, Houston, TX 77054, USA.
| |
Collapse
|
3
|
Chu SSH, Xing G, Ling H. The role of human Shu complex in ATP-dependent regulation of RAD51 filaments during homologous recombination-associated DNA damage response. J Biol Chem 2025; 301:110212. [PMID: 40345587 PMCID: PMC12167799 DOI: 10.1016/j.jbc.2025.110212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/25/2025] [Accepted: 05/04/2025] [Indexed: 05/11/2025] Open
Abstract
Error-free DNA lesion bypass is an important pathway in DNA damage tolerance. The Shu complex facilitates this process by promoting homologous recombination (HR) to bypass DNA damage. Biochemical analysis of the human Shu complex homolog, hSWS1-SWSAP1, offers valuable insights into the HR-associated DNA damage response. Here, we biochemically characterized the human Shu complex and examined its interactions with RAD51 filaments, which are essential in HR. Using fluorescence polarization assays, we first revealed that hSWS1-SWSAP1 preferentially binds DNA with an exposed 5' end in the presence of adenine nucleotides. We then investigated and validated the DNA-stimulated ATPase activity of hSWS1-SWSAP1 through site-specific mutagenesis, revealing that DNA with an exposed 5' end is the most efficient in enhancing this activity. Furthermore, we showed that hSWS1-SWSAP1 initially interacts with RAD51 filaments at the 5' end and modulates the properties of the nucleoprotein filaments using fluorescence-based assays. Our findings revealed that hSWS1-SWSAP1 induces conformational changes in RAD51 filaments in an ATP hydrolysis-dependent manner, while its stabilization of the filaments depends on ATP binding. This work provides mechanistic insights into the regulation of RAD51 filaments in HR-associated DNA damage tolerance.
Collapse
Affiliation(s)
- Sam S H Chu
- Department of Biochemistry, University of Western Ontario, London, Ontario, Canada
| | - Guangxin Xing
- Department of Biochemistry, University of Western Ontario, London, Ontario, Canada
| | - Hong Ling
- Department of Biochemistry, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
4
|
Chu SSH, Xing G, Jha VK, Ling H. The Shu complex is an ATPase that regulates Rad51 filaments during homologous recombination in the DNA damage response. DNA Repair (Amst) 2025; 145:103792. [PMID: 39647428 DOI: 10.1016/j.dnarep.2024.103792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 11/15/2024] [Accepted: 11/24/2024] [Indexed: 12/10/2024]
Abstract
Rad51 filaments are Rad51-coated single-stranded DNA and essential in homologous recombination (HR). The yeast Shu complex (Shu) is a conserved regulator of homologous recombination, working through its modulation on Rad51 filaments to direct HR-associated DNA damage response. However, the biochemical properties of Shu remain unclear, which hinders molecular insights into Shu's role in HR and the DNA damage response. In this work, we biochemically characterized Shu and analyzed its molecular actions on single-stranded DNA and Rad51 filaments. First, we revealed that Shu preferentially binds fork-shaped DNA with 20nt ssDNA components. Then, we identified and validated, through site-specific mutagenesis, that Shu is an ATPase and hydrolyzes ATP in a DNA-dependent manner. Furthermore, we showed that Shu interacts with ssDNA and Rad51 filaments and alters the properties of ssDNA and the filaments with a 5'-3' polarity. The alterations depend on the ATP hydrolysis of Shu, suggesting that the ATPase activity of Shu is important in regulating its functions. The preference of Shu for acting on the 5' end of Rad51 filaments aligns with the observation that Shu promotes lesion bypass at the lagging strand of a replication fork. Our work on Shu, a prototype modulator of Rad51 filaments in eukaryotes, provides a general molecular mechanism for Rad51-mediated error-free DNA lesion bypass.
Collapse
Affiliation(s)
- Sam S H Chu
- Department of Biochemistry, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Guangxin Xing
- Department of Biochemistry, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Vikash K Jha
- Department of Biochemistry, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Hong Ling
- Department of Biochemistry, University of Western Ontario, London, Ontario N6A 5C1, Canada.
| |
Collapse
|
5
|
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
|
6
|
Longo MA, Ahmed SM, Chen Y, Tsai CL, Namjoshi S, Wang X, Perera RL, Arvai A, Lee M, Kong LR, Engl W, Shyuan W, Zhao ZW, Venkitaraman AR, Tainer JA, Schlacher K. BRCA2 C-terminal clamp restructures RAD51 dimers to bind B-DNA for replication fork stability. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.21.614229. [PMID: 39345573 PMCID: PMC11429943 DOI: 10.1101/2024.09.21.614229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Tumor suppressor protein BRCA2 acts with RAD51 in replication-fork protection (FP) and homology-directed DNA break repair (HDR). Critical for cancer etiology and therapy resistance, BRCA2 C-terminus was thought to stabilize RAD51-filaments after they assemble on single-stranded (ss)DNA. Here we determined the detailed crystal structure for BRCA2 C-terminal interaction-domain (TR2i) with ATP-bound RAD51 prior to DNA binding. In contrast to recombinogenic RAD51-filaments comprising extended ATP-bound RAD51 dimers, TR2i unexpectedly reshapes ATP-RAD51 into a unique dimer conformation accommodating double-stranded B-DNA binding unsuited for HDR initiation. Structural, biochemical, and molecular results with interface-guided mutations uncover TR2i's FP mechanism. Proline-driven secondary-structure stabilizes residue triads and spans the RAD51 dimer engaging pivotal interactions of RAD51 M210 and BRCA2 S3291/P3292, the cyclin-dependent kinase (CDK) phosphorylation site that toggles between FP during S-phase and HDR in G2. TR2i evidently acts as an allosteric clamp switching RAD51 from ssDNA to double-stranded and B-DNA binding enforcing FP over HDR.
Collapse
|
7
|
Park SJ, Park SJ, Kwon YW, Choi EH. Synergistic combination of RAD51-SCR7 improves CRISPR-Cas9 genome editing efficiency by preventing R-loop accumulation. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102274. [PMID: 39161621 PMCID: PMC11331969 DOI: 10.1016/j.omtn.2024.102274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 07/13/2024] [Indexed: 08/21/2024]
Abstract
CRISPR-Cas9 has emerged as a powerful tool for genome editing. However, Cas9 genome editing faces challenges, including low efficiency and off-target effects. Here, we report that combined treatment with RAD51, a key factor in homologous recombination, and SCR7, a DNA ligase IV small-molecule inhibitor, enhances CRISPR-Cas9-mediated genome-editing efficiency in human embryonic kidney 293T and human induced pluripotent stem cells, as confirmed by cyro- transmission electron microscopy and functional analyses. First, our findings reveal the crucial role of RAD51 in homologous recombination (HR)-mediated DNA repair process. Elevated levels of exogenous RAD51 promote a post-replication step via single-strand DNA gap repair process, ensuring the completion of DNA replication. Second, using the all-in-one CRISPR-Cas9-RAD51 system, highly expressed RAD51 improved the multiple endogenous gene knockin/knockout efficiency and insertion/deletion (InDel) mutation by activating the HR-based repair pathway in concert with SCR7. Sanger sequencing shows distinct outcomes for RAD51-SCR7 in the ratio of InDel mutations in multiple genome sites. Third, RAD51-SCR7 combination can induce efficient R-loop resolution and DNA repair by enhanced HR process, which leads to DNA replication stalling and thus is advantageous to CRISPR-Cas9-based stable genome editing. Our study suggests promising applications in genome editing by enhancing CRISPR-Cas9 efficiency through RAD51 and SCR7, offering potential advancements in biotechnology and therapeutics.
Collapse
Affiliation(s)
- Sun-Ji Park
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Deagu 41061, South Korea
| | - Seo Jung Park
- New Drug Development Center, Osong Medical Innovation Foundation, Cheongju 28160, South Korea
| | - Yang Woo Kwon
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Deagu 41061, South Korea
| | - Eui-Hwan Choi
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Deagu 41061, South Korea
| |
Collapse
|
8
|
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
|
9
|
Hengel SR, Oppenheimer KG, Smith CM, Schaich MA, Rein HL, Martino J, Darrah KE, Witham M, Ezekwenna OC, Burton KR, Van Houten B, Spies M, Bernstein KA. The human Shu complex promotes RAD51 activity by modulating RPA dynamics on ssDNA. Nat Commun 2024; 15:7197. [PMID: 39169038 PMCID: PMC11339404 DOI: 10.1038/s41467-024-51595-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/09/2024] [Indexed: 08/23/2024] Open
Abstract
Templated DNA repair that occurs during homologous recombination and replication stress relies on RAD51. RAD51 activity is positively regulated by BRCA2 and the RAD51 paralogs. The Shu complex is a RAD51 paralog-containing complex consisting of SWSAP1, SWS1, and SPIDR. We demonstrate that SWSAP1-SWS1 binds RAD51, maintains RAD51 filament stability, and enables strand exchange. Using single-molecule confocal fluorescence microscopy combined with optical tweezers, we show that SWSAP1-SWS1 decorates RAD51 filaments proficient for homologous recombination. We also find SWSAP1-SWS1 enhances RPA diffusion on ssDNA. Importantly, we show human sgSWSAP1 and sgSWS1 knockout cells are sensitive to pharmacological inhibition of PARP and APE1. Lastly, we identify cancer variants in SWSAP1 that alter Shu complex formation. Together, we show that SWSAP1-SWS1 stimulates RAD51-dependent high-fidelity repair and may be an important new cancer therapeutic target.
Collapse
Affiliation(s)
- Sarah R Hengel
- University of Pittsburgh, School of Medicine, Department of Pharmacology and Chemical Biology, UPMC-Hillman Cancer Center, Pittsburgh, PA, USA.
