1
|
Tossetta G, Fantone S, Procopio AD, Pugnaloni A, Gualtieri AF, Marzioni D. Effects of mineral fibres in an in vitro placental syncytiotrophoblast model. Curr Res Toxicol 2025; 8:100231. [PMID: 40237000 PMCID: PMC11999340 DOI: 10.1016/j.crtox.2025.100231] [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: 11/20/2024] [Revised: 03/18/2025] [Accepted: 03/24/2025] [Indexed: 04/17/2025] Open
Abstract
It is known that mineral fibres can be found in placental tissues, but their effect is not known on these tissues. BeWo in vitro model of syncytiotrophoblast, the outer layer of maternal-foetal barrier, is necessary to understand if mineral fibres can alter placental cell turnover and consequently to influence the outcome of pregnancy. We performed in vitro experiments using chrysotile UICC (UICC), chrysotile Valmalenco (VM) and erionite (ERI) to investigate the potential cytotoxic effects of these mineral fibres on BeWo cells. We demonstrated that all fibres are toxic while only UICC fibres caused a DNA damage that the cells were not able to repair through RAD51 activity. In addition, we demonstrated that DNA replication is not altered while cyclin D1 showed a significant decrease in VM and UICC suggesting that the cell cycle is altered in G1 phase. Moreover, UICC increased active form of caspase 3 demonstrating that apoptosis can be induced in BeWo cells. We suggest that although morphological changes are not visible in BeWo cells treated with these mineral fibres, DNA damage can lead to altered placenta physiology that can be seen late when the damage at the foetal tissues has already occurred.
Collapse
Affiliation(s)
- Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
| | | | - Antonio Domenico Procopio
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
- Clinical Laboratory and Molecular Diagnostic, IRCCS INRCA, Ancona, Italy
| | - Armanda Pugnaloni
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| | | | - Daniela Marzioni
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
- IRCCS INRCA, Ancona, Italy
| |
Collapse
|
2
|
Ashraf R, Polasek-Sedlackova H, Marini V, Prochazkova J, Hasanova Z, Zacpalova M, Boudova M, Krejci L. RECQ4-MUS81 interaction contributes to telomere maintenance with implications to Rothmund-Thomson syndrome. Nat Commun 2025; 16:1302. [PMID: 39900600 PMCID: PMC11791078 DOI: 10.1038/s41467-025-56518-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/21/2025] [Indexed: 02/05/2025] Open
Abstract
Replication stress, particularly in hard-to-replicate regions such as telomeres and centromeres, leads to the accumulation of replication intermediates that must be processed to ensure proper chromosome segregation. In this study, we identify a critical role for the interaction between RECQ4 and MUS81 in managing such stress. We show that RECQ4 physically interacts with MUS81, targeting it to specific DNA substrates and enhancing its endonuclease activity. Loss of this interaction, results in significant chromosomal segregation defects, including the accumulation of micronuclei, anaphase bridges, and ultrafine bridges (UFBs). Our data further demonstrate that the RECQ4-MUS81 interaction plays an important role in ALT-positive cells, where MUS81 foci primarily colocalise with telomeres, highlighting its role in telomere maintenance. We also observe that a mutation associated with Rothmund-Thomson syndrome, which produces a truncated RECQ4 unable to interact with MUS81, recapitulates these chromosome instability phenotypes. This underscores the importance of RECQ4-MUS81 in safeguarding genome integrity and suggests potential implications for human disease. Our findings demonstrate the RECQ4-MUS81 interaction as a key mechanism in alleviating replication stress at hard-to-replicate regions and highlight its relevance in pathological conditions such as RTS.
Collapse
Affiliation(s)
- Raghib Ashraf
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5/A4, 625 00, Brno, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5/A7, Brno, 62500, Czech Republic
| | - Hana Polasek-Sedlackova
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5/A4, 625 00, Brno, Czech Republic
- Institute of Biophysics, Czech Academy of Sciences, Kralovopolska 135, Brno, 61200, Czech Republic
| | - Victoria Marini
- Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5/A7, Brno, 62500, Czech Republic
| | - Jana Prochazkova
- Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5/A7, Brno, 62500, Czech Republic
| | - Zdenka Hasanova
- Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5/A7, Brno, 62500, Czech Republic
| | - Magdalena Zacpalova
- Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5/A7, Brno, 62500, Czech Republic
| | - Michala Boudova
- Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5/A7, Brno, 62500, Czech Republic
| | - Lumir Krejci
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5/A4, 625 00, Brno, Czech Republic.
- Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5/A7, Brno, 62500, Czech Republic.
| |
Collapse
|
3
|
Roos J, Manolikakes G, Schlomann U, Klinke A, Schopfer FJ, Neumann CA, Maier TJ. Nitro-fatty acids: promising agents for the development of new cancer therapeutics. Trends Pharmacol Sci 2024; 45:1061-1080. [PMID: 39490362 DOI: 10.1016/j.tips.2024.09.009] [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: 06/14/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 11/05/2024]
Abstract
Nitro-fatty acids (NO2-FAs) are endogenous pleiotropic lipid mediators regarded as promising drug candidates for treating inflammatory and fibrotic diseases. Over the past two decades, the anti-inflammatory and cytoprotective actions of NO2-FAs and several molecular targets have been identified. More recently, preclinical studies have demonstrated their potential as prospective cancer therapeutics with favorable safety and tumor-selective profiles. In this review, we describe the mechanisms of action, with a focus on NO2-FA antineoplastic and chemosensitizing effects. We also address the potential therapeutic applications of endogenous and structurally modified NO2-FAs species in cancer treatment.
Collapse
Affiliation(s)
- Jessica Roos
- Division of Immunology, Paul-Ehrlich-Institute, Federal Institute for Vaccines and Biomedicines, Langen, 63225, Hesse, Germany.
| | - Georg Manolikakes
- Department of Chemistry, RPTU Kaiserslautern-Landau, Kaiserslautern, 67663, Rhineland-Palatinate, Germany
| | - Uwe Schlomann
- Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum NRW, University Hospital of the Ruhr-Universität Bochum, 32545 Bad Oeynhausen, Germany
| | - Anna Klinke
- Clinic for General and Interventional Cardiology/Angiology, Herz- und Diabeteszentrum NRW, University Hospital of the Ruhr-Universität Bochum, 32545 Bad Oeynhausen, Germany
| | - Francisco J Schopfer
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute (VMI), University of Pittsburgh, Pittsburgh, PA, USA; Pittsburgh Liver Research Center (PLRC), University of Pittsburgh, Pittsburgh, PA, USA; Center for Metabolism and Mitochondrial Medicine (C3M) University of Pittsburgh, Pittsburgh, PA, USA
| | - Carola A Neumann
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee-Women's Research Institute, Pittsburgh, PA, USA
| | - Thorsten J Maier
- Division of Immunology, Paul-Ehrlich-Institute, Federal Institute for Vaccines and Biomedicines, Langen, 63225, Hesse, Germany
| |
Collapse
|
4
|
Maarouf A, Iqbal F, Sanaullah S, Locatelli M, Atanasiu AT, Kolbin D, Hommais C, Mühlemann JK, Bonin K, Bloom K, Liu J, Vidi PA. RAD51 regulates eukaryotic chromatin motions in the absence of DNA damage. Mol Biol Cell 2024; 35:ar136. [PMID: 39292916 PMCID: PMC11617103 DOI: 10.1091/mbc.e24-04-0188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/12/2024] [Accepted: 09/13/2024] [Indexed: 09/20/2024] Open
Abstract
In yeasts and higher eukaryotes, chromatin motions may be tuned to genomic functions, with transcriptional activation and the DNA damage response both leading to profound changes in chromatin dynamics. The RAD51 recombinase is a key mediator of chromatin mobility following DNA damage. As functions of RAD51 beyond DNA repair are being discovered, we asked whether RAD51 modulates chromatin dynamics in the absence of DNA damage and found that inhibition or depletion of RAD51 alters chromatin motions in undamaged cells. Inhibition of RAD51 increased nucleosome clustering. Predictions from polymer models are that chromatin clusters reduce chain mobility and, indeed, we measured reduced motion of individual chromatin loci in cells treated with a RAD51 inhibitor. This effect was conserved in mammalian cells, yeasts, and plant cells. In contrast, RAD51 depletion or inhibition increased global chromatin motions at the microscale. The results uncover a role for RAD51 in regulating local and global chromatin dynamics independently from DNA damage and highlight the importance of considering different physical scales when studying chromatin dynamics.
Collapse
Affiliation(s)
- Amine Maarouf
- Institut de Cancérologie de l'Ouest, Angers F-49055, France
| | - Fadil Iqbal
- Department of Physics, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202
| | - Sarvath Sanaullah
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Maëlle Locatelli
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Andrew T. Atanasiu
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Daniel Kolbin
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Chloé Hommais
- Institut de Cancérologie de l'Ouest, Angers F-49055, France
| | - Joëlle K. Mühlemann
- Climate Resilient Crop Production Laboratory, Division of Crop Biotechnics, Department of Biosystems, Katholieke Universiteit (KU) Leuven, Leuven 3000, Belgium
| | - Keith Bonin
- Department of Physics, Wake Forest University, Winston-Salem, NC 27109
| | - Kerry Bloom
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Jing Liu
- Department of Physics and Astronomy, Purdue University, West Lafayette, IN 47907
| | - Pierre-Alexandre Vidi
- Institut de Cancérologie de l'Ouest, Angers F-49055, France
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157
| |
Collapse
|
5
|
Liu T, Pan G, Zhang J, Wang J, Guo X, Chen Y, Wang X, Cui X, Liu H, Jiang F. Molecular basis of CX-5461-induced DNA damage response in primary vascular smooth muscle cells. Heliyon 2024; 10:e37227. [PMID: 39296007 PMCID: PMC11407941 DOI: 10.1016/j.heliyon.2024.e37227] [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: 02/10/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/21/2024] Open
Abstract
Our previous studies have shown that the novel selective RNA polymerase I inhibitor CX-5461 suppresses proliferation of vascular smooth muscle cells, mainly by inducing DNA damage response (DDR), including activations of ataxia telangiectasia mutated (ATM)/ATM and Rad3-related (ATR) and p53. Currently, there is no information about the molecular mechanism(s) underlying CX-5461-induced DDR in vascular cells, while the results obtained in cancer cells and immortalized cell lines are controversial. In this study, we examined the responses of various DDR pathways to CX-5461 treatment in primary aortic smooth muscle cells isolated from normal adult Sprague Dawley rats. We demonstrated that CX-5461-induced DDR was not associated with activations of the nucleotide excision repair, DNA mismatch repair, or the non-homologous end joining pathways, while the homologous recombination pathway was activated. However, the alkaline comet assay did not show massive DNA double strand breaks in CX-5461-treated cells. Instead, CX-5461-induced DDR appeared to be related to induction of DNA replication stress, which was not attributable to increased formation of G-quadruplex or R-loop structures, but might be explained by the increased replication-transcription conflict. CX-5461-induced DDR was not exclusively confined to rDNA within the nucleolar compartment; the extra-nucleolar DDR might represent a distinct secondary response related to the downregulated Rad51 expression in CX-5461-treated cells. In summary, we suggest that DNA replication stress may be the primary molecular event leading to downstream ATM/ATR and p53 activations in CX-5461-treated vascular smooth muscle cells. Our results provide further insights into the molecular basis of the beneficial effects of CX-5461 in proliferative vascular diseases.
