1
|
Sharma R, Mishra A, Bhardwaj M, Singh G, Indira Harahap LV, Vanjani S, Pan CH, Nepali K. Medicinal chemistry breakthroughs on ATM, ATR, and DNA-PK inhibitors as prospective cancer therapeutics. J Enzyme Inhib Med Chem 2025; 40:2489720. [PMID: 40256842 PMCID: PMC12013171 DOI: 10.1080/14756366.2025.2489720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 04/22/2025] Open
Abstract
This review discusses the critical roles of Ataxia Telangiectasia Mutated Kinase (ATM), ATM and Rad3-related Kinase (ATR), and DNA-dependent protein kinase (DNA-PK) in the DNA damage response (DDR) and their implications in cancer. Emphasis is placed on the intricate interplay between these kinases, highlighting their collaborative and distinct roles in maintaining genomic integrity and promoting tumour development under dysregulated conditions. Furthermore, the review covers ongoing clinical trials, patent literature, and medicinal chemistry campaigns on ATM/ATR/DNA-PK inhibitors as antitumor agents. Notably, the medicinal chemistry campaigns employed robust drug design strategies and aimed at assembling new structural templates with amplified DDR kinase inhibitory ability, as well as outwitting the pharmacokinetic liabilities of the existing DDR kinase inhibitors. Given the success attained through such endeavours, the clinical pipeline of DNA repair kinase inhibitors is anticipated to be supplemented by a reasonable number of tractable entries (DDR kinase inhibitors) soon.
Collapse
Affiliation(s)
- Ram Sharma
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Anshul Mishra
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Monika Bhardwaj
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Gurpreet Singh
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, India
| | | | - Sakshi Vanjani
- Molecular Medicine, University of South Florida, Tampa, FL, USA
| | - Chun Hsu Pan
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Kunal Nepali
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
2
|
Barut G, Çelik A. Gene expression profiles related to apoptosis and levels of DNA oxidative damage in primary dermal fibroblast cells (ATCC® PCS-201-012TM) treated with zirconium oxide nanoparticles. Food Chem Toxicol 2025; 201:115446. [PMID: 40252907 DOI: 10.1016/j.fct.2025.115446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 03/11/2025] [Accepted: 04/10/2025] [Indexed: 04/21/2025]
Abstract
Nanoparticles have attracted growing interest in recent years. They are small and can easily penetrate into cells. We investigated the genotoxic, cytotoxic, and apoptotic effects of zirconium nanoparticles on dermal fibroblast cells, the comet assay, Xcelligence system, and apoptotic gene expression, respectively. The comet assay analysis showed a non-signifcant increase in the genetic damage index and the percentage of damaged cells in the groups exposed to 10 and 20-nm zirconium oxide nanoparticles. Xcelligence system analysis observed a decrease in the indices of cells exposed to 10 and 20 nm zirconium oxide nanoparticles in the 48th-hour, 72nd-hour, and 96th-hour data. It was observed that caspase 3 and caspase 8 gene expression levels were suppressed in cells exposed to 10 and 20 nm zirconium oxide nanoparticles. Compared to the negative control group, this suppression was significant in the 10 nm groups (p < 0.01) while it was not significant in the 20 nm groups. Although zirconium oxide nanoparticles do not show toxicity and genotoxicity at a given concentration, but the overall mechanism is still not clear regarding the consequences of these nanoparticles use and its efect on living system. However, our apoptotic gene expression studies concluded that the nanoparticle size, particularly 10 nm, had an impact on gene expression.
Collapse
Affiliation(s)
- Gizem Barut
- Department of Biology, Postgraduate School of Natural and Applied Science, Mersin University, Mersin, Turkey
| | - Ayla Çelik
- Department of Biology, Faculty of Science Mersin University, Mersin, Turkey.
| |
Collapse
|
3
|
Gao S, Huang X, Zhang X, Yuan Z, Chen H, Li Z, El-Mesery HS, Shi J, Zou X. Empowering protein single-molecule sequencing: nanopore technology toward sensing gene sequences. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2025; 17:3902-3924. [PMID: 40331275 DOI: 10.1039/d5ay00572h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
The investigation of proteins at the single-molecule level is urgent to reveal the relationship between their structure and function. Unlike traditional techniques for attaining the overall average effect of group systems, nanopore sensing mode can provide information on the characteristics of proteins at the single-molecule level. Assisting with the intensity, frequency, and period of current changes, nanopore sequencing technology is rapidly advancing due to its merits, including fast readout, high accuracy, low cost, and portability. In particular, the single-molecule nanopore sequencing mode enables in-depth studies of DNA-protein interactions, protein conformation, DNA sequencing, and microbial assay, including genome sequencing of new species. This review summarizes the sensing mechanisms of nanopore sequencing technology in DNA damage, DNA methylation, RNA sequencing, and protein post-translational modifications and unfolding, covering both biological and solid-state nanopores. Due to these significant advantages, nanopore sequencing provides new insights into complex biological processes and enables more precise real-time monitoring of molecular changes. Its applications extend to clinical diagnostics, environmental monitoring, food safety, and forensic analysis. Moreover, the review outlines the present challenges faced by nanopore sequencing patterns, such as the choice of raw reagents and the design of special construction, offering a deep understanding of nanoporous single-molecule sensing toward protein sequence information and structure prediction.
Collapse
Affiliation(s)
- Shujie Gao
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, P. R. China.
- Faculty of Agricultural Engineering, Jiangsu University, Zhenjiang 212013, P. R. China
| | - Xiaowei Huang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, P. R. China.
| | - Xinai Zhang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, P. R. China.
| | - Zhecong Yuan
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, P. R. China.
| | - Haili Chen
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, P. R. China.
| | - Zhihua Li
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, P. R. China.
| | - Hany S El-Mesery
- School of Energy and Power Engineering, Jiangsu University, Zhenjiang 212013, P. R. China
| | - Jiyong Shi
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, P. R. China.
| | - Xiaobo Zou
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, P. R. China.
| |
Collapse
|
4
|
Li H, Liu Y, Wang M, Xu S, Liu J, Qin X, Li B, Sun J, Liu Y, Jin F. FANCI is involved in the malignant progression of glioma cells by regulating the Akt/Bcl-2 signaling pathway. Discov Oncol 2025; 16:753. [PMID: 40358883 PMCID: PMC12075725 DOI: 10.1007/s12672-025-02284-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 04/01/2025] [Indexed: 05/15/2025] Open
Abstract
INTRODUCTION Fanconi anemia supplementation group I (FANCI), one of the Fanconi family proteins, may be closely related to the malignant progression of glioma cells. Here, we sought to validate the expression of FANCI in glioma cells and its role in regulating the Akt signaling pathway in the malignant progression of glioma cells. METHODS The expression of FANCI in glioma cells was analyzed by bioinformatics and microarray. Lentivirus transfection was used to regulate the expression of FANCI in glioma cells. CCK-8, EdU, Transwell, and flow cytometry were used to observe the effect of FANCI on the malignant progression of glioma cells. RESULTS We found that low expression of FANCI inhibited the proliferation, migration and invasion of human glioma cells, and promoted cell apoptosis. However, overexpression of FANCI produced the opposite effect. We also found that low expression of FANCI inhibited the expression of P-Akt and B-cell lymphoma-2 (Bcl-2). Finally, we validated these in vitro results in a xenograft mouse model. CONCLUSION FANCI can inhibit the development of glioma by inhibiting Akt/Bcl-2 signaling pathway.
Collapse
Affiliation(s)
- Hanyun Li
- Cheeloo College of Medicine, Shandong University, Jinan, China
- Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Yinrui Liu
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | | | - Shaoshao Xu
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jilan Liu
- Department of Medical Research Center, Affiliated Hospital of Jining Medical University, Jining, China
| | - Xianyun Qin
- Department of Medical Research Center, Affiliated Hospital of Jining Medical University, Jining, China
| | - Banban Li
- Department of Hematology, The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
| | - Jikui Sun
- Department of Neurosurgery, Huanhu Hospital, Tianjin Medical University, Tianjin, China.
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgery Institute, Tianjin Huanhu Hospital, Tianjin, China.
| | - Yan Liu
- School of Public Health, Jining Medical University, Jining, China.
| | - Feng Jin
- Department of Neurosurgery, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, China.
- Shandong Provincial Key Medical and Health Laboratory of Neuro-oncology of Innovative Integrated Medicine, Qingdao Central Hospital, Qingdao, China.
| |
Collapse
|
5
|
Li MY, Niu H, Jiang J, Wu XY, Ying YL, Long YT. Real-Time Recording the Dynamic Catalytic Heterogeneity of Enzymatic Reactions Using a Nanopore. J Am Chem Soc 2025. [PMID: 40354520 DOI: 10.1021/jacs.5c02535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Enzymatic reactions in cells control the diversity of biomolecular composition, structure, and function, by virtue of their dynamics and heterogeneity. Here, we describe the use of a protein nanopore to monitor, in real time, the action of Exonuclease I (Exo I) on its substrate (homogeneous and heterogeneous short single-stranded DNA, ssDNA) on a single-reactant molecule basis. The nanopore-based single-molecule measurement, combined with a transition kinetic analysis, determines the temporal dynamics and heterogeneous cleavage and release pathways of ssDNA by Exo I. The results demonstrate a stepwise cleavage that is sequence-dependent on short ssDNA molecules (<15 nt), which differs from the kinetic model based on bulk measurements. In addition, we show that damaged DNA irreversibly changes the enzymatic reaction processes by Exo I. Thus, nanopores might prove to be useful for studying multienzyme cascade reactions at the single-molecule level.
Collapse
Affiliation(s)
- Meng-Yin Li
- Molecular Sensing and Imaging Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, P. R. China
| | - Hongyan Niu
- Molecular Sensing and Imaging Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Jie Jiang
- Molecular Sensing and Imaging Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Xue-Yuan Wu
- Molecular Sensing and Imaging Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Yi-Lun Ying
- Molecular Sensing and Imaging Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, P. R. China
| | - Yi-Tao Long
- Molecular Sensing and Imaging Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| |
Collapse
|
6
|
Foote K, Rienks M, Schmidt L, Theofilatos K, Yasmin, Ozols M, Eckersley A, Shah A, Figg N, Finigan A, O’Shaughnessy K, Wilkinson I, Mayr M, Bennett M. Oxidative DNA damage promotes vascular ageing associated with changes in extracellular matrix-regulating proteins. Cardiovasc Res 2025; 121:614-628. [PMID: 38717632 PMCID: PMC12054627 DOI: 10.1093/cvr/cvae091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 01/31/2024] [Accepted: 02/29/2024] [Indexed: 02/13/2025] Open
Abstract
AIMS Vascular ageing is characterized by vessel stiffening, with increased deposition of extracellular matrix (ECM) proteins including collagens. Oxidative DNA damage occurs in vascular ageing, but how it regulates ECM proteins and vascular stiffening is unknown. We sought to determine the relationship between oxidative DNA damage and ECM regulatory proteins in vascular ageing. METHODS AND RESULTS We examined oxidative DNA damage, the major base excision repair (BER) enzyme 8-Oxoguanine DNA Glycosylase (Ogg1) and its regulators, multiple physiological markers of ageing, and ECM proteomics in mice from 22 to 72 w. Vascular ageing was associated with increased oxidative DNA damage, and decreased expression of Ogg1, its active acetylated form, its acetylation regulatory proteins P300 and CBP, and the transcription factor Foxo3a. Vascular stiffness was examined in vivo in control, Ogg1-/-, or mice with vascular smooth muscle cell-specific expression of Ogg1+ (Ogg1) or an inactive mutation (Ogg1KR). Ogg1-/- and Ogg1KR mice showed reduced arterial compliance and distensibility, and increased stiffness and pulse pressure, whereas Ogg1 expression normalized all parameters to 72 w. ECM proteomics identified major changes in collagens with ageing, and downregulation of the ECM regulatory proteins Protein 6-lysyl oxidase (LOX) and WNT1-inducible-signaling pathway protein 2 (WISP2). Ogg1 overexpression upregulated LOX and WISP2 both in vitro and in vivo, and downregulated Transforming growth factor β1 (TGFb1) and Collagen 4α1 in vivo compared with Ogg1KR. Foxo3a activation induced Lox, while Wnt3 induction of Wisp2 also upregulated LOX and Foxo3a, and downregulated TGFβ1 and fibronectin 1. In humans, 8-oxo-G increased with vascular stiffness, while active OGG1 reduced with both age and stiffness. CONCLUSION Vascular ageing is associated with oxidative DNA damage, downregulation of major BER proteins, and changes in multiple ECM structural and regulatory proteins. Ogg1 protects against vascular ageing, associated with changes in ECM regulatory proteins including LOX and WISP2.
Collapse
MESH Headings
- Animals
- DNA Glycosylases/genetics
- DNA Glycosylases/metabolism
- DNA Glycosylases/deficiency
- Oxidative Stress
- DNA Damage
- Forkhead Box Protein O3/metabolism
- Extracellular Matrix Proteins/metabolism
- Extracellular Matrix Proteins/genetics
- Aging/metabolism
- Aging/pathology
- Aging/genetics
- Vascular Stiffness
- Mice, Knockout
- Mice, Inbred C57BL
- Male
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Humans
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/enzymology
- Signal Transduction
- Cells, Cultured
- Acetylation
- Extracellular Matrix/metabolism
- Mice
Collapse
Affiliation(s)
- Kirsty Foote
- Section of Cardiorespiratory Medicine, University of Cambridge, Victor Phillip Dahdaleh Heart & Lung Research Institute, Papworth Road, Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
| | - Marieke Rienks
- Cardiovascular Division, King’s College London, The James Black Centre, 2nd Floor, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Lukas Schmidt
- Cardiovascular Division, King’s College London, The James Black Centre, 2nd Floor, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Konstantinos Theofilatos
- Cardiovascular Division, King’s College London, The James Black Centre, 2nd Floor, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Yasmin
- Department of Medicine, Experimental Medicine and Therapeutics, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 2QQ, UK
| | - Matiss Ozols
- Human Genetics, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Saffron Walden CB10 1RQ, UK
| | - Alexander Eckersley
- Division of Musculoskeletal & Dermatological Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Aarti Shah
- Section of Cardiorespiratory Medicine, University of Cambridge, Victor Phillip Dahdaleh Heart & Lung Research Institute, Papworth Road, Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
| | - Nichola Figg
- Section of Cardiorespiratory Medicine, University of Cambridge, Victor Phillip Dahdaleh Heart & Lung Research Institute, Papworth Road, Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
| | - Alison Finigan
- Section of Cardiorespiratory Medicine, University of Cambridge, Victor Phillip Dahdaleh Heart & Lung Research Institute, Papworth Road, Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
| | - Kevin O’Shaughnessy
- Department of Medicine, Experimental Medicine and Therapeutics, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 2QQ, UK
| | - Ian Wilkinson
- Department of Medicine, Experimental Medicine and Therapeutics, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 2QQ, UK
| | - Manuel Mayr
- Cardiovascular Division, King’s College London, The James Black Centre, 2nd Floor, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Martin Bennett
- Section of Cardiorespiratory Medicine, University of Cambridge, Victor Phillip Dahdaleh Heart & Lung Research Institute, Papworth Road, Cambridge Biomedical Campus, Cambridge CB2 0BB, UK
| |
Collapse
|
7
|
Elkhalil A, Whited A, Ghose P. SQST-1/p62-regulated SKN-1/Nrf mediates a phagocytic stress response via transcriptional activation of lyst-1/LYST. PLoS Genet 2025; 21:e1011696. [PMID: 40315422 PMCID: PMC12068719 DOI: 10.1371/journal.pgen.1011696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 05/12/2025] [Accepted: 04/19/2025] [Indexed: 05/04/2025] Open
Abstract
Cells may be intrinsically fated to die to sculpt tissues during development or to maintain homeostasis. Cells can also die in response to various stressors, injury or pathological conditions. Additionally, cells of the metazoan body are often highly specialized with distinct domains that differ both structurally and with respect to their neighbors. Specialized cells can also die, as in normal brain development or pathological states and their different regions may be eliminated via different programs. Clearance of different types of cell debris must be performed quickly and efficiently to prevent autoimmunity and secondary necrosis of neighboring cells. Moreover, all cells, including those programmed to die, may be subject to various stressors. Some largely unexplored questions include whether predestined cell elimination during development could be altered by stress, if adaptive stress responses exist and if polarized cells may need compartment-specific stress-adaptive programs. We leveraged Compartmentalized Cell Elimination (CCE) in the nematode C. elegans to explore these questions. CCE is a developmental cell death program whereby three segments of two embryonic polarized cell types are eliminated differently. We have previously employed this in vivo genetic system to uncover a cell compartment-specific, cell non-autonomous clearance function of the fusogen EFF-1 in phagosome closure during corpse internalization. Here, we introduce an adaptive response that serves to aid developmental phagocytosis as a part of CCE during stress. We employ a combination of forward and reverse genetics, CRISPR/Cas9 gene editing, stress response assays and advanced fluorescence microscopy. Specifically, we report that, under heat stress, the selective autophagy receptor SQST-1/p62 promotes the nuclear translocation of the oxidative stress-related transcription factor SKN-1/Nrf via negative regulation of WDR-23. This in turn allows SKN-1/Nrf to transcribe lyst-1/LYST (lysosomal trafficking associated gene) which subsequently promotes the phagocytic resolution of the developmentally-killed internalized cell even under stress conditions.