- Tufts University, Department of Biology, Medford, MA, USA.
| | - Katherine G Oppenheimer
- University of Pittsburgh, School of Medicine, Department of Pharmacology and Chemical Biology, UPMC-Hillman Cancer Center, Pittsburgh, PA, USA
| | - Chelsea M Smith
- University of Pittsburgh, School of Medicine, Department of Pharmacology and Chemical Biology, UPMC-Hillman Cancer Center, Pittsburgh, PA, USA
- University of North Carolina at Chapel Hill, Department of Pathology and Laboratory Medicine, Chapel Hill, NC, USA
| | - Matthew A Schaich
- University of Pittsburgh, School of Medicine, Department of Pharmacology and Chemical Biology, UPMC-Hillman Cancer Center, Pittsburgh, PA, USA
| | - Hayley L Rein
- University of Pittsburgh, School of Medicine, Department of Pharmacology and Chemical Biology, UPMC-Hillman Cancer Center, Pittsburgh, PA, USA
| | - Julieta Martino
- University of Pittsburgh, School of Medicine, Department of Pharmacology and Chemical Biology, UPMC-Hillman Cancer Center, Pittsburgh, PA, USA
- GeneDx, Gaithersburg, MD, USA
| | - Kristie E Darrah
- University of Pennsylvania School of Medicine, Penn Center for Genome Integrity, Department of Biochemistry and Biophysics, 421 Curie Boulevard, Philadelphia, PA, USA
| | - Maggie Witham
- Tufts University, Department of Biology, Medford, MA, USA
| | | | - Kyle R Burton
- Tufts University, Department of Biology, Medford, MA, USA
| | - Bennett Van Houten
- University of Pittsburgh, School of Medicine, Department of Pharmacology and Chemical Biology, UPMC-Hillman Cancer Center, Pittsburgh, PA, USA
| | - Maria Spies
- University of Iowa, Department of Biochemistry and Molecular Biology, Iowa City, IA, USA
| | - Kara A Bernstein
- University of Pittsburgh, School of Medicine, Department of Pharmacology and Chemical Biology, UPMC-Hillman Cancer Center, Pittsburgh, PA, USA.
- University of Pennsylvania School of Medicine, Penn Center for Genome Integrity, Department of Biochemistry and Biophysics, 421 Curie Boulevard, Philadelphia, PA, USA.
| |
Collapse
|
10
|
Wang X, Zhao X, Yu Z, Fan T, Guo Y, Liang J, Wang Y, Zhan J, Chen G, Zhou C, Zhang X, Li X, Chen X. Rtt105 stimulates Rad51-ssDNA assembly and orchestrates Rad51 and RPA actions to promote homologous recombination repair. Proc Natl Acad Sci U S A 2024; 121:e2402262121. [PMID: 39145931 PMCID: PMC11348298 DOI: 10.1073/pnas.2402262121] [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: 02/02/2024] [Accepted: 07/09/2024] [Indexed: 08/16/2024] Open
Abstract
Homologous recombination (HR) is essential for the maintenance of genome stability. During HR, Replication Protein A (RPA) rapidly coats the 3'-tailed single-strand DNA (ssDNA) generated by end resection. Then, the ssDNA-bound RPA must be timely replaced by Rad51 recombinase to form Rad51 nucleoprotein filaments that drive homology search and HR repair. How cells regulate Rad51 assembly dynamics and coordinate RPA and Rad51 actions to ensure proper HR remains poorly understood. Here, we identified that Rtt105, a Ty1 transposon regulator, acts to stimulate Rad51 assembly and orchestrate RPA and Rad51 actions during HR. We found that Rtt105 interacts with Rad51 in vitro and in vivo and restrains the adenosine 5' triphosphate (ATP) hydrolysis activity of Rad51. We showed that Rtt105 directly stimulates dynamic Rad51-ssDNA assembly, strand exchange, and D-loop formation in vitro. Notably, we found that Rtt105 physically regulates the binding of Rad51 and RPA to ssDNA via different motifs and that both regulations are necessary and epistatic in promoting Rad51 nucleation, strand exchange, and HR repair. Consequently, disrupting either of the interactions impaired HR and conferred DNA damage sensitivity, underscoring the importance of Rtt105 in orchestrating the actions of Rad51 and RPA. Our work reveals additional layers of mechanisms regulating Rad51 filament dynamics and the coordination of HR.
Collapse
Affiliation(s)
- Xuejie Wang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Department of Radiation Oncology, Renmin Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan430072, China
| | - Xiaocong Zhao
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Department of Radiation Oncology, Renmin Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan430072, China
| | - Zhengshi Yu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Department of Radiation Oncology, Renmin Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan430072, China
| | - Tianai Fan
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Department of Radiation Oncology, Renmin Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan430072, China
| | - Yunjing Guo
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Department of Radiation Oncology, Renmin Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan430072, China
| | - Jianqiang Liang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Department of Radiation Oncology, Renmin Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan430072, China
| | - Yanyan Wang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Department of Radiation Oncology, Renmin Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan430072, China
| | - Jingfei Zhan
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Department of Radiation Oncology, Renmin Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan430072, China
| | - Guifang Chen
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Department of Radiation Oncology, Renmin Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan430072, China
| | - Chun Zhou
- School of Public Health, Zhejiang University School of Medicine, Hangzhou310058, China
| | - Xinghua Zhang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Department of Radiation Oncology, Renmin Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan430072, China
| | - Xiangpan Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Department of Radiation Oncology, Renmin Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan430072, China
| | - Xuefeng Chen
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Department of Radiation Oncology, Renmin Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan430072, China
| |
Collapse
|
11
|
Norris J, Rogers L, Pytko K, Dannenberg R, Perreault S, Kaushik V, Kuppa S, Antony E, Hedglin M. Replication protein A dynamically re-organizes on primer/template junctions to permit DNA polymerase δ holoenzyme assembly and initiation of DNA synthesis. Nucleic Acids Res 2024; 52:7650-7664. [PMID: 38842913 PMCID: PMC11260492 DOI: 10.1093/nar/gkae475] [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: 08/28/2023] [Revised: 05/03/2024] [Accepted: 05/21/2024] [Indexed: 06/07/2024] Open
Abstract
DNA polymerase δ (pol δ) holoenzymes, comprised of pol δ and the processivity sliding clamp, PCNA, carry out DNA synthesis during lagging strand replication, initiation of leading strand replication, and the major DNA damage repair and tolerance pathways. Pol δ holoenzymes are assembled at primer/template (P/T) junctions and initiate DNA synthesis in a stepwise process involving the major single strand DNA (ssDNA)-binding protein complex, RPA, the processivity sliding clamp loader, RFC, PCNA and pol δ. During this process, the interactions of RPA, RFC and pol δ with a P/T junction all significantly overlap. A burning issue that has yet to be resolved is how these overlapping interactions are accommodated during this process. To address this, we design and utilize novel, ensemble FRET assays that continuously monitor the interactions of RPA, RFC, PCNA and pol δ with DNA as pol δ holoenzymes are assembled and initiate DNA synthesis. Results from the present study reveal that RPA remains engaged with P/T junctions throughout this process and the RPA•DNA complexes dynamically re-organize to allow successive binding of RFC and pol δ. These results have broad implications as they highlight and distinguish the functional consequences of dynamic RPA•DNA interactions in RPA-dependent DNA metabolic processes.
Collapse
Affiliation(s)
- Jessica L Norris
- Department of Chemistry, The Pennsylvania State University, University Park, PA 16802, USA
| | - Lindsey O Rogers
- Department of Chemistry, The Pennsylvania State University, University Park, PA 16802, USA
| | - Kara G Pytko
- Department of Chemistry, The Pennsylvania State University, University Park, PA 16802, USA
| | - Rachel L Dannenberg
- Department of Chemistry, The Pennsylvania State University, University Park, PA 16802, USA
| | - Samuel Perreault
- Department of Chemistry, The Pennsylvania State University, University Park, PA 16802, USA
| | - Vikas Kaushik
- The Saint Louis University School of Medicine, Department of Biochemistry and Molecular Biology, St. Louis, MO 63104, USA
| | - Sahiti Kuppa
- The Saint Louis University School of Medicine, Department of Biochemistry and Molecular Biology, St. Louis, MO 63104, USA
| | - Edwin Antony
- The Saint Louis University School of Medicine, Department of Biochemistry and Molecular Biology, St. Louis, MO 63104, USA
| | - Mark Hedglin
- Department of Chemistry, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
12
|
Son MY, Belan O, Spirek M, Cibulka J, Nikulenkov F, Kim YY, Hwang S, Myung K, Montagna C, Kim TM, Krejci L, Hasty P. RAD51 separation of function mutation disables replication fork maintenance but preserves DSB repair. iScience 2024; 27:109524. [PMID: 38577109 PMCID: PMC10993188 DOI: 10.1016/j.isci.2024.109524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 03/01/2023] [Accepted: 03/14/2024] [Indexed: 04/06/2024] Open
Abstract
Homologous recombination (HR) protects replication forks (RFs) and repairs DNA double-strand breaks (DSBs). Within HR, BRCA2 regulates RAD51 via two interaction regions: the BRC repeats to form filaments on single-stranded DNA and exon 27 (Ex27) to stabilize the filament. Here, we identified a RAD51 S181P mutant that selectively disrupted the RAD51-Ex27 association while maintaining interaction with BRC repeat and proficiently forming filaments capable of DNA binding and strand invasion. Interestingly, RAD51 S181P was defective for RF protection/restart but proficient for DSB repair. Our data suggest that Ex27-mediated stabilization of RAD51 filaments is required for the protection of RFs, while it seems dispensable for the repair of DSBs.
