1
|
Elazab IM, El-Feky OA, Khedr EG, El-Ashmawy NE. Prostate cancer and the cell cycle: Focusing on the role of microRNAs. Gene 2024; 928:148785. [PMID: 39053658 DOI: 10.1016/j.gene.2024.148785] [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: 03/25/2024] [Revised: 07/12/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024]
Abstract
Prostate cancer is the most frequent solid tumor in terms of incidence and ranks second only to lung cancer in terms of cancer mortality among men. It has a considerably high mortality rate; around 375,000 deaths occurred worldwide in 2020. In 2024, the American Cancer Society estimated that the number of new prostate cancer cases will be around 299,010 cases, and the estimated deaths will be around 32,250 deaths only in the USA. Cell cycle dysregulation is inevitable in cancer etiology and is targeted by various therapies in cancer treatment. MicroRNAs (miRNAs) are small, endogenous, non-coding regulatory molecules involved in both normal and abnormal cellular events. One of the cellular processes regulated by miRNAs is the cell cycle. Although there are some exceptions, tumor suppressor miRNAs could potentially arrest the cell cycle by downregulating several molecular machineries involved in catalyzing the cell cycle progression. In contrast, oncogenic miRNAs (oncomirs) help the cell cycle to progress by targeting various regulatory proteins such as retinoblastoma (Rb) or cell cycle inhibitors such as p21 or p27, and hence may contribute to prostate cancer progression; however, this is not always the case. In this review, we emphasize how a dysregulated miRNA expression profile is linked to an abnormal cell cycle progression in prostate cancer, which subsequently paves the way to a new therapeutic option for prostate cancer.
Collapse
Affiliation(s)
- Ibrahim M Elazab
- Department of Biochemistry, Faculty of Pharmacy, Tanta University, Al-Geish Street, Tanta, El-Gharbia, 31527, Egypt.
| | - Ola A El-Feky
- Department of Biochemistry, Faculty of Pharmacy, Tanta University, Al-Geish Street, Tanta, El-Gharbia, 31527, Egypt.
| | - Eman G Khedr
- Department of Biochemistry, Faculty of Pharmacy, Tanta University, Al-Geish Street, Tanta, El-Gharbia, 31527, Egypt.
| | - Nahla E El-Ashmawy
- Department of Biochemistry, Faculty of Pharmacy, Tanta University, Al-Geish Street, Tanta, El-Gharbia, 31527, Egypt; Department of Pharmacology and Biochemistry, Faculty of Pharmacy, The British University in Egypt, BUE, Cairo, 11837, Egypt.
| |
Collapse
|
2
|
Zhao H, Richardson C, Marriott I, Yang IH, Yan S. APE1 is a master regulator of the ATR-/ATM-mediated DNA damage response. DNA Repair (Amst) 2024; 144:103776. [PMID: 39461278 DOI: 10.1016/j.dnarep.2024.103776] [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: 09/18/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024]
Abstract
To maintain genomic integrity, cells have evolved several conserved DNA damage response (DDR) pathways in response to DNA damage and stress conditions. Apurinic/apyrimidinic endonuclease 1 (APE1) exhibits AP endonuclease, 3'-5' exonuclease, 3'-phosphodiesterase, and 3'-exoribonuclease activities and plays critical roles in the DNA repair and redox regulation of transcription. However, it remains unclear whether and how APE1 is involved in DDR pathways. In this perspective, we first updated our knowledge of APE1's functional domains and its nuclease activities and their specific associated substrates. We then summarized the newly discovered roles and mechanisms of action of APE1 in the global and nucleolar ATR-mediated DDR pathway. While the ATM-mediated DDR is well known to be activated by DNA double-strand breaks and oxidative stress, here we provided new perspectives as to how ATM DDR signaling is activated by indirect single-strand breaks (SSBs) resulting from genotoxic stress and defined SSB structures, and discuss how ATM kinase is directly activated and regulated by its activator, APE1. Together, accumulating body of new evidence supports the notion that APE1 is a master regulator protein of the ATR- and ATM-mediated DDR pathways. These new findings of APE1 in DDR signaling provide previously uncharacterized but critical functions and regulations of APE1 in genome integrity.
Collapse
Affiliation(s)
- Haichao Zhao
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Christine Richardson
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC 28223, USA; Center for Biomedical Engineering and Science, University of North Carolina at Charlotte, Charlotte, NC 28223, USA; School of Data Science, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Ian Marriott
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - In Hong Yang
- Center for Biomedical Engineering and Science, University of North Carolina at Charlotte, Charlotte, NC 28223, USA; Department of Mechanical Engineering and Engineering Science, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | - Shan Yan
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC 28223, USA; Center for Biomedical Engineering and Science, University of North Carolina at Charlotte, Charlotte, NC 28223, USA; School of Data Science, University of North Carolina at Charlotte, Charlotte, NC 28223, USA.
| |
Collapse
|
3
|
Zhen T, Sun T, Xiong B, Liu H, Wang L, Chen Y, Sun H. New insight into targeting the DNA damage response in the treatment of glioblastoma. Chin J Nat Med 2024; 22:869-886. [PMID: 39428180 DOI: 10.1016/s1875-5364(24)60694-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Indexed: 10/22/2024]
Abstract
Glioblastoma (GBM) is the most common invasive malignant tumor in human brain tumors, representing the most severe grade of gliomas. Despite existing therapeutic approaches, patient prognosis remains dismal, necessitating the exploration of novel strategies to enhance treatment efficacy and extend survival. Due to the restrictive nature of the blood-brain barrier (BBB), small-molecule inhibitors are prioritized in the treatment of central nervous system tumors. Among these, DNA damage response (DDR) inhibitors have garnered significant attention due to their potent therapeutic potential across various malignancies. This review provides a detailed analysis of DDR pathways as therapeutic targets in GBM, summarizes recent advancements, therapeutic strategies, and ongoing clinical trials, and offers perspectives on future directions in this rapidly evolving field. The goal is to present a comprehensive outlook on the potential of DDR inhibitors in improving GBM management and outcomes.
Collapse
Affiliation(s)
- Tengfei Zhen
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Tianyu Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Baichen Xiong
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Hui Liu
- Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Lei Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Haopeng Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
4
|
Modafferi S, Esposito F, Tavella S, Gioia U, Francia S. Traffic light at DSB-transit regulation between gene transcription and DNA repair. FEBS Lett 2024. [PMID: 39333024 DOI: 10.1002/1873-3468.15024] [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: 02/27/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 09/29/2024]
Abstract
Transcription of actively expressed genes is dampened for kilobases around DNA lesions via chromatin modifications. This is believed to favour repair and prevent genome instability. Nonetheless, mounting evidence suggests that transcription may be induced by DNA breakage, resulting in the local de novo synthesis of non-coding RNAs (ncRNAs). Such transcripts have been proposed to play important functions in both DNA damage signalling and repair. Here, we review the recently identified mechanistic details of transcriptional silencing at damaged chromatin, highlighting how post-translational histone modifications can also be modulated by the local synthesis of DNA damage-induced ncRNAs. Finally, we envision that these entangled transcriptional events at DNA breakages can be targeted to modulate DNA repair, with potential implications for locus-specific therapeutic strategies.
Collapse
Affiliation(s)
- Stefania Modafferi
- Istituto di Genetica Molecolare "Luigi Luca Cavalli Sforza"- Consiglio Nazionale delle Ricerche, Pavia, Italy
- PhD Program in Biomolecular Sciences and Biotechnology (SBB), Istituto Universitario di Studi Superiori (IUSS), Pavia, Italy
| | - Francesca Esposito
- Istituto di Genetica Molecolare "Luigi Luca Cavalli Sforza"- Consiglio Nazionale delle Ricerche, Pavia, Italy
- PhD Program in Genetics, Molecular and Cellular Biology (GMCB), University of Pavia, Pavia, Italy
| | - Sara Tavella
- Istituto di Genetica Molecolare "Luigi Luca Cavalli Sforza"- Consiglio Nazionale delle Ricerche, Pavia, Italy
- IFOM-ETS - The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Ubaldo Gioia
- Istituto di Genetica Molecolare "Luigi Luca Cavalli Sforza"- Consiglio Nazionale delle Ricerche, Pavia, Italy
- IFOM-ETS - The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Sofia Francia
- Istituto di Genetica Molecolare "Luigi Luca Cavalli Sforza"- Consiglio Nazionale delle Ricerche, Pavia, Italy
- IFOM-ETS - The AIRC Institute of Molecular Oncology, Milan, Italy
| |
Collapse
|
5
|
Qian C, Li X, Zhang J, Wang Y. Small Molecular Inhibitors That Target ATM for Drug Discovery: Current Research and Potential Prospective. J Med Chem 2024; 67:14742-14767. [PMID: 39149790 DOI: 10.1021/acs.jmedchem.4c01064] [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: 08/17/2024]
Abstract
The protein kinase ataxia telangiectasia mutated (ATM) is a constituent of the phosphatidylinositol 3-kinase-related kinase (PIKK) family, exerting a pivotal influence on diverse cellular processes, notably the signaling of double-strand DNA breaks (DSB) and stress response. The dysregulation of ATM is implicated in the pathogenesis of cancer and other diseases such as neurodegeneration. Hence, ATM is deemed a promising candidate for potential therapeutic interventions across a spectrum of diseases. Presently, while ATM small molecule inhibitors are not commercially available, various selective inhibitors have progressed to the clinical research phase. Specifically, AZD1390, WSD0628, SYH2051, and ZN-B-2262 are under investigation in clinical studies pertaining to glioblastoma multiforme and advanced solid tumors, respectively. In this Perspective, we encapsulate the structure, biological functions, and disease relevance of ATM. Subsequently, we concentrate on the design concepts and structure-activity relationships (SAR) of ATM inhibitors, delineating potential avenues for the development of more efficacious ATM-targeted inhibitors.
Collapse
Affiliation(s)
- Chunlin Qian
- Department of Respiratory and Critical Care Medicine and Targeted Tracer Research and Development Laboratory and Institute of Respiratory Health and State Key Laboratory of Respiratory Health and Multimorbidity and Laboratory of Neuro-system and Multimorbidity, West China Hospital, Sichuan University, Chengdu 610041, Sichuan China
| | - Xiaoxue Li
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan China
| | - Jifa Zhang
- Department of Respiratory and Critical Care Medicine and Targeted Tracer Research and Development Laboratory and Institute of Respiratory Health and State Key Laboratory of Respiratory Health and Multimorbidity and Laboratory of Neuro-system and Multimorbidity, West China Hospital, Sichuan University, Chengdu 610041, Sichuan China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, Sichuan China
| | - Yuxi Wang
- Department of Respiratory and Critical Care Medicine and Targeted Tracer Research and Development Laboratory and Institute of Respiratory Health and State Key Laboratory of Respiratory Health and Multimorbidity and Laboratory of Neuro-system and Multimorbidity, West China Hospital, Sichuan University, Chengdu 610041, Sichuan China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, Sichuan China
| |
Collapse
|
6
|
Bishop A, Romero JC, Tonapi S, Parihar M, Loranc E, Miller H, Lawrence L, Bassani N, Robledo D, Cao L, Nie J, Kanda K, Stoja A, Garcia N, Gorthi A, Stoveken B, Lane A, Fan T, Cassel T, Zha S, Musi N. ATM phosphorylation of CD98HC increases antiporter membrane localization and prevents chronic toxic glutamate accumulation in Ataxia telangiectasia. RESEARCH SQUARE 2024:rs.3.rs-4947457. [PMID: 39281865 PMCID: PMC11398575 DOI: 10.21203/rs.3.rs-4947457/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Ataxia telangiectasia (A-T) is a rare genetic disorder characterized by neurological defects, immunodeficiency, cancer predisposition, radiosensitivity, decreased blood vessel integrity, and diabetes. ATM, the protein mutated in A-T, responds to DNA damage and oxidative stress, but its functional relationship to the progressive clinical manifestation of A-T is not understood. CD98HC chaperones cystine/glutamate (x c - ) and cationic/neutral amino acid (y + L) antiporters to the cell membrane, and CD98HC phosphorylation by ATM accelerates membrane localization to acutely increase amino acid transport. Loss of ATM impacts tissues reliant on SLC family antiporters relevant to A-T phenotypes, such as endothelial cells (telangiectasia) and pancreatic α-cells (fatty liver and diabetes) with toxic glutamate accumulation. Bypassing the antiporters restores intracellular metabolic balance both in ATM-deficient cells and mouse models. These findings provide new insight into the long-known benefits of N-acetyl cysteine to A-T cells beyond oxidative stress through removing excess glutamate by production of glutathione.
Collapse
|
7
|
Burlet D, Huber AL, Tissier A, Petrilli V. Crosstalk between inflammasome sensors and DNA damage response pathways. FEBS J 2024; 291:3978-3988. [PMID: 38273453 DOI: 10.1111/febs.17060] [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: 09/05/2023] [Revised: 12/04/2023] [Accepted: 01/10/2024] [Indexed: 01/27/2024]
Abstract
Eukaryotic cells encounter diverse threats jeopardizing their integrity, prompting the development of defense mechanisms against these stressors. Among these mechanisms, inflammasomes are well-known for their roles in coordinating the inflammatory response against infections. Extensive research has unveiled their multifaceted involvement in cellular processes beyond inflammation. Recent studies emphasize the intricate relationship between the inflammasome and the DNA damage response (DDR). They highlight how the DDR participates in inflammasome activation and the reciprocal impact of inflammasome on DDR and genome integrity preservation. Moreover, novel functions of inflammasome sensors in DDR pathways have emerged, broadening our understanding of their roles. Finally, this review delves into identifying common signals that drive the activation of inflammasome sensors alongside activation cues for the DNA damage response, offering potential insights into shared regulatory pathways between these critical cellular processes.