Collapse
Affiliation(s)
- Aladin Elkhalil
- The University of Texas at Arlington, Arlington, Texas, United States of America
| | - Alec Whited
- The University of Texas at Arlington, Arlington, Texas, United States of America
| | - Piya Ghose
- The University of Texas at Arlington, Arlington, Texas, United States of America
| |
Collapse
|
8
|
Aning OA, Dvirnas A, Nyblom M, Krog J, Carlson J, Johansson P, Ambjörnsson T, Westerlund F. Stained DNA Dot Detection (SD 3): An automated tool for quantifying fluorescent features along single stretched DNA molecules. DNA Repair (Amst) 2025; 149:103836. [PMID: 40300455 DOI: 10.1016/j.dnarep.2025.103836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 03/23/2025] [Accepted: 04/04/2025] [Indexed: 05/01/2025]
Abstract
The main information in DNA is its four-letter sequence that builds up the genetic information and that is traditionally read using sequencing methodologies. DNA can, however, also carry other important information, such as epigenetic marks and DNA damage. This information has recently been visualized along single DNA molecules using fluorescent labels. Quantifying fluorescent labels along DNA is done by counting the number of "dots" per length of each DNA molecule on DNA stretched on a glass surface. So far, a major challenge has been the lack of standardized data analysis tools. Focusing on DNA damage, we here present a Matlab-based automated software, Stained DNA Dot Detection (SD3), which uses a robust method for finding DNA molecules and estimating the number of dots along each molecule. We have validated SD3 by comparing the outcome to manual analysis using DNA extracted from cells exposed to H2O2 as a model system. Our results show that SD3 achieves high accuracy and reduced analysis time relative to manual counting. SD3 allows the user to define specific parameters regarding the DNA molecule and the location of dots to include during analysis via a user-friendly interface. We foresee that our open-source software can have broad use in the analysis of single DNA molecules and their modifications in research and in diagnostics.
Collapse
Affiliation(s)
- Obed A Aning
- Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Albertas Dvirnas
- Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden; Computational Science for Health and Environment, Centre for Environmental and Climate Science, Lund University, Lund, Sweden
| | - My Nyblom
- Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Jens Krog
- Computational Science for Health and Environment, Centre for Environmental and Climate Science, Lund University, Lund, Sweden
| | - Johanna Carlson
- Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Pegah Johansson
- Region Västra Götaland, Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Tobias Ambjörnsson
- Computational Science for Health and Environment, Centre for Environmental and Climate Science, Lund University, Lund, Sweden
| | - Fredrik Westerlund
- Department of Life Sciences, Chalmers University of Technology, Gothenburg, Sweden.
| |
Collapse
|
9
|
Almawash S. Revolutionary Cancer Therapy for Personalization and Improved Efficacy: Strategies to Overcome Resistance to Immune Checkpoint Inhibitor Therapy. Cancers (Basel) 2025; 17:880. [PMID: 40075727 PMCID: PMC11899125 DOI: 10.3390/cancers17050880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer remains a significant public health issue worldwide, standing as a primary contributor to global mortality, accounting for approximately 10 million fatalities in 2020 [...].
Collapse
Affiliation(s)
- Saud Almawash
- Department of Pharmaceutics, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia
| |
Collapse
|
10
|
Rehmutulla M, Zhang S, Yin J, Huang J, Xiao Y, Hu Z, Tong Q, Zhang Y. Ustusolate E and 11α-Hydroxy-Ustusolate E induce apoptosis in cancer cell lines by regulating the PI3K/AKT/mTOR and p-53 pathways. Chin J Nat Med 2025; 23:346-353. [PMID: 40122664 DOI: 10.1016/s1875-5364(25)60840-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 04/23/2024] [Accepted: 05/06/2024] [Indexed: 03/25/2025]
Abstract
Cancer represents a significant disease that profoundly impacts human health and longevity. Projections indicate a 47% increase in the global cancer burden by 2040 compared to 2020, accompanied by a further rise in the associated economic burden. Consequently, there is an urgent need to discover and develop new alternative drugs to mitigate the global impact of cancer. Natural products (NPs) play a crucial role in the identification and development of anticancer therapeutics. This study identified ustusolate E (UE) and its analog 11α-hydroxy-ustusolate E (HUE) from strain Aspergilluscalidoustus TJ403-EL05, and examined their antitumor activities and mechanisms of action. The findings demonstrate that both compounds significantly inhibited the proliferation and colony formation of AGS (human gastric cancer cells) and 786-O (human renal clear cell carcinoma cells), induced irreversible DNA damage, blocked the cell cycle at the G2/M phase, and further induced apoptosis in tumor cells. To the best of the authors' knowledge, this is the first report on the anticancer effects of UE and HUE and their underlying mechanisms. The present study suggests that HUE and UE could serve as lead compounds for the development of novel anticancer drugs.
Collapse
Affiliation(s)
- Mewlude Rehmutulla
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Sitian Zhang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jie Yin
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jianzheng Huang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yang Xiao
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhengxi Hu
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Qingyi Tong
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Yonghui Zhang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
11
|
Shanmugam N, Chatterjee S, Cisneros GA. Impact of a Cancer-Associated Mutation on Poly(ADP-ribose) Polymerase1 Inhibition. J Phys Chem B 2025; 129:2175-2186. [PMID: 39962867 PMCID: PMC12005076 DOI: 10.1021/acs.jpcb.4c07960] [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] [Indexed: 02/26/2025]
Abstract
Poly(ADP-ribose) polymerase1 (PARP1) plays a vital role in DNA repair, and its inhibition in cancer cells may cause cell apoptosis. In this study, we investigated the effects of a PARP1 variant, V762A, which is strongly associated with several cancers in humans, on the inhibition of PARP1 by three FDA-approved inhibitors: niraparib, rucaparib, and talazoparib. Specifically, we compared the inhibition of the mutant to that of wild-type (WT) PARP1. Additionally, we investigated how the mutation influences the binding of these inhibitors to PARP1. Our work suggests that while mutant PARP1 exhibits only minor differences in residual fluctuations, backbone deviations, and residue motion correlations compared to the WT under niraparib and rucaparib inhibitions, it shows significant and distinct differences in these features when inhibited by talazoparib. Among the three inhibitions, talazoparib inhibition uniquely lowers the average residue fluctuations in the mutant than the WT including lower fluctuations of mutant's N- and C-terminal residues in the catalytic domain, conserved H-Y-E traid residues, and donor loop (D-loop) residues which are important for catalysis more effectively than other inhibitions. However, talazoparib also significantly enhances destabilizing interactions between the mutation site in the HD domain in the mutant than WT. Further, among the three inhibitions, talazoparib inhibition uniquely and significantly disrupts the functional fluctuations of terminal regions in the mutant, which are otherwise present in the WT. The mutation and inhibition do not significantly affect PARP1's essential dynamics. Lastly, these inhibitors bind to the V762A mutant more effectively than to the WT, with similar binding free energies between them.
Collapse
Affiliation(s)
- Neel Shanmugam
- Department of Chemistry, University of North Texas, Denton, Texas 76201, United States; Present Address: Department of Computer Science, Columbia University, New York, New York 10027, United States
| | - Shubham Chatterjee
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - G. Andrés Cisneros
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States; Department of Physics, University of Texas at Dallas, Richardson, Texas 75080, United States
| |
Collapse
|
12
|
Meade AD, Maguire A, Bryant J, Cullen D, Medipally D, White L, Armstrong J, Dunne M, Noone E, Bradshaw S, Finn M, Shannon AM, Howe OL, Lyng FM. Detection of radiosensitive subpopulations ex-vivo with Raman microspectroscopy. Front Oncol 2025; 15:1470431. [PMID: 40083875 PMCID: PMC11903398 DOI: 10.3389/fonc.2025.1470431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 01/30/2025] [Indexed: 03/16/2025] Open
Abstract
Although significant advances in understanding the molecular drivers of acquired and inherited radiosensitivity have occurred in recent decades, a single analytical method which can detect and classify radiosensitivity remains elusive. Raman microspectroscopy has demonstrated capabilities in the objective classification of various diseases, and more recently in the detection and modelling of radiobiological effect. In this study, Raman spectroscopy is presented as a potential tool for the detection of radiosensitivity subpopulations represented by four lymphoblastoid cell lines derived from individuals with ataxia telangiectasia (2 lines), non-Hodgkins lymphoma, and Turner's syndrome. These are classified with respect to a population with mixed radiosensitivity, represented by lymphocytes drawn from both healthy controls, and prostate cancer patients. Raman spectroscopic measurements were made ex-vivo after exposure to X-ray doses of 0 Gy, 50 mGy and 500 mGy, in parallel to radiation-induced G2 chromosomal radiosensitivity scores, for all samples. Support vector machine models developed on the basis of the spectral data were capable of discrimination of radiosensitive populations before and after irradiation, with superior discrimination when spectra were subjected to a non-linear dimensionality reduction (UMAP) as opposed to a linear (PCA) approach. Models developed on spectral data acquired on samples irradiated in-vitro with a dose of 0Gy were found to provide the highest level of performance in discriminating between classes, with performances of F1 = 0.92 ± 0.06 achieved on a held-out test set. Overall, this study suggests that Raman spectroscopy may have potential as a tool for the detection of intrinsic radiosensitivity using liquid biopsies.
Collapse
Affiliation(s)
- Aidan D. Meade
- School of Physics, Clinical and Optometric Sciences, Technological University Dublin, Dublin, Ireland
| | - Adrian Maguire
- School of Physics, Clinical and Optometric Sciences, Technological University Dublin, Dublin, Ireland
- Radiation and Environmental Science Centre (RESC), Physical to Life Sciences Research Hub, FOCAS Building, Technological University Dublin, Dublin, Ireland
| | - Jane Bryant
- Radiation and Environmental Science Centre (RESC), Physical to Life Sciences Research Hub, FOCAS Building, Technological University Dublin, Dublin, Ireland
| | - Daniel Cullen
- Radiation and Environmental Science Centre (RESC), Physical to Life Sciences Research Hub, FOCAS Building, Technological University Dublin, Dublin, Ireland
- School of Biological, Health and Sport Sciences, Technological University (TU) Dublin, Dublin, Ireland
| | - Dinesh Medipally
- School of Physics, Clinical and Optometric Sciences, Technological University Dublin, Dublin, Ireland
- Radiation and Environmental Science Centre (RESC), Physical to Life Sciences Research Hub, FOCAS Building, Technological University Dublin, Dublin, Ireland
| | - Lisa White
- Radiation and Environmental Science Centre (RESC), Physical to Life Sciences Research Hub, FOCAS Building, Technological University Dublin, Dublin, Ireland
- School of Biological, Health and Sport Sciences, Technological University (TU) Dublin, Dublin, Ireland
| | - John Armstrong
- Department of Radiation Oncology, Saint Luke’s Radiation Oncology Network (SLRON), St Luke’s Hospital, Dublin, Ireland
| | - Mary Dunne
- Department of Radiation Oncology, Saint Luke’s Radiation Oncology Network (SLRON), St Luke’s Hospital, Dublin, Ireland
| | - Emma Noone
- Department of Radiation Oncology, Saint Luke’s Radiation Oncology Network (SLRON), St Luke’s Hospital, Dublin, Ireland
| | - Shirley Bradshaw
- Department of Radiation Oncology, Saint Luke’s Radiation Oncology Network (SLRON), St Luke’s Hospital, Dublin, Ireland
| | - Marie Finn
- Department of Radiation Oncology, Saint Luke’s Radiation Oncology Network (SLRON), St Luke’s Hospital, Dublin, Ireland
| | | | - Orla L. Howe
- Radiation and Environmental Science Centre (RESC), Physical to Life Sciences Research Hub, FOCAS Building, Technological University Dublin, Dublin, Ireland
- School of Biological, Health and Sport Sciences, Technological University (TU) Dublin, Dublin, Ireland
| | - Fiona M. Lyng
- School of Physics, Clinical and Optometric Sciences, Technological University Dublin, Dublin, Ireland
- Radiation and Environmental Science Centre (RESC), Physical to Life Sciences Research Hub, FOCAS Building, Technological University Dublin, Dublin, Ireland
| |
Collapse
|
13
|
Permatasari LI, Afifah NN, Ishmatullah MH, Intania R, Halimah E, Barliana MI. Genetic Navigation: A Narrative Review of XRCC1 Polymorphism Impact on Platinum-Based Chemotherapy Outcomes in NSCLC Patients. Cancer Manag Res 2025; 17:383-395. [PMID: 40034865 PMCID: PMC11874740 DOI: 10.2147/cmar.s501420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 02/01/2025] [Indexed: 03/05/2025] Open
Abstract
Non-small cell lung cancer (NSCLC) is the most common form of lung cancer, accounting for 85% of all cases, with a poor 5-year survival rate of less than 20%. The majority of NSCLC patients are diagnosed at an advanced stage, contributing to the low survival rate. Platinum-based chemotherapy, including cisplatin and carboplatin, remains the cornerstone of treatment for advanced NSCLC. However, DNA repair mechanisms often hinder treatment efficacy, notably Base Excision Repair (BER), mediated by the X-ray Repair Cross Complementing 1 (XRCC1) protein. This review aims to investigate the role of XRCC1 polymorphisms in platinum resistance, focusing on their impact on DNA repair efficiency. XRCC1's involvement in the BER pathway is critical for repairing DNA damage caused by platinum agents, and polymorphisms in XRCC1 have been linked to altered repair capacity, influencing clinical outcomes and resistance to platinum-based chemotherapy in NSCLC patients.