Collapse
Affiliation(s)
- Mi Young Son
- Department of Molecular Medicine, The Barshop Institute for Longevity and Aging Studies, The Cancer Therapy Research Center, UT Health San Antonio, San Antonio, TX 78229, USA
| | - Ondrej Belan
- Department of Biology, Masaryk University, 625 00 Brno, Czech Republic
| | - Mario Spirek
- Department of Biology, Masaryk University, 625 00 Brno, Czech Republic
- National Centre for Biomolecular Research, Masaryk University, 625 00 Brno, Czech Republic
| | - Jakub Cibulka
- Department of Biology, Masaryk University, 625 00 Brno, Czech Republic
| | - Fedor Nikulenkov
- Department of Biology, Masaryk University, 625 00 Brno, Czech Republic
| | - You Young Kim
- Center for Genomic Integrity Institute for Basic Science (IBS), Ulsan 44919, Republic of Korea
| | - Sunyoung Hwang
- Center for Genomic Integrity Institute for Basic Science (IBS), Ulsan 44919, Republic of Korea
| | - Kyungjae Myung
- Center for Genomic Integrity Institute for Basic Science (IBS), Ulsan 44919, Republic of Korea
| | - Cristina Montagna
- Department of Genetics, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, USA
| | - Tae Moon Kim
- Department of Molecular Medicine, The Barshop Institute for Longevity and Aging Studies, The Cancer Therapy Research Center, UT Health San Antonio, San Antonio, TX 78229, USA
- Center for Genomic Integrity Institute for Basic Science (IBS), Ulsan 44919, Republic of Korea
| | - Lumir Krejci
- Department of Biology, Masaryk University, 625 00 Brno, Czech Republic
- National Centre for Biomolecular Research, Masaryk University, 625 00 Brno, Czech Republic
| | - Paul Hasty
- Department of Molecular Medicine, The Barshop Institute for Longevity and Aging Studies, The Cancer Therapy Research Center, UT Health San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
13
|
Balakrishnan S, Adolph M, Tsai MS, Akizuki T, Gallagher K, Cortez D, Chazin WJ. Structure of RADX and mechanism for regulation of RAD51 nucleofilaments. Proc Natl Acad Sci U S A 2024; 121:e2316491121. [PMID: 38466836 PMCID: PMC10962997 DOI: 10.1073/pnas.2316491121] [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: 09/27/2023] [Accepted: 02/07/2024] [Indexed: 03/13/2024] Open
Abstract
Replication fork reversal is a fundamental process required for resolution of encounters with DNA damage. A key step in the stabilization and eventual resolution of reversed forks is formation of RAD51 nucleoprotein filaments on exposed single strand DNA (ssDNA). To avoid genome instability, RAD51 filaments are tightly controlled by a variety of positive and negative regulators. RADX (RPA-related RAD51-antagonist on the X chromosome) is a recently discovered negative regulator that binds tightly to ssDNA, directly interacts with RAD51, and regulates replication fork reversal and stabilization in a context-dependent manner. Here, we present a structure-based investigation of RADX's mechanism of action. Mass photometry experiments showed that RADX forms multiple oligomeric states in a concentration-dependent manner, with a predominance of trimers in the presence of ssDNA. The structure of RADX, which has no structurally characterized orthologs, was determined ab initio by cryo-electron microscopy (cryo-EM) from maps in the 2 to 4 Å range. The structure reveals the molecular basis for RADX oligomerization and the coupled multi-valent binding of ssDNA binding. The interaction of RADX with RAD51 filaments was imaged by negative stain EM, which showed a RADX oligomer at the end of filaments. Based on these results, we propose a model in which RADX functions by capping and restricting the end of RAD51 filaments.
Collapse
Affiliation(s)
- Swati Balakrishnan
- Center for Structural Biology, Vanderbilt University, Nashville, TN37240
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN37237
| | - Madison Adolph
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN37237
| | - Miaw-Sheue Tsai
- Biological Systems and Bioengineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA94720
| | - Tae Akizuki
- Center for Structural Biology, Vanderbilt University, Nashville, TN37240
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN37237
| | - Kaitlyn Gallagher
- Center for Structural Biology, Vanderbilt University, Nashville, TN37240
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN37237
| | - David Cortez
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN37237
| | - Walter J. Chazin
- Center for Structural Biology, Vanderbilt University, Nashville, TN37240
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN37237
- Department of Chemistry, Vanderbilt University, Nashville, TN37235
| |
Collapse
|
14
|
Ye Z, Xu S, Shi Y, Cheng X, Zhang Y, Roy S, Namjoshi S, Longo MA, Link TM, Schlacher K, Peng G, Yu D, Wang B, Tainer JA, Ahmed Z. GRB2 stabilizes RAD51 at reversed replication forks suppressing genomic instability and innate immunity against cancer. Nat Commun 2024; 15:2132. [PMID: 38459011 PMCID: PMC10923831 DOI: 10.1038/s41467-024-46283-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 02/20/2024] [Indexed: 03/10/2024] Open
Abstract
Growth factor receptor-bound protein 2 (GRB2) is a cytoplasmic adapter for tyrosine kinase signaling and a nuclear adapter for homology-directed-DNA repair. Here we find nuclear GRB2 protects DNA at stalled replication forks from MRE11-mediated degradation in the BRCA2 replication fork protection axis. Mechanistically, GRB2 binds and inhibits RAD51 ATPase activity to stabilize RAD51 on stalled replication forks. In GRB2-depleted cells, PARP inhibitor (PARPi) treatment releases DNA fragments from stalled forks into the cytoplasm that activate the cGAS-STING pathway to trigger pro-inflammatory cytokine production. Moreover in a syngeneic mouse metastatic ovarian cancer model, GRB2 depletion in the context of PARPi treatment reduced tumor burden and enabled high survival consistent with immune suppression of cancer growth. Collective findings unveil GRB2 function and mechanism for fork protection in the BRCA2-RAD51-MRE11 axis and suggest GRB2 as a potential therapeutic target and an enabling predictive biomarker for patient selection for PARPi and immunotherapy combination.
Collapse
Affiliation(s)
- Zu Ye
- Departments of Molecular and Cellular Oncology and Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Shengfeng Xu
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yin Shi
- Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xueqian Cheng
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yuan Zhang
- Departments of Molecular and Cellular Oncology and Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Sunetra Roy
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Sarita Namjoshi
- Departments of Molecular and Cellular Oncology and Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Michael A Longo
- Departments of Molecular and Cellular Oncology and Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Todd M Link
- Departments of Molecular and Cellular Oncology and Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Katharina Schlacher
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Guang Peng
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Dihua Yu
- Departments of Molecular and Cellular Oncology and Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Bin Wang
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - John A Tainer
- Departments of Molecular and Cellular Oncology and Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Zamal Ahmed
- Departments of Molecular and Cellular Oncology and Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
15
|
Hengel SR, Oppenheimer K, Smith C, Schaich MA, Rein HL, Martino J, Darrah K, Ezekwenna O, Burton K, Van Houten B, Spies M, Bernstein KA. The human Shu complex promotes RAD51 activity by modulating RPA dynamics on ssDNA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.14.580393. [PMID: 38405734 PMCID: PMC10888808 DOI: 10.1101/2024.02.14.580393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Templated DNA repair that occurs during homologous recombination and replication stress relies on RAD51. RAD51 activity is positively regulated by BRCA2 and the RAD51 paralogs. The Shu complex is a RAD51 paralog-containing complex consisting of SWSAP1 and SWS1. We demonstrate that SWSAP1-SWS1 binds RAD51, maintains RAD51 filament stability, and enables strand exchange. Using single molecule confocal fluorescence microscopy combined with optical tweezers, we show that SWSAP1-SWS1 decorates RAD51 filaments proficient for homologous recombination. We also find SWSAP1-SWS1 enhances RPA diffusion on ssDNA. Importantly, we show human sgSWSAP1 and sgSWS1 knockout cells are sensitive to pharmacological inhibition of PARP and APE1. Lastly, we identify cancer variants in SWSAP1 that alter SWS1 complex formation. Together, we show that SWSAP1-SWS1 stimulates RAD51-dependent high-fidelity repair and may be an important new cancer therapeutic target.
Collapse
|
16
|
Nifontova G, Charlier C, Ayadi N, Fleury F, Karaulov A, Sukhanova A, Nabiev I. Photonic Crystal Surface Mode Real-Time Imaging of RAD51 DNA Repair Protein Interaction with the ssDNA Substrate. BIOSENSORS 2024; 14:43. [PMID: 38248420 PMCID: PMC10813746 DOI: 10.3390/bios14010043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/08/2024] [Accepted: 01/11/2024] [Indexed: 01/23/2024]
Abstract
Photonic crystals (PCs) are promising tools for label-free sensing in drug discovery screening, diagnostics, and analysis of ligand-receptor interactions. Imaging of PC surface modes has emerged as a novel approach to the detection of multiple binding events at the sensor surface. PC surface modification and decoration with recognition units yield an interface providing the highly sensitive detection of cancer biomarkers, antibodies, and oligonucleotides. The RAD51 protein plays a central role in DNA repair via the homologous recombination pathway. This recombinase is essential for the genome stability and its overexpression is often correlated with aggressive cancer. RAD51 is therefore a potential target in the therapeutic strategy for cancer. Here, we report the designing of a PC-based array sensor for real-time monitoring of oligonucleotide-RAD51 recruitment by means of surface mode imaging and validation of the concept of this approach. Our data demonstrate that the designed biosensor ensures the highly sensitive multiplexed analysis of association-dissociation events and detection of the biomarker of DNA damage using a microfluidic PC array. The obtained results highlight the potential of the developed technique for testing the functionality of candidate drugs, discovering new molecular targets and drug entities. This paves the way to further adaption and bioanalytical use of the biosensor for high-content screening to identify new DNA repair inhibitor drugs targeting the RAD51 nucleoprotein filament or to discover new molecular targets.
Collapse
Affiliation(s)
- Galina Nifontova
- Laboratoire de Recherche en Nanosciences, LRN-EA4682, Structure Fédérative de Recherche Cap Santé, UFR de Pharmacie, Université de Reims Champagne-Ardenne, 51100 Reims, France;
| | - Cathy Charlier
- Nantes Université, CNRS, US2B, UMR 6286, IMPACT Platform and SFR Bonamy, 44000 Nantes, France;
| | - Nizar Ayadi
- Nantes Université, CNRS, US2B, UMR 6286, DNA Repair Group, 44000 Nantes, France; (N.A.); (F.F.)
| | - Fabrice Fleury
- Nantes Université, CNRS, US2B, UMR 6286, DNA Repair Group, 44000 Nantes, France; (N.A.); (F.F.)
| | - Alexander Karaulov
- Department of Clinical Immunology and Allergology, Institute of Molecular Medicine, Sechenov First Moscow State Medical University (Sechenov University), 119146 Moscow, Russia;
| | - Alyona Sukhanova
- Laboratoire de Recherche en Nanosciences, LRN-EA4682, Structure Fédérative de Recherche Cap Santé, UFR de Pharmacie, Université de Reims Champagne-Ardenne, 51100 Reims, France;
- Life Improvement by Future Technologies (LIFT) Center, 143025 Moscow, Russia
| | - Igor Nabiev
- Laboratoire de Recherche en Nanosciences, LRN-EA4682, Structure Fédérative de Recherche Cap Santé, UFR de Pharmacie, Université de Reims Champagne-Ardenne, 51100 Reims, France;
- Department of Clinical Immunology and Allergology, Institute of Molecular Medicine, Sechenov First Moscow State Medical University (Sechenov University), 119146 Moscow, Russia;
- Life Improvement by Future Technologies (LIFT) Center, 143025 Moscow, Russia
- Laboratory of Nano-Bioengineering, National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), 115522 Moscow, Russia
| |
Collapse
|
17
|
Nasheuer HP, Meaney AM, Hulshoff T, Thiele I, Onwubiko NO. Replication Protein A, the Main Eukaryotic Single-Stranded DNA Binding Protein, a Focal Point in Cellular DNA Metabolism. Int J Mol Sci 2024; 25:588. [PMID: 38203759 PMCID: PMC10779431 DOI: 10.3390/ijms25010588] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
Replication protein A (RPA) is a heterotrimeric protein complex and the main single-stranded DNA (ssDNA)-binding protein in eukaryotes. RPA has key functions in most of the DNA-associated metabolic pathways and DNA damage signalling. Its high affinity for ssDNA helps to stabilise ssDNA structures and protect the DNA sequence from nuclease attacks. RPA consists of multiple DNA-binding domains which are oligonucleotide/oligosaccharide-binding (OB)-folds that are responsible for DNA binding and interactions with proteins. These RPA-ssDNA and RPA-protein interactions are crucial for DNA replication, DNA repair, DNA damage signalling, and the conservation of the genetic information of cells. Proteins such as ATR use RPA to locate to regions of DNA damage for DNA damage signalling. The recruitment of nucleases and DNA exchange factors to sites of double-strand breaks are also an important RPA function to ensure effective DNA recombination to correct these DNA lesions. Due to its high affinity to ssDNA, RPA's removal from ssDNA is of central importance to allow these metabolic pathways to proceed, and processes to exchange RPA against downstream factors are established in all eukaryotes. These faceted and multi-layered functions of RPA as well as its role in a variety of human diseases will be discussed.