Collapse
Affiliation(s)
- Delphine Burlet
- INSERM U1052, Centre de Recherche en Cancérologie de Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, France
- Université de Lyon, Université Lyon 1, France
- Centre Léon Bérard, Lyon, France
| | - Anne-Laure Huber
- INSERM U1052, Centre de Recherche en Cancérologie de Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, France
- Université de Lyon, Université Lyon 1, France
- Centre Léon Bérard, Lyon, France
| | - Agnès Tissier
- INSERM U1052, Centre de Recherche en Cancérologie de Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, France
- Université de Lyon, Université Lyon 1, France
- Centre Léon Bérard, Lyon, France
| | - Virginie Petrilli
- INSERM U1052, Centre de Recherche en Cancérologie de Lyon, France
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, France
- Université de Lyon, Université Lyon 1, France
- Centre Léon Bérard, Lyon, France
| |
Collapse
|
8
|
Fu H, Zhong J, Zhao J, Huo L, Wang C, Ma D, Pan W, Sun L, Ren Z, Fan T, Wang Z, Wang W, Lei X, Yu G, Li J, Zhu Y, Geelen D, Liu B. Ultraviolet attenuates centromere-mediated meiotic genome stability and alters gametophytic ploidy consistency in flowering plants. THE NEW PHYTOLOGIST 2024; 243:2214-2234. [PMID: 39039772 DOI: 10.1111/nph.19978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 06/29/2024] [Indexed: 07/24/2024]
Abstract
Ultraviolet (UV) radiation influences development and genome stability in organisms; however, its impact on meiosis, a special cell division essential for the delivery of genetic information across generations in eukaryotes, has not yet been elucidated. In this study, by performing cytogenetic studies, we reported that UV radiation does not damage meiotic chromosome integrity but attenuates centromere-mediated chromosome stability and induces unreduced gametes in Arabidopsis thaliana. We showed that functional centromere-specific histone 3 (CENH3) is required for obligate crossover formation and plays a role in the protection of sister chromatid cohesion under UV stress. Moreover, we found that UV specifically alters the orientation and organization of spindles and phragmoplasts at meiosis II, resulting in meiotic restitution and unreduced gametes. We determined that UV-induced meiotic restitution does not rely on the UV Resistance Locus8-mediated UV perception and the Tapetal Development and Function1- and Aborted Microspores-dependent tapetum development, but possibly occurs via altered JASON function and downregulated Parallel Spindle1. This study provides evidence that UV radiation influences meiotic genome stability and gametophytic ploidy consistency in flowering plants.
Collapse
Affiliation(s)
- Huiqi Fu
- College of Life Sciences, South-Central Minzu University, Wuhan, 430074, China
| | - Jiaqi Zhong
- College of Life Sciences, South-Central Minzu University, Wuhan, 430074, China
| | - Jiayi Zhao
- College of Life Sciences, South-Central Minzu University, Wuhan, 430074, China
| | - Li Huo
- College of Life Sciences, South-Central Minzu University, Wuhan, 430074, China
| | - Chong Wang
- Shanghai Key Laboratory of Plant Molecular Sciences, Development Center of Plant Germplasm Resources, College of Life Sciences, Shanghai Normal University, Shanghai, 200234, China
| | - Dexuan Ma
- Shanghai Key Laboratory of Plant Molecular Sciences, Development Center of Plant Germplasm Resources, College of Life Sciences, Shanghai Normal University, Shanghai, 200234, China
| | - Wenjing Pan
- College of Life Sciences, South-Central Minzu University, Wuhan, 430074, China
| | - Limin Sun
- Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, Ghent, 9000, Belgium
| | - Ziming Ren
- Department of Landscape Architecture, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Tianyi Fan
- Department of Biochemistry, Institute of Plant Biology, School of Life Sciences, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, 200438, China
| | - Ze Wang
- College of Tropical Crops, Hainan University, Haikou, 570228, China
| | - Wenyi Wang
- College of Life Sciences, South-Central Minzu University, Wuhan, 430074, China
| | - Xiaoning Lei
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Guanghui Yu
- College of Life Sciences, South-Central Minzu University, Wuhan, 430074, China
| | - Jing Li
- College of Tropical Crops, Hainan University, Haikou, 570228, China
| | - Yan Zhu
- Department of Biochemistry, Institute of Plant Biology, School of Life Sciences, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, 200438, China
| | - Danny Geelen
- Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, Ghent, 9000, Belgium
| | - Bing Liu
- College of Life Sciences, South-Central Minzu University, Wuhan, 430074, China
| |
Collapse
|
9
|
Lim B, Matsui Y, Jung S, Djekidel MN, Qi W, Yuan ZF, Wang X, Yang X, Connolly N, Pilehroud AS, Pan H, Wang F, Pruett-Miller SM, Kavdia K, Pagala V, Fan Y, Peng J, Xu B, Peng JC. Phosphorylation of the DNA damage repair factor 53BP1 by ATM kinase controls neurodevelopmental programs in cortical brain organoids. PLoS Biol 2024; 22:e3002760. [PMID: 39226322 PMCID: PMC11398655 DOI: 10.1371/journal.pbio.3002760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 09/13/2024] [Accepted: 07/19/2024] [Indexed: 09/05/2024] Open
Abstract
53BP1 is a well-established DNA damage repair factor that has recently emerged to critically regulate gene expression for tumor suppression and neural development. However, its precise function and regulatory mechanisms remain unclear. Here, we showed that phosphorylation of 53BP1 at serine 25 by ATM is required for neural progenitor cell proliferation and neuronal differentiation in cortical brain organoids. Dynamic phosphorylation of 53BP1-serine 25 controls 53BP1 target genes governing neuronal differentiation and function, cellular response to stress, and apoptosis. Mechanistically, ATM and RNF168 govern 53BP1's binding to gene loci to directly affect gene regulation, especially at genes for neuronal differentiation and maturation. 53BP1 serine 25 phosphorylation effectively impedes its binding to bivalent or H3K27me3-occupied promoters, especially at genes regulating H3K4 methylation, neuronal functions, and cell proliferation. Beyond 53BP1, ATM-dependent phosphorylation displays wide-ranging effects, regulating factors in neuronal differentiation, cytoskeleton, p53 regulation, as well as key signaling pathways such as ATM, BDNF, and WNT during cortical organoid differentiation. Together, our data suggest that the interplay between 53BP1 and ATM orchestrates essential genetic programs for cell morphogenesis, tissue organization, and developmental pathways crucial for human cortical development.
Collapse
Affiliation(s)
- Bitna Lim
- Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Yurika Matsui
- Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Seunghyun Jung
- Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Mohamed Nadhir Djekidel
- Center for Applied Bioinformatics, St Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Wenjie Qi
- Center for Applied Bioinformatics, St Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Zuo-Fei Yuan
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Xusheng Wang
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Xiaoyang Yang
- Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Nina Connolly
- Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Abbas Shirinifard Pilehroud
- Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Haitao Pan
- Department of Biostatistics, St Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Fang Wang
- Department of Biostatistics, St Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Shondra M Pruett-Miller
- Department of Cell & Molecular Biology, St Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Kanisha Kavdia
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Vishwajeeth Pagala
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Yiping Fan
- Center for Applied Bioinformatics, St Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Junmin Peng
- Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, Tennessee, United States of America
- Department of Structural Biology, St Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Beisi Xu
- Center for Applied Bioinformatics, St Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Jamy C Peng
- Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| |
Collapse
|
10
|
Lee JH. Oxidative stress and the multifaceted roles of ATM in maintaining cellular redox homeostasis. Redox Biol 2024; 75:103269. [PMID: 39018798 PMCID: PMC11301354 DOI: 10.1016/j.redox.2024.103269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/12/2024] [Accepted: 07/13/2024] [Indexed: 07/19/2024] Open
Abstract
The ataxia telangiectasia mutated (ATM) protein kinase is best known as a master regulator of the DNA damage response. However, accumulating evidence has unveiled an equally vital function for ATM in sensing oxidative stress and orchestrating cellular antioxidant defenses to maintain redox homeostasis. ATM can be activated through a non-canonical pathway involving intermolecular disulfide crosslinking of the kinase dimers, distinct from its canonical activation by DNA double-strand breaks. Structural studies have elucidated the conformational changes that allow ATM to switch into an active redox-sensing state upon oxidation. Notably, loss of ATM function results in elevated reactive oxygen species (ROS) levels, altered antioxidant profiles, and mitochondrial dysfunction across multiple cell types and tissues. This oxidative stress arising from ATM deficiency has been implicated as a central driver of the neurodegenerative phenotypes in ataxia-telangiectasia (A-T) patients, potentially through mechanisms involving oxidative DNA damage, PARP hyperactivation, and widespread protein aggregation. Moreover, defective ATM oxidation sensing disrupts transcriptional programs and RNA metabolism, with detrimental impacts on neuronal homeostasis. Significantly, antioxidant therapy can ameliorate cellular and organismal abnormalities in various ATM-deficient models. This review synthesizes recent advances illuminating the multifaceted roles of ATM in preserving redox balance and mitigating oxidative insults, providing a unifying paradigm for understanding the complex pathogenesis of A-T disease.
Collapse
Affiliation(s)
- Ji-Hoon Lee
- Department of Biological Sciences, Research Center of Ecomimetics, Chonnam National University, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
11
|
Lee JH. Targeting the ATM pathway in cancer: Opportunities, challenges and personalized therapeutic strategies. Cancer Treat Rev 2024; 129:102808. [PMID: 39106770 DOI: 10.1016/j.ctrv.2024.102808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/09/2024]
Abstract
Ataxia telangiectasia mutated (ATM) kinase plays a pivotal role in orchestrating the DNA damage response, maintaining genomic stability, and regulating various cellular processes. This review provides a comprehensive analysis of ATM's structure, activation mechanisms, and various functions in cancer development, progression, and treatment. I discuss ATM's dual nature as both a tumor suppressor and potential promoter of cancer cell survival in certain contexts. The article explores the complex signaling pathways mediated by ATM, its interactions with other DNA repair mechanisms, and its influence on cell cycle checkpoints, apoptosis, and metabolism. I examine the clinical implications of ATM alterations, including their impact on cancer predisposition, prognosis, and treatment response. The review highlights recent advances in ATM-targeted therapies, discussing ongoing clinical trials of ATM inhibitors and their potential in combination with other treatment modalities. I also address the challenges in developing effective biomarkers for ATM activity and patient selection strategies for personalized cancer therapy. Finally, I outline future research directions, emphasizing the need for refined biomarker development, optimized combination therapies, and strategies to overcome potential resistance mechanisms. This comprehensive overview underscores the critical importance of ATM in cancer biology and its emerging potential as a therapeutic target in precision oncology.
Collapse
Affiliation(s)
- Ji-Hoon Lee
- Department of Biological Sciences, Research Center of Ecomimetics, Chonnam National University, Gwangju 61186, Republic of Korea.
| |
Collapse
|
12
|
Tamai T, Reginato G, Ojiri R, Morita I, Avrutis A, Cejka P, Shinohara M, Sugimoto K. Sae2 controls Mre11 endo- and exonuclease activities by different mechanisms. Nat Commun 2024; 15:7221. [PMID: 39174552 PMCID: PMC11341764 DOI: 10.1038/s41467-024-51493-5] [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: 10/12/2023] [Accepted: 08/09/2024] [Indexed: 08/24/2024] Open
Abstract
DNA double-strand breaks (DSBs) must be repaired to ensure cell survival and genomic integrity. In yeast, the Mre11-Rad50-Xrs2 complex (MRX) collaborates with Sae2 to initiate DSB repair. Sae2 stimulates two MRX nuclease activities, endonuclease and 3'-5' exonuclease. However, how Sae2 controls the two nuclease activities remains enigmatic. Using a combined genetic and biochemical approach, we identified a separation-of-function rad50 mutation, rad50-C47, that causes a defect in Sae2-dependent MRX 3'-5' exonuclease activity, but not endonuclease activity. We found that both the endo- and 3'-5' exonuclease activities are essential to release Spo11 from DNA ends, whereas only the endonuclease activity is required for hairpin removal. We also uncovered that MRX-Sae2 endonuclease introduces a cleavage at defined distances from the Spo11-blocked end with gradually decreasing efficiency. Our findings demonstrate that Sae2 stimulates the MRX endo- and exonuclease activities via Rad50 by different mechanisms, ensuring diverse actions of MRX-Sae2 nuclease at DNA ends.
Collapse
Affiliation(s)
- Tomoki Tamai
- Faculty of Advanced Bioscience, Graduate School of Agriculture, Kindai University, Nara City, Nara, 631-8505, Japan
| | - Giordano Reginato
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana (USI), 6500, Bellinzona, Switzerland
| | - Ryusei Ojiri
- Faculty of Advanced Bioscience, Graduate School of Agriculture, Kindai University, Nara City, Nara, 631-8505, Japan
| | - Issei Morita
- Faculty of Advanced Bioscience, Graduate School of Agriculture, Kindai University, Nara City, Nara, 631-8505, Japan
| | - Alexandra Avrutis
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers, the State University of New Jersey, Newark, NJ, 07103, USA
| | - Petr Cejka
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana (USI), 6500, Bellinzona, Switzerland.
| | - Miki Shinohara
- Faculty of Advanced Bioscience, Graduate School of Agriculture, Kindai University, Nara City, Nara, 631-8505, Japan.
- Agricultural Technology and Innovation Research Institute, Kindai University, Nara City, Nara, 631-8505, Japan.
| | - Katsunori Sugimoto
- Department of Microbiology, Biochemistry and Molecular Genetics, New Jersey Medical School, Rutgers, the State University of New Jersey, Newark, NJ, 07103, USA.
| |
Collapse
|
13
|
Zhao H, Li J, You Z, Lindsay HD, Yan S. Distinct regulation of ATM signaling by DNA single-strand breaks and APE1. Nat Commun 2024; 15:6517. [PMID: 39112456 PMCID: PMC11306256 DOI: 10.1038/s41467-024-50836-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 07/21/2024] [Indexed: 08/10/2024] Open
Abstract
In response to DNA double-strand breaks or oxidative stress, ATM-dependent DNA damage response (DDR) is activated to maintain genome integrity. However, it remains elusive whether and how DNA single-strand breaks (SSBs) activate ATM. Here, we provide direct evidence in Xenopus egg extracts that ATM-mediated DDR is activated by a defined SSB structure. Our mechanistic studies reveal that APE1 promotes the SSB-induced ATM DDR through APE1 exonuclease activity and ATM recruitment to SSB sites. APE1 protein can form oligomers to activate the ATM DDR in Xenopus egg extracts in the absence of DNA and can directly stimulate ATM kinase activity in vitro. Our findings reveal distinct mechanisms of the ATM-dependent DDR activation by SSBs in eukaryotic systems and identify APE1 as a direct activator of ATM kinase.
Collapse
Affiliation(s)
- Haichao Zhao
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, 28223, USA
| | - Jia Li
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, 28223, USA
| | - Zhongsheng You
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Howard D Lindsay
- Lancaster Medical School, Faculty of Health and Medicine, Lancaster University, Lancaster, LA1 4YQ, UK
| | - Shan Yan
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC, 28223, USA.
- School of Data Science, University of North Carolina at Charlotte, Charlotte, NC, 28223, USA.