Collapse
Affiliation(s)
- Lanny Indah Permatasari
- Department of Biological Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
| | - Nadiya Nurul Afifah
- Department of Biological Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
| | | | - Ruri Intania
- Department of Pulmonology and Respiratory Medicine, Dr. H.A. Rotinsulu Lung Hospital, Bandung, Indonesia
| | - Eli Halimah
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
| | - Melisa Intan Barliana
- Department of Biological Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
- Center of Excellence for Pharmaceutical Care Innovation, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
| |
Collapse
|
14
|
Kumar U, Singhal S, Khan AA, Alanazi AM, Gurjar P, Khandia R. Insights into genetic architecture and disease associations of genes associated with different human blood group systems using codon usage bias. J Biomol Struct Dyn 2025:1-21. [PMID: 39988946 DOI: 10.1080/07391102.2025.2466710] [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: 09/06/2024] [Accepted: 11/13/2024] [Indexed: 02/25/2025]
Abstract
The differential use of synonymous codons of an amino acid is an imperative evolutionary phenomenon, termed codon usage bias, that functions across various levels of organisms. It is accustomed to providing an understanding of a gene's differential architecture driven by functional regulation of gene expression. Numerous synonymous mutations are linked to various diseases, demonstrating that silent mutations can be deleterious. We employed bioinformatics methods to examine codon usage trends in 263 coding sequences of 44 blood group systems. The blood group systems were categorized into two groups based on association with a sort of neurodegenerative disorder. We performed a CUB study to investigate how multiple components, such as selection, mutation and biased nucleotide composition are accountable for the evolution of the transcripts of the blood group antigens. The compositional analysis implicated blood group genes were GC-rich. RSCU analysis showed G/C-ending codon choice among synonymous codons. Also, a distinct codon choice was found in both blood groups for serine and proline. Moreover, the leucine-coding CTG codon was found the most overrepresented in all the genes, indicating selectional pressure substantially impacts overall codon usage. This was also supported by biplot analysis. Additionally, CpC and GpG overrepresentation is in concordance with the results concerning neurodegenerative disorders where CpC has been attributed to non-CpG methylation and linked to several neurodegenerative ailments. Both the Z-test analysis and rare codon choice showed a substantial difference in codon usage by the genes of both groups.
Collapse
Affiliation(s)
- Utsang Kumar
- Department of Biochemistry and Genetics, Barkatullah University, Bhopal, Madhya Pradesh, India
| | - Shailja Singhal
- Department of Biochemistry and Genetics, Barkatullah University, Bhopal, Madhya Pradesh, India
| | - Azmat Ali Khan
- Pharmaceutical Biotechnology Laboratory, Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Amer M Alanazi
- Pharmaceutical Biotechnology Laboratory, Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Pankaj Gurjar
- Centre for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, Australia
| | - Rekha Khandia
- Department of Biochemistry and Genetics, Barkatullah University, Bhopal, Madhya Pradesh, India
| |
Collapse
|
15
|
Zachayus A, Loup-Forest J, Cura V, Poterszman A. Nucleotide Excision Repair: Insights into Canonical and Emerging Functions of the Transcription/DNA Repair Factor TFIIH. Genes (Basel) 2025; 16:231. [PMID: 40004560 PMCID: PMC11855273 DOI: 10.3390/genes16020231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/31/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Nucleotide excision repair (NER) is a universal cut-and-paste DNA repair mechanism that corrects bulky DNA lesions such as those caused by UV radiation, environmental mutagens, and some chemotherapy drugs. In this review, we focus on the human transcription/DNA repair factor TFIIH, a key player of the NER pathway in eukaryotes. This 10-subunit multiprotein complex notably verifies the presence of a lesion and opens the DNA around the damage via its XPB and XPD subunits, two proteins identified in patients suffering from Xeroderma Pigmentosum syndrome. Isolated as a class II gene transcription factor in the late 1980s, TFIIH is a prototypic molecular machine that plays an essential role in both DNA repair and transcription initiation and harbors a DNA helicase, a DNA translocase, and kinase activity. More recently, TFIIH subunits have been identified as participating in other cellular processes, including chromosome segregation during mitosis, maintenance of mitochondrial DNA integrity, and telomere replication.
Collapse
Affiliation(s)
- Amélie Zachayus
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France; (A.Z.); (J.L.-F.); (V.C.)
- Centre National de la Recherche Scientifique (CNRS), UMR 7104, 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France
- Institut National De La Sante et de la Recherche Médicale (Inserm), UMR S 1258, 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France
- Equipe Labellisée Ligue Contre le Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France
| | - Jules Loup-Forest
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France; (A.Z.); (J.L.-F.); (V.C.)
- Centre National de la Recherche Scientifique (CNRS), UMR 7104, 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France
- Institut National De La Sante et de la Recherche Médicale (Inserm), UMR S 1258, 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France
- Equipe Labellisée Ligue Contre le Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France
| | - Vincent Cura
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France; (A.Z.); (J.L.-F.); (V.C.)
- Centre National de la Recherche Scientifique (CNRS), UMR 7104, 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France
- Institut National De La Sante et de la Recherche Médicale (Inserm), UMR S 1258, 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France
- Equipe Labellisée Ligue Contre le Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France
| | - Arnaud Poterszman
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France; (A.Z.); (J.L.-F.); (V.C.)
- Centre National de la Recherche Scientifique (CNRS), UMR 7104, 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France
- Institut National De La Sante et de la Recherche Médicale (Inserm), UMR S 1258, 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France
- Equipe Labellisée Ligue Contre le Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France
| |
Collapse
|
16
|
Imani MM, Ashabi A, Rezaei F, Saffar Shahroudi A, Kashkouli S, Sadeghi E, Sadeghi M. Association of ERCC2/XPD polymorphisms and the risk of head and neck carcinoma: a systematic review, meta-analysis, trial sequential analysis, network analysis, and functional effects. BMC Oral Health 2025; 25:201. [PMID: 39923044 PMCID: PMC11806812 DOI: 10.1186/s12903-025-05476-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 01/10/2025] [Indexed: 02/10/2025] Open
Abstract
OBJECTIVES AND AIMS The combination of genetic and environmental factors may contribute to the carcinogenesis of HNC. Despite the reported associations between xeroderma pigmentosum group D (XPD) polymorphisms and HNC, the results have been inconsistent, with different studies reporting varying results. Therefore, our aim is to assess the association of three XPD polymorphisms (rs13181, rs1799793, and rs238406) in a comprehensive meta-analysis. MATERIALS AND METHODS An exhaustive literature review was performed across several databases, including PubMed, Web of Science, Scopus, and Cochrane Library, up to November 18, 2023, without any restrictions. The effect sizes were presented as the odds ratio (OR) with a 95% confidence interval (CI). RESULTS Thirty-nine articles including 56 studies were entered into the meta-analysis. Evaluating rs13181, rs1799793, and rs238406 polymorphisms in five genetic models, just significant associations were found for rs1799793 polymorphism in heterozygous and dominant models. The findings reported that the ethnicity and the cancer subtype for rs13181, the ethnicity, the sample size, and the control source for rs1799793, and the ethnicity and the control source for rs238406 polymorphisms were effective factors in the pooled results. Trial sequential analysis suggested that the studies included an insufficient number of individuals. Sensitivity analysis reported stability of pooled results. The XPD protein variants were predicted to be benign. CONCLUSIONS The study reveals a significant association between the rs1799793 polymorphism and HNC, but not rs13181 and rs238406 polymorphisms. Future studies should also aim to minimize the impact of confounding factors and heterogeneity to ensure more accurate results.
Collapse
Affiliation(s)
- Mohammad Moslem Imani
- Department of Orthodontics, School of Dentistry, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ali Ashabi
- Students Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Farzad Rezaei
- Department of Oral and Maxillofacial Surgery, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Atefe Saffar Shahroudi
- Dental Research Center, Dentistry Research Institute, Department of Orthodontics, Dental School, Tehran University of Medical Sciences, Tehran, Iran
| | - Sadegh Kashkouli
- Department of Prosthodontics, Dental School, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Edris Sadeghi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Masoud Sadeghi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
17
|
Guo G, Cui C, Hartanto H, Li J, Chen TH. DNA polymerase mediated CRISPR/Cas12a trans-cleavage for dual-mode quantification of uracil DNA glycosylase activity. Talanta 2025; 283:127089. [PMID: 39467442 DOI: 10.1016/j.talanta.2024.127089] [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: 06/17/2024] [Revised: 09/19/2024] [Accepted: 10/20/2024] [Indexed: 10/30/2024]
Abstract
Since an unusual expression of uracil-DNA glycosylase (UDG) is often associated with the pathogenesis of numerous disorders, the detection of UDG activity is regarded as a promising application in disease diagnosis. Here, we develop a DNA polymerase-mediated CRISPR/Cas12a trans cleavage strategy, which can achieve dual-mode determination of UDG activity. By introducing a hairpin DNA probe containing a single uracil base, the probe undergoes specific cleavage and elongation under the existence of UDG only, thus activating the trans cleavage of ssDNA regardless of its length and sequence. To accommodate different detection modes, the ssDNA was further modified by fluorophore-quencher pairs or designed for connecting magnetic microparticles (MMPs) and polystyrene microparticles (PMPs). Finally, the UDG activity is quantified by fluorescence signal and microparticle accumulation length on a microfluidic chip visible to the naked eye. This strategy provides a detectable minimum UDG concentration of 0.00047 U/mL for fluorescent mode and 0.0048 U/mL for microfluidic mode. Furthermore, we performed the UDG inhibition test and detected UDG activity in cell lysates, suggesting its potential for inhibitor screening and detection of UDG in biological samples.
Collapse
Affiliation(s)
- Guihuan Guo
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Hong Kong Special Administrative Region, China
| | - Chenyu Cui
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Hong Kong Special Administrative Region, China; Hong Kong Centre for Cerebro-Cardiovascular Health Engineering, Hong Kong Science Park, Hong Kong SAR, China
| | - Hogi Hartanto
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Hong Kong Special Administrative Region, China
| | - Jiaheng Li
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Hong Kong Special Administrative Region, China
| | - Ting-Hsuan Chen
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Hong Kong Special Administrative Region, China; Hong Kong Centre for Cerebro-Cardiovascular Health Engineering, Hong Kong Science Park, Hong Kong SAR, China; City University of Hong Kong Shenzhen Research Institute, Shenzhen, China; Tung Biomedical Sciences Centre, City University of Hong Kong, China.
| |
Collapse
|
18
|
Mutte SK, Barendse P, Ugarte PB, Swarts DC. Distribution of bacterial DNA repair proteins and their co-occurrence with immune systems. Cell Rep 2025; 44:115110. [PMID: 39752253 DOI: 10.1016/j.celrep.2024.115110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/20/2024] [Accepted: 12/03/2024] [Indexed: 02/01/2025] Open
Abstract
Bacteria encode various DNA repair pathways to maintain genome integrity. However, the high degree of homology between DNA repair proteins or their domains hampers accurate identification. Here, we describe a stringent search strategy to identify DNA repair proteins and provide a systematic analysis of taxonomic distribution and co-occurrence of DNA repair proteins involved in RecA-dependent homologous recombination. Our results reveal the widespread presence of RecA, SSB, and RecOR proteins and phyla-specific distribution for the DNA repair complexes RecBCD, AddAB, and AdnAB. Furthermore, we report co-occurrences of DNA repair proteins with immune systems, including specific CRISPR-Cas subtypes, prokaryotic Argonautes (pAgos), dGTPases, GAPS2, and Wadjet. Our results imply that while certain DNA repair proteins and immune systems might function in conjunction, no immune system strictly relies on a specific DNA repair protein. As such, these findings offer an updated perspective on the distribution of DNA repair systems and their connection to immune systems in bacteria.
Collapse
Affiliation(s)
- Sumanth K Mutte
- Laboratory of Biochemistry, Wageningen University, 6708 WE Wageningen, the Netherlands; MyGen Informatics, 6706 JE Wageningen, the Netherlands
| | - Patrick Barendse
- Laboratory of Biochemistry, Wageningen University, 6708 WE Wageningen, the Netherlands
| | | | - Daan C Swarts
- Laboratory of Biochemistry, Wageningen University, 6708 WE Wageningen, the Netherlands.
| |
Collapse
|
19
|
Dorival J, Yuan H, Walker AS, Tang GL, Eichman BF. Yatakemycin biosynthesis requires two deoxyribonucleases for toxin self-resistance. RSC Chem Biol 2025; 6:94-105. [PMID: 39649339 PMCID: PMC11621827 DOI: 10.1039/d4cb00203b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/29/2024] [Indexed: 12/10/2024] Open
Abstract
The highly active natural product yatakemycin (YTM) from Streptomyces sp. TP-A0356 is a potent DNA damaging agent with antimicrobial and antitumor properties. The YTM biosynthesis gene cluster (ytk) contains several toxin self-resistance genes. Of these, ytkR2 encodes a DNA glycosylase that is important for YTM production and host survival by excising lethal YTM-adenine lesions from the genome, presumably initiating a base excision repair (BER) pathway. However, the genes involved in repair of the resulting apurinic/apyrimidinic (AP) site as the second BER step have not been identified. Here, we show that ytkR4 and ytkR5 are essential for YTM production and encode deoxyribonucleases related to other known DNA repair nucleases. Purified YtkR4 and YtkR5 exhibit AP endonuclease activity specific for YtkR2-generated AP sites, providing a basis for BER of the toxic AP intermediate produced from YTM-adenine excision and consistent with co-evolution of ytkR2, ytkR4, and ytkR5. YtkR4 and YtkR5 also exhibit 3'-5' exonuclease activity with differing substrate specificities. The YtkR5 exonuclease is capable of digesting through a YTM-DNA lesion and may represent an alternative repair mechanism to BER. We also show that ytkR4 and ytkR5 homologs are often clustered together in putative gene clusters related to natural product production, consistent with non-redundant roles in repair of other DNA adducts derived from genotoxic natural products.
Collapse
Affiliation(s)
- Jonathan Dorival
- Department of Biological Sciences, Vanderbilt University Nashville Tennessee USA
| | - Hua Yuan
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences Shanghai 200032 China
| | - Allison S Walker
- Department of Biological Sciences, Vanderbilt University Nashville Tennessee USA
- Department of Chemistry, Vanderbilt University Nashville Tennessee USA
| | - Gong-Li Tang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences Shanghai 200032 China
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences 1 Sub-lane Xiangshan Hangzhou 310024 China
| | - Brandt F Eichman
- Department of Biological Sciences, Vanderbilt University Nashville Tennessee USA
- Department of Biochemistry, Vanderbilt University School of Medicine Nashville Tennessee USA
| |
Collapse
|
20
|
Guo XL, Wang YD, Liu YJ, Chu L, Zhu H, Hu Y, Wu RY, Xie HY, Yu J, Li SP, Xiong CY, Li RY, Ke F, Chen L, Chen GQ, Chen L, Bai F, Enver T, Li GH, Li HF, Hong DL. Fetal hepatocytes protect the HSPC genome via fetuin-A. Nature 2025; 637:402-411. [PMID: 39633051 PMCID: PMC11711094 DOI: 10.1038/s41586-024-08307-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 10/30/2024] [Indexed: 12/07/2024]
Abstract
The maintenance of genomic integrity in rapidly proliferating cells is a substantial challenge during embryonic development1-3. Although numerous cell-intrinsic mechanisms have been revealed4-7, little is known about genome-protective effects and influences of developmental tissue microenvironments on tissue-forming cells. Here we show that fetal liver hepatocytes provide protection to haematopoietic stem and progenitor cell (HSPC) genomes. Lineage tracing and depletion in mice demonstrated that delayed hepatocyte development in early fetal livers increased the chromosomal instability of newly colonizing HSPCs. In addition, HSPCs developed tolerance to genotoxins in hepatocyte-conditioned medium, suggesting that hepatocytes protect the HSPC genome in a paracrine manner. Proteomic analyses demonstrated the enrichment of fetuin-A in hepatocyte-conditioned medium but not in early fetal livers. Fetuin-A activates a Toll-like receptor pathway to prevent pathogenic R-loop accumulation in HSPCs undergoing DNA replication and gene transcription in the fetal liver. Numerous haematopoietic regulatory genes frequently involved in leukaemogenic mutations are associated with R-loop-enriched regions. In Fetua-knockout mice, HSPCs showed increased genome instability and susceptibility to malignancy induction. Moreover, low concentrations of fetuin-A correlated with the oncogenesis of childhood leukaemia. Therefore, we uncover a mechanism operating in developmental tissues that offers tissue-forming cell genome protection and is implicated in developmental-related diseases.