Collapse
Affiliation(s)
- Tengfei Liu
- Department of Pharmacology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
- Gerontology and Anti-Aging Research Laboratory, Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Guopin Pan
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, Xinxiang, Henan Province, China
| | - Jing Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Jianli Wang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Xiaosun Guo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Ye Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Xiaoyun Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Xiaopei Cui
- Gerontology and Anti-Aging Research Laboratory, Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Huiqing Liu
- Department of Pharmacology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Fan Jiang
- Gerontology and Anti-Aging Research Laboratory, Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| |
Collapse
|
6
|
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
|
7
|
Zainu A, Dupaigne P, Bouchouika S, Cau J, Clément JAJ, Auffret P, Ropars V, Charbonnier JB, de Massy B, Mercier R, Kumar R, Baudat F. FIGNL1-FIRRM is essential for meiotic recombination and prevents DNA damage-independent RAD51 and DMC1 loading. Nat Commun 2024; 15:7015. [PMID: 39147779 PMCID: PMC11327267 DOI: 10.1038/s41467-024-51458-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/07/2024] [Indexed: 08/17/2024] Open
Abstract
During meiosis, nucleoprotein filaments of the strand exchange proteins RAD51 and DMC1 are crucial for repairing SPO11-generated DNA double-strand breaks (DSBs) by homologous recombination (HR). A balanced activity of positive and negative RAD51/DMC1 regulators ensures proper recombination. Fidgetin-like 1 (FIGNL1) was previously shown to negatively regulate RAD51 in human cells. However, FIGNL1's role during meiotic recombination in mammals remains unknown. Here, we decipher the meiotic functions of FIGNL1 and FIGNL1 Interacting Regulator of Recombination and Mitosis (FIRRM) using male germline-specific conditional knock-out (cKO) mouse models. Both FIGNL1 and FIRRM are required for completing meiotic prophase in mouse spermatocytes. Despite efficient recruitment of DMC1 on ssDNA at meiotic DSB hotspots, the formation of late recombination intermediates is defective in Firrm cKO and Fignl1 cKO spermatocytes. Moreover, the FIGNL1-FIRRM complex limits RAD51 and DMC1 accumulation on intact chromatin, independently from the formation of SPO11-catalyzed DSBs. Purified human FIGNL1ΔN alters the RAD51/DMC1 nucleoprotein filament structure and inhibits strand invasion in vitro. Thus, this complex might regulate RAD51 and DMC1 association at sites of meiotic DSBs to promote proficient strand invasion and processing of recombination intermediates.
Collapse
Affiliation(s)
- Akbar Zainu
- Institut de Génétique Humaine, University of Montpellier, CNRS, Montpellier, France
| | - Pauline Dupaigne
- Genome Integrity and Cancers UMR9019 CNRS, Université Paris-Saclay, Gustave Roussy, Villejuif, France
| | - Soumya Bouchouika
- Institut de Génétique Humaine, University of Montpellier, CNRS, Montpellier, France
- Institut de Génétique Moléculaire de Montpellier, CNRS-UMR 5535, Univ Montpellier, Montpellier, France
| | - Julien Cau
- Biocampus Montpellier, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Julie A J Clément
- IHPE, Univ Montpellier, CNRS, IFREMER, Univ Perpignan Via Domitia, Perpignan, France
| | - Pauline Auffret
- Institut de Génétique Humaine, University of Montpellier, CNRS, Montpellier, France
- Ifremer, IRSI, Service de Bioinformatique (SeBiMER), Plouzané, France
| | - Virginie Ropars
- Institute for Integrative Biology of the Cell (I2BC), Université Paris-Saclay, CEA, CNRS, Gif-sur-Yvette, France
| | - Jean-Baptiste Charbonnier
- Institute for Integrative Biology of the Cell (I2BC), Université Paris-Saclay, CEA, CNRS, Gif-sur-Yvette, France
| | - Bernard de Massy
- Institut de Génétique Humaine, University of Montpellier, CNRS, Montpellier, France
| | - Raphael Mercier
- Department of Chromosome Biology, Max Planck Institute for Plant Breeding Research, Cologne, Germany
| | - Rajeev Kumar
- Institut Jean-Pierre Bourgin, INRAE, AgroParisTech, CNRS, Université Paris-Saclay, Versailles, France
| | - Frédéric Baudat
- Institut de Génétique Humaine, University of Montpellier, CNRS, Montpellier, France.
| |
Collapse
|
8
|
Ozawa S, Ojiro R, Tang Q, Zou X, Jin M, Yoshida T, Shibutani M. Involvement of multiple epigenetic mechanisms by altered DNA methylation from the early stage of renal carcinogenesis before proliferative lesion formation upon repeated administration of ochratoxin A. Toxicology 2024; 506:153875. [PMID: 38945198 DOI: 10.1016/j.tox.2024.153875] [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: 04/03/2024] [Revised: 06/20/2024] [Accepted: 06/27/2024] [Indexed: 07/02/2024]
Abstract
Ochratoxin A (OTA) is a rat renal carcinogen that induces karyomegaly and micronuclei in proximal tubular epithelial cells (PTECs). We previously performed comprehensive gene profiling of alterations in promoter-region methylation and gene expression in PTECs of rats treated with OTA for 13 weeks. The OTA-specific gene profile was obtained by excluding genes showing expression changes similar to those upon treatment with 3-chloro-1,2-propanediol, a renal carcinogen not inducing karyomegaly. In this study, we validated the candidate genes using methylated DNA enrichment PCR and real-time RT-PCR, and identified Gen1, Anxa3, Cdkn1a, and Osm as genes showing OTA-specific epigenetic changes. These genes and related molecules were subjected to gene expression and immunohistochemical analyses in the PTECs of rats treated with OTA, other renal carcinogens, or non-carcinogenic renal toxicants for 4 or 13 weeks. Cdkn1a upregulation and increase of p21WAF1/CIP1+ karyomegalic PTECs were observed with OTA, matching the findings associated with micronucleus-inducing carcinogens. This suggested that the increase of p21WAF1/CIP1+ karyomegalic PTECs is linked to micronucleus formation, which in turn accelerates chromosomal instability. The upregulation of Cdkn1a-related genes with OTA suggests the acquisition of a senescence-associated secretory phenotype, which promotes the establishment of a carcinogenic environment. Meanwhile, OTA specifically caused a decrease of GEN1+ PTECs reflecting Gen1 downregulation and an increase of ANXA3+ PTECs reflecting Anxa3 upregulation, as well as Osm upregulation. OTA may efficiently disrupt pathways for repairing the DNA double-strand breaks that it itself causes, via Gen1 downregulation, and enhance cell proliferation through the upregulation of Anxa3 and Osm. This may exacerbate the chromosomal instability from the early stage of OTA-induced renal carcinogenesis before proliferative lesions form. OTA may cause renal carcinogenesis involving multiple epigenetic mechanisms.
Collapse
Affiliation(s)
- Shunsuke Ozawa
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan.
| | - Ryota Ojiro
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan.
| | - Qian Tang
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan.
| | - Xinyu Zou
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan.
| | - Meilan Jin
- Laboratory of Veterinary Pathology, College of Veterinary Medicine, Southwest University, No. 2 Tiansheng Road, BeiBei District, Chongqing 400715, PR China.
| | - Toshinori Yoshida
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan.
| | - Makoto Shibutani
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan.
| |
Collapse
|
9
|
He X, Huang T, Wang Q, Bao L, Wang Z, Song H, Li Y, Zhou J, Zhao Y, Xie Y. A prominent role of LncRNA H19 in H. pylori CagA induced DNA damage response and cell malignancy. Sci Rep 2024; 14:14185. [PMID: 38902391 PMCID: PMC11190245 DOI: 10.1038/s41598-024-65221-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 06/18/2024] [Indexed: 06/22/2024] Open
Abstract
Helicobacter pylori (H. pylori), together with its CagA, has been implicated in causing DNA damage, cell cycle arrest, apoptosis, and the development of gastric cancer. Although lncRNA H19 is abundantly expressed in gastric cancer and functions as a pro-oncogene, it remains unclear whether lncRNA H19 contributes to the oncogenic process of H. pylori CagA. This study investigates the role of H19 in the DNA damage response and malignancy induced by H. pylori. It was observed that cells infected with CagA+ H. pylori strain (GZ7/cagA) showed significantly higher H19 expression, resulting in increased γH2A.X and p-ATM expression and decreased p53 and Rad51 expression. Faster cell migration and invasion was also observed, which was reversed by H19 knockdown in H. pylori. YWHAZ was identified as an H19 target protein, and its expression was increased in H19 knockdown cells. GZ7/cagA infection responded to the increased YWHAZ expression induced by H19 knockdown. In addition, H19 knockdown stimulated cells to enter the G2-phase and attenuated the effect of GZ7/cagA infection on the cellular S-phase barrier. The results suggest that H. pylori CagA can upregulate H19 expression, participate in the DNA damage response and promote cell migration and invasion, and possibly affect cell cycle arrest via regulation of YWHAZ.
Collapse
Affiliation(s)
- Xiaofeng He
- Key Laboratory of Endemic and Ethnic Minority Diseases, Ministry of Education and Key Laboratory of Molecular Biology, Guizhou Medical University, 4 Beijing Road, Guiyang, 550004, Guizhou, People's Republic of China
- Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, 563003, Guizhou, People's Republic of China
| | - Tingting Huang
- Key Laboratory of Endemic and Ethnic Minority Diseases, Ministry of Education and Key Laboratory of Molecular Biology, Guizhou Medical University, 4 Beijing Road, Guiyang, 550004, Guizhou, People's Republic of China
| | - Qinrong Wang
- Key Laboratory of Endemic and Ethnic Minority Diseases, Ministry of Education and Key Laboratory of Molecular Biology, Guizhou Medical University, 4 Beijing Road, Guiyang, 550004, Guizhou, People's Republic of China
| | - Liya Bao
- Hepatitis Laboratory, Department of Infectious Diseases, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, People's Republic of China
| | - Zhengrong Wang
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, 550004, Guizhou, People's Republic of China
| | - Hui Song
- Key Laboratory of Endemic and Ethnic Minority Diseases, Ministry of Education and Key Laboratory of Molecular Biology, Guizhou Medical University, 4 Beijing Road, Guiyang, 550004, Guizhou, People's Republic of China
| | - Yanhong Li
- Key Laboratory of Endemic and Ethnic Minority Diseases, Ministry of Education and Key Laboratory of Molecular Biology, Guizhou Medical University, 4 Beijing Road, Guiyang, 550004, Guizhou, People's Republic of China
| | - Jianjiang Zhou
- Key Laboratory of Endemic and Ethnic Minority Diseases, Ministry of Education and Key Laboratory of Molecular Biology, Guizhou Medical University, 4 Beijing Road, Guiyang, 550004, Guizhou, People's Republic of China.
| | - Yan Zhao
- Key Laboratory of Endemic and Ethnic Minority Diseases, Ministry of Education and Key Laboratory of Molecular Biology, Guizhou Medical University, 4 Beijing Road, Guiyang, 550004, Guizhou, People's Republic of China.
| | - Yuan Xie
- Key Laboratory of Endemic and Ethnic Minority Diseases, Ministry of Education and Key Laboratory of Molecular Biology, Guizhou Medical University, 4 Beijing Road, Guiyang, 550004, Guizhou, People's Republic of China.