- Center for Biomedical Engineering and Science, University of North Carolina at Charlotte, Charlotte, USA.
| |
Collapse
|
14
|
Martin OA, Sykes PJ, Lavin M, Engels E, Martin RF. What's Changed in 75 Years of RadRes? - An Australian Perspective on Selected Topics. Radiat Res 2024; 202:309-327. [PMID: 38966925 DOI: 10.1667/rade-24-00037.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/09/2024] [Indexed: 07/06/2024]
Abstract
Several scientific themes are reviewed in the context of the 75-year period relevant to this special platinum issue of Radiation Research. Two criteria have been considered in selecting the scientific themes. One is the exposure of the associated research activity in the annual meetings of the Radiation Research Society (RRS) and in the publications of the Society's Journal, thus reflecting the interest of members of RRS. The second criteria is a focus on contributions from Australian members of RRS. The first theme is the contribution of radiobiology to radiation oncology, featuring two prominent Australian radiation oncologists, the late Rod Withers and his younger colleague, Lester Peters. Two other themes are also linked to radiation oncology; preclinical research aimed at developing experimental radiotherapy modalities, namely microbeam radiotherapy (MRT) and Auger endoradiotherapy. The latter has a long history, in contrast to MRT, especially in Australia, given that the associated medical beamline at the Australian Synchrotron in Melbourne only opened in 2011. Another theme is DNA repair, which has a trajectory parallel to the 75-year period of interest, given the birth of molecular biology in the 1950s. The low-dose radiobiology theme has a similar timeline, predominantly prompted by the nuclear era, which is also connected to the radioprotector theme, although radioprotectors also have a long-established potential utility in cancer radiotherapy. Finally, two themes are associated with biodosimetry. One is the micronucleus assay, highlighting the pioneering contribution from Michael Fenech in Adelaide, South Australia, and the other is the γ-H2AX assay and its widespread clinical applications.
Collapse
Affiliation(s)
- Olga A Martin
- Centre of Medical Radiation Physics (CMRP), University of Wollongong, Wollongong, NSW, Australia
| | - Pamela J Sykes
- College of Medicine and Public Health, Flinders University and Medical Centre, Bedford Park, SA, Australia
| | - Martin Lavin
- Centre for Clinical Research, University of Queensland, QSL, Brisbane, Australia
| | - Elette Engels
- Centre of Medical Radiation Physics (CMRP), University of Wollongong, Wollongong, NSW, Australia
- Australian Synchrotron, Australian Nuclear Science and Technology Organisation (ANSTO), Clayton, VIC, Australia
| | - Roger F Martin
- School of Chemistry, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
15
|
Chow Z, Johnson J, Chauhan A, Jeong JC, Castle JT, Izumi T, Weiss H, Townsend CM, Schrader J, Anthony L, Yang ES, Evers BM, Rychahou P. Inhibition of ribonucleotide reductase subunit M2 enhances the radiosensitivity of metastatic pancreatic neuroendocrine tumor. Cancer Lett 2024; 596:216993. [PMID: 38801884 PMCID: PMC11299177 DOI: 10.1016/j.canlet.2024.216993] [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: 01/02/2024] [Revised: 04/18/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024]
Abstract
Ribonucleotide Reductase (RNR) is a rate-limiting enzyme in the production of deoxyribonucleoside triphosphates (dNTPs), which are essential substrates for DNA repair after radiation damage. We explored the radiosensitization property of RNR and investigated a selective RRM2 inhibitor, 3-AP, as a radiosensitizer in the treatment of metastatic pNETs. We investigated the role of RNR subunit, RRM2, in pancreatic neuroendocrine (pNET) cells and responses to radiation in vitro. We also evaluated the selective RRM2 subunit inhibitor, 3-AP, as a radiosensitizer to treat pNET metastases in vivo. Knockdown of RNR subunits demonstrated that RRM1 and RRM2 subunits, but not p53R3, play significant roles in cell proliferation. RRM2 inhibition activated DDR pathways through phosphorylation of ATM and DNA-PK protein kinases but not ATR. RRM2 inhibition also induced Chk1 and Chk2 phosphorylation, resulting in G1/S phase cell cycle arrest. RRM2 inhibition sensitized pNET cells to radiotherapy and induced apoptosis in vitro. In vivo, we utilized pNET subcutaneous and lung metastasis models to examine the rationale for RNR-targeted therapy and 3-AP as a radiosensitizer in treating pNETs. Combination treatment significantly increased apoptosis of BON (human pNET) xenografts and significantly reduced the burden of lung metastases. Together, our results demonstrate that selective RRM2 inhibition induced radiosensitivity of metastatic pNETs both in vitro and in vivo. Therefore, treatment with the selective RRM2 inhibitor, 3-AP, is a promising radiosensitizer in the therapeutic armamentarium for metastatic pNETs.
Collapse
Affiliation(s)
- Zeta Chow
- Markey Cancer Center, Lexington, KY, USA; Department of Radiation Medicine, University of Kentucky, Lexington, KY, USA
| | | | - Aman Chauhan
- Division of Medical Oncology, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Jong Cheol Jeong
- Markey Cancer Center, Lexington, KY, USA; Department of Internal Medicine, Division of Biomedical Informatics, University of Kentucky, Lexington, KY, USA
| | - Jennifer T Castle
- Markey Cancer Center, Lexington, KY, USA; Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Tadahide Izumi
- Markey Cancer Center, Lexington, KY, USA; Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| | - Heidi Weiss
- Markey Cancer Center, Lexington, KY, USA; Department of Internal Medicine, Division of Cancer Biostatistics, University of Kentucky, Lexington, KY, USA
| | - Courtney M Townsend
- Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA
| | - Jörg Schrader
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lowell Anthony
- Markey Cancer Center, Lexington, KY, USA; Department of Internal Medicine, Division of Medical Oncology, University of Kentucky, Lexington, KY, USA
| | - Eddy S Yang
- Markey Cancer Center, Lexington, KY, USA; Department of Radiation Medicine, University of Kentucky, Lexington, KY, USA
| | - B Mark Evers
- Markey Cancer Center, Lexington, KY, USA; Department of Surgery, University of Kentucky, Lexington, KY, USA
| | - Piotr Rychahou
- Markey Cancer Center, Lexington, KY, USA; Department of Surgery, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
16
|
Justice JL, Reed TJ, Phelan B, Greco TM, Hutton JE, Cristea IM. DNA-PK and ATM drive phosphorylation signatures that antagonistically regulate cytokine responses to herpesvirus infection or DNA damage. Cell Syst 2024; 15:339-361.e8. [PMID: 38593799 PMCID: PMC11098675 DOI: 10.1016/j.cels.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 01/09/2024] [Accepted: 03/15/2024] [Indexed: 04/11/2024]
Abstract
The DNA-dependent protein kinase, DNA-PK, is an essential regulator of DNA damage repair. DNA-PK-driven phosphorylation events and the activated DNA damage response (DDR) pathways are also components of antiviral intrinsic and innate immune responses. Yet, it is not clear whether and how the DNA-PK response differs between these two forms of nucleic acid stress-DNA damage and DNA virus infection. Here, we define DNA-PK substrates and the signature cellular phosphoproteome response to DNA damage or infection with the nuclear-replicating DNA herpesvirus, HSV-1. We establish that DNA-PK negatively regulates the ataxia-telangiectasia-mutated (ATM) DDR kinase during viral infection. In turn, ATM blocks the binding of DNA-PK and the nuclear DNA sensor IFI16 to viral DNA, thereby inhibiting cytokine responses. However, following DNA damage, DNA-PK enhances ATM activity, which is required for IFN-β expression. These findings demonstrate that the DDR autoregulates cytokine expression through the opposing modulation of DDR kinases.
Collapse
Affiliation(s)
- Joshua L Justice
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, USA
| | - Tavis J Reed
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, USA
| | - Brett Phelan
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, USA
| | - Todd M Greco
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, USA
| | - Josiah E Hutton
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, USA
| | - Ileana M Cristea
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, USA.
| |
Collapse
|
17
|
Liang M, Sheng L, Ke Y, Wu Z. The research progress on radiation resistance of cervical cancer. Front Oncol 2024; 14:1380448. [PMID: 38651153 PMCID: PMC11033433 DOI: 10.3389/fonc.2024.1380448] [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: 02/01/2024] [Accepted: 03/25/2024] [Indexed: 04/25/2024] Open
Abstract
Cervical carcinoma is the most prevalent gynecology malignant tumor and ranks as the fourth most common cancer worldwide, thus posing a significant threat to the lives and health of women. Advanced and early-stage cervical carcinoma patients with high-risk factors require adjuvant treatment following surgery, with radiotherapy being the primary approach. However, the tolerance of cervical cancer to radiotherapy has become a major obstacle in its treatment. Recent studies have demonstrated that radiation resistance in cervical cancer is closely associated with DNA damage repair pathways, the tumor microenvironment, tumor stem cells, hypoxia, cell cycle arrest, and epigenetic mechanisms, among other factors. The development of tumor radiation resistance involves complex interactions between multiple genes, pathways, and mechanisms, wherein each factor interacts through one or more signaling pathways. This paper provides an overview of research progress on an understanding of the mechanism underlying radiation resistance in cervical cancer.
Collapse
Affiliation(s)
| | | | - Yumin Ke
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Zhuna Wu
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| |
Collapse
|
18
|
Zhong Y, Wang G, Yang S, Zhang Y, Wang X. The role of DNA damage in neural stem cells ageing. J Cell Physiol 2024; 239:e31187. [PMID: 38219047 DOI: 10.1002/jcp.31187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 11/17/2023] [Accepted: 12/20/2023] [Indexed: 01/15/2024]
Abstract
Neural stem cells (NSCs) are pluripotent stem cells with the potential to differentiate into a variety of nerve cells. NSCs are susceptible to both intracellular and extracellular insults, thus causing DNA damage. Extracellular insults include ultraviolet, ionizing radiation, base analogs, modifiers, alkyl agents and others, while intracellular factors include Reactive oxygen species (ROS) radicals produced by mitochondria, mismatches that occur during DNA replication, deamination of bases, loss of bases, and more. When encountered with DNA damage, cells typically employ three coping strategies: DNA repair, damage tolerance, and apoptosis. NSCs, like many other stem cells, have the ability to divide, differentiate, and repair DNA damage to prevent mutations from being passed down to the next generation. However, when DNA damage accumulates over time, it will lead to a series of alterations in the metabolism of cells, which will cause cellular ageing. The ageing and exhaustion of neural stem cell will have serious effects on the body, such as neurodegenerative diseases. The purpose of this review is to examine the processes by which DNA damage leads to NSCs ageing and the mechanisms of DNA repair in NSCs.
Collapse
Affiliation(s)
- Yiming Zhong
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Guangming Wang
- School of Medicine, Postdoctoral Station of Clinical Medicine, Shanghai Tongji Hospital, Tongji University, Shanghai, China
| | - Shangzhi Yang
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yang Zhang
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xianli Wang
- School of Public Health, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
19
|
Westover KR, Jin P, Yao B. Bridging the gap: R-loop mediated genomic instability and its implications in neurological diseases. Epigenomics 2024; 16:589-608. [PMID: 38530068 PMCID: PMC11160457 DOI: 10.2217/epi-2023-0379] [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: 10/27/2023] [Accepted: 03/12/2024] [Indexed: 03/27/2024] Open
Abstract
R-loops, intricate three-stranded structures formed by RNA-DNA hybrids and an exposed non-template DNA strand, are fundamental to various biological phenomena. They carry out essential and contrasting functions within cellular mechanisms, underlining their critical role in maintaining cellular homeostasis. The specific cellular context that dictates R-loop formation determines their function, particularly emphasizing the necessity for their meticulous genomic regulation. Notably, the aberrant formation or misregulation of R-loops is implicated in numerous neurological disorders. This review focuses on the complex interactions between R-loops and double-strand DNA breaks, exploring how R-loop dysregulation potentially contributes to the pathogenesis of various brain disorders, which could provide novel insights into the molecular mechanisms underpinning neurological disease progression and identify potential therapeutic targets by highlighting these aspects.
Collapse
Affiliation(s)
- Katherine R Westover
- Department of Human Genetics, Emory University, School of Medicine, Atlanta, GA 30322, USA
| | - Peng Jin
- Department of Human Genetics, Emory University, School of Medicine, Atlanta, GA 30322, USA
| | - Bing Yao
- Department of Human Genetics, Emory University, School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
20
|
Zheng Y, Li Y, Zhou K, Li T, VanDusen NJ, Hua Y. Precise genome-editing in human diseases: mechanisms, strategies and applications. Signal Transduct Target Ther 2024; 9:47. [PMID: 38409199 PMCID: PMC10897424 DOI: 10.1038/s41392-024-01750-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 02/28/2024] Open
Abstract
Precise genome-editing platforms are versatile tools for generating specific, site-directed DNA insertions, deletions, and substitutions. The continuous enhancement of these tools has led to a revolution in the life sciences, which promises to deliver novel therapies for genetic disease. Precise genome-editing can be traced back to the 1950s with the discovery of DNA's double-helix and, after 70 years of development, has evolved from crude in vitro applications to a wide range of sophisticated capabilities, including in vivo applications. Nonetheless, precise genome-editing faces constraints such as modest efficiency, delivery challenges, and off-target effects. In this review, we explore precise genome-editing, with a focus on introduction of the landmark events in its history, various platforms, delivery systems, and applications. First, we discuss the landmark events in the history of precise genome-editing. Second, we describe the current state of precise genome-editing strategies and explain how these techniques offer unprecedented precision and versatility for modifying the human genome. Third, we introduce the current delivery systems used to deploy precise genome-editing components through DNA, RNA, and RNPs. Finally, we summarize the current applications of precise genome-editing in labeling endogenous genes, screening genetic variants, molecular recording, generating disease models, and gene therapy, including ex vivo therapy and in vivo therapy, and discuss potential future advances.
Collapse
Affiliation(s)
- Yanjiang Zheng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Kaiyu Zhou
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Tiange Li
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Nathan J VanDusen
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - Yimin Hua
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
21
|
Waters KL, Spratt DE. New Discoveries on Protein Recruitment and Regulation during the Early Stages of the DNA Damage Response Pathways. Int J Mol Sci 2024; 25:1676. [PMID: 38338953 PMCID: PMC10855619 DOI: 10.3390/ijms25031676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/26/2024] [Accepted: 01/28/2024] [Indexed: 02/12/2024] Open
Abstract
Maintaining genomic stability and properly repairing damaged DNA is essential to staying healthy and preserving cellular homeostasis. The five major pathways involved in repairing eukaryotic DNA include base excision repair (BER), nucleotide excision repair (NER), mismatch repair (MMR), non-homologous end joining (NHEJ), and homologous recombination (HR). When these pathways do not properly repair damaged DNA, genomic stability is compromised and can contribute to diseases such as cancer. It is essential that the causes of DNA damage and the consequent repair pathways are fully understood, yet the initial recruitment and regulation of DNA damage response proteins remains unclear. In this review, the causes of DNA damage, the various mechanisms of DNA damage repair, and the current research regarding the early steps of each major pathway were investigated.