Collapse
Affiliation(s)
- Xiao-Lin Guo
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Institute of Haematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi-Ding Wang
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Institute of Haematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan-Jun Liu
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Institute of Haematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Chu
- Department of Obstetrics and Gynecology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hua Zhu
- Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ye Hu
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Institute of Haematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ren-Yan Wu
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Institute of Haematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong-Yu Xie
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Institute of Haematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Juan Yu
- National laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Shui-Ping Li
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Institute of Haematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chao-Yang Xiong
- National laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Ruo-Yan Li
- Biomedical Pioneering Innovation Centre (BIOPIC) and Translational Cancer Research Centre, School of Life Sciences, First Hospital, Peking University, Beijing, China
| | - Fang Ke
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Institute of Haematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Chen
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guo-Qiang Chen
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Institute of Haematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Research Units of Stress and Tumor, Chinese Academy of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Hainan Academy of Medical Sciences, School of Basic Medicine and Life Science, Hainan Medical University, Haikou, China
| | - Liang Chen
- RNA Institute, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Fan Bai
- Biomedical Pioneering Innovation Centre (BIOPIC) and Translational Cancer Research Centre, School of Life Sciences, First Hospital, Peking University, Beijing, China
| | - Tariq Enver
- Department of Cancer Biology, UCL Cancer Institute, University College London, London, UK
| | - Guo-Hong Li
- National laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Huai-Fang Li
- Department of Obstetrics and Gynecology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Deng-Li Hong
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Institute of Haematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Research Units of Stress and Tumor, Chinese Academy of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Innovative Research Team of High-level Local Universities in Shanghai, Shanghai, China.
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
21
|
Liu W, Kovacs AH, Hou J. Cancer Cells in Sleep Mode: Wake Them to Eliminate or Keep Them Asleep Forever? Cells 2024; 13:2022. [PMID: 39682769 PMCID: PMC11640528 DOI: 10.3390/cells13232022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 11/03/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Cancer cell dormancy is a critical phase in cancer development, wherein cancer cells exist in a latent state marked by temporary but reversible growth arrest. This dormancy phase contributes to anticancer drug resistance, cancer recurrence, and metastasis. Treatment strategies aimed at cancer dormancy can be categorized into two contradictory approaches: inducing cancer cells into a dormant state or eliminating dormant cells. While the former seeks to establish permanent dormancy, the latter aims at eradicating this small population of dormant cells. In this review, we explore the current advancements in therapeutic methods targeting cancer cell dormancy and discuss future strategies. The concept of cancer cell dormancy has emerged as a promising avenue for novel cancer treatments, holding the potential for breakthroughs in the future.
Collapse
Affiliation(s)
- Wenjie Liu
- Department of Chemistry, Lakehead University, 955 Oliver Rd, Thunder Bay, ON P7B 5E1, Canada; (W.L.); (A.H.K.)
- Thunder Bay Regional Health Research Institute, 980 Oliver Road, Thunder Bay, ON P7B 6V4, Canada
| | - Antal H. Kovacs
- Department of Chemistry, Lakehead University, 955 Oliver Rd, Thunder Bay, ON P7B 5E1, Canada; (W.L.); (A.H.K.)
| | - Jinqiang Hou
- Department of Chemistry, Lakehead University, 955 Oliver Rd, Thunder Bay, ON P7B 5E1, Canada; (W.L.); (A.H.K.)
- Thunder Bay Regional Health Research Institute, 980 Oliver Road, Thunder Bay, ON P7B 6V4, Canada
| |
Collapse
|
22
|
Arya S, Khare R, Garg I, Srivastava S. Gene expression profiling in Venous thromboembolism: Insights from publicly available datasets. Comput Biol Chem 2024; 113:108246. [PMID: 39413445 DOI: 10.1016/j.compbiolchem.2024.108246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/03/2024] [Accepted: 10/09/2024] [Indexed: 10/18/2024]
Abstract
BACKGROUND Venous thromboembolism (VTE) is the third most common cardiovascular disease and is a major cause of mobility and mortality worldwide. VTE is a complex multifactorial disease and genetic mechanisms underlying its pathogenesis is yet to be completely elucidated. The aim of the present study was to identify hub genes and pathways involved in development and progression of blood clot during VTE using gene expression data from public repositories. METHODOLOGY Differential gene expression (DEG) data from two datasets, GSE48000 and GSE19151 were analysed using GEO2R tool. Gene expression data of VTE patients were compared to that of healthy controls using various bioinformatics tools. RESULTS When the differentially expressed genes of the two datasets were compared, it was found that 19 genes were up-regulated while 134 genes were down-regulated. Gene ontology (GO) and pathway analysis revealed that pathways such as complement and coagulation cascade and B-cell receptor signalling along with DNA methylation, DNA alkylation and inflammatory genes were significantly up-regulated in VTE patients. On the other hand, differentially down-regulated genes included mitochondrial translation elongation, termination and biosysthesis along with heme biosynthesis, erythrocyte differentiation and homeostasis. The top 5 up-regulated hub genes obtained by protein-protein interaction (PPI) network analysis included MYC, FOS, SGK1, CR2 and CXCR4, whereas the top 5 down-regulated hub genes included MRPL13, MRPL3, MRPL11, RPS29 and RPL9. The up-regulated hub genes are functionally involved in maintain vascular integrity and complementation cascade while the down-regulated hub genes were mostly mitochondrial ribosomal proteins. CONCLUSION Present study highlights significantly enriched pathways and genes associated with VTE development and prognosis. The data hereby obtained could be used for designing newer diagnostic and therapeutic tools for VTE management.
Collapse
Affiliation(s)
- Sunanda Arya
- Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi 110054, India
| | - Rashi Khare
- Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi 110054, India
| | - Iti Garg
- Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi 110054, India
| | - Swati Srivastava
- Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi 110054, India.
| |
Collapse
|
23
|
Fotakopoulos G, Montasr MM, Georgakopoulou VE, Gatos C, Foroglou N. Association Between Polymorphisms in DNA Repair Genes and Glioma Susceptibility: A Meta-Analysis of Four Single Nucleotide Polymorphisms (rs3212986, rs13181, rs25487, and rs861539). Cureus 2024; 16:e76084. [PMID: 39834980 PMCID: PMC11743921 DOI: 10.7759/cureus.76084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2024] [Indexed: 01/22/2025] Open
Abstract
Cases with central nervous system tumors represent a small amount of all tumors, and the diagnosis of high-grade gliomas (HGGs) is mostly difficult as they frequently show intratumoral morphological heterogeneity. Genetic factors, such as single nucleotide polymorphisms (SNPs), have an important role in modifying glioma susceptibility. We conducted a comprehensive meta-analysis to investigate the ERCC1 (rs3212986), ERCC2 (rs13181), XRCC1 (rs25487), and XRCC3 (rs861539) genes to see if they are any risk factors for glioma susceptibility. We identified 30 eligible studies investigating the PubMed records (up to January 2024) via a mishmash of the subsequent terms: brain tumors, glioma, glioblastoma, gene associations, SNPs, XRCC1, XRCC3, ERCC1, and ERCC2. The total number of patients was 23678 (9731 in cases (poor outcome) and 13947 in controls (good outcome)). This comprehensive meta-analysis declared a significant association between ERCC2 rs13181, XRCC1 rs25487, and the risk of glioma.
Collapse
Affiliation(s)
- George Fotakopoulos
- Department of Neurosurgery, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, GRC
| | - Mohamed M Montasr
- Department of Neurosurgery, General University Hospital of Larissa, Larissa, GRC
| | | | - Charalabos Gatos
- Department of Neurosurgery, General University Hospital of Larissa, Larissa, GRC
| | - Nikolaos Foroglou
- Department of Neurosurgery, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, GRC
| |
Collapse
|
24
|
Zalenski N, He Y, Suo Z. Mechanistic Basis for a Single Amino Acid Residue Mutation Causing Human DNA Ligase 1 Deficiency, A Rare Pediatric Disease. J Mol Biol 2024; 436:168813. [PMID: 39374888 DOI: 10.1016/j.jmb.2024.168813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/26/2024] [Accepted: 10/01/2024] [Indexed: 10/09/2024]
Abstract
In mammalian cells, DNA ligase 1 (LIG1) functions as the primary DNA ligase in both genomic replication and single-strand break repair. Several reported mutations in human LIG1, including R305Q, R641L, and R771W, cause LIG1 syndrome, a primary immunodeficiency. While the R641L and R771W mutations, respectively located in the nucleotidyl transferase and oligonucleotide binding domains, have been biochemically characterized and shown to reduce catalytic efficiency, the recently reported R305Q mutation within the DNA binding domain (DBD) remains mechanistically unexplored. The R641L and R771W mutations are known to decrease the catalytic activity of LIG1 by affecting both interdomain interactions and DNA binding during catalysis, without significantly impacting overall DNA affinity. To elucidate the molecular basis of the LIG1 syndrome-causing R305Q mutation, we purified this single-residue mutant protein and investigated its secondary structure, protein stability, DNA binding affinity, and catalytic efficiency. Our findings reveal that the R305Q mutation significantly impairs the function of LIG1 by disrupting the DBD-DNA interactions, leading to a 7-21-fold lower DNA binding affinity and a 33-300-fold reduced catalytic efficiency of LIG1. Additionally, the R305Q mutation slightly decreases LIG1's protein stability by 2 to 3.6 °C, on par with the effect observed previously with either the R641L or R771W mutant. Collectively, our results uncover a new mechanism whereby the R305Q mutation impairs LIG1-catalyzed nicked DNA ligation, resulting in LIG1 syndrome, and highlight the crucial roles of the DBD-DNA interactions in tight DNA binding and efficient LIG1 catalysis.
Collapse
Affiliation(s)
- Nikita Zalenski
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | - Yufan He
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | - Zucai Suo
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA.
| |
Collapse
|
25
|
Rossetti GG, Dommann N, Karamichali A, Dionellis VS, Asensio Aldave A, Yarahmadov T, Rodriguez-Carballo E, Keogh A, Candinas D, Stroka D, Halazonetis TD. In vivo DNA replication dynamics unveil aging-dependent replication stress. Cell 2024; 187:6220-6234.e13. [PMID: 39293447 DOI: 10.1016/j.cell.2024.08.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 03/16/2024] [Accepted: 08/19/2024] [Indexed: 09/20/2024]
Abstract
The genome duplication program is affected by multiple factors in vivo, including developmental cues, genotoxic stress, and aging. Here, we monitored DNA replication initiation dynamics in regenerating livers of young and old mice after partial hepatectomy to investigate the impact of aging. In young mice, the origin firing sites were well defined; the majority were located 10-50 kb upstream or downstream of expressed genes, and their position on the genome was conserved in human cells. Old mice displayed the same replication initiation sites, but origin firing was inefficient and accompanied by a replication stress response. Inhibitors of the ATR checkpoint kinase fully restored origin firing efficiency in the old mice but at the expense of an inflammatory response and without significantly enhancing the fraction of hepatocytes entering the cell cycle. These findings unveil aging-dependent replication stress and a crucial role of ATR in mitigating the stress-associated inflammation, a hallmark of aging.
Collapse
Affiliation(s)
- Giacomo G Rossetti
- Department of Molecular and Cellular Biology, University of Geneva, Geneva 1205, Switzerland
| | - Noëlle Dommann
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Angeliki Karamichali
- Department of Molecular and Cellular Biology, University of Geneva, Geneva 1205, Switzerland
| | - Vasilis S Dionellis
- Department of Molecular and Cellular Biology, University of Geneva, Geneva 1205, Switzerland
| | - Ainhoa Asensio Aldave
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Tural Yarahmadov
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland
| | | | - Adrian Keogh
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Daniel Candinas
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Deborah Stroka
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department for BioMedical Research, University of Bern, Bern, Switzerland.
| | - Thanos D Halazonetis
- Department of Molecular and Cellular Biology, University of Geneva, Geneva 1205, Switzerland.
| |
Collapse
|
26
|
He M, Jiang H, Li S, Xue M, Wang H, Zheng C, Tong J. The crosstalk between DNA-damage responses and innate immunity. Int Immunopharmacol 2024; 140:112768. [PMID: 39088918 DOI: 10.1016/j.intimp.2024.112768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/14/2024] [Accepted: 07/22/2024] [Indexed: 08/03/2024]
Abstract
DNA damage is typically caused during cell growth by DNA replication stress or exposure to endogenous or external toxins. The accumulation of damaged DNA causes genomic instability, which is the root cause of many serious disorders. Multiple cellular organisms utilize sophisticated signaling pathways against DNA damage, collectively known as DNA damage response (DDR) networks. Innate immune responses are activated following cellular abnormalities, including DNA damage. Interestingly, recent studies have indicated that there is an intimate relationship between the DDR network and innate immune responses. Diverse kinds of cytosolic DNA sensors, such as cGAS and STING, recognize damaged DNA and induce signals related to innate immune responses, which link defective DDR to innate immunity. Moreover, DDR components operate in immune signaling pathways to induce IFNs and/or a cascade of inflammatory cytokines via direct interactions with innate immune modulators. Consistently, defective DDR factors exacerbate the innate immune imbalance, resulting in severe diseases, including autoimmune disorders and tumorigenesis. Here, the latest progress in understanding crosstalk between the DDR network and innate immune responses is reviewed. Notably, the dual function of innate immune modulators in the DDR network may provide novel insights into understanding and developing targeted immunotherapies for DNA damage-related diseases, even carcinomas.
Collapse
Affiliation(s)
- Mei He
- College of Life Sciences, Hebei University, Baoding 071002, China; Institute of Life Science and Green Development, Hebei University, Baoding 071002, China
| | - Hua Jiang
- Department of Hematology, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200000, China
| | - Shun Li
- Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu 610041, China
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450001, China.
| | - Huiqing Wang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China.
| | - Chunfu Zheng
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada.
| | - Jie Tong
- College of Life Sciences, Hebei University, Baoding 071002, China.
| |
Collapse
|
27
|
Bai B, Ma Y, Liu D, Zhang Y, Zhang W, Shi R, Zhou Q. DNA damage caused by chemotherapy has duality, and traditional Chinese medicine may be a better choice to reduce its toxicity. Front Pharmacol 2024; 15:1483160. [PMID: 39502534 PMCID: PMC11534686 DOI: 10.3389/fphar.2024.1483160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/14/2024] [Indexed: 11/08/2024] Open
Abstract
Background DNA damage induced by chemotherapy has duality. It affects the efficacy of chemotherapy and constrains its application. An increasing number of studies have shown that traditional Chinese medicine (TCM) is highly effective in reducing side-effects induced by chemotherapy due to its natural, non-toxic and many sourced from food. Recent advancements have demonstrated survival rates are improved attributable to effective chemotherapy. DNA damage is the principal mechanism underlying chemotherapy. However, not all instances of DNA damage are beneficial. Chemotherapy induces DNA damage in normal cells, leading to side effects. It affects the efficacy of chemotherapy and constrains its application. Objectives This review aims to summarize the dual nature of DNA damage induced by chemotherapy and explore how TCM can mitigate chemotherapy-induced side effects. Results The review summarized the latest research progress in DNA damage caused by chemotherapy and the effect of alleviating side effects by TCM. It focused on advantages and disadvantages of chemotherapy, the mechanism of drugs and providing insights for rational and effective clinical treatment and serving as a basis for experiment. In this review, we described the mechanisms of DNA damage, associated chemotherapeutics, and their toxicity. Furthermore, we explored Chinese herb that can alleviate chemotherapy-induced side-effects. Conclusion We highlight key mechanisms of DNA damage caused by chemotherapeutics and discuss specific TCM herbs that have shown potential in reducing these side effects. It can provide reference for clinical and basic research.