Collapse
Affiliation(s)
- Heinz Peter Nasheuer
- Centre for Chromosome Biology, School of Biological and Chemical Sciences, Biochemistry, University of Galway, H91 TK33 Galway, Ireland
| | - Anna Marie Meaney
- Centre for Chromosome Biology, School of Biological and Chemical Sciences, Biochemistry, University of Galway, H91 TK33 Galway, Ireland
| | - Timothy Hulshoff
- Molecular Systems Physiology Group, School of Biological and Chemical Sciences, University of Galway, H91 TK33 Galway, Ireland
| | - Ines Thiele
- Molecular Systems Physiology Group, School of Biological and Chemical Sciences, University of Galway, H91 TK33 Galway, Ireland
| | - Nichodemus O. Onwubiko
- Centre for Chromosome Biology, School of Biological and Chemical Sciences, Biochemistry, University of Galway, H91 TK33 Galway, Ireland
| |
Collapse
|
18
|
Adolph MB, Garje AS, Balakrishnan S, Morati F, Modesti M, Chazin WJ, Cortez D. CRISPR-dependent Base Editing Screens Identify Separation of Function Mutants of RADX with Altered RAD51 Regulatory Activity. J Mol Biol 2023; 435:168236. [PMID: 37572935 PMCID: PMC10530557 DOI: 10.1016/j.jmb.2023.168236] [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: 06/19/2023] [Revised: 08/01/2023] [Accepted: 08/04/2023] [Indexed: 08/14/2023]
Abstract
RAD51 forms nucleoprotein filaments to promote homologous recombination, replication fork reversal, and fork protection. Numerous factors regulate the stability of these filaments and improper regulation leads to genomic instability and ultimately disease including cancer. RADX is a single stranded DNA binding protein that modulates RAD51 filament stability. Here, we utilize a CRISPR-dependent base editing screen to tile mutations across RADX to delineate motifs required for RADX function. We identified separation of function mutants of RADX that bind DNA and RAD51 but have a reduced ability to stimulate its ATP hydrolysis activity. Cells expressing these RADX mutants accumulate RAD51 on chromatin, exhibit replication defects, have reduced growth, accumulate DNA damage, and are hypersensitive to DNA damage and replication stress. These results indicate that RADX must promote RAD51 ATP turnover to regulate RAD51 and genome stability during DNA replication.
Collapse
Affiliation(s)
- Madison B Adolph
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Atharv S Garje
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Swati Balakrishnan
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA; Center for Structural Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Florian Morati
- Cancer Research Center of Marseille, Department of Genome Integrity, CNRS UMR7258, Inserm U1068, Institut Paoli-Calmettes, Aix Marseille Univ, Marseille, France
| | - Mauro Modesti
- Cancer Research Center of Marseille, Department of Genome Integrity, CNRS UMR7258, Inserm U1068, Institut Paoli-Calmettes, Aix Marseille Univ, Marseille, France
| | - Walter J Chazin
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA; Center for Structural Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - David Cortez
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
19
|
Luo SC, Yeh MC, Lien YH, Yeh HY, Siao HL, Tu IP, Chi P, Ho MC. A RAD51-ADP double filament structure unveils the mechanism of filament dynamics in homologous recombination. Nat Commun 2023; 14:4993. [PMID: 37591853 PMCID: PMC10435448 DOI: 10.1038/s41467-023-40672-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 08/04/2023] [Indexed: 08/19/2023] Open
Abstract
ATP-dependent RAD51 recombinases play an essential role in eukaryotic homologous recombination by catalyzing a four-step process: 1) formation of a RAD51 single-filament assembly on ssDNA in the presence of ATP, 2) complementary DNA strand-exchange, 3) ATP hydrolysis transforming the RAD51 filament into an ADP-bound disassembly-competent state, and 4) RAD51 disassembly to provide access for DNA repairing enzymes. Of these steps, filament dynamics between the ATP- and ADP-bound states, and the RAD51 disassembly mechanism, are poorly understood due to the lack of near-atomic-resolution information of the ADP-bound RAD51-DNA filament structure. We report the cryo-EM structure of ADP-bound RAD51-DNA filaments at 3.1 Å resolution, revealing a unique RAD51 double-filament that wraps around ssDNA. Structural analysis, supported by ATP-chase and time-resolved cryo-EM experiments, reveals a collapsing mechanism involving two four-protomer movements along ssDNA for mechanical transition between RAD51 single- and double-filament without RAD51 dissociation. This mechanism enables elastic change of RAD51 filament length during structural transitions between ATP- and ADP-states.
Collapse
Affiliation(s)
- Shih-Chi Luo
- Institute of Biological Chemistry, Academia Sinica, 11529, Taipei, Taiwan
| | - Min-Chi Yeh
- Institute of Biological Chemistry, Academia Sinica, 11529, Taipei, Taiwan
- Institute of Biochemical Sciences, National Taiwan University, 10617, Taipei, Taiwan
| | - Yu-Hsiang Lien
- Institute of Statistical Science, Academia Sinica, 11529, Taipei, Taiwan
| | - Hsin-Yi Yeh
- Institute of Biochemical Sciences, National Taiwan University, 10617, Taipei, Taiwan
| | - Huei-Lun Siao
- Institute of Statistical Science, Academia Sinica, 11529, Taipei, Taiwan
| | - I-Ping Tu
- Institute of Statistical Science, Academia Sinica, 11529, Taipei, Taiwan
| | - Peter Chi
- Institute of Biological Chemistry, Academia Sinica, 11529, Taipei, Taiwan
- Institute of Biochemical Sciences, National Taiwan University, 10617, Taipei, Taiwan
| | - Meng-Chiao Ho
- Institute of Biological Chemistry, Academia Sinica, 11529, Taipei, Taiwan.
- Institute of Biochemical Sciences, National Taiwan University, 10617, Taipei, Taiwan.
| |
Collapse
|
20
|
Belan O, Greenhough L, Kuhlen L, Anand R, Kaczmarczyk A, Gruszka DT, Yardimci H, Zhang X, Rueda DS, West SC, Boulton SJ. Visualization of direct and diffusion-assisted RAD51 nucleation by full-length human BRCA2 protein. Mol Cell 2023; 83:2925-2940.e8. [PMID: 37499663 PMCID: PMC7615647 DOI: 10.1016/j.molcel.2023.06.031] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 06/09/2023] [Accepted: 06/26/2023] [Indexed: 07/29/2023]
Abstract
Homologous recombination (HR) is essential for error-free repair of DNA double-strand breaks, perturbed replication forks (RFs), and post-replicative single-stranded DNA (ssDNA) gaps. To initiate HR, the recombination mediator and tumor suppressor protein BRCA2 facilitates nucleation of RAD51 on ssDNA prior to stimulation of RAD51 filament growth by RAD51 paralogs. Although ssDNA binding by BRCA2 has been implicated in RAD51 nucleation, the function of double-stranded DNA (dsDNA) binding by BRCA2 remains unclear. Here, we exploit single-molecule (SM) imaging to visualize BRCA2-mediated RAD51 nucleation in real time using purified proteins. We report that BRCA2 nucleates and stabilizes RAD51 on ssDNA either directly or through an unappreciated diffusion-assisted delivery mechanism involving binding to and sliding along dsDNA, which requires the cooperative action of multiple dsDNA-binding modules in BRCA2. Collectively, our work reveals two distinct mechanisms of BRCA2-dependent RAD51 loading onto ssDNA, which we propose are critical for its diverse functions in maintaining genome stability and cancer suppression.
Collapse
Affiliation(s)
- Ondrej Belan
- DSB Repair Metabolism Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Luke Greenhough
- DNA Recombination and Repair Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Lucas Kuhlen
- Section of Structural Biology, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, South Kensington, London SW7 2AZ, UK
| | - Roopesh Anand
- DSB Repair Metabolism Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Artur Kaczmarczyk
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London W12 0NN, UK; Single Molecule Imaging Group, MRC-London Institute of Medical Sciences, London W12 0NN, UK
| | - Dominika T Gruszka
- Single Molecule Imaging of Genome Duplication and Maintenance Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Hasan Yardimci
- Single Molecule Imaging of Genome Duplication and Maintenance Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Xiaodong Zhang
- Section of Structural Biology, Faculty of Medicine, Imperial College London, Sir Alexander Fleming Building, South Kensington, London SW7 2AZ, UK
| | - David S Rueda
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London W12 0NN, UK; Single Molecule Imaging Group, MRC-London Institute of Medical Sciences, London W12 0NN, UK
| | - Stephen C West
- DNA Recombination and Repair Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Simon J Boulton
- DSB Repair Metabolism Laboratory, The Francis Crick Institute, London NW1 1AT, UK.
| |
Collapse
|
21
|
Ding J, Li X, Shen J, Zhao Y, Zhong S, Lai L, Niu H, Qi Z. ssDNA accessibility of Rad51 is regulated by orchestrating multiple RPA dynamics. Nat Commun 2023; 14:3864. [PMID: 37391417 PMCID: PMC10313831 DOI: 10.1038/s41467-023-39579-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 06/20/2023] [Indexed: 07/02/2023] Open
Abstract
The eukaryotic single-stranded DNA (ssDNA)-binding protein Replication Protein A (RPA) plays a crucial role in various DNA metabolic pathways, including DNA replication and repair, by dynamically associating with ssDNA. While the binding of a single RPA molecule to ssDNA has been thoroughly studied, the accessibility of ssDNA is largely governed by the bimolecular behavior of RPA, the biophysical nature of which remains unclear. In this study, we develop a three-step low-complexity ssDNA Curtains method, which, when combined with biochemical assays and a Markov chain model in non-equilibrium physics, allow us to decipher the dynamics of multiple RPA binding to long ssDNA. Interestingly, our results suggest that Rad52, the mediator protein, can modulate the ssDNA accessibility of Rad51, which is nucleated on RPA coated ssDNA through dynamic ssDNA exposure between neighboring RPA molecules. We find that this process is controlled by the shifting between the protection mode and action mode of RPA ssDNA binding, where tighter RPA spacing and lower ssDNA accessibility are favored under RPA protection mode, which can be facilitated by the Rfa2 WH domain and inhibited by Rad52 RPA interaction.