Collapse
Affiliation(s)
| | - Donald E. Spratt
- Gustaf H. Carlson School of Chemistry and Biochemistry, Clark University, 950 Main St., Worcester, MA 01610, USA;
| |
Collapse
|
22
|
Zhong A, Cheng CS, Lu RQ, Guo L. Suppression of NBS1 Upregulates CyclinB to Induce Olaparib Sensitivity in Ovarian Cancer. Technol Cancer Res Treat 2024; 23:15330338231212085. [PMID: 38192153 PMCID: PMC10777771 DOI: 10.1177/15330338231212085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 09/17/2023] [Accepted: 10/18/2023] [Indexed: 01/10/2024] Open
Abstract
Background: Deficiencies in DNA damage repair responses promote chemotherapy sensitivity of tumor cells. The Nibrin homolog encoding gene Nijmegen Breakage Syndrome 1 (NBS1) is a crucial component of the MRE11-RAD50-NBN complex (MRN complex) and is involved in the response to DNA double-strand breaks (DSBs) repair that has emerged as an attractive strategy to overcome tumor drug resistance, but the functional relationship between NBS1 regulated DNA damage repair and cell cycle checkpoints has not been fully elucidated. Methods: In this study, lentivirus-mediated RNAi was used to construct NBS1-downregulated cells. Flow cytometry, qPCR, and immunohistochemistry were used to explore the regulatory relationship between NBS1 and CyclinB in vivo and in vitro. Results: Our findings suggest that NBS1 deficiency leads to defective homologous recombination repair. Inhibition of NBS1 expression activates CHK1 and CyclinB signaling pathways leading to cell cycle arrest and sensitizes ovarian cancer cells to Olaparib treatment in vitro and in vivo. NBS1-deficient ovarian cancer cells tend to maintain sensitivity to chemotherapeutic drugs through activation of cell cycle checkpoints. Conclusions: NBS1 may be a potential therapeutic target for epithelial ovarian cancer as it plays a role in the regulation of the DNA damage response and cell cycle checkpoints. Suppression of NBS1 upregulates CyclinB to induce Olaparib sensitivity in ovarian cancer.
Collapse
Affiliation(s)
- Ailing Zhong
- Department of Clinical Laboratory, Fudan University, Shanghai Cancer Center, Shanghai, China
| | - Chien-shan Cheng
- Department of Integrative Oncology, Fudan University, Shanghai Cancer Center, Shanghai, China
| | - Ren quan Lu
- Department of Clinical Laboratory, Fudan University, Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lin Guo
- Department of Clinical Laboratory, Fudan University, Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
23
|
Dias Nunes J, Demeestere I, Devos M. BRCA Mutations and Fertility Preservation. Int J Mol Sci 2023; 25:204. [PMID: 38203374 PMCID: PMC10778779 DOI: 10.3390/ijms25010204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Hereditary cancers mostly affect the adolescent and young adult population (AYA) at reproductive age. Mutations in BReast CAncer (BRCA) genes are responsible for the majority of cases of hereditary breast and ovarian cancer. BRCA1 and BRCA2 act as tumor suppressor genes as they are key regulators of DNA repair through homologous recombination. Evidence of the accumulation of DNA double-strand break has been reported in aging oocytes, while BRCA expression decreases, leading to the hypothesis that BRCA mutation may impact fertility. Moreover, patients exposed to anticancer treatments are at higher risk of fertility-related issues, and BRCA mutations could exacerbate the treatment-induced depletion of the ovarian reserve. In this review, we summarized the functions of both genes and reported the current knowledge on the impact of BRCA mutations on ovarian ageing, premature ovarian insufficiency, female fertility preservation strategies and insights about male infertility. Altogether, this review provides relevant up-to-date information on the impact of BRCA1/2 mutations on fertility. Notably, BRCA-mutated patients should be adequately counselled for fertility preservation strategies, considering their higher sensitivity to chemotherapy gonadotoxic effects.
Collapse
Affiliation(s)
- Joana Dias Nunes
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; (J.D.N.); (M.D.)
| | - Isabelle Demeestere
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; (J.D.N.); (M.D.)
- Fertility Clinic, HUB-Erasme Hospital, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Melody Devos
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; (J.D.N.); (M.D.)
| |
Collapse
|
24
|
El Nachef L, Al-Choboq J, Bourguignon M, Foray N. Response of Fibroblasts from Menkes' and Wilson's Copper Metabolism-Related Disorders to Ionizing Radiation: Influence of the Nucleo-Shuttling of the ATM Protein Kinase. Biomolecules 2023; 13:1746. [PMID: 38136617 PMCID: PMC10741441 DOI: 10.3390/biom13121746] [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: 10/19/2023] [Revised: 11/18/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023] Open
Abstract
Menkes' disease (MD) and Wilson's disease (WD) are two major copper (Cu) metabolism-related disorders caused by mutations of the ATP7A and ATP7B ATPase gene, respectively. While Cu is involved in DNA strand breaks signaling and repair, the response of cells from both diseases to ionizing radiation, a common DNA strand breaks inducer, has not been investigated yet. To this aim, three MD and two WD skin fibroblasts lines were irradiated at two Gy X-rays and clonogenic cell survival, micronuclei, anti-γH2AX, -pATM, and -MRE11 immunofluorescence assays were applied to evaluate the DNA double-strand breaks (DSB) recognition and repair. MD and WD cells appeared moderately radiosensitive with a delay in the radiation-induced ATM nucleo-shuttling (RIANS) associated with impairments in the DSB recognition. Such delayed RIANS was notably caused in both MD and WD cells by a highly expressed ATP7B protein that forms complexes with ATM monomers in cytoplasm. Interestingly, a Cu pre-treatment of cells may influence the activity of the MRE11 nuclease and modulate the radiobiological phenotype. Lastly, some high-passage MD cells cultured in routine may transform spontaneously becoming immortalized. Altogether, our findings suggest that exposure to ionizing radiation may impact on clinical features of MD and WD, which requires cautiousness when affected patients are submitted to radiodiagnosis and, eventually, radiotherapy.
Collapse
Affiliation(s)
- Laura El Nachef
- INSERM U1296 Unit “Radiation: Defense, Health, Environment”, Centre Léon-Bérard, 69008 Lyon, France; (L.E.N.); (J.A.-C.); (M.B.)
| | - Joëlle Al-Choboq
- INSERM U1296 Unit “Radiation: Defense, Health, Environment”, Centre Léon-Bérard, 69008 Lyon, France; (L.E.N.); (J.A.-C.); (M.B.)
| | - Michel Bourguignon
- INSERM U1296 Unit “Radiation: Defense, Health, Environment”, Centre Léon-Bérard, 69008 Lyon, France; (L.E.N.); (J.A.-C.); (M.B.)
- Department of Biophysics and Nuclear Medicine, Université Paris Saclay Versailles St Quentin en Yvelines, 78035 Versailles, France
| | - Nicolas Foray
- INSERM U1296 Unit “Radiation: Defense, Health, Environment”, Centre Léon-Bérard, 69008 Lyon, France; (L.E.N.); (J.A.-C.); (M.B.)
| |
Collapse
|
25
|
Schuhwerk H, Brabletz T. Mutual regulation of TGFβ-induced oncogenic EMT, cell cycle progression and the DDR. Semin Cancer Biol 2023; 97:86-103. [PMID: 38029866 DOI: 10.1016/j.semcancer.2023.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 10/06/2023] [Accepted: 11/23/2023] [Indexed: 12/01/2023]
Abstract
TGFβ signaling and the DNA damage response (DDR) are two cellular toolboxes with a strong impact on cancer biology. While TGFβ as a pleiotropic cytokine affects essentially all hallmarks of cancer, the multifunctional DDR mostly orchestrates cell cycle progression, DNA repair, chromatin remodeling and cell death. One oncogenic effect of TGFβ is the partial activation of epithelial-to-mesenchymal transition (EMT), conferring invasiveness, cellular plasticity and resistance to various noxae. Several reports show that both individual networks as well as their interface affect chemo-/radiotherapies. However, the underlying mechanisms remain poorly resolved. EMT often correlates with TGFβ-induced slowing of proliferation, yet numerous studies demonstrate that particularly the co-activated EMT transcription factors counteract anti-proliferative signaling in a partially non-redundant manner. Collectively, evidence piled up over decades underscore a multifaceted, reciprocal inter-connection of TGFβ signaling / EMT with the DDR / cell cycle progression, which we will discuss here. Altogether, we conclude that full cell cycle arrest is barely compatible with the propagation of oncogenic EMT traits and further propose that 'EMT-linked DDR plasticity' is a crucial, yet intricate facet of malignancy, decisively affecting metastasis formation and therapy resistance.
Collapse
Affiliation(s)
- Harald Schuhwerk
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany.
| | - Thomas Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany; Comprehensive Cancer Center Erlangen-EMN, Erlangen University Hospital, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
26
|
Ferretti LP, Böhi F, Leslie Pedrioli DM, Cheng PF, Ferrari E, Baumgaertner P, Alvarado-Diaz A, Sella F, Cereghetti A, Turko P, Wright RH, De Bock K, Speiser DE, Ferrari R, Levesque MP, Hottiger MO. Combinatorial Treatment with PARP and MAPK Inhibitors Overcomes Phenotype Switch-Driven Drug Resistance in Advanced Melanoma. Cancer Res 2023; 83:3974-3988. [PMID: 37729428 DOI: 10.1158/0008-5472.can-23-0485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 07/07/2023] [Accepted: 09/15/2023] [Indexed: 09/22/2023]
Abstract
Metastatic melanoma is either intrinsically resistant or rapidly acquires resistance to targeted therapy treatments, such as MAPK inhibitors (MAPKi). A leading cause of resistance to targeted therapy is a dynamic transition of melanoma cells from a proliferative to a highly invasive state, a phenomenon called phenotype switching. Mechanisms regulating phenotype switching represent potential targets for improving treatment of patients with melanoma. Using a drug screen targeting chromatin regulators in patient-derived three-dimensional MAPKi-resistant melanoma cell cultures, we discovered that PARP inhibitors (PARPi) restore sensitivity to MAPKis, independent of DNA damage repair pathways. Integrated transcriptomic, proteomic, and epigenomic analyses demonstrated that PARPis induce lysosomal autophagic cell death, accompanied by enhanced mitochondrial lipid metabolism that ultimately increases antigen presentation and sensitivity to T-cell cytotoxicity. Moreover, transcriptomic and epigenetic rearrangements induced by PARP inhibition reversed epithelial-mesenchymal transition-like phenotype switching, which redirected melanoma cells toward a proliferative and MAPKi-sensitive state. The combination of PARP and MAPKis synergistically induced cancer cell death both in vitro and in vivo in patient-derived xenograft models. Therefore, this study provides a scientific rationale for treating patients with melanoma with PARPis in combination with MAPKis to abrogate acquired therapy resistance. SIGNIFICANCE PARP inhibitors can overcome resistance to MAPK inhibitors by activating autophagic cell death and reversing phenotype switching, suggesting that this synergistic combination could help improve the prognosis of patients with melanoma.
Collapse
Affiliation(s)
- Lorenza P Ferretti
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland
| | - Flurina Böhi
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland
| | | | - Phil F Cheng
- Department of Dermatology, University of Zurich, University Hospital Zurich, Schlieren, Switzerland
| | - Elena Ferrari
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland
| | - Petra Baumgaertner
- Department of Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Abdiel Alvarado-Diaz
- Department of Health Sciences and Technology, ETH Zürich, Schwerzenbach, Switzerland
| | - Federica Sella
- Department of Dermatology, University of Zurich, University Hospital Zurich, Schlieren, Switzerland
| | - Alessandra Cereghetti
- Department of Dermatology, University of Zurich, University Hospital Zurich, Schlieren, Switzerland
| | - Patrick Turko
- Department of Dermatology, University of Zurich, University Hospital Zurich, Schlieren, Switzerland
| | - Roni H Wright
- Department of Basic Sciences, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Sant Cugat del Vallès, Barcelona
| | - Katrien De Bock
- Department of Health Sciences and Technology, ETH Zürich, Schwerzenbach, Switzerland
| | - Daniel E Speiser
- Department of Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Roberto Ferrari
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Mitchell P Levesque
- Department of Dermatology, University of Zurich, University Hospital Zurich, Schlieren, Switzerland
| | - Michael O Hottiger
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland
| |
Collapse
|
27
|
Song X, Kirtipal N, Lee S, Malý P, Bharadwaj S. Current therapeutic targets and multifaceted physiological impacts of caffeine. Phytother Res 2023; 37:5558-5598. [PMID: 37679309 DOI: 10.1002/ptr.8000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 08/04/2023] [Accepted: 08/17/2023] [Indexed: 09/09/2023]
Abstract
Caffeine, which shares consubstantial structural similarity with purine adenosine, has been demonstrated as a nonselective adenosine receptor antagonist for eliciting most of the biological functions at physiologically relevant dosages. Accumulating evidence supports caffeine's beneficial effects against different disorders, such as total cardiovascular diseases and type 2 diabetes. Conversely, paradoxical effects are also linked to caffeine ingestion in humans including hypertension-hypotension and tachycardia-bradycardia. These observations suggest the association of caffeine action with its ingested concentration and/or concurrent interaction with preferential molecular targets to direct explicit events in the human body. Thus, a coherent analysis of the functional targets of caffeine, relevant to normal physiology, and disease pathophysiology, is required to understand the pharmacology of caffeine. This review provides a broad overview of the experimentally validated targets of caffeine, particularly those of therapeutic interest, and the impacts of caffeine on organ-specific physiology and pathophysiology. Overall, the available empirical and epidemiological evidence supports the dose-dependent functional activities of caffeine and advocates for further studies to get insights into the caffeine-induced changes under specific conditions, such as asthma, DNA repair, and cancer, in view of its therapeutic applications.