Collapse
Affiliation(s)
- Bufan Bai
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yingrui Ma
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Deng Liu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yifei Zhang
- Department of Intensive Care Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weihong Zhang
- Breast Surgery Department, Baoshan Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Rong Shi
- Department of Intensive Care Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qianmei Zhou
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Dongfang Hospital Affiliated to Shanghai Tongji University, Shanghai, China
| |
Collapse
|
28
|
Yuan XQ, Lei YM, Li YH, Zhou XM, Yang X, Chai YQ, Yuan R, Zhuo Y. Sequentially Activated-Dumbbell DNA Nanodevices for Accurate Detection of Uracil-DNA Glycosylase via PER-Based Orthogonal Signal Outputs. Anal Chem 2024; 96:17013-17020. [PMID: 39392054 DOI: 10.1021/acs.analchem.4c04477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Accurate and reliable detection of uracil-DNA glycosylase (UDG) activity is crucial for clinical diagnosis and prognosis assessment. However, current techniques for accurately monitoring UDG activity still face significant challenges due to the single input or output signal modes. Here, we develop a sequentially activated-dumbbell DNA nanodevice (SEAD) that enables precise and reliable evaluation of UDG activity through primer exchange reactions (PER)-based orthogonal signal output. The SEAD incorporates a double-hairpin structure with a stem containing two deoxyuridine (dU) sites for target recognition and two preblocked primer binding regions for target amplification and signal output. Upon UDG recognition of dU, the SEAD can be cleaved by apurinic/apyrimidinic endonuclease 1 (APE1), generating two different hairpins with exposed primer binding regions. These hairpins serve as templates to initiate the parallel PER, enabling the extending of two different amplification products: a long single-stranded DNA (ssDNA) with repetitive sequences and a short ferrocene-labeled ssDNA with complementary sequences. These products further self-assemble into DNA nano-strings in an orthogonal manner that act as an electrochemiluminescence signal switch, enabling precise detection of low-abundance UDG. This work develops a sequential input and orthogonal output strategy for accurately monitoring UDG activity, highlighting the significant potential in cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Xiao-Qing Yuan
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Yan-Mei Lei
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Ying-Huan Li
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Xue-Mei Zhou
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Xia Yang
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Ya-Qin Chai
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Ruo Yuan
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Ying Zhuo
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, Institute of Developmental Biology and Regenerative Medicine, College of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| |
Collapse
|
29
|
Chang Y, Yu C, Dai X, Sun H, Tang T. Association of dietary inflammatory index and dietary oxidative balance score with gastrointestinal cancers in NHANES 2005-2018. BMC Public Health 2024; 24:2760. [PMID: 39385181 PMCID: PMC11465896 DOI: 10.1186/s12889-024-20268-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/03/2024] [Indexed: 10/11/2024] Open
Abstract
BACKGROUND&AIMS Gastrointestinal (GI) cancers, including gastric, liver, esophageal, pancreatic, and colorectal cancers, represent significant global health burdens. Emerging evidence suggests that dietary patterns, particularly their inflammatory and oxidative properties, may influence cancer risk. The Dietary Inflammatory Index (DII) and Dietary Oxidative Balance Score (DOBS) assess the inflammatory and oxidative effects of diets, respectively. This study aims to explore the association between DII, DOBS, and the combined risk of GI cancers, and investigates the potential mediating roles of serum albumin and red cell distribution width (RDW). METHODS Data from 26,320 participants in the NHANES 2005-2018 cycles were analyzed. DII was calculated based on 28 dietary components, and DOBS included 17 nutrients (3 pro-oxidants and 14 antioxidants). Logistic regression models assessed the associations between DII, DOBS, and GI cancers. Restricted cubic spline (RCS) models examined dose-response relationships. Mediation analysis evaluated the roles of serum albumin and RDW. Subgroup analyses explored interactions with demographic and health-related factors. RESULTS Higher DII was associated with increased GI cancer risk (OR: 1.26, 95% CI: 1.07-1.49 per unit increase), while higher DOBS was associated with reduced risk (OR: 0.90, 95% CI: 0.76-0.99 per unit increase). RCS analysis indicated a significant nonlinear relationship between DII and GI cancer risk. Serum albumin and RDW partially mediated the associations between DII, DOBS, and GI cancers. Subgroup analyses showed stronger associations for DII among certain demographics, and significant interactions were found between DII and BMI. For DOBS, significant interactions were observed with age and BMI. CONCLUSION This study reveals significant associations between dietary inflammatory and oxidative balance scores and GI cancer risk. Higher DII is linked to increased risk, while higher DOBS is protective. The mediating roles of serum albumin and RDW provide insights into underlying mechanisms. These findings underscore the potential of dietary modifications in GI cancer prevention and management, emphasizing the importance of anti-inflammatory and antioxidant-rich diets.
Collapse
Affiliation(s)
- Yu Chang
- The First Hospital of Jilin University, No.1 Xinmin Street, Changchun, 130012, China
| | - Chanjiao Yu
- The First Hospital of Jilin University, No.1 Xinmin Street, Changchun, 130012, China
| | - Xianyu Dai
- The First Hospital of Jilin University, No.1 Xinmin Street, Changchun, 130012, China
| | - Haibo Sun
- The First Hospital of Jilin University, No.1 Xinmin Street, Changchun, 130012, China.
| | - Tongyu Tang
- The First Hospital of Jilin University, No.1 Xinmin Street, Changchun, 130012, China.
| |
Collapse
|
30
|
Loeffler CML, El Nahhas OSM, Muti HS, Carrero ZI, Seibel T, van Treeck M, Cifci D, Gustav M, Bretz K, Gaisa NT, Lehmann KV, Leary A, Selenica P, Reis-Filho JS, Ortiz-Bruechle N, Kather JN. Prediction of homologous recombination deficiency from routine histology with attention-based multiple instance learning in nine different tumor types. BMC Biol 2024; 22:225. [PMID: 39379982 PMCID: PMC11462727 DOI: 10.1186/s12915-024-02022-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 09/26/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Homologous recombination deficiency (HRD) is recognized as a pan-cancer predictive biomarker that potentially indicates who could benefit from treatment with PARP inhibitors (PARPi). Despite its clinical significance, HRD testing is highly complex. Here, we investigated in a proof-of-concept study whether Deep Learning (DL) can predict HRD status solely based on routine hematoxylin & eosin (H&E) histology images across nine different cancer types. METHODS We developed a deep learning pipeline with attention-weighted multiple instance learning (attMIL) to predict HRD status from histology images. As part of our approach, we calculated a genomic scar HRD score by combining loss of heterozygosity (LOH), telomeric allelic imbalance (TAI), and large-scale state transitions (LST) from whole genome sequencing (WGS) data of n = 5209 patients across two independent cohorts. The model's effectiveness was evaluated using the area under the receiver operating characteristic curve (AUROC), focusing on its accuracy in predicting genomic HRD against a clinically recognized cutoff value. RESULTS Our study demonstrated the predictability of genomic HRD status in endometrial, pancreatic, and lung cancers reaching cross-validated AUROCs of 0.79, 0.58, and 0.66, respectively. These predictions generalized well to an external cohort, with AUROCs of 0.93, 0.81, and 0.73. Moreover, a breast cancer-trained image-based HRD classifier yielded an AUROC of 0.78 in the internal validation cohort and was able to predict HRD in endometrial, prostate, and pancreatic cancer with AUROCs of 0.87, 0.84, and 0.67, indicating that a shared HRD-like phenotype occurs across these tumor entities. CONCLUSIONS This study establishes that HRD can be directly predicted from H&E slides using attMIL, demonstrating its applicability across nine different tumor types.
Collapse
Affiliation(s)
- Chiara Maria Lavinia Loeffler
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
- Department of Medicine I, Faculty of Medicine Carl Gustav Carus, University Hospitaland, Technische Universität Dresden , Dresden, Germany
| | - Omar S M El Nahhas
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Hannah Sophie Muti
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
- Department for Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Zunamys I Carrero
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Tobias Seibel
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Marko van Treeck
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Didem Cifci
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Marco Gustav
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Kevin Bretz
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Nadine T Gaisa
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
- Joint Research Center Computational Biomedicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Kjong-Van Lehmann
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
- Joint Research Center Computational Biomedicine, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology, Aachen Bonn Cologne Duesseldorf (CIO ABCD), Duesseldorf, Germany
- Cancer Research Center Cologne-Essen, University Hospital Cologne, Cologne, Germany
| | - Alexandra Leary
- Gynecological Cancer Unit, Department of Medicine, Institut Gustave Roussy, Villejuif, France
| | - Pier Selenica
- Experimental Pathology, Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jorge S Reis-Filho
- Experimental Pathology, Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nadina Ortiz-Bruechle
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology, Aachen Bonn Cologne Duesseldorf (CIO ABCD), Duesseldorf, Germany
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany.
- Department of Medicine I, Faculty of Medicine Carl Gustav Carus, University Hospitaland, Technische Universität Dresden , Dresden, Germany.
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK.
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
31
|
Kuper J, Hove T, Maidl S, Neitz H, Sauer F, Kempf M, Schroeder T, Greiter E, Höbartner C, Kisker C. XPD stalled on cross-linked DNA provides insight into damage verification. Nat Struct Mol Biol 2024; 31:1580-1588. [PMID: 38806694 PMCID: PMC11479942 DOI: 10.1038/s41594-024-01323-5] [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: 03/11/2024] [Accepted: 04/24/2024] [Indexed: 05/30/2024]
Abstract
The superfamily 2 helicase XPD is a central component of the general transcription factor II H (TFIIH), which is essential for transcription and nucleotide excision DNA repair (NER). Within these two processes, the helicase function of XPD is vital for NER but not for transcription initiation, where XPD acts only as a scaffold for other factors. Using cryo-EM, we deciphered one of the most enigmatic steps in XPD helicase action: the active separation of double-stranded DNA (dsDNA) and its stalling upon approaching a DNA interstrand cross-link, a highly toxic form of DNA damage. The structure shows how dsDNA is separated and reveals a highly unusual involvement of the Arch domain in active dsDNA separation. Combined with mutagenesis and biochemical analyses, we identified distinct functional regions important for helicase activity. Surprisingly, those areas also affect core TFIIH translocase activity, revealing a yet unencountered function of XPD within the TFIIH scaffold. In summary, our data provide a universal basis for NER bubble formation, XPD damage verification and XPG incision.
Collapse
Affiliation(s)
- Jochen Kuper
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany.
| | - Tamsanqa Hove
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
| | - Sarah Maidl
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
| | - Hermann Neitz
- Institute of Organic Chemistry, University of Würzburg, Würzburg, Germany
| | - Florian Sauer
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
| | - Maximilian Kempf
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
| | - Till Schroeder
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
| | - Elke Greiter
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
| | - Claudia Höbartner
- Institute of Organic Chemistry, University of Würzburg, Würzburg, Germany
- Center for Nanosystems Chemistry (CNC), University of Würzburg, Würzburg, Germany
| | - Caroline Kisker
- Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany.
| |
Collapse
|
32
|
Wang SW, Zheng QY, Hong WF, Tang BF, Hsu SJ, Zhang Y, Zheng XB, Zeng ZC, Gao C, Ke AW, Du SS. Mechanism of immune activation mediated by genomic instability and its implication in radiotherapy combined with immune checkpoint inhibitors. Radiother Oncol 2024; 199:110424. [PMID: 38997092 DOI: 10.1016/j.radonc.2024.110424] [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: 04/07/2024] [Revised: 06/27/2024] [Accepted: 07/05/2024] [Indexed: 07/14/2024]
Abstract
Various genetic and epigenetic changes associated with genomic instability (GI), including DNA damage repair defects, chromosomal instability, and mitochondrial GI, contribute to development and progression of cancer. These alterations not only result in DNA leakage into the cytoplasm, either directly or through micronuclei, but also trigger downstream inflammatory signals, such as the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway. Apart from directly inducing DNA damage to eliminate cancer cells, radiotherapy (RT) exerts its antitumor effects through intracellular DNA damage sensing mechanisms, leading to the activation of downstream inflammatory signaling pathways. This not only enables local tumor control but also reshapes the immune microenvironment, triggering systemic immune responses. The combination of RT and immunotherapy has emerged as a promising approach to increase the probability of abscopal effects, where distant tumors respond to treatment due to the systemic immunomodulatory effects. This review emphasizes the importance of GI in cancer biology and elucidates the mechanisms by which RT induces GI remodeling of the immune microenvironment. By elucidating the mechanisms of GI and RT-induced immune responses, we aim to emphasize the crucial importance of this approach in modern oncology. Understanding the impact of GI on tumor biological behavior and therapeutic response, as well as the possibility of activating systemic anti-tumor immunity through RT, will pave the way for the development of new treatment strategies and improve prognosis for patients.
Collapse
Affiliation(s)
- Si-Wei Wang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Zhongshan Hospital, Liver Cancer Institute, Fudan University, Shanghai 200030, China
| | - Qiu-Yi Zheng
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Wei-Feng Hong
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Bu-Fu Tang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Shu-Jung Hsu
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Yang Zhang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Xiao-Bin Zheng
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Zhao-Chong Zeng
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Chao Gao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Zhongshan Hospital, Liver Cancer Institute, Fudan University, Shanghai 200030, China.
| | - Ai-Wu Ke
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Zhongshan Hospital, Liver Cancer Institute, Fudan University, Shanghai 200030, China.
| | - Shi-Suo Du
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China.
| |
Collapse
|
33
|
Gonzalo-Hansen C, Steurer B, Janssens RC, Zhou D, van Sluis M, Lans H, Marteijn JA. Differential processing of RNA polymerase II at DNA damage correlates with transcription-coupled repair syndrome severity. Nucleic Acids Res 2024; 52:9596-9612. [PMID: 39021334 PMCID: PMC11381366 DOI: 10.1093/nar/gkae618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 06/25/2024] [Accepted: 07/10/2024] [Indexed: 07/20/2024] Open
Abstract
DNA damage severely impedes gene transcription by RNA polymerase II (Pol II), causing cellular dysfunction. Transcription-Coupled Nucleotide Excision Repair (TC-NER) specifically removes such transcription-blocking damage. TC-NER initiation relies on the CSB, CSA and UVSSA proteins; loss of any results in complete TC-NER deficiency. Strikingly, UVSSA deficiency results in UV-Sensitive Syndrome (UVSS), with mild cutaneous symptoms, while loss of CSA or CSB activity results in the severe Cockayne Syndrome (CS), characterized by neurodegeneration and premature aging. Thus far the underlying mechanism for these contrasting phenotypes remains unclear. Live-cell imaging approaches reveal that in TC-NER proficient cells, lesion-stalled Pol II is swiftly resolved, while in CSA and CSB knockout (KO) cells, elongating Pol II remains damage-bound, likely obstructing other DNA transacting processes and shielding the damage from alternative repair pathways. In contrast, in UVSSA KO cells, Pol II is cleared from the damage via VCP-mediated proteasomal degradation which is fully dependent on the CRL4CSA ubiquitin ligase activity. This Pol II degradation might provide access for alternative repair mechanisms, such as GG-NER, to remove the damage. Collectively, our data indicate that the inability to clear lesion-stalled Pol II from the chromatin, rather than TC-NER deficiency, causes the severe phenotypes observed in CS.
Collapse
Affiliation(s)
- Camila Gonzalo-Hansen
- Department of Molecular Genetics, Oncode Institute, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Barbara Steurer
- Department of Molecular Genetics, Oncode Institute, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Roel C Janssens
- Department of Molecular Genetics, Oncode Institute, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Di Zhou
- Department of Molecular Genetics, Oncode Institute, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Marjolein van Sluis
- Department of Molecular Genetics, Oncode Institute, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Hannes Lans
- Department of Molecular Genetics, Oncode Institute, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jurgen A Marteijn
- Department of Molecular Genetics, Oncode Institute, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
34
|
Mendes IC, Dos Reis Bertoldo W, Miranda-Junior AS, Assis AVD, Repolês BM, Ferreira WRR, Chame DF, Souza DDL, Pavani RS, Macedo AM, Franco GR, Serra E, Perdomo V, Menck CFM, da Silva Leandro G, Fragoso SP, Barbosa Elias MCQ, Machado CR. DNA lesions that block transcription induce the death of Trypanosoma cruzi via ATR activation, which is dependent on the presence of R-loops. DNA Repair (Amst) 2024; 141:103726. [PMID: 39096697 DOI: 10.1016/j.dnarep.2024.103726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 06/25/2024] [Accepted: 07/07/2024] [Indexed: 08/05/2024]
Abstract
Trypanosoma cruzi is the etiological agent of Chagas disease and a peculiar eukaryote with unique biological characteristics. DNA damage can block RNA polymerase, activating transcription-coupled nucleotide excision repair (TC-NER), a DNA repair pathway specialized in lesions that compromise transcription. If transcriptional stress is unresolved, arrested RNA polymerase can activate programmed cell death. Nonetheless, how this parasite modulates these processes is unknown. Here, we demonstrate that T. cruzi cell death after UV irradiation, a genotoxic agent that generates lesions resolved by TC-NER, depends on active transcription and is signaled mainly by an apoptotic-like pathway. Pre-treated parasites with α-amanitin, a selective RNA polymerase II inhibitor, become resistant to such cell death. Similarly, the gamma pre-irradiated cells are more resistant to UV when the transcription processes are absent. The Cockayne Syndrome B protein (CSB) recognizes blocked RNA polymerase and can initiate TC-NER. Curiously, CSB overexpression increases parasites' cell death shortly after UV exposure. On the other hand, at the same time after irradiation, the single-knockout CSB cells show resistance to the same treatment. UV-induced fast death is signalized by the exposition of phosphatidylserine to the outer layer of the membrane, indicating a cell death mainly by an apoptotic-like pathway. Furthermore, such death is suppressed in WT parasites pre-treated with inhibitors of ataxia telangiectasia and Rad3-related (ATR), a key DDR kinase. Signaling for UV radiation death may be related to R-loops since the overexpression of genes associated with the resolution of these structures suppress it. Together, results suggest that transcription blockage triggered by UV radiation activates an ATR-dependent apoptosis-like mechanism in T. cruzi, with the participation of CSB protein in this process.