Collapse
Affiliation(s)
- Jiawei Ding
- Center for Quantitative Biology, Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Xiangting Li
- Department of Computational Medicine, University of California, Los Angeles, CA, USA
| | - Jiangchuan Shen
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN, USA
| | - Yiling Zhao
- Center for Quantitative Biology, Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Shuchen Zhong
- BNLMS, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Luhua Lai
- Center for Quantitative Biology, Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- BNLMS, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Hengyao Niu
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN, USA.
| | - Zhi Qi
- Center for Quantitative Biology, Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
| |
Collapse
|
22
|
Knadler C, Graham V W, Rolfsmeier M, Haseltine CA. Divalent metal cofactors differentially modulate RadA-mediated strand invasion and exchange in Saccharolobus solfataricus. Biosci Rep 2023; 43:BSR20221807. [PMID: 36601994 PMCID: PMC9950535 DOI: 10.1042/bsr20221807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/20/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
Central to the universal process of recombination, RecA family proteins form nucleoprotein filaments to catalyze production of heteroduplex DNA between substrate ssDNAs and template dsDNAs. ATP binding assists the filament in assuming the necessary conformation for forming heteroduplex DNA, but hydrolysis is not required. ATP hydrolysis has two identified roles which are not universally conserved: promotion of filament dissociation and enhancing flexibility of the filament. In this work, we examine ATP utilization of the RecA family recombinase SsoRadA from Saccharolobus solfataricus to determine its function in recombinase-mediated heteroduplex DNA formation. Wild-type SsoRadA protein and two ATPase mutant proteins were evaluated for the effects of three divalent metal cofactors. We found that unlike other archaeal RadA proteins, SsoRadA-mediated strand exchange is not enhanced by Ca2+. Instead, the S. solfataricus recombinase can utilize Mn2+ to stimulate strand invasion and reduce ADP-binding stability. Additionally, reduction of SsoRadA ATPase activity by Walker Box mutation or cofactor alteration resulted in a loss of large, complete strand exchange products. Depletion of ADP was found to improve initial strand invasion but also led to a similar loss of large strand exchange events. Our results indicate that overall, SsoRadA is distinct in its use of divalent cofactors but its activity with Mn2+ shows similarity to human RAD51 protein with Ca2+.
Collapse
Affiliation(s)
- Corey J. Knadler
- School of Molecular Biosciences, Washington State University, Pullman, Washington 99164-7520, U.S.A
| | - William J. Graham V
- School of Molecular Biosciences, Washington State University, Pullman, Washington 99164-7520, U.S.A
| | - Michael L. Rolfsmeier
- School of Molecular Biosciences, Washington State University, Pullman, Washington 99164-7520, U.S.A
| | - Cynthia A. Haseltine
- School of Molecular Biosciences, Washington State University, Pullman, Washington 99164-7520, U.S.A
| |
Collapse
|
23
|
Altmannova V, Spirek M, Orlic L, Jēkabsons A, Clarence T, Henggeler A, Mlcouskova J, Chaleil RA, Matos J, Krejci L. The role of bivalent ions in the regulation of D-loop extension mediated by DMC1 during meiotic recombination. iScience 2022; 25:105439. [PMID: 36388968 PMCID: PMC9641244 DOI: 10.1016/j.isci.2022.105439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 09/06/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022] Open
Abstract
During meiosis, programmed DNA double-strand breaks (DSBs) are repaired by homologous recombination. DMC1, a conserved recombinase, plays a central role in this process. DMC1 promotes DNA strand exchange between homologous chromosomes, thus creating the physical linkage between them. Its function is regulated not only by several accessory proteins but also by bivalent ions. Here, we show that whereas calcium ions in the presence of ATP cause a conformational change within DMC1, stimulating its DNA binding and D-loop formation, they inhibit the extension of the invading strand within the D-loop. Based on structural studies, we have generated mutants of two highly conserved amino acids - E162 and D317 - in human DMC1, which are deficient in calcium regulation. In vivo studies of their yeast homologues further showed that they exhibit severe defects in meiosis, thus emphasizing the importance of calcium ions in the regulation of DMC1 function and meiotic recombination.
Collapse
Affiliation(s)
- Veronika Altmannova
- Department of Biology, Masaryk University, Brno 62500, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, Brno 65691, Czech Republic
| | - Mario Spirek
- Department of Biology, Masaryk University, Brno 62500, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, Brno 65691, Czech Republic
| | - Lucija Orlic
- Max Perutz Labs, University of Vienna, Dr. Bohr-Gasse 9 1030 Vienna, Austria
| | - Atis Jēkabsons
- Department of Biology, Masaryk University, Brno 62500, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, Brno 65691, Czech Republic
| | - Tereza Clarence
- Biomolecular Modelling Laboratory, The Francis Crick Institute, London, UK
| | - Adrian Henggeler
- Max Perutz Labs, University of Vienna, Dr. Bohr-Gasse 9 1030 Vienna, Austria
| | - Jarmila Mlcouskova
- International Clinical Research Center, St. Anne’s University Hospital, Brno 65691, Czech Republic
| | | | - Joao Matos
- Max Perutz Labs, University of Vienna, Dr. Bohr-Gasse 9 1030 Vienna, Austria
| | - Lumir Krejci
- Department of Biology, Masaryk University, Brno 62500, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, Brno 65691, Czech Republic
- National Center for Biomolecular Research, Masaryk University, Brno 62500, Czech Republic
| |
Collapse
|
24
|
Czajkowski D, Szmyd R, Gee HE. Impact of DNA damage response defects in cancer cells on response to immunotherapy and radiotherapy. J Med Imaging Radiat Oncol 2022; 66:546-559. [PMID: 35460184 PMCID: PMC9321602 DOI: 10.1111/1754-9485.13413] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/03/2022] [Accepted: 04/06/2022] [Indexed: 11/30/2022]
Abstract
The DNA damage response (DDR) is a complex set of downstream pathways triggered in response to DNA damage to maintain genomic stability. Many tumours exhibit mutations which inactivate components of the DDR, making them prone to the accumulation of DNA defects. These can both facilitate the development of tumours and provide potential targets for novel therapeutic interventions. The inhibition of the DDR has been shown to induce radiosensitivity in certain cancers, rendering them susceptible to treatment with radiotherapy and improving the therapeutic window. Moreover, DDR defects are a strong predictor of patient response to immune checkpoint inhibition (ICI). The ability to target the DDR selectively has the potential to expand the tumour neoantigen repertoire, thus increasing tumour immunogenicity and facilitating a CD8+ T and NK cell response against cancer cells. Combinatorial approaches, which seek to integrate DDR inhibition with radiotherapy and immunotherapy, have shown promise in early trials. Further studies are necessary to understand these synergies and establish reliable biomarkers.
Collapse
Affiliation(s)
| | - Radosław Szmyd
- Genome Integrity Unit, Children's Medical Research Institute, University of Sydney, Sydney, New South Wales, Australia.,Sydney West Radiation Oncology Network, Crown Princess Mary Cancer Centre Westmead, Sydney, New South Wales, Australia
| | - Harriet E Gee
- University of Sydney, Sydney, New South Wales, Australia.,Genome Integrity Unit, Children's Medical Research Institute, University of Sydney, Sydney, New South Wales, Australia.,Sydney West Radiation Oncology Network, Crown Princess Mary Cancer Centre Westmead, Sydney, New South Wales, Australia
| |
Collapse
|
25
|
Ali A, Xiao W, Babar ME, Bi Y. Double-Stranded Break Repair in Mammalian Cells and Precise Genome Editing. Genes (Basel) 2022; 13:genes13050737. [PMID: 35627122 PMCID: PMC9142082 DOI: 10.3390/genes13050737] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 12/16/2022] Open
Abstract
In mammalian cells, double-strand breaks (DSBs) are repaired predominantly by error-prone non-homologous end joining (NHEJ), but less prevalently by error-free template-dependent homologous recombination (HR). DSB repair pathway selection is the bedrock for genome editing. NHEJ results in random mutations when repairing DSB, while HR induces high-fidelity sequence-specific variations, but with an undesirable low efficiency. In this review, we first discuss the latest insights into the action mode of NHEJ and HR in a panoramic view. We then propose the future direction of genome editing by virtue of these advancements. We suggest that by switching NHEJ to HR, full fidelity genome editing and robust gene knock-in could be enabled. We also envision that RNA molecules could be repurposed by RNA-templated DSB repair to mediate precise genetic editing.
Collapse
Affiliation(s)
- Akhtar Ali
- Key Laboratory of Animal Embryo and Molecular Breeding of Hubei Province, Institute of Animal Science and Veterinary Medicine, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; (A.A.); (W.X.)
- Department of Biotechnology, Virtual University of Pakistan, Lahore 54000, Pakistan
| | - Wei Xiao
- Key Laboratory of Animal Embryo and Molecular Breeding of Hubei Province, Institute of Animal Science and Veterinary Medicine, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; (A.A.); (W.X.)
| | - Masroor Ellahi Babar
- The University of Agriculture Dera Ismail Khan, Dera Ismail Khan 29220, Pakistan;
| | - Yanzhen Bi
- Key Laboratory of Animal Embryo and Molecular Breeding of Hubei Province, Institute of Animal Science and Veterinary Medicine, Hubei Academy of Agricultural Sciences, Wuhan 430064, China; (A.A.); (W.X.)