Collapse
Affiliation(s)
- Xinjie Song
- Zhejiang Provincial Key Lab for Chemical and Biological Processing Technology of Farm Product, School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou, China
| | - Nikhil Kirtipal
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Sunjae Lee
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Petr Malý
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences v.v.i, BIOCEV Research Center, Vestec, Czech Republic
| | - Shiv Bharadwaj
- Laboratory of Ligand Engineering, Institute of Biotechnology of the Czech Academy of Sciences v.v.i, BIOCEV Research Center, Vestec, Czech Republic
| |
Collapse
|
28
|
Terlizzi C, De Rosa V, Iommelli F, Pezone A, Altobelli GG, Maddalena M, Dimitrov J, De Rosa C, Della Corte CM, Avvedimento VE, Del Vecchio S. ATM inhibition blocks glucose metabolism and amplifies the sensitivity of resistant lung cancer cell lines to oncogene driver inhibitors. Cancer Metab 2023; 11:20. [PMID: 37932830 PMCID: PMC10629204 DOI: 10.1186/s40170-023-00320-4] [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: 01/13/2023] [Accepted: 10/14/2023] [Indexed: 11/08/2023] Open
Abstract
BACKGROUND ATM is a multifunctional serine/threonine kinase that in addition to its well-established role in DNA repair mechanisms is involved in a number of signaling pathways including regulation of oxidative stress response and metabolic diversion of glucose through the pentose phosphate pathway. Oncogene-driven tumorigenesis often implies the metabolic switch from oxidative phosphorylation to glycolysis which provides metabolic intermediates to sustain cell proliferation. The aim of our study is to elucidate the role of ATM in the regulation of glucose metabolism in oncogene-driven cancer cells and to test whether ATM may be a suitable target for anticancer therapy. METHODS Two oncogene-driven NSCLC cell lines, namely H1975 and H1993 cells, were treated with ATM inhibitor, KU55933, alone or in combination with oncogene driver inhibitors, WZ4002 or crizotinib. Key glycolytic enzymes, mitochondrial complex subunits (OXPHOS), cyclin D1, and apoptotic markers were analyzed by Western blotting. Drug-induced toxicity was assessed by MTS assay using stand-alone or combined treatment with KU55933 and driver inhibitors. Glucose consumption, pyruvate, citrate, and succinate levels were also analyzed in response to KU55933 treatment. Both cell lines were transfected with ATM-targeted siRNA or non-targeting siRNA and then exposed to treatment with driver inhibitors. RESULTS ATM inhibition deregulates and inhibits glucose metabolism by reducing HKII, p-PKM2Tyr105, p-PKM2Ser37, E1α subunit of pyruvate dehydrogenase complex, and all subunits of mitochondrial complexes except ATP synthase. Accordingly, glucose uptake and pyruvate concentrations were reduced in response to ATM inhibition, whereas citrate and succinate levels were increased in both cell lines indicating the supply of alternative metabolic substrates. Silencing of ATM resulted in similar changes in glycolytic cascade and OXPHOS levels. Furthermore, the driver inhibitors amplified the effects of ATM downregulation on glucose metabolism, and the combined treatment with ATM inhibitors enhanced the cytotoxic effect of driver inhibitors alone by increasing the apoptotic response. CONCLUSIONS Inhibition of ATM reduced both glycolytic enzymes and OXPHOS levels in oncogene-driven cancer cells and enhanced apoptosis induced by driver inhibitors thus highlighting the possibility to use ATM and the driver inhibitors in combined regimens of anticancer therapy in vivo.
Collapse
Affiliation(s)
- Cristina Terlizzi
- Department of Advanced Biomedical Sciences, University "Federico II", 80131, Naples, Italy
| | - Viviana De Rosa
- Institute of Biostructures and Bioimaging, National Research Council, Naples, Italy
| | - Francesca Iommelli
- Institute of Biostructures and Bioimaging, National Research Council, Naples, Italy
| | - Antonio Pezone
- Department of Biology, University "Federico II", Naples, Italy
| | - Giovanna G Altobelli
- Department of Advanced Biomedical Sciences, University "Federico II", 80131, Naples, Italy
| | - Maurizio Maddalena
- Department of Advanced Biomedical Sciences, University "Federico II", 80131, Naples, Italy
| | - Jelena Dimitrov
- Department of Advanced Biomedical Sciences, University "Federico II", 80131, Naples, Italy
| | - Caterina De Rosa
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | | | | | - Silvana Del Vecchio
- Department of Advanced Biomedical Sciences, University "Federico II", 80131, Naples, Italy.
| |
Collapse
|
29
|
Jin Y, Li Y, He S, Ge Y, Zhao Y, Zhu K, He A, Li S, Yan S, Cao C. ATM participates in fine particulate matter-induced airway inflammation through regulating DNA damage and DNA damage response. ENVIRONMENTAL TOXICOLOGY 2023; 38:2668-2678. [PMID: 37483094 DOI: 10.1002/tox.23901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 05/20/2023] [Accepted: 07/06/2023] [Indexed: 07/25/2023]
Abstract
The relationship between fine particulate matter (PM2.5) and chronic airway inflammatory diseases, such as chronic obstructive pulmonary disease and asthma, have garnered public attention, while the detailed mechanisms of PM2.5-induced airway inflammation remain unclear. This study reveals that PM2.5 induces airway inflammation both in vivo and in vitro, and, moreover, identifies DNA damage and DNA damage repair (DDR) as results of this exposure. Ataxia telangiectasia-mutated heterozygous (ATM+/- ) and wild-type C57BL/6 (WT) mice were exposed to PM2.5. The results show that, following exposure to PM2.5, the number of neutrophils in broncho alveolar lavage fluid and the mRNA expression of CXCL-1 in lung tissues of the ATM+/- mice were lower than those of the WT mice. The mRNA expression of FANCD2 and FANCI were also down-regulated. Human bronchial epithelial (HBE) cells were transfected with ATM-siRNA to induce down-regulation of ATM gene expression and were subsequently stimulated with PM2.5. The results show that the mRNA expression of TNF-α decreased in the ATM-siRNA-transfected cells. The mRNA expression of CXCL-1 and CXCL-2 in peritoneal macrophages, derived from ATM-null mice in which experiments showed that the protein expression of FANCD2 and FANCI decreased, were also decreased after PM2.5 exposure in ATM-siRNA-transfected HBE cells. In conclusion, PM2.5-induced airway inflammation is alleviated in ATM+/- mice compared with WT mice. ATM promotes PM2.5-induced airway inflammation, which may be attributed to the regulation of DNA damage and DDR.
Collapse
Affiliation(s)
- Yan Jin
- Department of Respiratory and Critical Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, China
- Department of Respiratory and Critical Care Medicine, Municipal Hospital Affiliated to Taizhou University, Taizhou, China
| | - Yiting Li
- Department of Respiratory and Critical Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Shiyi He
- Department of Respiratory and Critical Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Yijun Ge
- Department of Respiratory and Critical Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Yun Zhao
- Department of Respiratory and Critical Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Ke Zhu
- Department of Respiratory and Critical Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Andong He
- Department of Respiratory and Critical Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Siyu Li
- Department of Respiratory and Critical Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Siyu Yan
- Department of Respiratory and Critical Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Chao Cao
- Department of Respiratory and Critical Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
30
|
Scherthan H, Geiger B, Ridinger D, Müller J, Riccobono D, Bestvater F, Port M, Hausmann M. Nano-Architecture of Persistent Focal DNA Damage Regions in the Minipig Epidermis Weeks after Acute γ-Irradiation. Biomolecules 2023; 13:1518. [PMID: 37892200 PMCID: PMC10605239 DOI: 10.3390/biom13101518] [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: 07/25/2023] [Revised: 09/22/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
Exposure to high acute doses of ionizing radiation (IR) can induce cutaneous radiation syndrome. Weeks after such radiation insults, keratinocyte nuclei of the epidermis exhibit persisting genomic lesions that present as focal accumulations of DNA double-strand break (DSB) damage marker proteins. Knowledge about the nanostructure of these genomic lesions is scarce. Here, we compared the chromatin nano-architecture with respect to DNA damage response (DDR) factors in persistent genomic DNA damage regions and healthy chromatin in epidermis sections of two minipigs 28 days after lumbar irradiation with ~50 Gy γ-rays, using single-molecule localization microscopy (SMLM) combined with geometric and topological mathematical analyses. SMLM analysis of fluorochrome-stained paraffin sections revealed, within keratinocyte nuclei with perisitent DNA damage, the nano-arrangements of pATM, 53BP1 and Mre11 DDR proteins in γ-H2AX-positive focal chromatin areas (termed macro-foci). It was found that persistent macro-foci contained on average ~70% of 53BP1, ~23% of MRE11 and ~25% of pATM single molecule signals of a nucleus. MRE11 and pATM fluorescent tags were organized in focal nanoclusters peaking at about 40 nm diameter, while 53BP1 tags formed nanoclusters that made up super-foci of about 300 nm in size. Relative to undamaged nuclear chromatin, the enrichment of DDR protein signal tags in γ-H2AX macro-foci was on average 8.7-fold (±3) for 53BP1, 3.4-fold (±1.3) for MRE11 and 3.6-fold (±1.8) for pATM. The persistent macro-foci of minipig epidermis displayed a ~2-fold enrichment of DDR proteins, relative to DSB foci of lymphoblastoid control cells 30 min after 0.5 Gy X-ray exposure. A lasting accumulation of damage signaling and sensing molecules such as pATM and 53BP1, as well as the DSB end-processing protein MRE11 in the persistent macro-foci suggests the presence of diverse DNA damages which pose an insurmountable problem for DSB repair.
Collapse
Affiliation(s)
- Harry Scherthan
- Bundeswehr Institute for Radiobiology Affiliated to the University of Ulm, Neuherbergstr. 11, D-80937 München, Germany (M.P.)
| | - Beatrice Geiger
- Kirchhoff-Institute for Physics, Heidelberg University, Im Neuenheimer Feld 227, D-69120 Heidelberg, Germany (D.R.)
| | - David Ridinger
- Kirchhoff-Institute for Physics, Heidelberg University, Im Neuenheimer Feld 227, D-69120 Heidelberg, Germany (D.R.)
| | - Jessica Müller
- Bundeswehr Institute for Radiobiology Affiliated to the University of Ulm, Neuherbergstr. 11, D-80937 München, Germany (M.P.)
| | - Diane Riccobono
- Département des Effets Biologiques des Rayonnements, French Armed Forces Biomedical Research Institute, UMR 1296, BP 73, 91223 Brétigny-sur-Orge, France;
| | - Felix Bestvater
- Core Facility Light Microscopy, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany;
| | - Matthias Port
- Bundeswehr Institute for Radiobiology Affiliated to the University of Ulm, Neuherbergstr. 11, D-80937 München, Germany (M.P.)
| | - Michael Hausmann
- Kirchhoff-Institute for Physics, Heidelberg University, Im Neuenheimer Feld 227, D-69120 Heidelberg, Germany (D.R.)
| |
Collapse
|
31
|
Reichlmeir M, Canet-Pons J, Koepf G, Nurieva W, Duecker RP, Doering C, Abell K, Key J, Stokes MP, Zielen S, Schubert R, Ivics Z, Auburger G. In Cerebellar Atrophy of 12-Month-Old ATM-Null Mice, Transcriptome Upregulations Concern Most Neurotransmission and Neuropeptide Pathways, While Downregulations Affect Prominently Itpr1, Usp2 and Non-Coding RNA. Cells 2023; 12:2399. [PMID: 37830614 PMCID: PMC10572167 DOI: 10.3390/cells12192399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 09/29/2023] [Accepted: 10/01/2023] [Indexed: 10/14/2023] Open
Abstract
The autosomal recessive disorder Ataxia-Telangiectasia is caused by a dysfunction of the stress response protein, ATM. In the nucleus of proliferating cells, ATM senses DNA double-strand breaks and coordinates their repair. This role explains T-cell dysfunction and tumour risk. However, it remains unclear whether this function is relevant for postmitotic neurons and underlies cerebellar atrophy, since ATM is cytoplasmic in postmitotic neurons. Here, we used ATM-null mice that survived early immune deficits via bone-marrow transplantation, and that reached initial neurodegeneration stages at 12 months of age. Global cerebellar transcriptomics demonstrated that ATM depletion triggered upregulations in most neurotransmission and neuropeptide systems. Downregulated transcripts were found for the ATM interactome component Usp2, many non-coding RNAs, ataxia genes Itpr1, Grid2, immediate early genes and immunity factors. Allelic splice changes affected prominently the neuropeptide machinery, e.g., Oprm1. Validation experiments with stressors were performed in human neuroblastoma cells, where ATM was localised only to cytoplasm, similar to the brain. Effect confirmation in SH-SY5Y cells occurred after ATM depletion and osmotic stress better than nutrient/oxidative stress, but not after ATM kinase inhibition or DNA stressor bleomycin. Overall, we provide pioneer observations from a faithful A-T mouse model, which suggest general changes in synaptic and dense-core vesicle stress adaptation.
Collapse
Affiliation(s)
- Marina Reichlmeir
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Exp. Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (M.R.); (J.C.-P.); (J.K.)
| | - Júlia Canet-Pons
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Exp. Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (M.R.); (J.C.-P.); (J.K.)
| | - Gabriele Koepf
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Exp. Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (M.R.); (J.C.-P.); (J.K.)
| | - Wasifa Nurieva
- Transposition and Genome Engineering, Research Centre of the Division of Hematology, Gene and Cell Therapy, Paul Ehrlich Institute, 63225 Langen, Germany; (W.N.); (Z.I.)
| | - Ruth Pia Duecker
- Division of Pediatrics, Pulmonology, Allergology, Infectious Diseases and Gastroenterology, Children’s Hospital, University Hospital, Goethe-University, 60590 Frankfurt am Main, Germany; (R.P.D.); (S.Z.); (R.S.)
| | - Claudia Doering
- Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, 60590 Frankfurt am Main, Germany;
| | - Kathryn Abell
- Cell Signaling Technology, Inc., Danvers, MA 01923, USA; (K.A.); (M.P.S.)
| | - Jana Key
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Exp. Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (M.R.); (J.C.-P.); (J.K.)
| | - Matthew P. Stokes
- Cell Signaling Technology, Inc., Danvers, MA 01923, USA; (K.A.); (M.P.S.)
| | - Stefan Zielen
- Division of Pediatrics, Pulmonology, Allergology, Infectious Diseases and Gastroenterology, Children’s Hospital, University Hospital, Goethe-University, 60590 Frankfurt am Main, Germany; (R.P.D.); (S.Z.); (R.S.)