| |
Collapse
|
10
|
Bidany-Mizrahi T, Shweiki A, Maroun K, Abu-Tair L, Mali B, Aqeilan RI. Unveiling the relationship between WWOX and BRCA1 in mammary tumorigenicity and in DNA repair pathway selection. Cell Death Discov 2024; 10:145. [PMID: 38499540 PMCID: PMC10948869 DOI: 10.1038/s41420-024-01878-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 02/07/2024] [Accepted: 02/20/2024] [Indexed: 03/20/2024] Open
Abstract
Breast cancer is the leading cause of cancer-related deaths in women worldwide, with the basal-like or triple-negative breast cancer (TNBC) subtype being particularly aggressive and challenging to treat. Understanding the molecular mechanisms driving the development and progression of TNBC is essential. We previously showed that WW domain-containing oxidoreductase (WWOX) is commonly inactivated in TNBC and is implicated in the DNA damage response (DDR) through ATM and ATR activation. In this study, we investigated the interplay between WWOX and BRCA1, both frequently inactivated in TNBC, on mammary tumor development and on DNA double-strand break (DSB) repair choice. We generated and characterized a transgenic mouse model (K14-Cre;Brca1fl/fl;Wwoxfl/fl) and observed that mice lacking both WWOX and BRCA1 developed basal-like mammary tumors and exhibited a decrease in 53BP1 foci and an increase in RAD51 foci, suggesting impaired DSB repair. We examined human TNBC cell lines harboring wild-type and mutant BRCA1 and found that WWOX expression promoted NHEJ repair in cells with wild-type BRCA1. Our findings suggest that WWOX and BRCA1 play an important role in DSB repair pathway choice in mammary epithelial cells, underscoring their functional interaction and significance in breast carcinogenesis.
Collapse
Affiliation(s)
- Tirza Bidany-Mizrahi
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Aya Shweiki
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Kian Maroun
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Lina Abu-Tair
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Bella Mali
- Department of Pathology, Hadassah University Hospital, Jerusalem, Israel
| | - Rami I Aqeilan
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
- Cyprus Cancer Research Institute (CCRI), Nicosia, Cyprus.
| |
Collapse
|
11
|
Fantone S, Tossetta G, Cianfruglia L, Frontini A, Armeni T, Procopio AD, Pugnaloni A, Gualtieri AF, Marzioni D. Mechanisms of action of mineral fibres in a placental syncytiotrophoblast model: An in vitro toxicology study. Chem Biol Interact 2024; 390:110895. [PMID: 38301883 DOI: 10.1016/j.cbi.2024.110895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/09/2024] [Accepted: 01/29/2024] [Indexed: 02/03/2024]
Abstract
Asbestos has been widely used due to its unique characteristics. It is known that exposure to asbestos causes serious damage to health but one species, chrysolite, is still used because it is considered less toxic and not biopersistent in some countries. The aim of our study was to investigate if cellular process underlying the proliferation, differentiation and cell death of placental tissues could be modify in presence of asbestos fibres (50 μg/ml final concentration), long chrysolite fibres (CHR-L) and short chrysolite fibres (CHR-S), using BeWo cell line, an in vitro model that mimics the syncytiotrophoblast (STB), the outer layer of placental villi. Our data demonstrated that none of the fibres analysed alter syncytiotrophoblast formation but all of them induce ROS formation and reduced cell proliferation. Moreover, we showed that only CHR-L fibre induced was able to induce irreversible DNA alterations that carried cells to apoptosis. In fact, BeWo cells exposed to CHR-L fibre showed a significant increase in cleaved CASP3 protein, a marker of apoptosis. These data suggest that CHR-L may induce death of the placental villi leading to impaired placental development. The impairment of placental development is the basis of many gestational pathologies such as preeclampsia and intrauterine growth retardation. Since these pathologies are very dangerous for foetal and maternal life, we suggest to the gynaecologists to carefully evaluate the area of maternal residence, the working environment, the food used, and the materials used daily to avoid contact with these fibres as much as possible.
Collapse
Affiliation(s)
- Sonia Fantone
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Laura Cianfruglia
- Department of Clinical Sciences, Polytechnic University of Marche, 60126, Ancona, Italy
| | - Andrea Frontini
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Tatiana Armeni
- Department of Clinical Sciences, Polytechnic University of Marche, 60126, Ancona, Italy
| | - Antonio D Procopio
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy; Clinical Laboratory and Molecular Diagnostic, IRCCS INRCA, Ancona, Italy
| | - Armanda Pugnaloni
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| | - Alessandro F Gualtieri
- Chemical and Earth Sciences Department, University of Modena and Reggio Emilia, Modena, Italy
| | - Daniela Marzioni
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy.
| |
Collapse
|
12
|
Mai Y, Lin T, Zhang L, Yang W, Liu S, Wang M, Liu P, Li Z, Luo W. RAD54B inhibits vascular endothelial senescence via suppression of CHK1/p53/p21 pathway. Can J Physiol Pharmacol 2024; 102:137-149. [PMID: 37748205 DOI: 10.1139/cjpp-2023-0192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
RAD54B belongs to the SNF2/SWI2 superfamily, participating in homologous recombination repair. DNA damage is the central driver of aging, but there is no direct evidence of an association between RAD54B and vascular aging. The present study sought to investigate the role and mechanisms of RAD54B in endothelial senescence. In senescent animal models, including spontaneously hypertensive rats, normal aging mice, and D-gal-induced senescent mice, and senescent cell models induced by H2O2, D-gal, and culture, RAD54B was remarkably downregulated. Knockdown of RAD54B increased the expression of p53 and p21, increased the ratio of SA-β-gal-positive cells, and decreased the proportion of EdU-positive cells. Conversely, overexpression of RAD54B reversed the senescent phenotypes stimulated by H2O2 and delayed replicative endothelial senescence. Mechanistically, silencing RAD54B compensatorily increased the expression of RAD51/XRCC4, which remained unchanged in H2O2-induced senescence. RAD54B lacking the SNF2 domain could still reverse the increasing expression of p53/p21 induced by H2O2. RAD54B reduced γH2A.X expression and inhibited the expression and phosphorylation of CHK1. In conclusion, RAD54B exerts a direct protective effect against DNA damage through enhancing homologous recombination repair in endothelial senescence, resulting in inhibition of the downstream CHK1/p53/p21 pathway, suggesting that RAD54B may be a potential therapeutic target for vascular aging-associated diseases.
Collapse
Affiliation(s)
- Yanqi Mai
- Laboratory of Pharmacology and Toxicology, School of Pharmaceutical Sciences; National and Local United Engineering Lab of Druggability and New Drugs Evaluation; Guangdong Engineering Laboratory of Druggability and New Drug Evaluation; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, P. R. China
| | - Tong Lin
- Laboratory of Pharmacology and Toxicology, School of Pharmaceutical Sciences; National and Local United Engineering Lab of Druggability and New Drugs Evaluation; Guangdong Engineering Laboratory of Druggability and New Drug Evaluation; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, P. R. China
- Department of Pharmacy, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, P. R. China
| | - Lili Zhang
- Laboratory of Pharmacology and Toxicology, School of Pharmaceutical Sciences; National and Local United Engineering Lab of Druggability and New Drugs Evaluation; Guangdong Engineering Laboratory of Druggability and New Drug Evaluation; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, P. R. China
| | - Wanqi Yang
- Laboratory of Pharmacology and Toxicology, School of Pharmaceutical Sciences; National and Local United Engineering Lab of Druggability and New Drugs Evaluation; Guangdong Engineering Laboratory of Druggability and New Drug Evaluation; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, P. R. China
| | - Sitong Liu
- Laboratory of Pharmacology and Toxicology, School of Pharmaceutical Sciences; National and Local United Engineering Lab of Druggability and New Drugs Evaluation; Guangdong Engineering Laboratory of Druggability and New Drug Evaluation; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, P. R. China
| | - Minghui Wang
- Laboratory of Pharmacology and Toxicology, School of Pharmaceutical Sciences; National and Local United Engineering Lab of Druggability and New Drugs Evaluation; Guangdong Engineering Laboratory of Druggability and New Drug Evaluation; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, P. R. China
| | - Peiqing Liu
- Laboratory of Pharmacology and Toxicology, School of Pharmaceutical Sciences; National and Local United Engineering Lab of Druggability and New Drugs Evaluation; Guangdong Engineering Laboratory of Druggability and New Drug Evaluation; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, P. R. China
| | - Zhuoming Li
- Laboratory of Pharmacology and Toxicology, School of Pharmaceutical Sciences; National and Local United Engineering Lab of Druggability and New Drugs Evaluation; Guangdong Engineering Laboratory of Druggability and New Drug Evaluation; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, P. R. China
| | - Wenwei Luo
- Laboratory of Pharmacology and Toxicology, School of Pharmaceutical Sciences; National and Local United Engineering Lab of Druggability and New Drugs Evaluation; Guangdong Engineering Laboratory of Druggability and New Drug Evaluation; Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou, P. R. China
- Department of Pharmacy, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, P. R. China
| |
Collapse
|
13
|
Wu N, Song Y, Zhu Y, Pang L, Chen Z, Chen X. Rad51 and Systemic Inflammatory Indicators as Novel Prognostic Markers in Esophageal Squamous Cell Carcinoma. Technol Cancer Res Treat 2024; 23:15330338231216333. [PMID: 38258400 PMCID: PMC10807337 DOI: 10.1177/15330338231216333] [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: 04/06/2023] [Revised: 10/18/2023] [Accepted: 11/07/2023] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND RAD51 is a central protein involved in homologous recombination, which has been linked to cancer development and progression. systemic inflammatory indicator markers such as neutrophil-to-lymphocyte ratio and lymphocyte-to-monocyte ratio have also been implicated in cancer. However, the relationship between Rad51 and these inflammatory markers in esophageal cancer patients undergoing esophagectomy is not yet understood. METHODS We retrospectively observed 320 esophageal cancer patients who underwent esophagectomy. We collected clinical characteristics, postoperative complications, and survival analysis data and analyzed the relationship between Rad51 expression, inflammatory markers, and prognosis. RESULTS We found significant linear relationships among the inflammatory markers. There were also close relationships between Rad51 expression and neutrophil-to-lymphocyte ratio or C-reactive protein. Patients with low lymphocyte percentage were more likely to have low Rad51 expression (P = .026), high C-reactive protein (P = .007), and high neutrophil-to-lymphocyte ratio (P = .006). Low lymphocyte-to-monocyte ratio was associated with poor overall survival and was an independent prognostic factor (HR = 2.214; 95% confidence interval: 1.044-4.695, P = .038). In patients without lymph node metastases, low albumin (HR= 0.131; 95% confidence interval: 0.025-0.687, P = .016), high neutrophil-to-lymphocyte ratio (HR = 0.002; 95% confidence interval: 0.000-0.221, P = .009), and high Rad51 expression (HR = 14.394; 95% confidence interval: 2.217-97.402, P = .006) were associated with poor overall survival. CONCLUSIONS Our study found a close correlation between elevated Rad51 expression and inflammatory markers. High Rad51 expression, high neutrophil-to-lymphocyte ratio, and low lymphocyte-to-monocyte ratio are associated with lower survival rates. The combined assessment of Rad51 and inflammatory markers can be useful for preoperative assessment and prognostic evaluation in esophageal squamous cell carcinoma patients.