- Correspondence: ; Tel.: +86-151-0714-8708
| |
Collapse
|
26
|
Anand R, Buechelmaier E, Belan O, Newton M, Vancevska A, Kaczmarczyk A, Takaki T, Rueda DS, Powell SN, Boulton SJ. HELQ is a dual-function DSB repair enzyme modulated by RPA and RAD51. Nature 2022; 601:268-273. [PMID: 34937945 PMCID: PMC8755542 DOI: 10.1038/s41586-021-04261-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 11/17/2021] [Indexed: 02/04/2023]
Abstract
DNA double-stranded breaks (DSBs) are deleterious lesions, and their incorrect repair can drive cancer development1. HELQ is a superfamily 2 helicase with 3' to 5' polarity, and its disruption in mice confers germ cells loss, infertility and increased predisposition to ovarian and pituitary tumours2-4. At the cellular level, defects in HELQ result in hypersensitivity to cisplatin and mitomycin C, and persistence of RAD51 foci after DNA damage3,5. Notably, HELQ binds to RPA and the RAD51-paralogue BCDX2 complex, but the relevance of these interactions and how HELQ functions in DSB repair remains unclear3,5,6. Here we show that HELQ helicase activity and a previously unappreciated DNA strand annealing function are differentially regulated by RPA and RAD51. Using biochemistry analyses and single-molecule imaging, we establish that RAD51 forms a complex with and strongly stimulates HELQ as it translocates during DNA unwinding. By contrast, RPA inhibits DNA unwinding by HELQ but strongly stimulates DNA strand annealing. Mechanistically, we show that HELQ possesses an intrinsic ability to capture RPA-bound DNA strands and then displace RPA to facilitate annealing of complementary sequences. Finally, we show that HELQ deficiency in cells compromises single-strand annealing and microhomology-mediated end-joining pathways and leads to bias towards long-tract gene conversion tracts during homologous recombination. Thus, our results implicate HELQ in multiple arms of DSB repair through co-factor-dependent modulation of intrinsic translocase and DNA strand annealing activities.
Collapse
Affiliation(s)
- Roopesh Anand
- DSB Repair Metabolism Laboratory, The Francis Crick Institute, London, UK
| | - Erika Buechelmaier
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Ondrej Belan
- DSB Repair Metabolism Laboratory, The Francis Crick Institute, London, UK
| | - Matthew Newton
- DSB Repair Metabolism Laboratory, The Francis Crick Institute, London, UK
| | | | - Artur Kaczmarczyk
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
- Single Molecule Imaging Group, MRC-London Institute of Medical Sciences, London, UK
| | - Tohru Takaki
- DSB Repair Metabolism Laboratory, The Francis Crick Institute, London, UK
| | - David S 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.
| | - Simon N Powell
- Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Simon J Boulton
- DSB Repair Metabolism Laboratory, The Francis Crick Institute, London, UK.
| |
Collapse
|
27
|
Špírek M, Taylor MRG, Belan O, Boulton SJ, Krejci L. Nucleotide proofreading functions by nematode RAD51 paralogs facilitate optimal RAD51 filament function. Nat Commun 2021; 12:5545. [PMID: 34545070 PMCID: PMC8452638 DOI: 10.1038/s41467-021-25830-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 09/02/2021] [Indexed: 12/30/2022] Open
Abstract
The RAD51 recombinase assembles as helical nucleoprotein filaments on single-stranded DNA (ssDNA) and mediates invasion and strand exchange with homologous duplex DNA (dsDNA) during homologous recombination (HR), as well as protection and restart of stalled replication forks. Strand invasion by RAD51-ssDNA complexes depends on ATP binding. However, RAD51 can bind ssDNA in non-productive ADP-bound or nucleotide-free states, and ATP-RAD51-ssDNA complexes hydrolyse ATP over time. Here, we define unappreciated mechanisms by which the RAD51 paralog complex RFS-1/RIP-1 limits the accumulation of RAD-51-ssDNA complexes with unfavorable nucleotide content. We find RAD51 paralogs promote the turnover of ADP-bound RAD-51 from ssDNA, in striking contrast to their ability to stabilize productive ATP-bound RAD-51 nucleoprotein filaments. In addition, RFS-1/RIP-1 inhibits binding of nucleotide-free RAD-51 to ssDNA. We propose that ‘nucleotide proofreading’ activities of RAD51 paralogs co-operate to ensure the enrichment of active, ATP-bound RAD-51 filaments on ssDNA to promote HR. A RAD51 paralog complex, RFS-1/RIP-1, is shown to control ssDNA binding and dissociation by RAD-51 differentially in the presence and absence of nucleotide cofactors. These nucleotide proofreading activities drive a preferential accumulation of RAD-51-ssDNA complexes with optimal nucleotide content.
Collapse
Affiliation(s)
- Mário Špírek
- International Clinical Research Center, St. Anne's University Hospital in Brno, 62500, Brno, Czech Republic.,Department of Biology Masaryk University, 62500, Brno, Czech Republic
| | | | - Ondrej Belan
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK.,National Centre for Biomolecular Research, Masaryk University, 62500, Brno, Czech Republic
| | - Simon J Boulton
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Lumir Krejci
- International Clinical Research Center, St. Anne's University Hospital in Brno, 62500, Brno, Czech Republic. .,Department of Biology Masaryk University, 62500, Brno, Czech Republic. .,National Centre for Biomolecular Research, Masaryk University, 62500, Brno, Czech Republic.
| |
Collapse
|
28
|
Carver A, Zhang X. Rad51 filament dynamics and its antagonistic modulators. Semin Cell Dev Biol 2021; 113:3-13. [PMID: 32631783 DOI: 10.1016/j.semcdb.2020.06.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/10/2020] [Accepted: 06/20/2020] [Indexed: 02/07/2023]
Abstract
Rad51 recombinase is the central player in homologous recombination, the faithful repair pathway for double-strand breaks and key event during meiosis. Rad51 forms nucleoprotein filaments on single-stranded DNA, exposed by a double-strand break. These filaments are responsible for homology search and strand invasion, which lead to homology-directed repair. Due to its central roles in DNA repair and genome stability, Rad51 is modulated by multiple factors and post-translational modifications. In this review, we summarize our current understanding of the dynamics of Rad51 filaments, the roles of other factors and their modes of action in modulating key stages of Rad51 filaments: formation, stability and disassembly.
Collapse
Affiliation(s)
- Alexander Carver
- Section of Structural Biology, Department of Infectious Diseases, Sir Alexander Fleming Building, Imperial College London, SW7 2AZ, UK
| | - Xiaodong Zhang
- Section of Structural Biology, Department of Infectious Diseases, Sir Alexander Fleming Building, Imperial College London, SW7 2AZ, UK.
| |
Collapse
|
29
|
Xue C, Molnarova L, Steinfeld JB, Zhao W, Ma C, Spirek M, Kaniecki K, Kwon Y, Beláň O, Krejci K, Boulton S, Sung P, Greene EC, Krejci L. Single-molecule visualization of human RECQ5 interactions with single-stranded DNA recombination intermediates. Nucleic Acids Res 2021; 49:285-305. [PMID: 33332547 PMCID: PMC7797033 DOI: 10.1093/nar/gkaa1184] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 11/03/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022] Open
Abstract
RECQ5 is one of five RecQ helicases found in humans and is thought to participate in homologous DNA recombination by acting as a negative regulator of the recombinase protein RAD51. Here, we use kinetic and single molecule imaging methods to monitor RECQ5 behavior on various nucleoprotein complexes. Our data demonstrate that RECQ5 can act as an ATP-dependent single-stranded DNA (ssDNA) motor protein and can translocate on ssDNA that is bound by replication protein A (RPA). RECQ5 can also translocate on RAD51-coated ssDNA and readily dismantles RAD51-ssDNA filaments. RECQ5 interacts with RAD51 through protein-protein contacts, and disruption of this interface through a RECQ5-F666A mutation reduces translocation velocity by ∼50%. However, RECQ5 readily removes the ATP hydrolysis-deficient mutant RAD51-K133R from ssDNA, suggesting that filament disruption is not coupled to the RAD51 ATP hydrolysis cycle. RECQ5 also readily removes RAD51-I287T, a RAD51 mutant with enhanced ssDNA-binding activity, from ssDNA. Surprisingly, RECQ5 can bind to double-stranded DNA (dsDNA), but it is unable to translocate. Similarly, RECQ5 cannot dismantle RAD51-bound heteroduplex joint molecules. Our results suggest that the roles of RECQ5 in genome maintenance may be regulated in part at the level of substrate specificity.
Collapse
Affiliation(s)
- Chaoyou Xue
- Department of Biochemistry & Molecular Biophysics, Columbia University, New York, NY 10032, USA
| | - Lucia Molnarova
- Department of Biology, Masaryk University, Brno 62500, Czech Republic
| | - Justin B Steinfeld
- Department of Biochemistry & Molecular Biophysics, Columbia University, New York, NY 10032, USA
| | - Weixing Zhao
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, TX 78229, USA
| | - Chujian Ma
- Department of Biochemistry & Molecular Biophysics, Columbia University, New York, NY 10032, USA
| | - Mario Spirek
- Department of Biology, Masaryk University, Brno 62500, Czech Republic
| | - Kyle Kaniecki
- Department of Biochemistry & Molecular Biophysics, Columbia University, New York, NY 10032, USA
| | - Youngho Kwon
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, TX 78229, USA
| | - Ondrej Beláň
- DSB Repair Metabolism Lab, The Francis Crick Institute, Midland Road, London NW1 1AT, UK
| | - Katerina Krejci
- Department of Biology, Masaryk University, Brno 62500, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno 65691, Czech Republic
| | - Simon J Boulton
- DSB Repair Metabolism Lab, The Francis Crick Institute, Midland Road, London NW1 1AT, UK
| | - Patrick Sung
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, TX 78229, USA
| | - Eric C Greene
- Department of Biochemistry & Molecular Biophysics, Columbia University, New York, NY 10032, USA
| | - Lumir Krejci
- Department of Biology, Masaryk University, Brno 62500, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno 65691, Czech Republic
- National Centre for Biomolecular Research, Masaryk, Brno 62500, Czech Republic
| |
Collapse
|
30
|
TREX2 Exonuclease Causes Spontaneous Mutations and Stress-Induced Replication Fork Defects in Cells Expressing RAD51 K133A. Cell Rep 2020; 33:108543. [PMID: 33357432 PMCID: PMC7896812 DOI: 10.1016/j.celrep.2020.108543] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 04/23/2020] [Accepted: 12/01/2020] [Indexed: 01/01/2023] Open
Abstract
DNA damage tolerance (DDT) and homologous recombination (HR) stabilize replication forks (RFs). RAD18/UBC13/three prime repair exonuclease 2 (TREX2)-mediated proliferating cell nuclear antigen (PCNA) ubiquitination is central to DDT, an error-prone lesion bypass pathway. RAD51 is the recombinase for HR. The RAD51 K133A mutation increased spontaneous mutations and stress-induced RF stalls and nascent strand degradation. Here, we report in RAD51K133A cells that this phenotype is reduced by expressing a TREX2 H188A mutation that deletes its exonuclease activity. In RAD51K133A cells, knocking out RAD18 or overexpressing PCNA reduces spontaneous mutations, while expressing ubiquitination-incompetent PCNAK164R increases mutations, indicating DDT as causal. Deleting TREX2 in cells deficient for the RF maintenance proteins poly(ADP-ribose) polymerase 1 (PARP1) or FANCB increased nascent strand degradation that was rescued by TREX2H188A, implying that TREX2 prohibits degradation independent of catalytic activity. A possible explanation for this occurrence is that TREX2H188A associates with UBC13 and ubiquitinates PCNA, suggesting a dual role for TREX2 in RF maintenance.