Collapse
Affiliation(s)
- Isabela Cecilia Mendes
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Willian Dos Reis Bertoldo
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Adalberto Sales Miranda-Junior
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Antônio Vinícius de Assis
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Bruno Marçal Repolês
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Wesley Roger Rodrigues Ferreira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Daniela Ferreira Chame
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Daniela De Laet Souza
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Raphael Souza Pavani
- Laboratório Especial de Ciclo Celular, Instituto Butantan, São Paulo, MG, São Paulo, SP 05503-900, Brazil
| | - Andrea Mara Macedo
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Glória Regina Franco
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Esteban Serra
- Instituto de Biología Molecular y Celular de Rosario, CONICET, 2000 Rosario, Santa Fe, Argentina; Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, 2000 Rosario, Santa Fe, Argentina
| | - Virginia Perdomo
- Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, 2000 Rosario, Santa Fe, Argentina
| | - Carlos Frederico Martins Menck
- Departamento de Microbiologia, Instituto de Ciências Biomédicas (ICB), Universidade de São Paulo (USP), São Paulo, SP 05508-900, Brazil
| | - Giovana da Silva Leandro
- Departamento de Microbiologia, Instituto de Ciências Biomédicas (ICB), Universidade de São Paulo (USP), São Paulo, SP 05508-900, Brazil
| | | | | | - Carlos Renato Machado
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil.
| |
Collapse
|
35
|
Gao Z, Luan X, Wang X, Han T, Li X, Li Z, Li P, Zhou Z. DNA damage response-related ncRNAs as regulators of therapy resistance in cancer. Front Pharmacol 2024; 15:1390300. [PMID: 39253383 PMCID: PMC11381396 DOI: 10.3389/fphar.2024.1390300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 08/14/2024] [Indexed: 09/11/2024] Open
Abstract
The DNA damage repair (DDR) pathway is a complex signaling cascade that can sense DNA damage and trigger cellular responses to DNA damage to maintain genome stability and integrity. A typical hallmark of cancer is genomic instability or nonintegrity, which is closely related to the accumulation of DNA damage within cancer cells. The treatment principles of radiotherapy and chemotherapy for cancer are based on their cytotoxic effects on DNA damage, which are accompanied by severe and unnecessary side effects on normal tissues, including dysregulation of the DDR and induced therapeutic tolerance. As a driving factor for oncogenes or tumor suppressor genes, noncoding RNA (ncRNA) have been shown to play an important role in cancer cell resistance to radiotherapy and chemotherapy. Recently, it has been found that ncRNA can regulate tumor treatment tolerance by altering the DDR induced by radiotherapy or chemotherapy in cancer cells, indicating that ncRNA are potential regulatory factors targeting the DDR to reverse tumor treatment tolerance. This review provides an overview of the basic information and functions of the DDR and ncRNAs in the tolerance or sensitivity of tumors to chemotherapy and radiation therapy. We focused on the impact of ncRNA (mainly microRNA [miRNA], long noncoding RNA [lncRNA], and circular RNA [circRNA]) on cancer treatment by regulating the DDR and the underlying molecular mechanisms of their effects. These findings provide a theoretical basis and new insights for tumor-targeted therapy and the development of novel drugs targeting the DDR or ncRNAs.
Collapse
Affiliation(s)
- Ziru Gao
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Xinchi Luan
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Xuezhe Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Tianyue Han
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Xiaoyuan Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Zeyang Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Zhixia Zhou
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, China
| |
Collapse
|
36
|
Ye BJ, Li DF, Li XY, Hao JL, Liu DJ, Yu H, Zhang CD. Methylation synthetic lethality: Exploiting selective drug targets for cancer therapy. Cancer Lett 2024; 597:217010. [PMID: 38849016 DOI: 10.1016/j.canlet.2024.217010] [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: 04/28/2024] [Revised: 05/26/2024] [Accepted: 05/30/2024] [Indexed: 06/09/2024]
Abstract
In cancer, synthetic lethality refers to the drug-induced inactivation of one gene and the inhibition of another in cancer cells by a drug, resulting in the death of only cancer cells; however, this effect is not present in normal cells, leading to targeted killing of cancer cells. Recent intensive epigenetic research has revealed that aberrant epigenetic changes are more frequently observed than gene mutations in certain cancers. Recently, numerous studies have reported various methylation synthetic lethal combinations involving DNA damage repair genes, metabolic pathway genes, and paralogs with significant results in cellular models, some of which have already entered clinical trials with promising results. This review systematically introduces the advantages of methylation synthetic lethality and describes the lethal mechanisms of methylation synthetic lethal combinations that have recently demonstrated success in cellular models. Furthermore, we discuss the future opportunities and challenges of methylation synthetic lethality in targeted anticancer therapies.
Collapse
Affiliation(s)
- Bing-Jie Ye
- Clinical Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Di-Fei Li
- Clinical Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Xin-Yun Li
- Clinical Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Jia-Lin Hao
- Central Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Di-Jie Liu
- Central Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Hang Yu
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Chun-Dong Zhang
- Central Laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| |
Collapse
|
37
|
Shen LP, Zhang WC, Deng JR, Qi ZH, Lin ZW, Wang ZD. Advances in the mechanism of small nucleolar RNA and its role in DNA damage response. Mil Med Res 2024; 11:53. [PMID: 39118131 PMCID: PMC11308251 DOI: 10.1186/s40779-024-00553-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
Small nucleolar RNAs (snoRNAs) were previously regarded as a class of functionally conserved housekeeping genes, primarily involved in the regulation of ribosome biogenesis by ribosomal RNA (rRNA) modification. However, some of them are involved in several biological processes via complex molecular mechanisms. DNA damage response (DDR) is a conserved mechanism for maintaining genomic stability to prevent the occurrence of various human diseases. It has recently been revealed that snoRNAs are involved in DDR at multiple levels, indicating their relevant theoretical and clinical significance in this field. The present review systematically addresses four main points, including the biosynthesis and classification of snoRNAs, the mechanisms through which snoRNAs regulate target molecules, snoRNAs in the process of DDR, and the significance of snoRNA in disease diagnosis and treatment. It focuses on the potential functions of snoRNAs in DDR to help in the discovery of the roles of snoRNAs in maintaining genome stability and pathological processes.
Collapse
Affiliation(s)
- Li-Ping Shen
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Wen-Cheng Zhang
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Jia-Rong Deng
- Graduate Collaborative Training Base of Academy of Military Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Zhen-Hua Qi
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Zhong-Wu Lin
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Zhi-Dong Wang
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, China.
- Graduate Collaborative Training Base of Academy of Military Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
38
|
Panebianco M, Cereda V, D’Andrea MR. Combination of the PARPi and ARSi in advanced castration resistant prostate cancer: a review of the recent phase III trials. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:997-1010. [PMID: 39351435 PMCID: PMC11438558 DOI: 10.37349/etat.2024.00260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 05/31/2024] [Indexed: 10/04/2024] Open
Abstract
Tumors with an impaired ability to repair DNA double-strand breaks by homologous recombination, including those with alterations in breast cancer 1 and 2 (BRCA1 and BRCA2) genes, are very sensitive to blocking DNA single-strand repair by inhibition of the poly (ADP-ribose) polymerase (PARP) enzyme. This provides the basis for a synthetic deadly strategy in the treatment of different types of cancer, such as prostate cancer (PCa). The phase 3 PROfound study was the first to lead to olaparib approval in patients with metastatic castration resistant PCa (mCRPC) and BRCA genes mutations. In recent years, the benefit of combination therapy consisted of a PARP inhibitor (PARPi) plus an androgen receptor signalling inhibitor (ARSi), was evaluated as first-line treatment of mCRPC, regardless of the mutational state of genes, participating in the homologous recombination repair (HRR). This review explores the role of PARPi in PCa and analyses the data of latest clinical trials exploring the PARPi-ARSi combinations, and how these results could change our clinical practice.
Collapse
Affiliation(s)
| | - Vittore Cereda
- Medical Oncology of ASL Roma 4 Hospital, 00053 Civitavecchia, Italy
| | | |
Collapse
|
39
|
Yan L, Shi J, Zhu J. Cellular and molecular events in colorectal cancer: biological mechanisms, cell death pathways, drug resistance and signalling network interactions. Discov Oncol 2024; 15:294. [PMID: 39031216 PMCID: PMC11265098 DOI: 10.1007/s12672-024-01163-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 07/15/2024] [Indexed: 07/22/2024] Open
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related deaths worldwide, affecting millions each year. It emerges from the colon or rectum, parts of the digestive system, and is closely linked to both genetic and environmental factors. In CRC, genetic mutations such as APC, KRAS, and TP53, along with epigenetic changes like DNA methylation and histone modifications, play crucial roles in tumor development and treatment responses. This paper delves into the complex biological underpinnings of CRC, highlighting the pivotal roles of genetic alterations, cell death pathways, and the intricate network of signaling interactions that contribute to the disease's progression. It explores the dysregulation of apoptosis, autophagy, and other cell death mechanisms, underscoring the aberrant activation of these pathways in CRC. Additionally, the paper examines how mutations in key molecular pathways, including Wnt, EGFR/MAPK, and PI3K, fuel CRC development, and how these alterations can serve as both diagnostic and prognostic markers. The dual function of autophagy in CRC, acting as a tumor suppressor or promoter depending on the context, is also scrutinized. Through a comprehensive analysis of cellular and molecular events, this research aims to deepen our understanding of CRC and pave the way for more effective diagnostics, prognostics, and therapeutic strategies.
Collapse
Affiliation(s)
- Lei Yan
- Medical Department, The Central Hospital of Shaoyang Affiliated to University of South China, Shaoyang, China
| | - Jia Shi
- Department of Obstetrics and Gynecology, The Central Hospital of Shaoyang Affiliated to University of South China, Shaoyang, China
| | - Jiazuo Zhu
- Department of Oncology, Xuancheng City Central Hospital, No. 117 Tong Road, Xuancheng, Anhui, China.
| |
Collapse
|
40
|
Cui C, Guo G, Chen TH. Toehold region triggered CRISPR/Cas12a trans-cleavage for detection of uracil-DNA glycosylase activity. Biotechnol J 2024; 19:e2400097. [PMID: 38987221 DOI: 10.1002/biot.202400097] [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: 02/19/2024] [Revised: 05/20/2024] [Accepted: 05/27/2024] [Indexed: 07/12/2024]
Abstract
DNA glycosylases are a group of enzymes that play a crucial role in the DNA repair process by recognizing and removing damaged or incorrect bases from DNA molecules, which maintains the integrity of the genetic information. The abnormal expression of uracil-DNA glycosylase (UDG), one of significant DNA glycosylases in the base-excision repair pathway, is linked to numerous diseases. Here, we proposed a simple UDG activity detection method based on toehold region triggered CRISPR/Cas12a trans-cleavage. The toehold region on hairpin DNA probe (HP) produced by UDG could induce the trans-cleavage of ssDNA with fluorophore and quencher, generating an obvious fluorescence signal. This protospacer adjacent motif (PAM)-free approach achieves remarkable sensitivity and specificity in detecting UDG, with a detection limit as low as 0.000368 U mL-1. Moreover, this method is able to screen inhibitors and measure UDG in complex biological samples. These advantages render it highly promising for applications in clinical diagnosis and drug discovery.
Collapse
Affiliation(s)
- Chenyu Cui
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
- Hong Kong Centre for Cerebro-cardiovascular Health Engineering, Hong Kong Science Park, Hong Kong, China
| | - Guihuan Guo
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Ting-Hsuan Chen
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
| |
Collapse
|
41
|
Garcia-Mouronte E, Pérez-González LA, Naharro-Rodriguez J, Fernández Guarino M. Understanding Active Photoprotection: DNA-Repair Enzymes and Antioxidants. Life (Basel) 2024; 14:822. [PMID: 39063576 PMCID: PMC11277730 DOI: 10.3390/life14070822] [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: 05/31/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
The detrimental effects of ultraviolet radiation (UVR) on human skin are well-documented, encompassing DNA damage, oxidative stress, and an increased risk of carcinogenesis. Conventional photoprotective measures predominantly rely on filters, which scatter or absorb UV radiation, yet fail to address the cellular damage incurred post-exposure. To fill this gap, antioxidant molecules and DNA-repair enzymes have been extensively researched, offering a paradigm shift towards active photoprotection capable of both preventing and reversing UV-induced damage. In the current review, we focused on "active photoprotection", assessing the state-of-the-art, latest advancements and scientific data from clinical trials and in vivo models concerning the use of DNA-repair enzymes and naturally occurring antioxidant molecules.
Collapse
Affiliation(s)
- Emilio Garcia-Mouronte
- Dermatology Department, Hospital Universitario Ramon y Cajal, Carretera M-607 km 9.1, 28034 Madrid, Spain; (L.A.P.-G.); (M.F.G.)
| | | | - Jorge Naharro-Rodriguez
- Dermatology Department, Hospital Universitario Ramon y Cajal, Carretera M-607 km 9.1, 28034 Madrid, Spain; (L.A.P.-G.); (M.F.G.)
| | | |
Collapse
|
42
|
Badja C, Momen S, Koh GCC, Boushaki S, Roumeliotis TI, Kozik Z, Jones I, Bousgouni V, Dias JML, Krokidis MG, Young J, Chen H, Yang M, Docquier F, Memari Y, Valcarcel-Zimenez L, Gupta K, Kong LR, Fawcett H, Robert F, Zhao S, Degasperi A, Kumar Y, Davies H, Harris R, Frezza C, Chatgilialoglu C, Sarkany R, Lehmann A, Bakal C, Choudhary J, Fassihi H, Nik-Zainal S. Insights from multi-omic modeling of neurodegeneration in xeroderma pigmentosum using an induced pluripotent stem cell system. Cell Rep 2024; 43:114243. [PMID: 38805398 DOI: 10.1016/j.celrep.2024.114243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 04/27/2024] [Accepted: 05/02/2024] [Indexed: 05/30/2024] Open
Abstract
Xeroderma pigmentosum (XP) is caused by defective nucleotide excision repair of DNA damage. This results in hypersensitivity to ultraviolet light and increased skin cancer risk, as sunlight-induced photoproducts remain unrepaired. However, many XP patients also display early-onset neurodegeneration, which leads to premature death. The mechanism of neurodegeneration is unknown. Here, we investigate XP neurodegeneration using pluripotent stem cells derived from XP patients and healthy relatives, performing functional multi-omics on samples during neuronal differentiation. We show substantially increased levels of 5',8-cyclopurine and 8-oxopurine in XP neuronal DNA secondary to marked oxidative stress. Furthermore, we find that the endoplasmic reticulum stress response is upregulated and reversal of the mutant genotype is associated with phenotypic rescue. Critically, XP neurons exhibit inappropriate downregulation of the protein clearance ubiquitin-proteasome system (UPS). Chemical enhancement of UPS activity in XP neuronal models improves phenotypes, albeit inadequately. Although more work is required, this study presents insights with intervention potential.