Collapse
Affiliation(s)
- Ning Wu
- Department of Cardiothoracic Surgery, HuaShan Hospital of Fudan University, Shanghai, China
| | - Yang Song
- Department of Cardiothoracic Surgery, HuaShan Hospital of Fudan University, Shanghai, China
| | - Yongjun Zhu
- Department of Cardiothoracic Surgery, HuaShan Hospital of Fudan University, Shanghai, China
| | - Liewen Pang
- Department of Cardiothoracic Surgery, HuaShan Hospital of Fudan University, Shanghai, China
| | - Zhiming Chen
- Department of Cardiothoracic Surgery, HuaShan Hospital of Fudan University, Shanghai, China
| | - Xiaofeng Chen
- Department of Cardiothoracic Surgery, HuaShan Hospital of Fudan University, Shanghai, China
| |
Collapse
|
14
|
Ouyang J, Feng Y, Zhang Y, Liu Y, Li S, Wang J, Tan L, Zou L. Integration of metabolomics and transcriptomics reveals metformin suppresses thyroid cancer progression via inhibiting glycolysis and restraining DNA replication. Biomed Pharmacother 2023; 168:115659. [PMID: 37864896 DOI: 10.1016/j.biopha.2023.115659] [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: 06/07/2023] [Revised: 09/26/2023] [Accepted: 10/04/2023] [Indexed: 10/23/2023] Open
Abstract
The anti-tumoral effects of metformin have been widely studied in several types of cancer, including thyroid cancer; however, the underlying molecular mechanisms remain poorly understood. As an oral hypoglycemic drug, metformin facilitates glucose catabolism and disrupts metabolic homeostasis. Metabolic reprogramming, particularly cellular glucose metabolism, is an important characteristic of malignant tumors. This study aimed to explore the therapeutic effects of metformin in thyroid cancer and the underlying metabolic mechanism. In the present study, it was shown that metformin reduced cell viability, invasion, migration, and EMT, and induced apoptosis and cell cycle G1 phase arrest in thyroid cancer. Transcriptome analysis demonstrated that the differentially expressed genes induced by metformin were involved in several signaling pathways including apoptosis singling pathways, TGF-β signaling, and cell cycle regulation in human thyroid cancer cell lines. In addition, the helicase activity of the CDC45-MCM2-7-GINS complex and DNA replication related genes such as RPA2, RAD51, and PCNA were downregulated in metformin-treated thyroid cancer cells. Moreover, metabolomics analysis showed that metformin-induced significant alterations in metabolic pathways such as glutathione metabolism and polyamine synthesis. Integrative analysis of transcriptomes and metabolomics revealed that metformin suppressed glycolysis by downregulating the key glycolytic enzymes LDHA and PKM2 and upregulating IDH1 expression in thyroid cancer. Furthermore, the anti-tumor role of metformin in thyroid cancer in vivo was shown. Together these results show that metformin plays an anti-tumor role by inhibiting glycolysis and restraining DNA replication in thyroid cancer.
Collapse
Affiliation(s)
- Jielin Ouyang
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan 410005, PR China; Central Laboratory of Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, PR China; Key Laboratory of Molecular Epidemiology of Hunan Province, Hunan Normal University, Changsha, Hunan 410013, PR China
| | - Yang Feng
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan 410005, PR China; Central Laboratory of Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, PR China; Key Laboratory of Molecular Epidemiology of Hunan Province, Hunan Normal University, Changsha, Hunan 410013, PR China
| | - Yiyuan Zhang
- Central Laboratory of Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, PR China
| | - Yarong Liu
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan 410005, PR China; Central Laboratory of Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, PR China; Key Laboratory of Molecular Epidemiology of Hunan Province, Hunan Normal University, Changsha, Hunan 410013, PR China
| | - Shutong Li
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan 410005, PR China; Central Laboratory of Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, PR China; Key Laboratory of Molecular Epidemiology of Hunan Province, Hunan Normal University, Changsha, Hunan 410013, PR China
| | - Jingjing Wang
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan 410005, PR China; Central Laboratory of Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, PR China
| | - Lihong Tan
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan 410005, PR China; Central Laboratory of Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, PR China.
| | - Lianhong Zou
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital), Changsha, Hunan 410005, PR China; Central Laboratory of Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, PR China.
| |
Collapse
|
15
|
Zhou X, Sekino Y, Li HT, Fu G, Yang Z, Zhao S, Gujar H, Zu X, Weisenberger DJ, Gill IS, Tulpule V, D’souza A, Quinn DI, Han B, Liang G. SETD2 Deficiency Confers Sensitivity to Dual Inhibition of DNA Methylation and PARP in Kidney Cancer. Cancer Res 2023; 83:3813-3826. [PMID: 37695044 PMCID: PMC10843145 DOI: 10.1158/0008-5472.can-23-0401] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 07/18/2023] [Accepted: 09/06/2023] [Indexed: 09/12/2023]
Abstract
SETD2 deficiency alters the epigenetic landscape by causing depletion of H3K36me3 and plays an important role in diverse forms of cancer, most notably in aggressive and metastatic clear-cell renal cell carcinomas (ccRCC). Development of an effective treatment scheme targeting SETD2-compromised cancer is urgently needed. Considering that SETD2 is involved in DNA methylation and DNA repair, a combination treatment approach using DNA hypomethylating agents (HMA) and PARP inhibitors (PARPi) could have strong antitumor activity in SETD2-deficient kidney cancer. We tested the effects of the DNA HMA 5-aza-2'-dexoxydytidine (DAC), the PARPi talazoparib (BMN-673), and both in combination in human ccRCC models with or without SETD2 deficiency. The combination treatment of DAC and BMN-673 synergistically increased cytotoxicity in vitro in SETD2-deficient ccRCC cell lines but not in SETD2-proficient cell lines. DAC and BMN-673 led to apoptotic induction, increased DNA damage, insufficient DNA damage repair, and increased genomic instability. Furthermore, the combination treatment elevated immune responses, upregulated STING, and enhanced viral mimicry by activating transposable elements. Finally, the combination effectively suppressed the growth of SETD2-deficient ccRCC in in vivo mouse models. Together, these findings indicate that combining HMA and PARPi is a promising potential therapeutic strategy for treating SETD2-compromised ccRCC. SIGNIFICANCE SETD2 deficiency creates a vulnerable epigenetic status that is targetable using a DNA hypomethylating agent and PARP inhibitor combination to suppress renal cell carcinoma, identifying a precision medicine-based approach for SETD2-compromised cancers.
Collapse
Affiliation(s)
- Xinyi Zhou
- Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Urology, Xiangya Hospital, Central South University, Hunan, Changsha 410008, China
| | - Yohei Sekino
- Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hong-Tao Li
- Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Guanghou Fu
- Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Zhi Yang
- Department of Surgery, Keck School of Medicine of USC, Los Angeles, California; Department of Surgery and Biomedical Engineering, Keck School of Medicine USC, Los Angeles, CA, USA
| | - Shuqing Zhao
- Department of Surgery, Keck School of Medicine of USC, Los Angeles, California; Department of Surgery and Biomedical Engineering, Keck School of Medicine USC, Los Angeles, CA, USA
| | - Hemant Gujar
- Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Xiongbing Zu
- Department of Urology, Xiangya Hospital, Central South University, Hunan, Changsha 410008, China
| | - Daniel J Weisenberger
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Inderbir S. Gill
- Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Varsha Tulpule
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Anishka D’souza
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - David I Quinn
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Bo Han
- Department of Surgery, Keck School of Medicine of USC, Los Angeles, California; Department of Surgery and Biomedical Engineering, Keck School of Medicine USC, Los Angeles, CA, USA
| | - Gangning Liang
- Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
16
|
Genetta T, Hurwitz J, Clark E, Herold B, Khalil S, Abbas T, Larner J. ZEB1 promotes non-homologous end joining double-strand break repair. Nucleic Acids Res 2023; 51:9863-9879. [PMID: 37665026 PMCID: PMC10570029 DOI: 10.1093/nar/gkad723] [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: 07/20/2022] [Revised: 07/31/2023] [Accepted: 08/21/2023] [Indexed: 09/05/2023] Open
Abstract
Repair of DSB induced by IR is primarily carried out by Non-Homologous End Joining (NHEJ), a pathway in which 53BP1 plays a key role. We have discovered that the EMT-inducing transcriptional repressor ZEB1 (i) interacts with 53BP1 and that this interaction occurs rapidly and is significantly amplified following exposure of cells to IR; (ii) is required for the localization of 53BP1 to a subset of double-stranded breaks, and for physiological DSB repair; (iii) co-localizes with 53BP1 at IR-induced foci (IRIF); (iv) promotes NHEJ and inhibits Homologous Recombination (HR); (v) depletion increases resection at DSBs and (vi) confers PARP inhibitor (PARPi) sensitivity on BRCA1-deficient cells. Lastly, ZEB1's effects on repair pathway choice, resection, and PARPi sensitivity all rely on its homeodomain. In contrast to the well-characterized therapeutic resistance of high ZEB1-expressing cancer cells, the novel ZEB1-53BP1-shieldin resection axis described here exposes a therapeutic vulnerability: ZEB1 levels in BRCA1-deficient tumors may serve as a predictive biomarker of response to PARPis.
Collapse
Affiliation(s)
- Thomas L Genetta
- Dept. of Radiation Oncology, University of Virginia School of Medicine, PO Box 800383, Charlottesville, VA 22908, USA
| | - Joshua C Hurwitz
- Dept. of Radiation Oncology, University of Virginia School of Medicine, PO Box 800383, Charlottesville, VA 22908, USA
| | - Evan A Clark
- Dept. of Radiation Oncology, University of Virginia School of Medicine, PO Box 800383, Charlottesville, VA 22908, USA
| | - Benjamin T Herold
- Dept. of Radiation Oncology, University of Virginia School of Medicine, PO Box 800383, Charlottesville, VA 22908, USA
| | - Shadi Khalil
- Dept. of Radiation Oncology, University of Virginia School of Medicine, PO Box 800383, Charlottesville, VA 22908, USA
| | - Tarek Abbas
- Dept. of Radiation Oncology, University of Virginia School of Medicine, PO Box 800383, Charlottesville, VA 22908, USA
- Dept. of Biochemistry and Molecular Genetics University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - James M Larner
- Dept. of Radiation Oncology, University of Virginia School of Medicine, PO Box 800383, Charlottesville, VA 22908, USA
| |
Collapse
|
17
|
Li Q, Qian W, Zhang Y, Hu L, Chen S, Xia Y. A new wave of innovations within the DNA damage response. Signal Transduct Target Ther 2023; 8:338. [PMID: 37679326 PMCID: PMC10485079 DOI: 10.1038/s41392-023-01548-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 06/01/2023] [Accepted: 06/27/2023] [Indexed: 09/09/2023] Open
Abstract
Genome instability has been identified as one of the enabling hallmarks in cancer. DNA damage response (DDR) network is responsible for maintenance of genome integrity in cells. As cancer cells frequently carry DDR gene deficiencies or suffer from replicative stress, targeting DDR processes could induce excessive DNA damages (or unrepaired DNA) that eventually lead to cell death. Poly (ADP-ribose) polymerase (PARP) inhibitors have brought impressive benefit to patients with breast cancer gene (BRCA) mutation or homologous recombination deficiency (HRD), which proves the concept of synthetic lethality in cancer treatment. Moreover, the other two scenarios of DDR inhibitor application, replication stress and combination with chemo- or radio- therapy, are under active clinical exploration. In this review, we revisited the progress of DDR targeting therapy beyond the launched first-generation PARP inhibitors. Next generation PARP1 selective inhibitors, which could maintain the efficacy while mitigating side effects, may diversify the application scenarios of PARP inhibitor in clinic. Albeit with unavoidable on-mechanism toxicities, several small molecules targeting DNA damage checkpoints (gatekeepers) have shown great promise in preliminary clinical results, which may warrant further evaluations. In addition, inhibitors for other DNA repair pathways (caretakers) are also under active preclinical or clinical development. With these progresses and efforts, we envision that a new wave of innovations within DDR has come of age.