- Respiratory Research Institute, Medaimun GmbH, 60596 Frankfurt am Main, Germany
| | - Ralf Schubert
- Division of Pediatrics, Pulmonology, Allergology, Infectious Diseases and Gastroenterology, Children’s Hospital, University Hospital, Goethe-University, 60590 Frankfurt am Main, Germany; (R.P.D.); (S.Z.); (R.S.)
| | - Zoltán Ivics
- Transposition and Genome Engineering, Research Centre of the Division of Hematology, Gene and Cell Therapy, Paul Ehrlich Institute, 63225 Langen, Germany; (W.N.); (Z.I.)
| | - Georg Auburger
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Exp. Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (M.R.); (J.C.-P.); (J.K.)
| |
Collapse
|
32
|
Kiweler N, Schwarz H, Nguyen A, Matschos S, Mullins C, Piée-Staffa A, Brachetti C, Roos WP, Schneider G, Linnebacher M, Brenner W, Krämer OH. The epigenetic modifier HDAC2 and the checkpoint kinase ATM determine the responses of microsatellite instable colorectal cancer cells to 5-fluorouracil. Cell Biol Toxicol 2023; 39:2401-2419. [PMID: 35608750 PMCID: PMC10547618 DOI: 10.1007/s10565-022-09731-3] [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: 07/07/2021] [Accepted: 05/10/2022] [Indexed: 11/02/2022]
Abstract
The epigenetic modifier histone deacetylase-2 (HDAC2) is frequently dysregulated in colon cancer cells. Microsatellite instability (MSI), an unfaithful replication of DNA at nucleotide repeats, occurs in about 15% of human colon tumors. MSI promotes a genetic frameshift and consequently a loss of HDAC2 in up to 43% of these tumors. We show that long-term and short-term cultures of colorectal cancers with MSI contain subpopulations of cells lacking HDAC2. These can be isolated as single cell-derived, proliferating populations. Xenografted patient-derived colon cancer tissues with MSI also show variable patterns of HDAC2 expression in mice. HDAC2-positive and HDAC2-negative RKO cells respond similarly to pharmacological inhibitors of the class I HDACs HDAC1/HDAC2/HDAC3. In contrast to this similarity, HDAC2-negative and HDAC2-positive RKO cells undergo differential cell cycle arrest and apoptosis induction in response to the frequently used chemotherapeutic 5-fluorouracil, which becomes incorporated into and damages RNA and DNA. 5-fluorouracil causes an enrichment of HDAC2-negative RKO cells in vitro and in a subset of primary colorectal tumors in mice. 5-fluorouracil induces the phosphorylation of KAP1, a target of the checkpoint kinase ataxia-telangiectasia mutated (ATM), stronger in HDAC2-negative cells than in their HDAC2-positive counterparts. Pharmacological inhibition of ATM sensitizes RKO cells to cytotoxic effects of 5-fluorouracil. These findings demonstrate that HDAC2 and ATM modulate the responses of colorectal cancer cells towards 5-FU.
Collapse
Affiliation(s)
- Nicole Kiweler
- Department of Toxicology, University Medical Center Mainz, 55131 Mainz, Germany
- Present Address: Department of Cancer Research, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg
| | - Helena Schwarz
- Department of Toxicology, University Medical Center Mainz, 55131 Mainz, Germany
| | - Alexandra Nguyen
- Department of Toxicology, University Medical Center Mainz, 55131 Mainz, Germany
| | - Stephanie Matschos
- Department of General Surgery, Molecular Oncology and Immunotherapy, Schillingallee 35, 18057 Rostock, Germany
| | - Christina Mullins
- Department of General Surgery, Molecular Oncology and Immunotherapy, Schillingallee 35, 18057 Rostock, Germany
| | - Andrea Piée-Staffa
- Department of Toxicology, University Medical Center Mainz, 55131 Mainz, Germany
| | - Christina Brachetti
- Department of Toxicology, University Medical Center Mainz, 55131 Mainz, Germany
| | - Wynand P. Roos
- Department of Toxicology, University Medical Center Mainz, 55131 Mainz, Germany
| | - Günter Schneider
- Klinikum Rechts Der Isar, Medical Clinic and Polyclinic II, Technical University Munich, 81675 Munich, Germany
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Michael Linnebacher
- Department of General Surgery, Molecular Oncology and Immunotherapy, Schillingallee 35, 18057 Rostock, Germany
| | - Walburgis Brenner
- Clinic for Obstetrics and Women’s Health, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Oliver H. Krämer
- Department of Toxicology, University Medical Center Mainz, 55131 Mainz, Germany
| |
Collapse
|
33
|
Lin M, Xian H, Chen Z, Wang S, Liu M, Liang W, Tang Q, Liu Y, Huang W, Che D, Guo C, Idiiatullina E, Fang R, Al-Azab M, Chang J, Wang R, Li X, Zuo X, Zhang Y, Zhao J, Tang Y, Jin S, He Z, Feng D, Lu L, Zhang K, Wu Y, Bai F, Lew AM, Cui J, Wu Y, Gu X, Zhang Y. MCM8-mediated mitophagy protects vascular health in response to nitric oxide signaling in a mouse model of Kawasaki disease. NATURE CARDIOVASCULAR RESEARCH 2023; 2:778-792. [PMID: 39195969 DOI: 10.1038/s44161-023-00314-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 07/03/2023] [Indexed: 08/29/2024]
Abstract
Mitophagy is a major quality control pathway that removes unwanted or dysfunctional mitochondria and plays an essential role in vascular health. Here we show that MCM8 expression is significantly decreased in children with Kawasaki disease (KD) who developed coronary artery aneurysms. Mechanistically, we discovered that nitric oxide signaling promotes TRIM21-mediated MCM8 ubiquitination, which disrupts its interaction with MCM9 and promotes its cytosolic export. In the cytosol, MCM8 relocates to the mitochondria pore-forming proteins and promotes their ubiquitination by TRIM21. In addition, MCM8 directly recruits LC3 via its LC3-interacting region (LIR) motif and initiates mitophagy. This suppresses mitochondrial DNA-mediated activation of type I interferon via cGAS and STING. Mice that are deficient in Mcm8, Trim21 and Nos2 or reconstituted with the East-Asian-specific MCM8-P276 variant develop more severe coronary artery vasculopathy in the Lactobacillus casei extract-induced KD model. Collectively, the data suggest that MCM8 protects vascular health in the KD setting.
Collapse
Affiliation(s)
- Meng Lin
- Clinical Biological Resource Bank and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, and State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
- Pediatric Immunity and Healthcare Biomedical Co., Ltd, Guangzhou, China
| | - Huifang Xian
- Clinical Biological Resource Bank and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, and State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Zhanghua Chen
- Clinical Biological Resource Bank and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, and State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Shang Wang
- Clinical Biological Resource Bank and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, and State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Ming Liu
- Clinical Biological Resource Bank and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, and State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Weiwei Liang
- Clinical Biological Resource Bank and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, and State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Qin Tang
- Clinical Biological Resource Bank and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, and State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Yao Liu
- Clinical Biological Resource Bank and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, and State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Wanming Huang
- Clinical Biological Resource Bank and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, and State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Di Che
- Clinical Biological Resource Bank and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, and State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Caiqin Guo
- Clinical Biological Resource Bank and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, and State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Elina Idiiatullina
- Clinical Biological Resource Bank and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, and State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Rongli Fang
- Clinical Biological Resource Bank and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, and State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Mahmoud Al-Azab
- Clinical Biological Resource Bank and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, and State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Jingjie Chang
- Clinical Biological Resource Bank and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, and State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Rongze Wang
- Clinical Biological Resource Bank and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, and State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Xiaojun Li
- Clinical Biological Resource Bank and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, and State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Xiaoyu Zuo
- Clinical Biological Resource Bank and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, and State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Yan Zhang
- Clinical Biological Resource Bank and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, and State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Jincun Zhao
- Clinical Biological Resource Bank and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, and State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Yaping Tang
- Clinical Biological Resource Bank and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, and State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China
| | - Shouheng Jin
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zhengjie He
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Du Feng
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Liwei Lu
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, China
- Chongqing International Institute for Immunology, Chongqing, China
| | - Kang Zhang
- Guangzhou National Laboratory, Guangzhou, China
| | - Yan Wu
- Department of Pathogen Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Fan Bai
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University, Beijing, China
| | - Andrew M Lew
- Walter and Eliza Hall Institute of Medical Research and Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Jun Cui
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yuzhang Wu
- Chongqing International Institute for Immunology, Chongqing, China.
| | - Xiaoqiong Gu
- Clinical Biological Resource Bank and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, and State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China.
| | - Yuxia Zhang
- Clinical Biological Resource Bank and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, and State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, China.
- Chongqing International Institute for Immunology, Chongqing, China.
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
34
|
Obata H, Ogawa M, Zalutsky MR. DNA Repair Inhibitors: Potential Targets and Partners for Targeted Radionuclide Therapy. Pharmaceutics 2023; 15:1926. [PMID: 37514113 PMCID: PMC10384049 DOI: 10.3390/pharmaceutics15071926] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
The present review aims to explore the potential targets/partners for future targeted radionuclide therapy (TRT) strategies, wherein cancer cells often are not killed effectively, despite receiving a high average tumor radiation dose. Here, we shall discuss the key factors in the cancer genome, especially those related to DNA damage response/repair and maintenance systems for escaping cell death in cancer cells. To overcome the current limitations of TRT effectiveness due to radiation/drug-tolerant cells and tumor heterogeneity, and to make TRT more effective, we propose that a promising strategy would be to target the DNA maintenance factors that are crucial for cancer survival. Considering their cancer-specific DNA damage response/repair ability and dysregulated transcription/epigenetic system, key factors such as PARP, ATM/ATR, amplified/overexpressed transcription factors, and DNA methyltransferases have the potential to be molecular targets for Auger electron therapy; moreover, their inhibition by non-radioactive molecules could be a partnering component for enhancing the therapeutic response of TRT.
Collapse
Affiliation(s)
- Honoka Obata
- Department of Advanced Nuclear Medicine Sciences, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
- Department of Molecular Imaging and Theranostics, National Institutes for Quantum Science and Technology (QST), 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
- Departments of Radiology and Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
- Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo 060-0812, Japan
| | - Mikako Ogawa
- Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo 060-0812, Japan
| | - Michael R Zalutsky
- Departments of Radiology and Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
35
|
Agostini F, Bisaglia M, Plotegher N. Linking ROS Levels to Autophagy: The Key Role of AMPK. Antioxidants (Basel) 2023; 12:1406. [PMID: 37507945 PMCID: PMC10376219 DOI: 10.3390/antiox12071406] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/04/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Oxygen reactive species (ROS) are a group of molecules generated from the incomplete reduction of oxygen. Due to their high reactivity, ROS can interact with and influence the function of multiple targets, which include DNA, lipids, and proteins. Among the proteins affected by ROS, AMP-activated protein kinase (AMPK) is considered a major sensor of the intracellular energetic status and a crucial hub involved in the regulation of key cellular processes, like autophagy and lysosomal function. Thanks to these features, AMPK has been recently demonstrated to be able to perceive signals related to the variation of mitochondrial dynamics and to transduce them to the lysosomes, influencing the autophagic flux. Since ROS production is largely dependent on mitochondrial activity, through the modulation of AMPK these molecules may represent important signaling agents which participate in the crosstalk between mitochondria and lysosomes, allowing the coordination of these organelles' functions. In this review, we will describe the mechanisms through which ROS activate AMPK and the signaling pathways that allow this protein to affect the autophagic process. The picture that emerges from the literature is that AMPK regulation is highly tissue-specific and that different pools of AMPK can be localized at specific intracellular compartments, thus differentially responding to altered ROS levels. For this reason, future studies will be highly advisable to discriminate the specific contribution of the activation of different AMPK subpopulations to the autophagic pathway.
Collapse
Affiliation(s)
- Francesco Agostini
- Department of Biology, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy
| | - Marco Bisaglia
- Department of Biology, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy
- Study Center for Neurodegeneration (CESNE), 35121 Padova, Italy
| | - Nicoletta Plotegher
- Department of Biology, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy
- Study Center for Neurodegeneration (CESNE), 35121 Padova, Italy
| |
Collapse
|
36
|
Berthel E, Pujo-Menjouet L, Le Reun E, Sonzogni L, Al-Choboq J, Chekroun A, Granzotto A, Devic C, Ferlazzo ML, Pereira S, Bourguignon M, Foray N. Toward an Early Diagnosis for Alzheimer's Disease Based on the Perinuclear Localization of the ATM Protein. Cells 2023; 12:1747. [PMID: 37443782 PMCID: PMC10340316 DOI: 10.3390/cells12131747] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/09/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative dementia, for which the molecular origins, genetic predisposition and therapeutic approach are still debated. In the 1980s, cells from AD patients were reported to be sensitive to ionizing radiation. In order to examine the molecular basis of this radiosensitivity, the ATM-dependent DNA double-strand breaks (DSB) signaling and repair were investigated by applying an approach based on the radiation-induced ataxia telangiectasia-mutated (ATM) protein nucleoshuttling (RIANS) model. Early after irradiation, all ten AD fibroblast cell lines tested showed impaired DSB recognition and delayed RIANS. AD fibroblasts specifically showed spontaneous perinuclear localization of phosphorylated ATM (pATM) forms. To our knowledge, such observation has never been reported before, and by considering the role of the ATM kinase in the stress response, it may introduce a novel interpretation of accelerated aging. Our data and a mathematical approach through a brand-new model suggest that, in response to a progressive and cumulative stress, cytoplasmic ATM monomers phosphorylate the APOE protein (pAPOE) close to the nuclear membrane and aggregate around the nucleus, preventing their entry in the nucleus and thus the recognition and repair of spontaneous DSB, which contributes to the aging process. Our findings suggest that pATM and/or pAPOE may serve as biomarkers for an early reliable diagnosis of AD on any fibroblast sample.
Collapse
Affiliation(s)
- Elise Berthel
- Institut National de la Santé et de la Recherche Médicale, U1296 Research Unit «Radiation: Defense, Health, Environment», Centre Léon-Bérard, 28 Rue Laennec, 69008 Lyon, France; (E.B.); (E.L.R.); (L.S.); (J.A.-C.); (A.G.); (M.L.F.); (M.B.)