Collapse
Affiliation(s)
- Cherif Badja
- Department of Medical Genetics, Box 238, Level 6, Addenbrooke's Treatment Centre, Cambridge Biomedical Research Campus, Cambridge CB2 0QQ, UK; Early Cancer Institute, Department of Oncology, Box 197, Hutchison Research Centre, Cambridge Biomedical Research Campus, Cambridge CB2 0XZ, UK.
| | - Sophie Momen
- Department of Medical Genetics, Box 238, Level 6, Addenbrooke's Treatment Centre, Cambridge Biomedical Research Campus, Cambridge CB2 0QQ, UK; Early Cancer Institute, Department of Oncology, Box 197, Hutchison Research Centre, Cambridge Biomedical Research Campus, Cambridge CB2 0XZ, UK
| | - Gene Ching Chiek Koh
- Department of Medical Genetics, Box 238, Level 6, Addenbrooke's Treatment Centre, Cambridge Biomedical Research Campus, Cambridge CB2 0QQ, UK; Early Cancer Institute, Department of Oncology, Box 197, Hutchison Research Centre, Cambridge Biomedical Research Campus, Cambridge CB2 0XZ, UK
| | - Soraya Boushaki
- Department of Medical Genetics, Box 238, Level 6, Addenbrooke's Treatment Centre, Cambridge Biomedical Research Campus, Cambridge CB2 0QQ, UK; Early Cancer Institute, Department of Oncology, Box 197, Hutchison Research Centre, Cambridge Biomedical Research Campus, Cambridge CB2 0XZ, UK
| | - Theodoros I Roumeliotis
- Functional Proteomics Group, Institute of Cancer Research, Chester Betty Labs, 237 Fulham Road, London SW3 6JB, UK
| | - Zuza Kozik
- Functional Proteomics Group, Institute of Cancer Research, Chester Betty Labs, 237 Fulham Road, London SW3 6JB, UK
| | - Ian Jones
- Dynamical Cell Systems Laboratory, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Vicky Bousgouni
- Dynamical Cell Systems Laboratory, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - João M L Dias
- Department of Medical Genetics, Box 238, Level 6, Addenbrooke's Treatment Centre, Cambridge Biomedical Research Campus, Cambridge CB2 0QQ, UK; Early Cancer Institute, Department of Oncology, Box 197, Hutchison Research Centre, Cambridge Biomedical Research Campus, Cambridge CB2 0XZ, UK
| | - Marios G Krokidis
- Institute of Nanoscience and Nanotechnology, N.C.S.R. "Demokritos", Agia Paraskevi Attikis, 15310 Athens, Greece; Bioinformatics and Human Electrophysiology Laboratory, Department of Informatics, Ionian University, 49100 Corfu, Greece
| | - Jamie Young
- Department of Medical Genetics, Box 238, Level 6, Addenbrooke's Treatment Centre, Cambridge Biomedical Research Campus, Cambridge CB2 0QQ, UK; Early Cancer Institute, Department of Oncology, Box 197, Hutchison Research Centre, Cambridge Biomedical Research Campus, Cambridge CB2 0XZ, UK
| | - Hongwei Chen
- Wellcome Sanger Institute, Hinxton CB10 1RQ, UK; Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Ming Yang
- Medical Research Council Cancer Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XZ, UK; CECAD Research Center, Faculty of Medicine, University Hospital Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - France Docquier
- Department of Medical Genetics, Box 238, Level 6, Addenbrooke's Treatment Centre, Cambridge Biomedical Research Campus, Cambridge CB2 0QQ, UK
| | - Yasin Memari
- Department of Medical Genetics, Box 238, Level 6, Addenbrooke's Treatment Centre, Cambridge Biomedical Research Campus, Cambridge CB2 0QQ, UK; Early Cancer Institute, Department of Oncology, Box 197, Hutchison Research Centre, Cambridge Biomedical Research Campus, Cambridge CB2 0XZ, UK
| | - Lorea Valcarcel-Zimenez
- Medical Research Council Cancer Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XZ, UK; CECAD Research Center, Faculty of Medicine, University Hospital Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Komal Gupta
- Medical Research Council Cancer Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XZ, UK
| | - Li Ren Kong
- Medical Research Council Cancer Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XZ, UK; NUS Centre for Cancer Research, N2CR, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; Cancer Science Institute of Singapore, Singapore 117599, Singapore
| | - Heather Fawcett
- Genome Damage and Stability Centre, University of Sussex, Brighton, UK
| | - Florian Robert
- Department of Medical Genetics, Box 238, Level 6, Addenbrooke's Treatment Centre, Cambridge Biomedical Research Campus, Cambridge CB2 0QQ, UK; Early Cancer Institute, Department of Oncology, Box 197, Hutchison Research Centre, Cambridge Biomedical Research Campus, Cambridge CB2 0XZ, UK
| | - Salome Zhao
- Department of Medical Genetics, Box 238, Level 6, Addenbrooke's Treatment Centre, Cambridge Biomedical Research Campus, Cambridge CB2 0QQ, UK; Early Cancer Institute, Department of Oncology, Box 197, Hutchison Research Centre, Cambridge Biomedical Research Campus, Cambridge CB2 0XZ, UK
| | - Andrea Degasperi
- Early Cancer Institute, Department of Oncology, Box 197, Hutchison Research Centre, Cambridge Biomedical Research Campus, Cambridge CB2 0XZ, UK
| | - Yogesh Kumar
- Early Cancer Institute, Department of Oncology, Box 197, Hutchison Research Centre, Cambridge Biomedical Research Campus, Cambridge CB2 0XZ, UK
| | - Helen Davies
- Department of Medical Genetics, Box 238, Level 6, Addenbrooke's Treatment Centre, Cambridge Biomedical Research Campus, Cambridge CB2 0QQ, UK; Early Cancer Institute, Department of Oncology, Box 197, Hutchison Research Centre, Cambridge Biomedical Research Campus, Cambridge CB2 0XZ, UK
| | - Rebecca Harris
- Department of Medical Genetics, Box 238, Level 6, Addenbrooke's Treatment Centre, Cambridge Biomedical Research Campus, Cambridge CB2 0QQ, UK
| | - Christian Frezza
- Medical Research Council Cancer Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XZ, UK; CECAD Research Center, Faculty of Medicine, University Hospital Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Chryssostomos Chatgilialoglu
- Istituto per la Sintesi Organica e la Fotoreattività, Consiglio Nazionale delle Ricerche, Via P. Gobetti 101, 40129 Bologna, Italy; Center for Advanced Technologies, Adam Mickiewicz University, 61-614 Poznan, Poland
| | - Robert Sarkany
- National Xeroderma Pigmentosum Service, St John's Institute of Dermatology, Guy's and St Thomas' Foundation Trust, London SE1 7EH, UK
| | - Alan Lehmann
- Genome Damage and Stability Centre, University of Sussex, Brighton, UK
| | - Chris Bakal
- Dynamical Cell Systems Laboratory, Division of Cancer Biology, Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK
| | - Jyoti Choudhary
- Functional Proteomics Group, Institute of Cancer Research, Chester Betty Labs, 237 Fulham Road, London SW3 6JB, UK
| | - Hiva Fassihi
- National Xeroderma Pigmentosum Service, St John's Institute of Dermatology, Guy's and St Thomas' Foundation Trust, London SE1 7EH, UK
| | - Serena Nik-Zainal
- Department of Medical Genetics, Box 238, Level 6, Addenbrooke's Treatment Centre, Cambridge Biomedical Research Campus, Cambridge CB2 0QQ, UK; Early Cancer Institute, Department of Oncology, Box 197, Hutchison Research Centre, Cambridge Biomedical Research Campus, Cambridge CB2 0XZ, UK.
| |
Collapse
|
43
|
Pepe F, Guerini-Rocco E, Fassan M, Fusco N, Vacirca D, Ranghiero A, Venetis K, Rappa A, Taormina SV, Russo G, Rebellato E, Munari G, Moreno-Manuel A, De Angelis C, Zamagni C, Valabrega G, Malapelle U, Troncone G, Barberis M, Iaccarino A. In-house homologous recombination deficiency testing in ovarian cancer: a multi-institutional Italian pilot study. J Clin Pathol 2024; 77:478-485. [PMID: 37072171 PMCID: PMC11228191 DOI: 10.1136/jcp-2023-208852] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 03/29/2023] [Indexed: 04/20/2023]
Abstract
AIMS Poly (ADP-ribose) polymerase (PARP) inhibitors (PARPIs) represent a standard of care for the clinical management of high-grade serous ovarian cancer (HGSOC). The recognition of homologous recombination deficiency (HRD) has emerged as a predictive biomarker of response for first-line PARPIs treatment in patients with HGOSC. On the other hand, this test is extremely complex and therefore it is often externalised. Regrettably, the reliability of outsourced HRD testing can be troubled by inconclusive results and high rejection rates. In this methodological study, we assessed the technical feasibility, interassay and interlaboratory reproducibility of in-house HRD testing using three different commercially available next-generation sequencing assays. METHODS A total of n=20 epithelial ovarian cancer samples previously analysed with MyChoice CDx were subjected to HRD retesting using three different platforms in three different major pathology laboratories, that is, SOPHiA DDM HRD Solution, HRD focus and Oncomine homologous recombination repair pathway predesigned panel. Concordance was calculated by Cohen's (dual) and Fleiss (triple) κ coefficients. RESULTS In-house BRCA1/2 molecular testing yielded a concordance rate >90.0% among all participating centres. HRD scores were successfully calculated by each institution with a concordance rate of 76.5%. Concerning the external gold standard test, the overall percentage of agreement ranged from 80.0% to 90.0% with a positive percentage agreement ranging from 75.0% to 80.0% and a negative percentage agreement ranging from 80.0% to 100%. CONCLUSIONS In-house testing for HRD can be reliably performed with commercially available next-generation sequencing assays.
Collapse
Affiliation(s)
- Francesco Pepe
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131, Naples, Italy
| | - Elena Guerini-Rocco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122, Milan, Italy
| | - Matteo Fassan
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, Padova, Italy
- Istituto Oncologico Veneto IRCCS, Padova, Italy
| | - Nicola Fusco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122, Milan, Italy
| | - Davide Vacirca
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141, Milan, Italy
| | - Alberto Ranghiero
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141, Milan, Italy
| | - Konstantinos Venetis
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141, Milan, Italy
| | - Alessandra Rappa
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141, Milan, Italy
| | - Sergio Vincenzo Taormina
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141, Milan, Italy
| | - Gianluca Russo
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131, Naples, Italy
| | - Elena Rebellato
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, Padova, Italy
| | | | - Andrea Moreno-Manuel
- Molecular Oncology Laboratory, Fundación Investigación Hospital General Universitario de Valencia, 46014, Valencia, Spain
- TRIAL Mixed Unit, Centro Investigación Príncipe Felipe-Fundación Investigación Hospital General Universitario de Valencia, Valencia, Spain
- Centro de Investigación Biomédica en Red Cáncer, CIBERONC, Madrid, Spain
| | - Carmine De Angelis
- Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy
| | - Claudio Zamagni
- Unit of Oncology, IRCCS Azienda Ospedaliero, Universitaria di Bologna, Bologna, Italy
| | - Giorgio Valabrega
- Department of Oncology, University of Torino at Ordine Mauriziano Hospital, Turin, Italy
| | - Umberto Malapelle
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131, Naples, Italy
| | - Giancarlo Troncone
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131, Naples, Italy
| | - Massimo Barberis
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Via Giuseppe Ripamonti 435, 20141, Milan, Italy
| | - Antonino Iaccarino
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131, Naples, Italy
| |
Collapse
|
44
|
Fernandes I, Chehade R, MacKay H. PARP inhibitors in non-ovarian gynecologic cancers. Ther Adv Med Oncol 2024; 16:17588359241255174. [PMID: 38882441 PMCID: PMC11179472 DOI: 10.1177/17588359241255174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 04/25/2024] [Indexed: 06/18/2024] Open
Abstract
Poly(ADP-ribose) polymerase (PARP) inhibitors (PARPis) have transformed the treatment of ovarian cancer, particularly benefiting patients whose tumors harbor genomic events that result in impaired homologous recombination (HR) repair. The use of PARPi over recent years has expanded to include subpopulations of patients with breast, pancreatic, and prostate cancers. Their potential to benefit patients with non-ovarian gynecologic cancers is being recognized. This review examines the underlying biological rationale for exploring PARPi in non-ovarian gynecologic cancers. We consider the clinical data and place this in the context of the current treatment landscape. We review the development of PARPi strategies for treating patients with endometrial, cervical, uterine leiomyosarcoma, and vulvar cancers. Furthermore, we discuss future directions and the importance of understanding HR deficiency in the context of each cancer type.
Collapse
Affiliation(s)
| | - Rania Chehade
- Sunnybrook Odette Cancer Centre, Toronto, ON, Canada
| | - Helen MacKay
- Sunnybrook Odette Cancer Centre, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON M4N 3M5, Canada
| |
Collapse
|
45
|
Zhang H, Jiang L, Du X, Qian Z, Wu G, Jiang Y, Mao Z. The cGAS-Ku80 complex regulates the balance between two end joining subpathways. Cell Death Differ 2024; 31:792-803. [PMID: 38664591 PMCID: PMC11164703 DOI: 10.1038/s41418-024-01296-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 06/12/2024] Open
Abstract
As the major DNA sensor that activates the STING-TBK1 signaling cascade, cGAS is mainly present in the cytosol. A number of recent reports have indicated that cGAS also plays critical roles in the nucleus. Our previous work demonstrated for the first time that cGAS is translocated to the nucleus upon the occurrence of DNA damage and inhibits homologous recombination (HR), one of the two major pathways of DNA double strand break (DSB) repair. However, whether nuclear cGAS regulates the other DSB repair pathway, nonhomologous end joining (NHEJ), which can be further divided into the less error-prone canonical NHEJ (c-NHEJ) and more mutagenic alternative NHEJ (alt-NHEJ) subpathways, has not been characterized. Here, we demonstrated that cGAS tipped the balance of the two NHEJ subpathways toward c-NHEJ. Mechanistically, the cGAS-Ku80 complex enhanced the interaction between DNA-PKcs and the deubiquitinase USP7 to improve DNA-PKcs protein stability, thereby promoting c-NHEJ. In contrast, the cGAS-Ku80 complex suppressed alt-NHEJ by directly binding to the promoter of Polθ to suppress its transcription. Together, these findings reveal a novel function of nuclear cGAS in regulating DSB repair, suggesting that the presence of cGAS in the nucleus is also important in the maintenance of genome integrity.