Collapse
Affiliation(s)
- Qi Li
- Domestic Discovery Service Unit, WuXi AppTec, 200131, Shanghai, China
| | - Wenyuan Qian
- Domestic Discovery Service Unit, WuXi AppTec, 200131, Shanghai, China
| | - Yang Zhang
- Domestic Discovery Service Unit, WuXi AppTec, 200131, Shanghai, China
| | - Lihong Hu
- Domestic Discovery Service Unit, WuXi AppTec, 200131, Shanghai, China
| | - Shuhui Chen
- Domestic Discovery Service Unit, WuXi AppTec, 200131, Shanghai, China
| | - Yuanfeng Xia
- Domestic Discovery Service Unit, WuXi AppTec, 200131, Shanghai, China.
| |
Collapse
|
18
|
Hara T, Nakaoka H, Miyoshi T, Ishikawa F. The CST complex facilitates cell survival under oxidative genotoxic stress. PLoS One 2023; 18:e0289304. [PMID: 37590191 PMCID: PMC10434909 DOI: 10.1371/journal.pone.0289304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 07/15/2023] [Indexed: 08/19/2023] Open
Abstract
Genomic DNA is constantly exposed to a variety of genotoxic stresses, and it is crucial for organisms to be equipped with mechanisms for repairing the damaged genome. Previously, it was demonstrated that the mammalian CST (CTC1-STN1-TEN1) complex, which was originally identified as a single-stranded DNA-binding trimeric protein complex essential for telomere maintenance, is required for survival in response to hydroxyurea (HU), which induces DNA replication fork stalling. It is still unclear, however, how the CST complex is involved in the repair of diverse types of DNA damage induced by oxidizing agents such as H2O2. STN1 knockdown (KD) sensitized HeLa cells to high doses of H2O2. While H2O2 induced DNA strand breaks throughout the cell cycle, STN1 KD cells were as resistant as control cells to H2O2 treatment when challenged in the G1 phase of the cell cycle, but they were sensitive when exposed to H2O2 in S/G2/M phase. STN1 KD cells showed a failure of DNA synthesis and RAD51 foci formation upon H2O2 treatment. Chemical inhibition of RAD51 in shSTN1 cells did not exacerbate the sensitivity to H2O2, implying that the CST complex and RAD51 act in the same pathway. Collectively, our results suggest that the CST complex is required for maintaining genomic stability in response to oxidative DNA damage, possibly through RAD51-dependent DNA repair/protection mechanisms.
Collapse
Affiliation(s)
- Tomohiko Hara
- Department of Gene Mechanisms, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Hidenori Nakaoka
- Department of Gene Mechanisms, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Tomoicihiro Miyoshi
- Department of Gene Mechanisms, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Laboratory for Retrotransposon Dynamics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Fuyuki Ishikawa
- Department of Gene Mechanisms, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| |
Collapse
|
19
|
Yang B, Zhang W, Sun L, Lu B, Yin C, Zhang Y, Jiang H. Creatine kinase brain-type regulates BCAR1 phosphorylation to facilitate DNA damage repair. iScience 2023; 26:106684. [PMID: 37182100 PMCID: PMC10173731 DOI: 10.1016/j.isci.2023.106684] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/07/2023] [Accepted: 04/13/2023] [Indexed: 05/16/2023] Open
Abstract
Creatine kinase (CK) is an essential metabolic enzyme mediating creatine/phosphocreatine interconversion and shuttle to replenish ATP for energy needs. Ablation of CK causes a deficiency in energy supply that eventually results in reduced muscle burst activity and neurological disorders in mice. Besides the well-established role of CK in energy-buffering, the mechanism underlying the non-metabolic function of CK is poorly understood. Here we demonstrate that creatine kinase brain-type (CKB) may function as a protein kinase to regulate BCAR1 Y327 phosphorylation that enhances the association between BCAR1 and RBBP4. Then the complex of BCAR1 and RPPB4 binds to the promoter region of DNA damage repair gene RAD51 and activates its transcription by modulating histone H4K16 acetylation to ultimately promote DNA damage repair. These findings reveal the possible role of CKB independently of its metabolic function and depict the potential pathway of CKB-BCAR1-RBBP4 operating in DNA damage repair.
Collapse
Affiliation(s)
- Bo Yang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Wentao Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Le Sun
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bin Lu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Changsong Yin
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Yaoyang Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hong Jiang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Corresponding author
| |
Collapse
|
20
|
Andriuskevicius T, Dubenko A, Makovets S. The Inability to Disassemble Rad51 Nucleoprotein Filaments Leads to Aberrant Mitosis and Cell Death. Biomedicines 2023; 11:1450. [PMID: 37239121 PMCID: PMC10216663 DOI: 10.3390/biomedicines11051450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 04/30/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
The proper maintenance of genetic material is essential for the survival of living organisms. One of the main safeguards of genome stability is homologous recombination involved in the faithful repair of DNA double-strand breaks, the restoration of collapsed replication forks, and the bypass of replication barriers. Homologous recombination relies on the formation of Rad51 nucleoprotein filaments which are responsible for the homology-based interactions between DNA strands. Here, we demonstrate that without the regulation of these filaments by Srs2 and Rad54, which are known to remove Rad51 from single-stranded and double-stranded DNA, respectively, the filaments strongly inhibit damage-associated DNA synthesis during DNA repair. Furthermore, this regulation is essential for cell survival under normal growth conditions, as in the srs2Δ rad54Δ mutants, unregulated Rad51 nucleoprotein filaments cause activation of the DNA damage checkpoint, formation of mitotic bridges, and loss of genetic material. These genome instability features may stem from the problems at stalled replication forks as the lack of Srs2 and Rad54 in the presence of Rad51 nucleoprotein filaments impedes cell recovery from replication stress. This study demonstrates that the timely and efficient disassembly of recombination machinery is essential for genome maintenance and cell survival.
Collapse
Affiliation(s)
| | | | - Svetlana Makovets
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Alexander Crum Brown Road, Edinburgh EH9 3FF, UK
| |
Collapse
|
21
|
Fukasawa T, Enomoto A, Yoshizaki-Ogawa A, Sato S, Miyagawa K, Yoshizaki A. The Role of Mammalian STK38 in DNA Damage Response and Targeting for Radio-Sensitization. Cancers (Basel) 2023; 15:cancers15072054. [PMID: 37046714 PMCID: PMC10093458 DOI: 10.3390/cancers15072054] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/13/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Protein kinases, found in the nucleus and cytoplasm, play essential roles in a multitude of cellular processes, including cell division, proliferation, apoptosis, and signal transduction. STK38 is a member of the protein kinase A (PKA)/PKG/PKC family implicated in regulating cell division and morphogenesis in yeast and C. elegans. However, its function remained largely unknown in mammals. In recent years, advances in research on STK38 and the identification of its substrates has led to a better understanding of its function and role in mammals. This review discusses the structure, expression, and regulation of activity as a kinase, its role in the DNA damage response, cross-talk with other signaling pathways, and its application for radio-sensitization.
Collapse
|
22
|
Kabirova E, Nurislamov A, Shadskiy A, Smirnov A, Popov A, Salnikov P, Battulin N, Fishman V. Function and Evolution of the Loop Extrusion Machinery in Animals. Int J Mol Sci 2023; 24:5017. [PMID: 36902449 PMCID: PMC10003631 DOI: 10.3390/ijms24055017] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 02/25/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Structural maintenance of chromosomes (SMC) complexes are essential proteins found in genomes of all cellular organisms. Essential functions of these proteins, such as mitotic chromosome formation and sister chromatid cohesion, were discovered a long time ago. Recent advances in chromatin biology showed that SMC proteins are involved in many other genomic processes, acting as active motors extruding DNA, which leads to the formation of chromatin loops. Some loops formed by SMC proteins are highly cell type and developmental stage specific, such as SMC-mediated DNA loops required for VDJ recombination in B-cell progenitors, or dosage compensation in Caenorhabditis elegans and X-chromosome inactivation in mice. In this review, we focus on the extrusion-based mechanisms that are common for multiple cell types and species. We will first describe an anatomy of SMC complexes and their accessory proteins. Next, we provide biochemical details of the extrusion process. We follow this by the sections describing the role of SMC complexes in gene regulation, DNA repair, and chromatin topology.
Collapse
Affiliation(s)
- Evelyn Kabirova
- Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Artem Nurislamov
- Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Artem Shadskiy
- Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Alexander Smirnov
- Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Andrey Popov
- Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Pavel Salnikov
- Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Nariman Battulin
- Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Veniamin Fishman
- Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
- Artificial Intelligence Research Institute (AIRI), 121108 Moscow, Russia
| |
Collapse
|
23
|
Pu J, Teng Z, Zhang T, Wang B, Zhang D, Yang Q, Yang Q, Sun X, Long W. Expression of Polyadenylate-binding Protein Cytoplasmic 1 (PABPC1) in Combination With RAD51 as Prognostic Biomarker in Patients Who Underwent Postoperative Chemotherapy for Esophageal Squamous Cell Carcinoma. Appl Immunohistochem Mol Morphol 2023; 31:189-195. [PMID: 36735495 DOI: 10.1097/pai.0000000000001100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 12/30/2022] [Indexed: 02/04/2023]
Abstract
Molecular markers in the prognosis of esophageal squamous cell carcinoma (ESCC) patients who received postoperative treatments are lacking. This research aims to evaluate the prognostic value of polyadenylate-binding protein cytoplasmic 1 (PABPC1) alone and in combination with RAD51 in ESCC patients who underwent postoperative chemotherapy (CT). A total of 103 ESCC patients who underwent postoperative CT and 103 matched ones who received surgery alone were analyzed in this study. PABPC1 and RAD51 expression was assessed in cancer samples by immunohistochemistry. PABPC1 high expression (PABPC1-HE) but not that of RAD51 was associated with poor patients' survival, regardless of the postoperative treatment or node status. Patients with PABPC1 low expression and RAD51 negative expression [RAD51- (PABPC1-LE/RAD51-)] tumor had good overall survival (OS) in both the CT treated and untreated groups. Patients with PABPC1-LE/RAD51+ and PABPC1-HE/RAD51+ tumors had longer OS in the CT treated group than in the untreated group. However, PABPC1-HE/RAD51- was associated with a poor outcome in both groups and the patients with PABPC1-HE/RAD51- tumor had hardly any benefit from CT in N+ status. PABPC1 alone and in combination with RAD51 was a prognostic biomarker for OS in ESCC patients who received postoperative CT.