Collapse
|
31
|
Shah SS, Hartono SR, Chédin F, Heyer WD. Bisulfite treatment and single-molecule real-time sequencing reveal D-loop length, position, and distribution. eLife 2020; 9:59111. [PMID: 33185185 PMCID: PMC7695462 DOI: 10.7554/elife.59111] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 10/27/2020] [Indexed: 01/01/2023] Open
Abstract
Displacement loops (D-loops) are signature intermediates formed during homologous recombination. Numerous factors regulate D-loop formation and disruption, thereby influencing crucial aspects of DNA repair, including donor choice and the possibility of crossover outcome. While D-loop detection methods exist, it is currently unfeasible to assess the relationship between D-loop editors and D-loop characteristics such as length and position. Here, we developed a novel in vitro assay to characterize the length and position of individual D-loops with near base-pair resolution and deep coverage, while also revealing their distribution in a population. Non-denaturing bisulfite treatment modifies the cytosines on the displaced strand of the D-loop to uracil, leaving a permanent signature for the displaced strand. Subsequent single-molecule real-time sequencing uncovers the cytosine conversion patch as a D-loop footprint. The D-loop Mapping Assay is widely applicable with different substrates and donor types and can be used to study factors that influence D-loop properties.
Collapse
Affiliation(s)
- Shanaya Shital Shah
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, United States
| | - Stella R Hartono
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, United States
| | - Frédéric Chédin
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, United States
| | - Wolf-Dietrich Heyer
- Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, United States.,Department of Molecular and Cellular Biology, University of California, Davis, Davis, United States
| |
Collapse
|
32
|
Feretzaki M, Pospisilova M, Valador Fernandes R, Lunardi T, Krejci L, Lingner J. RAD51-dependent recruitment of TERRA lncRNA to telomeres through R-loops. Nature 2020; 587:303-308. [PMID: 33057192 PMCID: PMC7116795 DOI: 10.1038/s41586-020-2815-6] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 07/16/2020] [Indexed: 12/12/2022]
Abstract
Telomeres which are present at the ends of eukaryotic chromosomes mediate genome stability and determine cellular lifespan1. Telomeric repeat containing RNAs (TERRA) are long noncoding RNAs transcribed from chromosome ends2,3, which regulate telomeric chromatin structure and telomere maintenance via telomerase and homology directed repair (HDR)4,5. The mechanisms by which TERRA is recruited to chromosome ends remain poorly defined. Here we develop a reporter system to dissect the underlying mechanisms and demonstrate that the UUAGGG-repeats of TERRA are both necessary and sufficient to target TERRA to chromosome ends. TERRA preferentially associates with short telomeres through the formation of telomeric DNA:RNA hybrid (R-loop) structures that can form in trans. Telomere association and R-loop formation triggers telomere fragility and is promoted by the RAD51 recombinase and its interacting partner BRCA2 but counteracted by RNA surveillance factors, RNaseH1 and TRF1. RAD51 physically interacts with TERRA and catalyzes R-loop formation with TERRA in vitro supporting a direct involvement of this DNA recombinase in TERRA recruitment by strand invasion. Together, our findings reveal a RAD51-dependent pathway that governs TERRA mediated R-loop formation post transcription providing a mechanism of how lncRNAs can be recruited to new loci in trans.
Collapse
Affiliation(s)
- Marianna Feretzaki
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Michaela Pospisilova
- Department of Biology and National Centre for Biomolecular Research, Masaryk University, Brno, Czech Republic
| | - Rita Valador Fernandes
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Thomas Lunardi
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Lumir Krejci
- Department of Biology and National Centre for Biomolecular Research, Masaryk University, Brno, Czech Republic. .,International Clinical Research Center, St Anne's University Hospital, Brno, Czech Republic.
| | - Joachim Lingner
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
33
|
Mechanism and significance of chromosome damage repair by homologous recombination. Essays Biochem 2020; 64:779-790. [DOI: 10.1042/ebc20190093] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/22/2020] [Accepted: 06/29/2020] [Indexed: 12/29/2022]
Abstract
Abstract
Homologous recombination (HR) is a major, conserved pathway of chromosome damage repair. It not only fulfills key functions in the removal of deleterious lesions such as DNA double-strand breaks (DSBs) and interstrand cross-links (ICLs), but also in replication fork repair and protection. Several familial and acquired cancer predisposition syndromes stem from defects in HR. In particular, individuals with mutations in HR genes exhibit predisposition to breast, ovarian, pancreatic, and prostate cancers, and they also show signs of accelerated aging. However, aberrant and untimely HR events can lead to the loss of heterozygosity, genomic rearrangements, and cytotoxic nucleoprotein intermediates. Thus, it is critically important that HR be tightly regulated. In addition to DNA repair, HR is also involved in meiotic chromosome segregation and telomere maintenance in cells that lack telomerase. In this review, we focus on the role of HR in DSB repair (DSBR) and summarize the current state of the field.
Collapse
|
34
|
Abstract
Accurate DNA repair and replication are critical for genomic stability and cancer prevention. RAD51 and its gene family are key regulators of DNA fidelity through diverse roles in double-strand break repair, replication stress, and meiosis. RAD51 is an ATPase that forms a nucleoprotein filament on single-stranded DNA. RAD51 has the function of finding and invading homologous DNA sequences to enable accurate and timely DNA repair. Its paralogs, which arose from ancient gene duplications of RAD51, have evolved to regulate and promote RAD51 function. Underscoring its importance, misregulation of RAD51, and its paralogs, is associated with diseases such as cancer and Fanconi anemia. In this review, we focus on the mammalian RAD51 structure and function and highlight the use of model systems to enable mechanistic understanding of RAD51 cellular roles. We also discuss how misregulation of the RAD51 gene family members contributes to disease and consider new approaches to pharmacologically inhibit RAD51.
Collapse
Affiliation(s)
- Braulio Bonilla
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA;
| | - Sarah R Hengel
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA;
| | - McKenzie K Grundy
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA;
| | - Kara A Bernstein
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA;
| |
Collapse
|
35
|
Sun Y, McCorvie TJ, Yates LA, Zhang X. Structural basis of homologous recombination. Cell Mol Life Sci 2020; 77:3-18. [PMID: 31748913 PMCID: PMC6957567 DOI: 10.1007/s00018-019-03365-1] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 10/10/2019] [Accepted: 10/31/2019] [Indexed: 12/12/2022]
Abstract
Homologous recombination (HR) is a pathway to faithfully repair DNA double-strand breaks (DSBs). At the core of this pathway is a DNA recombinase, which, as a nucleoprotein filament on ssDNA, pairs with homologous DNA as a template to repair the damaged site. In eukaryotes Rad51 is the recombinase capable of carrying out essential steps including strand invasion, homology search on the sister chromatid and strand exchange. Importantly, a tightly regulated process involving many protein factors has evolved to ensure proper localisation of this DNA repair machinery and its correct timing within the cell cycle. Dysregulation of any of the proteins involved can result in unchecked DNA damage, leading to uncontrolled cell division and cancer. Indeed, many are tumour suppressors and are key targets in the development of new cancer therapies. Over the past 40 years, our structural and mechanistic understanding of homologous recombination has steadily increased with notable recent advancements due to the advances in single particle cryo electron microscopy. These have resulted in higher resolution structural models of the signalling proteins ATM (ataxia telangiectasia mutated), and ATR (ataxia telangiectasia and Rad3-related protein), along with various structures of Rad51. However, structural information of the other major players involved, such as BRCA1 (breast cancer type 1 susceptibility protein) and BRCA2 (breast cancer type 2 susceptibility protein), has been limited to crystal structures of isolated domains and low-resolution electron microscopy reconstructions of the full-length proteins. Here we summarise the current structural understanding of homologous recombination, focusing on key proteins in recruitment and signalling events as well as the mediators for the Rad51 recombinase.
Collapse
Affiliation(s)
- Yueru Sun
- Section of Structural Biology, Department of Infectious Diseases, Imperial College, London, SW7 2AZ, UK
| | - Thomas J McCorvie
- Section of Structural Biology, Department of Infectious Diseases, Imperial College, London, SW7 2AZ, UK
| | - Luke A Yates
- Section of Structural Biology, Department of Infectious Diseases, Imperial College, London, SW7 2AZ, UK
| | - Xiaodong Zhang
- Section of Structural Biology, Department of Infectious Diseases, Imperial College, London, SW7 2AZ, UK.
| |
Collapse
|
36
|
Zhang S, Zhong X, Yuan H, Guo Y, Song D, Qi F, Zhu Z, Wang X, Guo Z. Interfering in apoptosis and DNA repair of cancer cells to conquer cisplatin resistance by platinum(iv) prodrugs. Chem Sci 2020. [DOI: 10.1039/d0sc00197j] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Platinum(iv) prodrugs targeting the DNA repair mechanism downregulate myeloid cell leukemia-1 (Mcl-1) and homologous recombination proteins (RAD51, BRCA2), thereby enhancing cytotoxicity against cisplatin-resistant cancer cells.