Collapse
Affiliation(s)
- Haiping Zhang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Lijun Jiang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xinyi Du
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhen Qian
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Guizhu Wu
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Ying Jiang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China.
| | - Zhiyong Mao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China.
| |
Collapse
|
46
|
Arat Çelik HE, Yılmaz S, Akşahin İC, Kök Kendirlioğlu B, Çörekli E, Dal Bekar NE, Çelik ÖF, Yorguner N, Targıtay Öztürk B, İşlekel H, Özerdem A, Akan P, Ceylan D, Tuna G. Oxidatively-induced DNA base damage and base excision repair abnormalities in siblings of individuals with bipolar disorder DNA damage and repair in bipolar disorder. Transl Psychiatry 2024; 14:207. [PMID: 38789433 PMCID: PMC11126633 DOI: 10.1038/s41398-024-02901-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 04/01/2024] [Accepted: 04/05/2024] [Indexed: 05/26/2024] Open
Abstract
Previous evidence suggests elevated levels of oxidatively-induced DNA damage, particularly 8-hydroxy-2'-deoxyguanosine (8-OH-dG), and abnormalities in the repair of 8-OH-dG by the base excision repair (BER) in bipolar disorder (BD). However, the genetic disposition of these abnormalities remains unknown. In this study, we aimed to investigate the levels of oxidatively-induced DNA damage and BER mechanisms in individuals with BD and their siblings, as compared to healthy controls (HCs). 46 individuals with BD, 41 siblings of individuals with BD, and 51 HCs were included in the study. Liquid chromatography-tandem mass spectrometry was employed to evaluate the levels of 8-OH-dG in urine, which were then normalized based on urine creatinine levels. The real-time-polymerase chain reaction was used to measure the expression levels of 8-oxoguanine DNA glycosylase 1 (OGG1), apurinic/apyrimidinic endonuclease 1 (APE1), poly ADP-ribose polymerase 1 (PARP1), and DNA polymerase beta (POLβ). The levels of 8-OH-dG were found to be elevated in both individuals with BD and their siblings when compared to the HCs. The OGG1 and APE1 expressions were downregulated, while POLβ expressions were upregulated in both the patient and sibling groups compared to the HCs. Age, smoking status, and the number of depressive episodes had an impact on APE1 expression levels in the patient group while body mass index, smoking status, and past psychiatric history had an impact on 8-OH-dG levels in siblings. Both individuals with BD and unaffected siblings presented similar abnormalities regarding oxidatively-induced DNA damage and BER, suggesting a link between abnormalities in DNA damage/BER mechanisms and familial susceptibility to BD. Our findings suggest that targeting the oxidatively-induced DNA damage and BER pathway could offer promising therapeutic strategies for reducing the risk of age-related diseases and comorbidities in individuals with a genetic predisposition to BD.
Collapse
Affiliation(s)
| | - Selda Yılmaz
- Department of Molecular Medicine, Institute of Health Sciences, Dokuz Eylul University, Izmir, Turkey
| | - İzel Cemre Akşahin
- Graduate School of Health Sciences, Koc University, Istanbul, Turkey
- Research Center for Translational Medicine (KUTTAM), School of Medicine, Koc University, Istanbul, Turkey
| | | | - Esma Çörekli
- Department of Psychiatry, School of Medicine, Maltepe University, Istanbul, Turkey
| | - Nazlı Ecem Dal Bekar
- Chair of Proteomics and Bioanalytics, School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Ömer Faruk Çelik
- Department of Medical Biochemistry, Sancaktepe Sehit Prof. Dr. Ilhan Varank Training and Research Hospital, Istanbul, Turkey
| | - Neşe Yorguner
- Department of Psychiatry, School of Medicine, Marmara University, Istanbul, Turkey
| | | | - Hüray İşlekel
- Department of Molecular Medicine, Institute of Health Sciences, Dokuz Eylul University, Izmir, Turkey
- Department of Medical Biochemistry, School of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Ayşegül Özerdem
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Pınar Akan
- Department of Neuroscience, Institute of Health Sciences, Dokuz Eylul University, Izmir, Turkey
- BioIzmir - Izmir Health Technologies Development and Accelerator Research and Application Center, Dokuz Eylul University, Izmir, Turkey
| | - Deniz Ceylan
- Research Center for Translational Medicine (KUTTAM), School of Medicine, Koc University, Istanbul, Turkey.
- Department of Psychiatry, School of Medicine, Koc University, Istanbul, Turkey.
| | - Gamze Tuna
- Department of Molecular Medicine, Institute of Health Sciences, Dokuz Eylul University, Izmir, Turkey
- BioIzmir - Izmir Health Technologies Development and Accelerator Research and Application Center, Dokuz Eylul University, Izmir, Turkey
| |
Collapse
|
47
|
González-Alvarez ME, Inyang I, Keating AF. Exposure to 7,12-dimethylbenz[a]anthracene impacts ovarian DNA damage sensing and repair proteins differently in lean and obese female mice and weight loss may mitigate obesity-induced ovarian dysfunction. Toxicol Appl Pharmacol 2024; 486:116930. [PMID: 38626870 DOI: 10.1016/j.taap.2024.116930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/28/2024] [Accepted: 04/10/2024] [Indexed: 05/21/2024]
Abstract
Obesity impairs oocyte quality, fertility, pregnancy maintenance, and is associated with offspring birth defects. The model ovotoxicant, 7,12-dimethylbenz[a]anthracene (DMBA), causes ovarian DNA damage and follicle loss. Both DMBA-induced chemical biotransformation and the DNA damage response are partially attenuated in obese relative to lean female mice but whether weight loss could improve the DNA damage response to DMBA exposure has not been explored. Thus, at six weeks of age, C57BL/6 J female mice were divided in three groups: 1) Lean (L; n = 20) fed a chow diet for 12 weeks, 2) obese (O; n = 20) fed a high fat high sugar (HFHS) diet for 12 weeks and, 3) slim-down (S; n = 20). The S group was fed with HFHS diet for 7 weeks until attaining a higher body relative to L mice on week 7.5 and switched to a chow diet for 5 weeks to achieve weight loss. Mice then received either corn oil (CT) or DMBA (D; 1 mg/kg) for 7 d via intraperitoneal injection (n = 10/treatment). Obesity increased (P < 0.05) kidney and spleen weight, and DMBA decreased uterine weight (P < 0.05). Ovarian weight was reduced (P < 0.05) in S mice, but DMBA exposure increased ovary weight in the S mice. LC-MS/MS identified 18, 64, and 7 ovarian proteins as altered (P < 0.05) by DMBA in the L, S and O groups, respectively. In S and O mice, 24 and 8 proteins differed, respectively, from L mice. These findings support weight loss as a strategy to modulate the ovarian genotoxicant response.
Collapse
Affiliation(s)
| | - Imaobong Inyang
- Department of Animal Science, Iowa State University, United States of America
| | - Aileen F Keating
- Department of Animal Science, Iowa State University, United States of America.
| |
Collapse
|
48
|
Kim J, Fahmy V, Haffty BG. Radiation therapy for triple-negative breast cancer: from molecular insights to clinical perspectives. Expert Rev Anticancer Ther 2024; 24:211-217. [PMID: 38502143 DOI: 10.1080/14737140.2024.2333320] [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: 10/31/2023] [Accepted: 03/18/2024] [Indexed: 03/20/2024]
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) lacks three common receptors, making traditional treatments less effective. This review highlights the importance of radiotherapy and emerging therapeutic strategies to enhance treatment outcomes in TNBC. AREAS COVERED We conducted a literature search on PubMed for publications from 2000 to 2023 to discuss the critical role of radiotherapy in managing TNBC, emphasizing its applications from locoregional control to improving survival rates. The review explores molecular mechanisms underlying TNBC's radiotherapy response, including DNA damage repair and apoptosis, with a focus on BRCA1/2 mutations and Poly (ADP-ribose) polymerase (PARP) inhibition. We summarize preclinical and clinical research on radiosensitization strategies, from gene-targeted therapies to immunotherapy combinations, and the impact of post-mastectomy radiation therapy on locoregional control. The potential of personalized treatment approaches, integrating molecular profiling, targeted radiosensitizers, and the synergistic effects of radiotherapy with immunotherapy, is also discussed. EXPERT OPINION Future TNBC treatment strategies should focus on precision medicine, integrating immunotherapy, developing novel radiosensitizers, and targeting biological pathways to overcome radioresistance. The integration of radiomics and artificial intelligence offers promising avenues for enhancing treatment personalization and efficacy, aiming to improve patient outcomes in TNBC.
Collapse
Affiliation(s)
- Jongmyung Kim
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
- Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Veronia Fahmy
- New York Institute of Technology College of Osteopathic Medicine, Glen Head, NY, USA
| | - Bruce G Haffty
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
- Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| |
Collapse
|
49
|
Grasso L, Fonzino A, Manzari C, Leonardi T, Picardi E, Gissi C, Lazzaro F, Pesole G, Muzi-Falconi M. Detection of ribonucleotides embedded in DNA by Nanopore sequencing. Commun Biol 2024; 7:491. [PMID: 38654143 PMCID: PMC11039623 DOI: 10.1038/s42003-024-06077-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 03/20/2024] [Indexed: 04/25/2024] Open
Abstract
Ribonucleotides represent the most common non-canonical nucleotides found in eukaryotic genomes. The sources of chromosome-embedded ribonucleotides and the mechanisms by which unrepaired rNMPs trigger genome instability and human pathologies are not fully understood. The available sequencing technologies only allow to indirectly deduce the genomic location of rNMPs. Oxford Nanopore Technologies (ONT) may overcome such limitation, revealing the sites of rNMPs incorporation in genomic DNA directly from raw sequencing signals. We synthesized two types of DNA molecules containing rNMPs at known or random positions and we developed data analysis pipelines for DNA-embedded ribonucleotides detection by ONT. We report that ONT can identify all four ribonucleotides incorporated in DNA by capturing rNMPs-specific alterations in nucleotide alignment features, current intensity, and dwell time. We propose that ONT may be successfully employed to directly map rNMPs in genomic DNA and we suggest a strategy to build an ad hoc basecaller to analyse native genomes.
Collapse
Grants
- IG-21806 Associazione Italiana per la Ricerca sul Cancro (Italian Association for Cancer Research)
- PRIN2017_2022KJHC7S Ministero dell'Istruzione, dell'Università e della Ricerca (Ministry of Education, University and Research)
- PRIN_2022JA8JY5 Ministero dell'Istruzione, dell'Università e della Ricerca (Ministry of Education, University and Research)
- CN_00000041 Ministero dell'Istruzione, dell'Università e della Ricerca (Ministry of Education, University and Research)
- PRIN2017_2022KJHC7S Ministero dell'Istruzione, dell'Università e della Ricerca (Ministry of Education, University and Research)
- National Research Centers: “High Performance Computing, Big Data and Quantum Computing” (Project no. CN_00000013)
- National Research Centers: “High Performance Computing, Big Data and Quantum Computing” extended Partnerships: MNESYS (Project no. PE_0000006) and Age-It (Project no. PE_00000015). ELIXIR-IT through the empowering project ELIXIRNextGenIT (Grant Code IR0000010).
Collapse
Affiliation(s)
- Lavinia Grasso
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Celoria 26, 20133, Milano, Italy
| | - Adriano Fonzino
- Dipartimento di Bioscienze, Biotecnologie e Ambiente, Università di Bari A. Moro, Via Orabona 4, 70126, Bari, Italy
| | - Caterina Manzari
- Dipartimento di Bioscienze, Biotecnologie e Ambiente, Università di Bari A. Moro, Via Orabona 4, 70126, Bari, Italy
| | - Tommaso Leonardi
- Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia, Via Adamello 16, 20139, Milano, Italy
| | - Ernesto Picardi
- Dipartimento di Bioscienze, Biotecnologie e Ambiente, Università di Bari A. Moro, Via Orabona 4, 70126, Bari, Italy
- Istituto di Biomembrane, Bioenergetica e Biotecnologie Molecolari, Consiglio Nazionale delle Ricerche, Via Amendola 122/O, 70126, Bari, Italy
| | - Carmela Gissi
- Dipartimento di Bioscienze, Biotecnologie e Ambiente, Università di Bari A. Moro, Via Orabona 4, 70126, Bari, Italy
- Istituto di Biomembrane, Bioenergetica e Biotecnologie Molecolari, Consiglio Nazionale delle Ricerche, Via Amendola 122/O, 70126, Bari, Italy
| | - Federico Lazzaro
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Celoria 26, 20133, Milano, Italy.
| | - Graziano Pesole
- Dipartimento di Bioscienze, Biotecnologie e Ambiente, Università di Bari A. Moro, Via Orabona 4, 70126, Bari, Italy.
- Istituto di Biomembrane, Bioenergetica e Biotecnologie Molecolari, Consiglio Nazionale delle Ricerche, Via Amendola 122/O, 70126, Bari, Italy.
| | - Marco Muzi-Falconi
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Celoria 26, 20133, Milano, Italy.
| |
Collapse
|
50
|
Arvanitaki ES, Goulielmaki E, Gkirtzimanaki K, Niotis G, Tsakani E, Nenedaki E, Rouska I, Kefalogianni M, Xydias D, Kalafatakis I, Psilodimitrakopoulos S, Karagogeos D, Schumacher B, Stratakis E, Garinis GA. Microglia-derived extracellular vesicles trigger age-related neurodegeneration upon DNA damage. Proc Natl Acad Sci U S A 2024; 121:e2317402121. [PMID: 38635632 PMCID: PMC11047102 DOI: 10.1073/pnas.2317402121] [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: 10/09/2023] [Accepted: 03/22/2024] [Indexed: 04/20/2024] Open
Abstract
DNA damage and neurodegenerative disorders are intimately linked but the underlying mechanism remains elusive. Here, we show that persistent DNA lesions in tissue-resident macrophages carrying an XPF-ERCC1 DNA repair defect trigger neuroinflammation and neuronal cell death in mice. We find that microglia accumulate dsDNAs and chromatin fragments in the cytosol, which are sensed thereby stimulating a viral-like immune response in Er1Cx/- and naturally aged murine brain. Cytosolic DNAs are packaged into extracellular vesicles (EVs) that are released from microglia and discharge their dsDNA cargo into IFN-responsive neurons triggering cell death. To remove cytosolic dsDNAs and prevent inflammation, we developed targeting EVs to deliver recombinant DNase I to Er1Cx/- brain microglia in vivo. We show that EV-mediated elimination of cytosolic dsDNAs is sufficient to prevent neuroinflammation, reduce neuronal apoptosis, and delay the onset of neurodegenerative symptoms in Er1Cx/- mice. Together, our findings unveil a causal mechanism leading to neuroinflammation and provide a rationalized therapeutic strategy against age-related neurodegeneration.
Collapse
Affiliation(s)
- Ermioni S. Arvanitaki
- Department of Biology, University of Crete, HeraklionGR71409, Crete, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, HeraklionGR70013, Crete, Greece
| | - Evi Goulielmaki
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, HeraklionGR70013, Crete, Greece
| | - Katerina Gkirtzimanaki
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, HeraklionGR70013, Crete, Greece
| | - George Niotis
- Department of Biology, University of Crete, HeraklionGR71409, Crete, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, HeraklionGR70013, Crete, Greece
| | - Edisona Tsakani
- Department of Biology, University of Crete, HeraklionGR71409, Crete, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, HeraklionGR70013, Crete, Greece
| | - Electra Nenedaki
- Department of Biology, University of Crete, HeraklionGR71409, Crete, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, HeraklionGR70013, Crete, Greece
| | - Iliana Rouska
- Department of Biology, University of Crete, HeraklionGR71409, Crete, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, HeraklionGR70013, Crete, Greece
| | - Mary Kefalogianni
- Department of Physics, University of Crete, HeraklionGR71003, Crete, Greece
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas, HeraklionGR71110, Crete, Greece
| | - Dionysios Xydias
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas, HeraklionGR71110, Crete, Greece
- Materials Science and Technology Department, University of Crete, HeraklionGR70013, Crete, Greece
| | - Ilias Kalafatakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, HeraklionGR70013, Crete, Greece
- Medical School, Division of Basic Sciences, University of Crete, HeraklionGR71003, Crete, Greece
| | - Sotiris Psilodimitrakopoulos
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas, HeraklionGR71110, Crete, Greece
| | - Domna Karagogeos
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, HeraklionGR70013, Crete, Greece
- Medical School, Division of Basic Sciences, University of Crete, HeraklionGR71003, Crete, Greece
| | - Björn Schumacher
- Institute for Genome Stability in Ageing and Disease, Medical Faculty, University and University Hospital of Cologne, Cologne50931, Germany
- Cologne Excellence Cluster for Cellular Stress Responses in Ageing-Associated Diseases (CECAD), Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne50931, Germany
| | - Emmanuel Stratakis
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas, HeraklionGR71110, Crete, Greece
| | - George A. Garinis
- Department of Biology, University of Crete, HeraklionGR71409, Crete, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, HeraklionGR70013, Crete, Greece
| |
Collapse
|