Collapse
Affiliation(s)
| | | | | | | | | | - Qin Yang
- Pathology, Affiliated Hospital of Southwest Medical University, Sichuan, China
| | - Qiao Yang
- Pathology, Affiliated Hospital of Southwest Medical University, Sichuan, China
| | - Xingwang Sun
- Pathology, Affiliated Hospital of Southwest Medical University, Sichuan, China
| | - Wenbo Long
- Pathology, Affiliated Hospital of Southwest Medical University, Sichuan, China
| |
Collapse
|
24
|
Kim K, Kim SH, Lee JY, Kim YN, Lee ST, Park E. RAD51/geminin/γH2AX immunohistochemical expression predicts platinum-based chemotherapy response in ovarian high-grade serous carcinoma. J Gynecol Oncol 2023:34.e45. [PMID: 36807748 DOI: 10.3802/jgo.2023.34.e45] [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: 09/26/2022] [Revised: 12/28/2022] [Accepted: 01/18/2023] [Indexed: 02/11/2023] Open
Abstract
OBJECTIVE The RAD51 assay is a recently developed functional assay for homologous recombination deficiency (HRD) that reflects real-time HRD status. We aimed to identify the applicability and predictive value of RAD51 immunohistochemical expression in pre- and post-neoadjuvant chemotherapy (NAC) samples of ovarian high-grade serous carcinoma (HGSC). METHODS We evaluated the immunohistochemical expression of RAD51/geminin/γH2AX in ovarian HGSC before and after NAC. RESULTS In pre-NAC tumors (n=51), 74.5% (39/51) showed at least 25% of γH2AX-positive tumor cells, suggesting endogenous DNA damage. The RAD51-high group (41.0%, 16/39) showed significantly worse progression-free survival (PFS) compared to the RAD51-low group (51.3%, 20/39) (p=0.032). In post-NAC tumors (n=50), the RAD51-high group (36.0%, 18/50) showed worse PFS (p=0.013) and tended to present worse overall survival (p=0.067) compared to the RAD51-low group (64.0%, 32/50). RAD51-high cases were more likely to progress than RAD51-low cases at both 6 months and 12 months (p=0.046 and p=0.019, respectively). Of 34 patients with matched pre- and post-NAC RAD51 results, 44% (15/34) of pre-NAC RAD51 results were changed in the post-NAC tissue, and the RAD51 high-to-high group showed the worst PFS, while the low-to-low group showed the best PFS (p=0.031). CONCLUSION High RAD51 expression was significantly associated with worse PFS in HGSC, and post-NAC RAD51 status showed higher association than pre-NAC RAD51 status. Moreover, RAD51 status can be evaluated in a significant proportion of treatment-naïve HGSC samples. As RAD51 status dynamically changes, sequential follow-up of RAD51 status might reflect the biological behavior of HGSCs.
Collapse
Affiliation(s)
- Kyeongmin Kim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.,Department of Pathology, Soonchunhyang University, Seoul, Korea
| | - Se Hoon Kim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jung-Yun Lee
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Yoo-Na Kim
- Department of Obstetrics and Gynecology, Institute of Women's Life Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Seung-Tae Lee
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Eunhyang Park
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
25
|
Chu C, Liu D, Wang D, Hu S, Zhang Y. Identification and development of TP53 mutation-associated Long non-coding RNAs signature for optimized prognosis assessment and treatment selection in hepatocellular carcinoma. Int J Immunopathol Pharmacol 2023; 37:3946320231211795. [PMID: 37942552 PMCID: PMC10637161 DOI: 10.1177/03946320231211795] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 10/17/2023] [Indexed: 11/10/2023] Open
Abstract
BACKGROUND The TP53 gene is estimated to be mutated in over 50% of tumors, with the majority of tumors exhibiting abnormal TP53 signaling pathways. However, the exploration of TP53 mutation-related LncRNAs in Hepatocellular carcinoma (HCC) remains incomplete. This study aims to identify such LncRNAs and enhance the prognostic accuracy for Hepatoma patients. MATERIAL AND METHODS Differential gene expression was identified using the "limma" package in R. Prognosis-related LncRNAs were identified via univariate Cox regression analysis, while a prognostic model was crafted using multivariate Cox regression analysis. Survival analysis was conducted using Kaplan-Meier curves. The precision of the prognostic model was assessed through ROC analysis. Subsequently, the Tumor Immune Dysfunction and Exclusion (TIDE) algorithm were executed on the TCGA dataset via the TIDE database. Fractions of 24 types of immune cell infiltration were obtained from NCI Cancer Research Data Commons using deconvolution techniques. The protein expression levels encoded by specific genes were obtained through the TPCA database. RESULTS In this research, we have identified 85 LncRNAs associated with TP53 mutations and developed a corresponding signature referred to as TP53MLncSig. Kaplan-Meier analysis revealed a lower 3-year survival rate in high-risk patients (46.9%) compared to low-risk patients (74.2%). The accuracy of the prognostic TP53MLncSig was further evaluated by calculating the area under the ROC curve. The analysis yielded a 5-year ROC score of 0.793, confirming its effectiveness. Furthermore, a higher score for TP53MLncSig was found to be associated with an increased response rate to immune checkpoint blocker (ICB) therapy (p = .005). Patients possessing high-risk classification exhibited lower levels of P53 protein expression and higher levels of genomic instability. CONCLUSION The present study aimed to identify and validate LncRNAs associated with TP53 mutations. We constructed a prognostic model that can predict chemosensitivity and response to ICB therapy in HCC patients. This novel approach sheds light on the role of LncRNAs in TP53 mutation and provides valuable resources for analyzing patient prognosis and treatment selection.
Collapse
Affiliation(s)
- Chenghao Chu
- Department of General Surgery, Anqing First People's Hospital Affiliated to Anhui Medical University, Anqing, China
- Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu, Malaysia
| | - Daoli Liu
- Department of General Surgery, Anqing First People's Hospital Affiliated to Anhui Medical University, Anqing, China
| | - Duofa Wang
- Department of General Surgery, Anqing First People's Hospital Affiliated to Anhui Medical University, Anqing, China
| | - Shuangjiu Hu
- Department of General Surgery, Anqing First People's Hospital Affiliated to Anhui Medical University, Anqing, China
| | - Yongwei Zhang
- Department of General Surgery, Anqing First People's Hospital Affiliated to Anhui Medical University, Anqing, China
- Department of Biomedical Sciences, Faculty of Medicine & Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu, Malaysia
| |
Collapse
|
26
|
RAD51 paralogs: Expanding roles in replication stress responses and repair. Curr Opin Pharmacol 2022; 67:102313. [PMID: 36343481 DOI: 10.1016/j.coph.2022.102313] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/26/2022] [Accepted: 09/29/2022] [Indexed: 11/06/2022]
Abstract
Mammalian RAD51 paralogs are essential for cell survival and are critical for RAD51-mediated repair of DNA double-strand breaks (DSBs) by homologous recombination (HR). However, the molecular mechanism by which RAD51 paralogs participate in HR is largely unclear. Germline mutations in RAD51 paralogs are associated with breast and ovarian cancers and Fanconi anemia-like disorder, underscoring the crucial roles of RAD51 paralogs in genome maintenance and tumor suppression. Despite their discovery over three decades ago, the essential functions of RAD51 paralogs in cell survival and genome stability remain obscure. Recent studies unravel DSB repair independent functions of RAD51 paralogs in replication stress responses. Here, we highlight the recent findings that uncovered the novel functions of RAD51 paralogs in replication fork progression, its stability, and restart and discuss RAD51 paralogs as a potential therapeutic target for cancer treatment.
Collapse
|
27
|
Wilson J, Loizou JI. Exploring the genetic space of the DNA damage response for cancer therapy through CRISPR-based screens. Mol Oncol 2022; 16:3778-3791. [PMID: 35708734 PMCID: PMC9627789 DOI: 10.1002/1878-0261.13272] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/11/2022] [Accepted: 06/14/2022] [Indexed: 12/24/2022] Open
Abstract
The concepts of synthetic lethality and viability have emerged as powerful approaches to identify vulnerabilities and resistances within the DNA damage response for the treatment of cancer. Historically, interactions between two genes have had a longstanding presence in genetics and have been identified through forward genetic screens that rely on the molecular basis of the characterized phenotypes, typically caused by mutations in single genes. While such complex genetic interactions between genes have been studied extensively in model organisms, they have only recently been prioritized as therapeutic strategies due to technological advancements in genetic screens. Here, we discuss synthetic lethal and viable interactions within the DNA damage response and present how CRISPR-based genetic screens and chemical compounds have allowed for the systematic identification and targeting of such interactions for the treatment of cancer.
Collapse
Affiliation(s)
- Jordan Wilson
- Center for Cancer Research, Comprehensive Cancer CentreMedical University of ViennaAustria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Joanna I. Loizou
- Center for Cancer Research, Comprehensive Cancer CentreMedical University of ViennaAustria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| |
Collapse
|
28
|
Petermann E. Conflicts with transcription make early replication late. Mol Cell 2022; 82:3315-3317. [PMID: 36113410 DOI: 10.1016/j.molcel.2022.08.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 08/17/2022] [Accepted: 08/17/2022] [Indexed: 11/19/2022]
Abstract
By sequencing sites of mitotic DNA synthesis in cells lacking homologous recombination, Groelly, Bhowmick, and colleagues show how conflicts between transcription and replication in early S phase can cause under-replicated DNA to persist into mitosis.
Collapse
Affiliation(s)
- Eva Petermann
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2TT, UK; Birmingham Centre for Genome Biology, University of Birmingham, Birmingham B15 2TT, UK.
| |
Collapse
|
29
|
Hicks T, Koury E, McCabe C, Williams C, Crahan C, Smolikove S. R-loop-induced irreparable DNA damage evades checkpoint detection in the C. elegans germline. Nucleic Acids Res 2022; 50:8041-8059. [PMID: 35871299 PMCID: PMC9371901 DOI: 10.1093/nar/gkac621] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 06/28/2022] [Accepted: 07/18/2022] [Indexed: 11/24/2022] Open
Abstract
Accumulation of DNA–RNA hybrids in the form of R-loops can result in replication–transcription conflict that leads to the formation of DNA double strand breaks (DSBs). Using null mutants for the two Caenorhabditis elegans genes encoding for RNaseH1 and RNaseH2, we identify novel effects of R-loop accumulation in the germline. R-loop accumulation leads, as expected, to replication stress, followed by the formation of DSBs. A subset of these DSBs are irreparable. However, unlike irreparable DSBs generated in other systems, which trigger permanent cell cycle arrest, germline irreparable DSBs are propagated to oocytes. Despite DNA damage checkpoint activation in the stem cell niche, the signaling cannot be sustained and nuclei with irreparable DNA damage progress into meiosis. Moreover, unlike other forms of DNA damage that increase germline apoptosis, R-loop-generated DSBs remain undetected by the apoptotic checkpoint. This coincides with attenuation of ATM/ATR signaling in mid-to-late meiotic prophase I. These data altogether indicate that in the germline, DSBs that are generated by R-loops can lead to irreparable DSBs that evade cellular machineries designed for damage recognition. These studies implicate germline R-loops as an especially dangerous driver of germline mutagenesis.