- NEOLYS Diagnostics, 7 Allée de l’Europe, 67960 Entzheim, France;
| | - Laurent Pujo-Menjouet
- Université Claude-Bernard Lyon 1, CNRS UMR5208, INRIA, Institut Camille-Jordan, 21 Avenue Claude Bernard, 69603 Villeurbanne, France; (L.P.-M.); (A.C.)
| | - Eymeric Le Reun
- Institut National de la Santé et de la Recherche Médicale, U1296 Research Unit «Radiation: Defense, Health, Environment», Centre Léon-Bérard, 28 Rue Laennec, 69008 Lyon, France; (E.B.); (E.L.R.); (L.S.); (J.A.-C.); (A.G.); (M.L.F.); (M.B.)
| | - Laurène Sonzogni
- Institut National de la Santé et de la Recherche Médicale, U1296 Research Unit «Radiation: Defense, Health, Environment», Centre Léon-Bérard, 28 Rue Laennec, 69008 Lyon, France; (E.B.); (E.L.R.); (L.S.); (J.A.-C.); (A.G.); (M.L.F.); (M.B.)
| | - Joëlle Al-Choboq
- Institut National de la Santé et de la Recherche Médicale, U1296 Research Unit «Radiation: Defense, Health, Environment», Centre Léon-Bérard, 28 Rue Laennec, 69008 Lyon, France; (E.B.); (E.L.R.); (L.S.); (J.A.-C.); (A.G.); (M.L.F.); (M.B.)
| | - Abdennasser Chekroun
- Université Claude-Bernard Lyon 1, CNRS UMR5208, INRIA, Institut Camille-Jordan, 21 Avenue Claude Bernard, 69603 Villeurbanne, France; (L.P.-M.); (A.C.)
| | - Adeline Granzotto
- Institut National de la Santé et de la Recherche Médicale, U1296 Research Unit «Radiation: Defense, Health, Environment», Centre Léon-Bérard, 28 Rue Laennec, 69008 Lyon, France; (E.B.); (E.L.R.); (L.S.); (J.A.-C.); (A.G.); (M.L.F.); (M.B.)
| | - Clément Devic
- Institut National de la Santé et de la Recherche Médicale, U1296 Research Unit «Radiation: Defense, Health, Environment», Centre Léon-Bérard, 28 Rue Laennec, 69008 Lyon, France; (E.B.); (E.L.R.); (L.S.); (J.A.-C.); (A.G.); (M.L.F.); (M.B.)
| | - Mélanie L. Ferlazzo
- Institut National de la Santé et de la Recherche Médicale, U1296 Research Unit «Radiation: Defense, Health, Environment», Centre Léon-Bérard, 28 Rue Laennec, 69008 Lyon, France; (E.B.); (E.L.R.); (L.S.); (J.A.-C.); (A.G.); (M.L.F.); (M.B.)
| | - Sandrine Pereira
- NEOLYS Diagnostics, 7 Allée de l’Europe, 67960 Entzheim, France;
| | - Michel Bourguignon
- Institut National de la Santé et de la Recherche Médicale, U1296 Research Unit «Radiation: Defense, Health, Environment», Centre Léon-Bérard, 28 Rue Laennec, 69008 Lyon, France; (E.B.); (E.L.R.); (L.S.); (J.A.-C.); (A.G.); (M.L.F.); (M.B.)
- Université Paris-Saclay, 78035 Versailles, France
| | - Nicolas Foray
- Institut National de la Santé et de la Recherche Médicale, U1296 Research Unit «Radiation: Defense, Health, Environment», Centre Léon-Bérard, 28 Rue Laennec, 69008 Lyon, France; (E.B.); (E.L.R.); (L.S.); (J.A.-C.); (A.G.); (M.L.F.); (M.B.)
| |
Collapse
|
37
|
Wu Y, Song Y, Wang R, Wang T. Molecular mechanisms of tumor resistance to radiotherapy. Mol Cancer 2023; 22:96. [PMID: 37322433 PMCID: PMC10268375 DOI: 10.1186/s12943-023-01801-2] [Citation(s) in RCA: 68] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 06/03/2023] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND Cancer is the most prevalent cause of death globally, and radiotherapy is considered the standard of care for most solid tumors, including lung, breast, esophageal, and colorectal cancers and glioblastoma. Resistance to radiation can lead to local treatment failure and even cancer recurrence. MAIN BODY In this review, we have extensively discussed several crucial aspects that cause resistance of cancer to radiation therapy, including radiation-induced DNA damage repair, cell cycle arrest, apoptosis escape, abundance of cancer stem cells, modification of cancer cells and their microenvironment, presence of exosomal and non-coding RNA, metabolic reprogramming, and ferroptosis. We aim to focus on the molecular mechanisms of cancer radiotherapy resistance in relation to these aspects and to discuss possible targets to improve treatment outcomes. CONCLUSIONS Studying the molecular mechanisms responsible for radiotherapy resistance and its interactions with the tumor environment will help improve cancer responses to radiotherapy. Our review provides a foundation to identify and overcome the obstacles to effective radiotherapy.
Collapse
Affiliation(s)
- Yu Wu
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042 Liaoning Province China
- School of Graduate, Dalian Medical University, Dalian, 116044 China
| | - Yingqiu Song
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042 Liaoning Province China
| | - Runze Wang
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042 Liaoning Province China
- School of Graduate, Dalian Medical University, Dalian, 116044 China
| | - Tianlu Wang
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042 Liaoning Province China
- Faculty of Medicine, Dalian University of Technology, Dalian, 116024 China
| |
Collapse
|
38
|
Vega-Muñoz I, Herrera-Estrella A, Martínez-de la Vega O, Heil M. ATM and ATR, two central players of the DNA damage response, are involved in the induction of systemic acquired resistance by extracellular DNA, but not the plant wound response. Front Immunol 2023; 14:1175786. [PMID: 37256140 PMCID: PMC10225592 DOI: 10.3389/fimmu.2023.1175786] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/27/2023] [Indexed: 06/01/2023] Open
Abstract
Background The plant immune response to DNA is highly self/nonself-specific. Self-DNA triggered stronger responses by early immune signals such as H2O2 formation than nonself-DNA from closely related plant species. Plants lack known DNA receptors. Therefore, we aimed to investigate whether a differential sensing of self-versus nonself DNA fragments as damage- versus pathogen-associated molecular patterns (DAMPs/PAMPs) or an activation of the DNA-damage response (DDR) represents the more promising framework to understand this phenomenon. Results We treated Arabidopsis thaliana Col-0 plants with sonicated self-DNA from other individuals of the same ecotype, nonself-DNA from another A. thaliana ecotype, or nonself-DNA from broccoli. We observed a highly self/nonself-DNA-specific induction of H2O2 formation and of jasmonic acid (JA, the hormone controlling the wound response to chewing herbivores) and salicylic acid (SA, the hormone controlling systemic acquired resistance, SAR, to biotrophic pathogens). Mutant lines lacking Ataxia Telangiectasia Mutated (ATM) or ATM AND RAD3-RELATED (ATR) - the two DDR master kinases - retained the differential induction of JA in response to DNA treatments but completely failed to induce H2O2 or SA. Moreover, we observed H2O2 formation in response to in situ-damaged self-DNA from plants that had been treated with bleomycin or SA or infected with virulent bacteria Pseudomonas syringae pv. tomato DC3000 or pv. glycinea carrying effector avrRpt2, but not to DNA from H2O2-treated plants or challenged with non-virulent P. syringae pv. glycinea lacking avrRpt2. Conclusion We conclude that both ATM and ATR are required for the complete activation of the plant immune response to extracellular DNA whereas an as-yet unknown mechanism allows for the self/nonself-differential activation of the JA-dependent wound response.
Collapse
Affiliation(s)
- Isaac Vega-Muñoz
- Laboratorio de Ecología de Plantas, Departamento de Ingeniería Genética, Centro de Investigación y de Estudios Avanzados (CINVESTAV)—Unidad Irapuato, Irapuato, GTO, Mexico
| | - Alfredo Herrera-Estrella
- Laboratorio Nacional de Genómica para la Biodiversidad, Centro de Investigación y de Estudios Avanzados (CINVESTAV)—Unidad de Genómica Avanzada, Irapuato, GTO, Mexico
| | - Octavio Martínez-de la Vega
- Laboratorio Nacional de Genómica para la Biodiversidad, Centro de Investigación y de Estudios Avanzados (CINVESTAV)—Unidad de Genómica Avanzada, Irapuato, GTO, Mexico
| | - Martin Heil
- Laboratorio de Ecología de Plantas, Departamento de Ingeniería Genética, Centro de Investigación y de Estudios Avanzados (CINVESTAV)—Unidad Irapuato, Irapuato, GTO, Mexico
| |
Collapse
|
39
|
Pavitra E, Kancharla J, Gupta VK, Prasad K, Sung JY, Kim J, Tej MB, Choi R, Lee JH, Han YK, Raju GSR, Bhaskar L, Huh YS. The role of NF-κB in breast cancer initiation, growth, metastasis, and resistance to chemotherapy. Biomed Pharmacother 2023; 163:114822. [PMID: 37146418 DOI: 10.1016/j.biopha.2023.114822] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/27/2023] [Accepted: 04/30/2023] [Indexed: 05/07/2023] Open
Abstract
Breast cancer (BC) is the second most fatal disease and is the prime cause of cancer allied female deaths. BC is caused by aberrant tumor suppressor genes and oncogenes regulated by transcription factors (TFs) like NF-κB. NF-κB is a pro-inflammatory TF that crucially alters the expressions of various genes associated with inflammation, cell progression, metastasis, and apoptosis and modulates a network of genes that underlie tumorigenesis. Herein, we focus on NF-κB signaling pathways, its regulators, and the rationale for targeting NF-κB. This review also includes TFs that maintain NF-κB crosstalk and their roles in promoting angiogenesis and metastasis. In addition, we discuss the importance of combination therapies, resistance to treatment, and potential novel therapeutic strategies including nanomedicine that targets NF-κB.
Collapse
Affiliation(s)
- Eluri Pavitra
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea; 3D Convergence Center, Inha University, Incheon 22212, Republic of Korea
| | - Jyothsna Kancharla
- Department of Bioscience and Biotechnology, Banasthali University, Vanasthali, Rajasthan 304022, India
| | - Vivek Kumar Gupta
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea
| | - Kiran Prasad
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur- 495009, Chhattisgarh, India
| | - Ju Yong Sung
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea
| | - Jigyeong Kim
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea
| | - Mandava Bhuvan Tej
- Department of Health care informatics, Sacred Heart University, 5151Park Avenue, Fair fields, CT06825, USA
| | - Rino Choi
- 3D Convergence Center, Inha University, Incheon 22212, Republic of Korea; Department of Materials Science and Engineering, Inha University, Incheon 22212, Republic of Korea
| | - Jeong-Hwan Lee
- 3D Convergence Center, Inha University, Incheon 22212, Republic of Korea; Department of Materials Science and Engineering, Inha University, Incheon 22212, Republic of Korea
| | - Young-Kyu Han
- Department of Energy and Materials Engineering, Dongguk University-Seoul, Seoul 04620, Republic of Korea
| | - Ganji Seeta Rama Raju
- Department of Energy and Materials Engineering, Dongguk University-Seoul, Seoul 04620, Republic of Korea.
| | - Lvks Bhaskar
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur- 495009, Chhattisgarh, India.
| | - Yun Suk Huh
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea.
| |
Collapse
|
40
|
Ajouaou Y, Magnani E, Madakashira B, Jenkins E, Sadler KC. atm Mutation and Oxidative Stress Enhance the Pre-Cancerous Effects of UHRF1 Overexpression in Zebrafish Livers. Cancers (Basel) 2023; 15:cancers15082302. [PMID: 37190230 DOI: 10.3390/cancers15082302] [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: 03/20/2023] [Accepted: 03/29/2023] [Indexed: 05/17/2023] Open
Abstract
The ataxia-telangiectasia mutated (atm) gene is activated in response to genotoxic stress and leads to activation of the tp53 tumor suppressor gene which induces either senescence or apoptosis as tumor suppressive mechanisms. Atm also serves non-canonical functions in the response to oxidative stress and chromatin reorganization. We previously reported that overexpression of the epigenetic regulator and oncogene Ubiquitin Like with PHD and Ring Finger Domains 1 (UHRF1) in zebrafish hepatocytes resulted in tp53-dependent hepatocyte senescence, a small liver and larval lethality. We investigated the role of atm on UHRF1-mediated phenotypes by generating zebrafish atm mutants. atm-/- adults were viable but had reduction in fertility. Embryos developed normally but were protected from lethality caused by etoposide or H2O2 exposure and failed to fully upregulate Tp53 targets or oxidative stress response genes in response to these treatments. In contrast to the finding that Tp53 prevents the small liver phenotype caused by UHRF1 overexpression, atm mutation and exposure to H2O2 further reduced the liver size in UHRF1 overexpressing larvae whereas treatment with the antioxidant N-acetyl cysteine suppressed this phenotype. We conclude that UHRF1 overexpression in hepatocytes causes oxidative stress, and that loss of atm further enhances this, triggering elimination of these precancerous cells, leading to a small liver.
Collapse
Affiliation(s)
- Yousra Ajouaou
- Program in Biology, New York University Abu Dhabi, Abu Dhabi P.O. 129188, United Arab Emirates
- Center for Genomics and Systems Biology (CGSB), New York University Abu Dhabi, Abu Dhabi P.O. 129188, United Arab Emirates
| | - Elena Magnani
- Program in Biology, New York University Abu Dhabi, Abu Dhabi P.O. 129188, United Arab Emirates
| | - Bhavani Madakashira
- Program in Biology, New York University Abu Dhabi, Abu Dhabi P.O. 129188, United Arab Emirates
| | - Eleanor Jenkins
- Program in Biology, New York University Abu Dhabi, Abu Dhabi P.O. 129188, United Arab Emirates
| | - Kirsten C Sadler
- Program in Biology, New York University Abu Dhabi, Abu Dhabi P.O. 129188, United Arab Emirates
- Center for Genomics and Systems Biology (CGSB), New York University Abu Dhabi, Abu Dhabi P.O. 129188, United Arab Emirates
| |
Collapse
|
41
|
Zhao J, Gui X, Ren Z, Fu H, Yang C, Wang W, Liu Q, Zhang M, Wang C, Schnittger A, Liu B. ATM-mediated double-strand break repair is required for meiotic genome stability at high temperature. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2023; 114:403-423. [PMID: 36786716 DOI: 10.1111/tpj.16145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 02/08/2023] [Indexed: 05/10/2023]
Abstract
In eukaryotes, meiotic recombination maintains genome stability and creates genetic diversity. The conserved Ataxia-Telangiectasia Mutated (ATM) kinase regulates multiple processes in meiotic homologous recombination, including DNA double-strand break (DSB) formation and repair, synaptonemal complex organization, and crossover formation and distribution. However, its function in plant meiotic recombination under stressful environmental conditions remains poorly understood. In this study, we demonstrate that ATM is required for the maintenance of meiotic genome stability under heat stress in Arabidopsis thaliana. Using cytogenetic approaches we determined that ATM does not mediate reduced DSB formation but does ensure successful DSB repair, and thus meiotic chromosome integrity, under heat stress. Further genetic analysis suggested that ATM mediates DSB repair at high temperature by acting downstream of the MRE11-RAD50-NBS1 (MRN) complex, and acts in a RAD51-independent but chromosome axis-dependent manner. This study extends our understanding on the role of ATM in DSB repair and the protection of genome stability in plants under high temperature stress.