Collapse
Affiliation(s)
- Shuren Zhang
- State Key Laboratory of Coordination Chemistry
- School of Chemistry and Chemical Engineering
- Chemistry and Biomedicine Innovation Centre
- Nanjing University
- Nanjing 210023
| | - Xuanmeng Zhong
- State Key Laboratory of Coordination Chemistry
- School of Chemistry and Chemical Engineering
- Chemistry and Biomedicine Innovation Centre
- Nanjing University
- Nanjing 210023
| | - Hao Yuan
- State Key Laboratory of Coordination Chemistry
- School of Chemistry and Chemical Engineering
- Chemistry and Biomedicine Innovation Centre
- Nanjing University
- Nanjing 210023
| | - Yan Guo
- State Key Laboratory of Coordination Chemistry
- School of Chemistry and Chemical Engineering
- Chemistry and Biomedicine Innovation Centre
- Nanjing University
- Nanjing 210023
| | - Dongfan Song
- State Key Laboratory of Coordination Chemistry
- School of Chemistry and Chemical Engineering
- Chemistry and Biomedicine Innovation Centre
- Nanjing University
- Nanjing 210023
| | - Fen Qi
- State Key Laboratory of Coordination Chemistry
- School of Chemistry and Chemical Engineering
- Chemistry and Biomedicine Innovation Centre
- Nanjing University
- Nanjing 210023
| | - Zhenzhu Zhu
- School of Food Science and Engineering
- Nanjing University of Finance & Economics
- Nanjing 210023
- P. R. China
| | - Xiaoyong Wang
- State Key Laboratory of Pharmaceutical Biotechnology
- School of Life Sciences
- Nanjing University
- Nanjing 210023
- P. R. China
| | - Zijian Guo
- State Key Laboratory of Coordination Chemistry
- School of Chemistry and Chemical Engineering
- Chemistry and Biomedicine Innovation Centre
- Nanjing University
- Nanjing 210023
| |
Collapse
|
37
|
Jin X, Hapsari ND, Lee S, Jo K. DNA binding fluorescent proteins as single-molecule probes. Analyst 2020; 145:4079-4095. [DOI: 10.1039/d0an00218f] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
DNA binding fluorescent proteins are useful probes for a broad range of biological applications.
Collapse
Affiliation(s)
- Xuelin Jin
- Department of Chemistry and Interdisciplinary Program of Integrated Biotechnology
- Sogang University
- Seoul
- Republic of Korea
| | - Natalia Diyah Hapsari
- Department of Chemistry and Interdisciplinary Program of Integrated Biotechnology
- Sogang University
- Seoul
- Republic of Korea
- Chemistry Education Program
| | - Seonghyun Lee
- Department of Chemistry and Interdisciplinary Program of Integrated Biotechnology
- Sogang University
- Seoul
- Republic of Korea
| | - Kyubong Jo
- Department of Chemistry and Interdisciplinary Program of Integrated Biotechnology
- Sogang University
- Seoul
- Republic of Korea
| |
Collapse
|
38
|
Son MY, Hasty P. Homologous recombination defects and how they affect replication fork maintenance. AIMS GENETICS 2019; 5:192-211. [PMID: 31435521 PMCID: PMC6690234 DOI: 10.3934/genet.2018.4.192] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 03/18/2019] [Indexed: 01/07/2023]
Abstract
Homologous recombination (HR) repairs DNA double strand breaks (DSBs) and stabilizes replication forks (RFs). RAD51 is the recombinase for the HR pathway. To preserve genomic integrity, RAD51 forms a filament on the 3' end of a DSB and on a single-stranded DNA (ssDNA) gap. But unregulated HR results in undesirable chromosomal rearrangements. This review describes the multiple mechanisms that regulate HR with a focus on those mechanisms that promote and contain RAD51 filaments to limit chromosomal rearrangements. If any of these pathways break down and HR becomes unregulated then disease, primarily cancer, can result.
Collapse
Affiliation(s)
- Mi Young Son
- Department of Molecular Medicine and Institute of Biotechnology, UT Health San Antonio, 15355 Lambda Drive, San Antonio, USA
| | - Paul Hasty
- Department of Molecular Medicine and Institute of Biotechnology, UT Health San Antonio, 15355 Lambda Drive, San Antonio, USA
- The Mays Cancer Center, USA
- Sam and Ann Barshop Institute for Longevity and Aging Studies, USA
| |
Collapse
|
39
|
van der Zon NL, Kanaar R, Wyman C. Variation in RAD51 details a hub of functions: opportunities to advance cancer diagnosis and therapy. F1000Res 2018; 7. [PMID: 30271574 PMCID: PMC6137408 DOI: 10.12688/f1000research.15650.2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/29/2018] [Indexed: 11/30/2022] Open
Abstract
Loss of genome stability is one of the hallmarks of the enabling characteristics of cancer development. Homologous recombination is a DNA repair process that often breaks down as a prelude to developing cancer. Conversely, homologous recombination can be the Achilles’ heel in common anti-cancer therapies, which are effective by inducing irreparable DNA damage. Here, we review recent structural and functional studies of RAD51, the protein that catalyzes the defining step of homologous recombination: homology recognition and DNA strand exchange. Specific mutations can be linked to structural changes and known essential functions. Additional RAD51 interactions and functions may be revealed. The identification of viable mutations in this essential protein may help define the range of activity and interactions needed. All of this information provides opportunities to fine-tune existing therapies based on homologous recombination status, guide diagnosis, and hopefully develop new clinical tools.
Collapse
Affiliation(s)
- Nick Ll van der Zon
- Department of Molecular Genetics, Erasmus MC, Rotterdam, PO Box 2040, 3000 CA, The Netherlands
| | - Roland Kanaar
- Department of Molecular Genetics, Erasmus MC, Rotterdam, PO Box 2040, 3000 CA, The Netherlands.,Oncode Institute, Erasmus MC, Rotterdam, PO Box 2040, 3000 CA, The Netherlands
| | - Claire Wyman
- Department of Molecular Genetics, Erasmus MC, Rotterdam, PO Box 2040, 3000 CA, The Netherlands.,Department of Radiation Oncology, Erasmus MC, Rotterdam, PO Box 2040, 3000 CA, The Netherlands
| |
Collapse
|
40
|
van der Zon NLL, Kanaar R, Wyman C. Variation in RAD51 details a hub of functions: opportunities to advance cancer diagnosis and therapy. F1000Res 2018; 7:F1000 Faculty Rev-1453. [PMID: 30271574 PMCID: PMC6137408 DOI: 10.12688/f1000research.15650.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/29/2018] [Indexed: 11/06/2023] Open
Abstract
Loss of genome stability is one of the hallmarks of the enabling characteristics of cancer development. Homologous recombination is a DNA repair process that often breaks down as a prelude to developing cancer. Conversely, homologous recombination can be the Achilles' heel in common anti-cancer therapies, which are effective by inducing irreparable DNA damage. Here, we review recent structural and functional studies of RAD51, the protein that catalyzes the defining step of homologous recombination: homology recognition and DNA strand exchange. Specific mutations can be linked to structural changes and known essential functions. Additional RAD51 interactions and functions may be revealed. The identification of viable mutations in this essential protein may help define the range of activity and interactions needed. All of this information provides opportunities to fine-tune existing therapies based on homologous recombination status, guide diagnosis, and hopefully develop new clinical tools.
Collapse
Affiliation(s)
- Nick LL van der Zon
- Department of Molecular Genetics, Erasmus MC, Rotterdam, PO Box 2040, 3000 CA, The Netherlands
| | - Roland Kanaar
- Department of Molecular Genetics, Erasmus MC, Rotterdam, PO Box 2040, 3000 CA, The Netherlands
- Oncode Institute, Erasmus MC, Rotterdam, PO Box 2040, 3000 CA, The Netherlands
| | - Claire Wyman
- Department of Molecular Genetics, Erasmus MC, Rotterdam, PO Box 2040, 3000 CA, The Netherlands
- Department of Radiation Oncology, Erasmus MC, Rotterdam, PO Box 2040, 3000 CA, The Netherlands
| |
Collapse
|
41
|
Wright WD, Shah SS, Heyer WD. Homologous recombination and the repair of DNA double-strand breaks. J Biol Chem 2018; 293:10524-10535. [PMID: 29599286 DOI: 10.1074/jbc.tm118.000372] [Citation(s) in RCA: 476] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Homologous recombination enables the cell to access and copy intact DNA sequence information in trans, particularly to repair DNA damage affecting both strands of the double helix. Here, we discuss the DNA transactions and enzymatic activities required for this elegantly orchestrated process in the context of the repair of DNA double-strand breaks in somatic cells. This includes homology search, DNA strand invasion, repair DNA synthesis, and restoration of intact chromosomes. Aspects of DNA topology affecting individual steps are highlighted. Overall, recombination is a dynamic pathway with multiple metastable and reversible intermediates designed to achieve DNA repair with high fidelity.
Collapse
Affiliation(s)
| | | | - Wolf-Dietrich Heyer
- From the Departments of Microbiology and Molecular Genetics and .,Molecular and Cellular Biology, University of California, Davis, Davis, California 95616-8665
| |
Collapse
|
42
|
Zhang S, Yuan H, Guo Y, Wang K, Wang X, Guo Z. Towards rational design of RAD51-targeting prodrugs: platinumIV–artesunate conjugates with enhanced cytotoxicity against BRCA-proficient ovarian and breast cancer cells. Chem Commun (Camb) 2018; 54:11717-11720. [DOI: 10.1039/c8cc06576d] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
PtIV–Artesunate prodrugs target the homologous recombination protein RAD51 and exhibit higher cytotoxicity against BRCA-proficient ovarian and breast cancer cells than cisplatin.
Collapse
Affiliation(s)
- Shuren Zhang
- State Key Laboratory of Coordination Chemistry
- School of Chemistry and Chemical Engineering
- Nanjing University
- Nanjing 210023
- P. R. China
| | - Hao Yuan
- State Key Laboratory of Coordination Chemistry
- School of Chemistry and Chemical Engineering
- Nanjing University
- Nanjing 210023
- P. R. China
| | - Yan Guo
- State Key Laboratory of Coordination Chemistry
- School of Chemistry and Chemical Engineering
- Nanjing University
- Nanjing 210023
- P. R. China
| | - Kun Wang
- State Key Laboratory of Coordination Chemistry
- School of Chemistry and Chemical Engineering
- Nanjing University
- Nanjing 210023
- P. R. China
| | - Xiaoyong Wang
- State Key Laboratory of Pharmaceutical Biotechnology
- School of Life Sciences
- Nanjing University
- Nanjing 210023
- P. R. China
| | - Zijian Guo
- State Key Laboratory of Coordination Chemistry
- School of Chemistry and Chemical Engineering
- Nanjing University
- Nanjing 210023
- P. R. China
| |
Collapse
|