Collapse
Affiliation(s)
- Tara Hicks
- Department of Biology, The University of Iowa , IA City, IA 52242, USA
| | - Emily Koury
- Department of Biology, The University of Iowa , IA City, IA 52242, USA
| | - Caleb McCabe
- Department of Biology, The University of Iowa , IA City, IA 52242, USA
| | - Cameron Williams
- Department of Biology, The University of Iowa , IA City, IA 52242, USA
| | - Caroline Crahan
- Department of Biology, The University of Iowa , IA City, IA 52242, USA
| | - Sarit Smolikove
- Department of Biology, The University of Iowa , IA City, IA 52242, USA
| |
Collapse
|
30
|
Yu R, Hu Y, Zhang S, Li X, Tang M, Yang M, Wu X, Li Z, Liao X, Xu Y, Li M, Chen S, Qian W, Gong LY, Song L, Li J. LncRNA CTBP1-DT-encoded microprotein DDUP sustains DNA damage response signalling to trigger dual DNA repair mechanisms. Nucleic Acids Res 2022; 50:8060-8079. [PMID: 35849344 PMCID: PMC9371908 DOI: 10.1093/nar/gkac611] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 06/21/2022] [Accepted: 07/01/2022] [Indexed: 11/14/2022] Open
Abstract
Sustaining DNA damage response (DDR) signalling via retention of DDR factors at damaged sites is important for transmitting damage-sensing and repair signals. Herein, we found that DNA damage provoked the association of ribosomes with IRES region in lncRNA CTBP1-DT, which overcame the negative effect of upstream open reading frames (uORFs), and elicited the novel microprotein DNA damage-upregulated protein (DDUP) translation via a cap-independent translation mechanism. Activated ATR kinase-mediated phosphorylation of DDUP induced a drastic 'dense-to-loose' conformational change, which sustained the RAD18/RAD51C and RAD18/PCNA complex at damaged sites and initiated RAD51C-mediated homologous recombination and PCNA-mediated post-replication repair mechanisms. Importantly, treatment with ATR inhibitor abolished the effect of DDUP on chromatin retention of RAD51C and PCNA, thereby leading to hypersensitivity of cancer cells to DNA-damaging chemotherapeutics. Taken together, our results uncover a plausible mechanism underlying the DDR sustaining and might represent an attractive therapeutic strategy in improvement of DNA damage-based anticancer therapies.
Collapse
Affiliation(s)
- Ruyuan Yu
- Program of Cancer Research, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, China.,Department of Biochemistry, Zhongshan school of medicine, Sun Yat-sen University, China
| | - Yameng Hu
- Program of Cancer Research, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, China.,Department of Biochemistry, Zhongshan school of medicine, Sun Yat-sen University, China
| | - Shuxia Zhang
- Program of Cancer Research, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, China.,Department of Biochemistry, Zhongshan school of medicine, Sun Yat-sen University, China
| | - Xincheng Li
- Program of Cancer Research, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, China.,Department of Biochemistry, Zhongshan school of medicine, Sun Yat-sen University, China
| | - Miaoling Tang
- Program of Cancer Research, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, China.,Department of Biochemistry, Zhongshan school of medicine, Sun Yat-sen University, China
| | - Meisongzhu Yang
- Program of Cancer Research, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, China.,Department of Biochemistry, Zhongshan school of medicine, Sun Yat-sen University, China
| | - Xingui Wu
- Program of Cancer Research, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, China.,Department of Biochemistry, Zhongshan school of medicine, Sun Yat-sen University, China
| | - Ziwen Li
- Program of Cancer Research, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, China.,Department of Biochemistry, Zhongshan school of medicine, Sun Yat-sen University, China
| | - Xinyi Liao
- Program of Cancer Research, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, China.,Department of Biochemistry, Zhongshan school of medicine, Sun Yat-sen University, China
| | - Yingru Xu
- Program of Cancer Research, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, China.,Department of Biochemistry, Zhongshan school of medicine, Sun Yat-sen University, China
| | - Man Li
- Program of Cancer Research, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, China.,Department of Biochemistry, Zhongshan school of medicine, Sun Yat-sen University, China
| | - Suwen Chen
- Program of Cancer Research, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, China.,Department of Biochemistry, Zhongshan school of medicine, Sun Yat-sen University, China
| | - Wanying Qian
- Program of Cancer Research, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, China.,Department of Biochemistry, Zhongshan school of medicine, Sun Yat-sen University, China
| | - Li-Yun Gong
- Guangdong Provincial Key Laboratory for Genome Stability and Disease Prevention, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Health Science Center, Shenzhen University, China
| | - Libing Song
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, China
| | - Jun Li
- Program of Cancer Research, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, China.,Department of Biochemistry, Zhongshan school of medicine, Sun Yat-sen University, China
| |
Collapse
|
31
|
Wu X, Wu J, Wang L, Yang W, Wang B, Yang H. CircRNAs in Malignant Tumor Radiation: The New Frontier as Radiotherapy Biomarkers. Front Oncol 2022; 12:854678. [PMID: 35372031 PMCID: PMC8966018 DOI: 10.3389/fonc.2022.854678] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/18/2022] [Indexed: 12/14/2022] Open
Abstract
World Health Organization (WHO) data show that of the top 20 factors that threaten human life and health, cancer is at the forefront, and the therapeutic approaches for cancer consist of surgery, radiotherapy, chemotherapy and immunotherapy. For most highly metastatic and recurrent cancer, radiation therapy is an essential modality to mitigate tumor burden and improve patient survival. Despite the great accomplishments that have been made in clinical therapy, an inevitable challenge in effective treatment is radioresistance, the mechanisms of which have not yet been completely elucidated. In addition, radiosensitization methods based on molecular mechanisms and targets, and clinical applications are still inadequate. Evidence indicates that circular RNAs (circRNAs) are important components in altering tumor progression, and in influencing resistance and susceptibility to radiotherapy. This review summarizes the reasons for tumor radiotherapy resistance induced by circRNAs, and clarifies the molecular mechanisms and targets of action. Moreover, we determine the potential value of circRNAs as clinical indicators in radiotherapy, providing a theoretical basis for circRNAs-based strategies for cancer radiotherapy.
Collapse
Affiliation(s)
- Xixi Wu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Junying Wu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China.,Department of Clinical Laboratory, The Children's Hospital of Soochow University, Suzhou, China
| | - Lingxia Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Wei Yang
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Bo Wang
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Huan Yang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China.,State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| |
Collapse
|
32
|
Huang C, Zhou S, Zhang C, Jin Y, Xu G, Zhou L, Ding G, Pang T, Jia S, Cao L. ZC3H13-mediated N6-methyladenosine modification of PHF10 is impaired by fisetin which inhibits the DNA damage response in pancreatic cancer. Cancer Lett 2022; 530:16-28. [PMID: 35033590 DOI: 10.1016/j.canlet.2022.01.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 01/11/2022] [Accepted: 01/11/2022] [Indexed: 12/13/2022]
Abstract
DNA damage repair is a major barrier for chemotherapy efficacy of pancreatic ductal adenocarcinoma (PDAC), including the efficacy of platinum-based and gemcitabine/nab-paclitaxel treatments. N6-methyladenosine modifications (m6A) have recently been reported to play a role in homologous recombination (HR) repair of DNA double strand breaks (DSBs); however, the mechanism of action remains unknown. Our previous work indicated that fisetin may be a promising anti-tumour agent that induces DNA damage. In this study, we reported that fisetin induced DSBs and suppressed HR repair through m6A modification in PDAC cells. The m6A writer ZC3H13 and PHF10, which is a subunit of the PBAF chromatin remodelling complex, were identified as the main molecules affected by fisetin treatment. To our knowledge, it's the first time that PHF10 was found and involved in the DNA damage response. PHF10 loss-of-function resulted in elevated recruitment of γH2AX, RAD51, and 53BP1 to DSB sites and decreased HR repair efficiency. Moreover, ZC3H13 knockdown downregulated the m6A methylation of PHF10 and decreased PHF10 translation in a YTHDF1-dependent manner. In conclusion, our study demonstrates that fisetin enhanced DSBs via ZC3Hl3-mediated m6A modification of PHF10, which may provide insight into novel therapeutic approaches for PDAC.
Collapse
Affiliation(s)
- Chaojie Huang
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China.
| | - Senhao Zhou
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China.
| | - Chaolei Zhang
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China.
| | - Yifeng Jin
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China.
| | - Gao Xu
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China.
| | - Liangjing Zhou
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China.
| | - Guoping Ding
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China.
| | - Tianshu Pang
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China.
| | - Shengnan Jia
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China.
| | - Liping Cao
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China; Zhejiang Engineering Research Center of Cognitive Healthcare, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China.
| |
Collapse
|
33
|
Qin J, Huang T, Wang J, Xu L, Dang Q, Xu X, Liu H, Liu Z, Shao C, Zhang X. RAD51 is essential for spermatogenesis and male fertility in mice. Cell Death Dis 2022; 8:118. [PMID: 35292640 PMCID: PMC8924220 DOI: 10.1038/s41420-022-00921-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 02/15/2022] [Accepted: 02/24/2022] [Indexed: 11/21/2022]
Abstract
The recombinase RAD51 catalyzes the DNA strand exchange reaction in homologous recombination (HR) during both mitosis and meiosis. However, the physiological role of RAD51 during spermatogenesis remains unclear since RAD51 null mutation is embryonic lethal in mice. In this study, we generated a conditional knockout mouse model to study the role of RAD51 in spermatogenesis. Conditional disruption of RAD51 in germ cells by Vasa-Cre led to spermatogonial loss and Sertoli cell-only syndrome. Furthermore, tamoxifen-inducible RAD51 knockout by UBC-CreERT2 confirmed that RAD51 deletion led to early spermatogenic cells loss and apoptosis. Notably, inducible knockout of RAD51 in adult mice caused defects in meiosis, with accumulated meiotic double-strand breaks (DSBs), reduced numbers of pachytene spermatocytes and less crossover formation. Our study revealed an essential role for Rad51 in the maintenance of spermatogonia as well as meiotic progression in mice.
Collapse
Affiliation(s)
- Junchao Qin
- Key Laboratory of Experimental Teratology, Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tao Huang
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jing Wang
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Limei Xu
- Key Laboratory of Experimental Teratology, Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qianli Dang
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiuhua Xu
- Department of Reproductive Medicine, Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hongbin Liu
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhaojian Liu
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Changshun Shao
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China.
| | - Xiyu Zhang
- Key Laboratory of Experimental Teratology, Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
34
|
Chen B, Xu F, Gao Y, Hu G, Zhu K, Lu H, Xu A, Chen S, Wu L, Zhao G. DNA damage-induced translocation of mitochondrial factor HIGD1A into the nucleus regulates homologous recombination and radio/chemo-sensitivity. Oncogene 2022; 41:1918-1930. [DOI: 10.1038/s41388-022-02226-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 01/17/2022] [Accepted: 02/01/2022] [Indexed: 12/13/2022]
|
35
|
Design, synthesis and mechanism studies of novel dual PARP1/BRD4 inhibitors against pancreatic cancer. Eur J Med Chem 2022; 230:114116. [DOI: 10.1016/j.ejmech.2022.114116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 12/08/2021] [Accepted: 01/09/2022] [Indexed: 11/23/2022]
|
36
|
Qiu M, Tu L, Zhao M, Yang M, Qi J, Xie Y, Gu J. Ataxia-televangelist mutated (ATM)/ ATR serine/threonine kinase (ATR)-mediated RAD51 recombinase (RAD51) promotes osteogenic differentiation and inhibits osteoclastogenesis in osteoporosis. Bioengineered 2022; 13:4201-4211. [PMID: 35176943 PMCID: PMC8974111 DOI: 10.1080/21655979.2022.2026729] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Osteoporosis is a metabolic bone disease that significantly affects the quality of life and can even lead to death. In this study, we aimed to investigate the role of RAD51 recombinase (RAD51) in osteoblast and osteoclast differentiation. We analyzed differentially expressed genes using microarray analysis. The osteogenic differentiation capability was analyzed by alkaline phosphatase (ALP) staining and alizarin red staining assays. Osteogenesis and osteoclast related genes expression was detected using quantitative real-time PCR (qPCR) and Western blotting. The phosphorylation of Ataxia-telangiectasia mutated (ATM) and ATR serine/threonine kinase (ATR) was tested using Western blotting. The effect of RAD51 on osteoporosis was also explored in vivo. The results showed that RAD51 was downregulated in osteoporosis, but upregulated in differentiated osteoblasts. Overexpression of RAD51 enhanced the differentiation of osteoblasts and suppressed the formation of osteoclasts. Furthermore, p-ATM and p-ATR levels were upregulated in osteoblasts and downregulated in osteoclasts. RAD51 expression was reduced by the ATM/ATR pathway inhibitor AZ20. AZ20 treatment inhibited osteoblastogenesis and promoted osteoclastogenesis, whereas RAD51 reversed the effects induced by AZ20. Moreover, RAD51 improved bone microarchitecture in vivo. Taken together, ATM/ATR signaling-mediated RAD51 promoted osteogenic differentiation and suppressed osteoclastogenesis. These findings reveal a critical role for RAD51 in osteoporosis.
Collapse
Affiliation(s)
- Minli Qiu
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Liudan Tu
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Minjing Zhao
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Mingcan Yang
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jun Qi
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Ya Xie
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jieruo Gu
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
37
|
Lan B, Zeng S, Zhang S, Ren X, Xing Y, Kutschick I, Pfeffer S, Frey B, Britzen-Laurent N, Grützmann R, Cordes N, Pilarsky C. CRISPR-Cas9 Screen Identifies DYRK1A as a Target for Radiotherapy Sensitization in Pancreatic Cancer. Cancers (Basel) 2022; 14:cancers14020326. [PMID: 35053488 PMCID: PMC8773906 DOI: 10.3390/cancers14020326] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/03/2022] [Accepted: 01/04/2022] [Indexed: 12/17/2022] Open
Abstract
Simple Summary Pancreatic cancer is the fourth leading cause of cancer-related death in Western countries. Although several therapeutic strategies have been developed for pancreatic cancer, radiation therapy has not yet yielded satisfactory results. Unraveling the mechanism of radioresistance in pancreatic cancer and developing new therapeutic targets has become a major challenge. Therefore, we applied kinome-wide CRISPR-Cas9 loss-of-function screening combined with the 3D cell culture method and identified DYRK1A as a sensitive target for radiotherapy. Additionally, we confirmed that DYRK1A-targeted inhibitors could enhance the efficacy of radiotherapy. Our results further support the use of CRISPR-Cas9 screening to identify novel therapeutic targets and develop new strategies to enhance radiotherapy efficacy in pancreatic cancer. Abstract Although radiation therapy has recently made great advances in cancer treatment, the majority of patients diagnosed with pancreatic cancer (PC) cannot achieve satisfactory outcomes due to intrinsic and acquired radioresistance. Identifying the molecular mechanisms that impair the efficacy of radiotherapy and targeting these pathways are essential to improve the radiation response of PC patients. Our goal is to identify sensitive targets for pancreatic cancer radiotherapy (RT) using the kinome-wide CRISPR-Cas9 loss-of-function screen and enhance the therapeutic effect through the development and application of targeted inhibitors combined with radiotherapy. We transduced pancreatic cancer cells with a protein kinase library; 2D and 3D library cells were irradiated daily with a single dose of up to 2 Gy for 4 weeks for a total of 40 Gy using an X-ray generator. Sufficient DNA was collected for next-generation deep sequencing to identify candidate genes. In this study, we identified several cell cycle checkpoint kinases and DNA damage related kinases in 2D- and 3D-cultivated cells, including DYRK1A, whose loss of function sensitizes cells to radiotherapy. Additionally, we demonstrated that the harmine-targeted suppression of DYRK1A used in conjunction with radiotherapy increases DNA double-strand breaks (DSBs) and impairs homologous repair (HR), resulting in more cancer cell death. Our results support the use of CRISPR-Cas9 screening to identify new therapeutic targets, develop radiosensitizers, and provide novel strategies for overcoming the tolerance of pancreatic cancer to radiotherapy.
Collapse
Affiliation(s)
- Bin Lan
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (B.L.); (S.Z.); (S.Z.); (X.R.); (Y.X.); (I.K.); (S.P.); (N.B.-L.); (R.G.)
| | - Siyuan Zeng
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (B.L.); (S.Z.); (S.Z.); (X.R.); (Y.X.); (I.K.); (S.P.); (N.B.-L.); (R.G.)
| | - Shuman Zhang
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (B.L.); (S.Z.); (S.Z.); (X.R.); (Y.X.); (I.K.); (S.P.); (N.B.-L.); (R.G.)
| | - Xiaofan Ren
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (B.L.); (S.Z.); (S.Z.); (X.R.); (Y.X.); (I.K.); (S.P.); (N.B.-L.); (R.G.)
| | - Yuming Xing
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (B.L.); (S.Z.); (S.Z.); (X.R.); (Y.X.); (I.K.); (S.P.); (N.B.-L.); (R.G.)
| | - Isabella Kutschick
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (B.L.); (S.Z.); (S.Z.); (X.R.); (Y.X.); (I.K.); (S.P.); (N.B.-L.); (R.G.)
| | - Susanne Pfeffer
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (B.L.); (S.Z.); (S.Z.); (X.R.); (Y.X.); (I.K.); (S.P.); (N.B.-L.); (R.G.)
| | - Benjamin Frey
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany;
| | - Nathalie Britzen-Laurent
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (B.L.); (S.Z.); (S.Z.); (X.R.); (Y.X.); (I.K.); (S.P.); (N.B.-L.); (R.G.)
| | - Robert Grützmann
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (B.L.); (S.Z.); (S.Z.); (X.R.); (Y.X.); (I.K.); (S.P.); (N.B.-L.); (R.G.)
| | - Nils Cordes
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus Technische Universität Dresden, 01307 Dresden, Germany;
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, 01328 Dresden, Germany
- German Cancer Consortium, Partner Site Dresden: German Cancer Research Center, 69120 Heidelberg, Germany
- Department of Radiotherapy and Radiation Oncology, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Christian Pilarsky
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (B.L.); (S.Z.); (S.Z.); (X.R.); (Y.X.); (I.K.); (S.P.); (N.B.-L.); (R.G.)
- Correspondence:
| |
Collapse
|
38
|
Zhang Z, Zhang HJ. Glycometabolic rearrangements-aerobic glycolysis in pancreatic ductal adenocarcinoma (PDAC): roles, regulatory networks, and therapeutic potential. Expert Opin Ther Targets 2021; 25:1077-1093. [PMID: 34874212 DOI: 10.1080/14728222.2021.2015321] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Glycometabolic rearrangements (aerobic glycolysis) is a hallmark of pancreatic ductal adenocarcinoma (PDAC) and contributes to tumorigenesis and progression through numerous mechanisms. The targeting of aerobic glycolysis is recognized as a potential therapeutic strategy which offers the possibility of improving treatment outcomes for PDAC patients. AREAS COVERED In this review, the role of aerobic glycolysis and its regulatory networks in PDAC are discussed. The targeting of aerobic glycolysis in PDAC is examined, and its therapeutic potential is evaluated. The relevant literature published from 2001 to 2021 was searched in databases including PubMed, Scopus, and Embase. EXPERT OPINION Regulatory networks of aerobic glycolysis in PDAC are based on key factors such as c-Myc, hypoxia-inducible factor 1α, the mammalian target of rapamycin pathway, and non-coding RNAs. Experimental evidence suggests that modulators or inhibitors of aerobic glycolysis promote therapeutic effects in preclinical tumor models. Nevertheless, successful clinical translation of drugs that target aerobic glycolysis in PDAC is an obstacle. Moreover, it is necessary to identify the potential targets for future interventions from regulatory networks to design efficacious and safer agents.
Collapse
Affiliation(s)
- Zhong Zhang
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, Nanjing, People's Republic of China
| | - Hai-Jun Zhang
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, Nanjing, People's Republic of China
| |
Collapse
|
39
|
Morati F, Modesti M. Insights into the control of RAD51 nucleoprotein filament dynamics from single-molecule studies. Curr Opin Genet Dev 2021; 71:182-187. [PMID: 34571340 DOI: 10.1016/j.gde.2021.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 09/05/2021] [Accepted: 09/10/2021] [Indexed: 11/26/2022]
Abstract
Genomic integrity depends on the RecA/RAD51 protein family. Discovered over five decades ago with the founder bacterial RecA protein, eukaryotic RAD51 is an ATP-dependent DNA strand transferase implicated in DNA double-strand break and single-strand gap repair, and in dealing with stressed DNA replication forks. RAD51 assembles as a nucleoprotein filament around single-stranded DNA to promote homology recognition in a duplex DNA and subsequent strand exchange. While the intrinsic dynamics of the RAD51 nucleoprotein filament has been extensively studied, a plethora of accessory factors control its dynamics. Understanding how modulators control filament dynamics is at the heart of current research efforts. Here, we describe recent advances in RAD51 control mechanisms obtained specifically using fluorescence-based single-molecule techniques.
Collapse
Affiliation(s)
- Florian Morati
- Cancer Research Center of Marseille, CNRS UMR7258, Inserm U1068, Institut Paoli-Calmettes, Aix-Marseille Université UM105, Marseille, France
| | - Mauro Modesti
- Cancer Research Center of Marseille, CNRS UMR7258, Inserm U1068, Institut Paoli-Calmettes, Aix-Marseille Université UM105, Marseille, France.
| |
Collapse
|
40
|
Wassing IE, Graham E, Saayman X, Rampazzo L, Ralf C, Bassett A, Esashi F. The RAD51 recombinase protects mitotic chromatin in human cells. Nat Commun 2021; 12:5380. [PMID: 34508092 PMCID: PMC8433380 DOI: 10.1038/s41467-021-25643-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 08/19/2021] [Indexed: 11/09/2022] Open
Abstract
The RAD51 recombinase plays critical roles in safeguarding genome integrity, which is fundamentally important for all living cells. While interphase functions of RAD51 in maintaining genome stability are well-characterised, its role in mitosis remains contentious. In this study, we show that RAD51 protects under-replicated DNA in mitotic human cells and, in this way, promotes mitotic DNA synthesis (MiDAS) and successful chromosome segregation. In cells experiencing mild replication stress, MiDAS was detected irrespective of mitotically generated DNA damage. MiDAS broadly required de novo RAD51 recruitment to single-stranded DNA, which was supported by the phosphorylation of RAD51 by the key mitotic regulator Polo-like kinase 1. Importantly, acute inhibition of MiDAS delayed anaphase onset and induced centromere fragility, suggesting a mechanism that prevents the satisfaction of the spindle assembly checkpoint while chromosomal replication remains incomplete. This study hence identifies an unexpected function of RAD51 in promoting genomic stability in mitosis.
Collapse
Affiliation(s)
- Isabel E Wassing
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Emily Graham
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Xanita Saayman
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Lucia Rampazzo
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Christine Ralf
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | | | - Fumiko Esashi
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK.
| |
Collapse
|