Collapse
Affiliation(s)
- Jiayi Zhao
- 8-A506, Arameiosis Lab, South-Central Minzu University, Wuhan, 430074, China
| | - Xin Gui
- 8-A506, Arameiosis Lab, South-Central Minzu University, Wuhan, 430074, China
| | - Ziming Ren
- Department of Landscape Architecture, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Huiqi Fu
- 8-A506, Arameiosis Lab, South-Central Minzu University, Wuhan, 430074, China
| | - Chao Yang
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China
- Department of Developmental Biology, University of Hamburg, Hamburg, 22609, Germany
| | - Wenyi Wang
- 8-A506, Arameiosis Lab, South-Central Minzu University, Wuhan, 430074, China
| | - Qingpei Liu
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, 430074, China
| | - Min Zhang
- 8-A506, Arameiosis Lab, South-Central Minzu University, Wuhan, 430074, China
| | - Chong Wang
- Shanghai Key Laboratory of Plant Molecular Sciences, Development Center of Plant Germplasm Resources, College of Life Sciences, Shanghai Normal University, Shanghai, 200234, China
| | - Arp Schnittger
- Department of Developmental Biology, University of Hamburg, Hamburg, 22609, Germany
| | - Bing Liu
- 8-A506, Arameiosis Lab, South-Central Minzu University, Wuhan, 430074, China
| |
Collapse
|
42
|
Fukasawa T, Enomoto A, Yoshizaki-Ogawa A, Sato S, Miyagawa K, Yoshizaki A. The Role of Mammalian STK38 in DNA Damage Response and Targeting for Radio-Sensitization. Cancers (Basel) 2023; 15:cancers15072054. [PMID: 37046714 PMCID: PMC10093458 DOI: 10.3390/cancers15072054] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/13/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Protein kinases, found in the nucleus and cytoplasm, play essential roles in a multitude of cellular processes, including cell division, proliferation, apoptosis, and signal transduction. STK38 is a member of the protein kinase A (PKA)/PKG/PKC family implicated in regulating cell division and morphogenesis in yeast and C. elegans. However, its function remained largely unknown in mammals. In recent years, advances in research on STK38 and the identification of its substrates has led to a better understanding of its function and role in mammals. This review discusses the structure, expression, and regulation of activity as a kinase, its role in the DNA damage response, cross-talk with other signaling pathways, and its application for radio-sensitization.
Collapse
|
43
|
Li X, Chung CI, Yang J, Chaudhuri S, Munster PN, Shu X. ATM-SPARK: A GFP phase separation-based activity reporter of ATM. SCIENCE ADVANCES 2023; 9:eade3760. [PMID: 36857446 PMCID: PMC9977181 DOI: 10.1126/sciadv.ade3760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 01/26/2023] [Indexed: 06/18/2023]
Abstract
The kinase ataxia telangiectasia mutated (ATM) plays a key role in the DNA damage response (DDR). It is thus essential to visualize spatiotemporal dynamics of ATM activity during DDR. Here, we designed a robust ATM activity reporter based on phosphorylation-inducible green fluorescent protein phase separation, dubbed ATM-SPARK (separation of phases-based activity reporter of kinase). Upon ATM activation, it undergoes phase separation via multivalent interactions, forming intensely bright droplets. The reporter visualizes spatiotemporal dynamics of endogenous ATM activity in living cells, and its signal is proportional to the amount of DNA damage. ATM-SPARK also enables high-throughput screening of biological and small-molecule regulators. We identified the protein phosphatase 4 that blocks ATM activity. We also identified BGT226 as a potent ATM inhibitor with a median inhibitory concentration of ~3.8 nanomolars. Furthermore, BGT226 sensitizes cancer cells to the radiomimetic drug neocarzinostatin, suggesting that BGT226 might be combined with radiotherapeutic treatment. ATM-SPARK achieves large dynamic range, bright fluorescence, and simple signal pattern.
Collapse
Affiliation(s)
- Xiaoquan Li
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Chan-I Chung
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - JunJiao Yang
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Sibapriya Chaudhuri
- Division of Hematology and Oncology, University of California, San Francisco, San Francisco, CA, USA
| | - Pamela N. Munster
- Division of Hematology and Oncology, University of California, San Francisco, San Francisco, CA, USA
| | - Xiaokun Shu
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
44
|
Huynh O, Ruis K, Montales K, Michael WM. NBS1 binds directly to TOPBP1 via disparate interactions between the NBS1 BRCT1 domain and the TOPBP1 BRCT1 and BRCT2 domains. DNA Repair (Amst) 2023; 123:103461. [PMID: 36738687 PMCID: PMC9992324 DOI: 10.1016/j.dnarep.2023.103461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 01/20/2023] [Accepted: 01/26/2023] [Indexed: 02/05/2023]
Abstract
The TOPBP1 and NBS1 proteins are key components of DNA repair and DNA-based signaling systems. TOPBP1 is a multi-BRCT domain containing protein that plays important roles in checkpoint signaling, DNA replication, and DNA repair. Likewise, NBS1, which is a component of the MRE11-RAD50-NBS1 (MRN) complex, functions in both checkpoint signaling and DNA repair. NBS1 also contains BRCT domains, and previous works have shown that TOPBP1 and NBS1 interact with one another. In this work we examine the interaction between TOPBP1 and NBS1 in detail. We report that NBS1 uses its BRCT1 domain to interact with TOPBP1's BRCT1 domain and, separately, with TOPBP1's BRCT2 domain. Thus, NBS1 can make two distinct contacts with TOPBP1. We report that recombinant TOPBP1 and NBS1 proteins bind one another in a purified system, showing that the interaction is direct and does not require post-translational modifications. Surprisingly, we also report that intact BRCT domains are not required for these interactions, as truncated versions of the domains are sufficient to confer binding. For TOPBP1, we find that small 24-29 amino acid sequences within BRCT1 or BRCT2 allow binding to NBS1, in a transferrable manner. These data expand our knowledge of how the crucial DNA damage response proteins TOPBP1 and NBS1 interact with one another and set the stage for functional analysis of the two disparate binding sites for NBS1 on TOPBP1.
Collapse
Affiliation(s)
- Oanh Huynh
- Molecular and Computational Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Kenna Ruis
- Molecular and Computational Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Katrina Montales
- Molecular and Computational Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - W Matthew Michael
- Molecular and Computational Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
45
|
Zhou T, Zhang LY, He JZ, Miao ZM, Li YY, Zhang YM, Liu ZW, Zhang SZ, Chen Y, Zhou GC, Liu YQ. Review: Mechanisms and perspective treatment of radioresistance in non-small cell lung cancer. Front Immunol 2023; 14:1133899. [PMID: 36865554 PMCID: PMC9971010 DOI: 10.3389/fimmu.2023.1133899] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 01/31/2023] [Indexed: 02/16/2023] Open
Abstract
Radiotherapy is the major treatment of non-small cell lung cancer (NSCLC). The radioresistance and toxicity are the main obstacles that leading to therapeutic failure and poor prognosis. Oncogenic mutation, cancer stem cells (CSCs), tumor hypoxia, DNA damage repair, epithelial-mesenchymal transition (EMT), and tumor microenvironment (TME) may dominate the occurrence of radioresistance at different stages of radiotherapy. Chemotherapy drugs, targeted drugs, and immune checkpoint inhibitors are combined with radiotherapy to treat NSCLC to improve the efficacy. This article reviews the potential mechanism of radioresistance in NSCLC, and discusses the current drug research to overcome radioresistance and the advantages of Traditional Chinese medicine (TCM) in improving the efficacy and reducing the toxicity of radiotherapy.
Collapse
Affiliation(s)
- Ting Zhou
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China,Experimental & Training Teaching Centers, Gansu University of Chinese Medicine, Lanzhou, China
| | - Li-Ying Zhang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China,College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Jian-Zheng He
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China,College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Zhi-Ming Miao
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yang-Yang Li
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yi-Ming Zhang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
| | - Zhi-Wei Liu
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
| | - Shang-Zu Zhang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yan Chen
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
| | - Gu-Cheng Zhou
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China
| | - Yong-Qi Liu
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou, China,College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China,Key Laboratory of Dunhuang Medicine and Transformation at Provincial and Ministerial Level, Gansu University of Chinese Medicine, Lanzhou, China,*Correspondence: Yong-Qi Liu,
| |
Collapse
|
46
|
Liu W, Zheng M, Zhang R, Jiang Q, Du G, Wu Y, Yang C, Li F, Li W, Wang L, Wu J, Shi L, Li W, Zhang K, Zhou Z, Liu R, Gao Y, Huang X, Fan S, Zhi X, Jiang D, Chen C. RNF126-Mediated MRE11 Ubiquitination Activates the DNA Damage Response and Confers Resistance of Triple-Negative Breast Cancer to Radiotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2203884. [PMID: 36563124 PMCID: PMC9929257 DOI: 10.1002/advs.202203884] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 12/05/2022] [Indexed: 05/27/2023]
Abstract
Triple-negative breast cancer (TNBC) has higher molecular heterogeneity and metastatic potential and the poorest prognosis. Because of limited therapeutics against TNBC, irradiation (IR) therapy is still a common treatment option for patients with lymph nodes or brain metastasis. Thus, it is urgent to develop strategies to enhance the sensitivity of TNBC tumors to low-dose IR. Here, the authors report that E3 ubiquitin ligase Ring finger protein 126 (RNF126) is important for IR-induced ATR-CHK1 pathway activation to enhance DNA damage repair (DDR). Mechanistically, RNF126 physically associates with the MRE11-RAD50-NBS1 (MRN) complex and ubiquitinates MRE11 at K339 and K480 to increase its DNA exonuclease activity, subsequent RPA binding, and ATR phosphorylation, promoting sustained DDR in a homologous recombination repair-prone manner. Accordingly, depletion of RNF126 leads to increased genomic instability and radiation sensitivity in both TNBC cells and mice. Furthermore, it is found that RNF126 expression is induced by IR activating the HER2-AKT-NF-κB pathway and targeting RNF126 expression with dihydroartemisinin significantly improves the sensitivity of TNBC tumors in the brain to IR treatment in vivo. Together, these results reveal that RNF126-mediated MRE11 ubiquitination is a critical regulator of the DDR, which provides a promising target for improving the sensitivity of TNBC to radiotherapy.
Collapse
Affiliation(s)
- Wenjing Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunming650201China
- Kunming College of Life SciencesUniversity of the Chinese Academy of SciencesKunming650204China
- The Third Affiliated HospitalKunming Medical UniversityKunming650118China
| | - Min Zheng
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunming650201China
- Kunming College of Life SciencesUniversity of the Chinese Academy of SciencesKunming650204China
| | - Rou Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunming650201China
| | - Qiuyun Jiang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunming650201China
- Kunming College of Life SciencesUniversity of the Chinese Academy of SciencesKunming650204China
| | - Guangshi Du
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunming650201China
- Kunming College of Life SciencesUniversity of the Chinese Academy of SciencesKunming650204China
| | - Yingying Wu
- Department of the PathologyFirst Affiliated Hospital of Kunming Medical UniversityKunming650032China
| | - Chuanyu Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunming650201China
- Kunming College of Life SciencesUniversity of the Chinese Academy of SciencesKunming650204China
| | - Fubing Li
- Academy of Biomedical EngineeringKunming Medical UniversityKunming650500China
| | - Wei Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunming650201China
- Kunming College of Life SciencesUniversity of the Chinese Academy of SciencesKunming650204China
| | - Luzhen Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunming650201China
- School of Life ScienceUniversity of Science & Technology of ChinaHefei230027China
| | - Jiao Wu
- The Third Affiliated HospitalKunming Medical UniversityKunming650118China
| | - Lei Shi
- Department of Biochemistry and Molecular BiologyTianjin Medical UniversityTianjin300070China
| | - Wenhui Li
- The Third Affiliated HospitalKunming Medical UniversityKunming650118China
| | - Kai Zhang
- Department of Biochemistry and Molecular BiologyTianjin Medical UniversityTianjin300070China
| | - Zhongmei Zhou
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunming650201China
| | - Rong Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunming650201China
- Translational Cancer Research CenterPeking University First HospitalBeijing100034China
| | - Yingzheng Gao
- Department of the Central LaboratorySecond Affiliated Hospital of Kunming Medical UniversityKunming650032China
| | - Xinwei Huang
- Department of the Central LaboratorySecond Affiliated Hospital of Kunming Medical UniversityKunming650032China
| | - Songqing Fan
- Department of Pathologythe Second Xiangya HospitalCentral South UniversityChangsha410000China
| | - Xu Zhi
- Center for Reproductive MedicineDepartment of Obstetrics and GynecologyPeking University Third HospitalBeijing100191China
| | - Dewei Jiang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunming650201China
- Kunming College of Life SciencesUniversity of the Chinese Academy of SciencesKunming650204China
| | - Ceshi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan ProvinceKunming Institute of ZoologyChinese Academy of SciencesKunming650201China
- The Third Affiliated HospitalKunming Medical UniversityKunming650118China
- Academy of Biomedical EngineeringKunming Medical UniversityKunming650500China
| |
Collapse
|
47
|
Role of T Cells in the Pathogenesis of Rheumatoid Arthritis: Focus on Immunometabolism Dysfunctions. Inflammation 2023; 46:88-102. [PMID: 36215002 DOI: 10.1007/s10753-022-01751-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 09/20/2022] [Accepted: 09/30/2022] [Indexed: 11/05/2022]
Abstract
Evidence demonstrated that metabolic-associated T cell abnormalities could be detected in the early stage of RA development. In this context, molecular evaluations have revealed changes in metabolic pathways, leading to the aggressive phenotype of RA T cells. A growing list of genes is downregulated or upregulated in RA T cells, and most of these genes with abnormal expression fall into the category of metabolic pathways. It has been shown that RA T cells shunt glucose towards the pentose phosphate pathway (PPP), which is associated with a high level of nicotinamide adenine dinucleotide phosphate (NADPH) and intermediate molecules. An increased level of NADPH inhibits ATM activation and thereby increases the proliferation capabilities of the RA T cells. Defects in the DNA repair nuclease MRE11A cause failures in repairing mitochondrial DNA, resulting in inhibiting the fatty acid oxidation pathway and further elevated cytoplasmic lipid droplets. Accumulated lipid droplets employ to generate lipid membranes for the cell building program and are also used to form the front-end membrane ruffles that are accomplices with invasive phenotypes of RA T cells. Metabolic pathway involvement in RA pathogenesis expands the pathogenic concept of the disease beyond the common view of autoimmunity triggered by autoantigen recognition. Increased knowledge about metabolic pathways' implications in RA pathogenesis paves the way to understand better the environment/gene interactions and host/microbiota interactions and introduce potential therapeutic approaches. This review summarized emerging data about the roles of T cells in RA pathogenesis with a focus on immunometabolism dysfunctions and how these metabolic alterations can affect the disease process.
Collapse
|
48
|
ATM deficiency aggravates the progression of liver fibrosis induced by carbon tetrachloride in mice. Toxicology 2023; 484:153397. [PMID: 36526012 DOI: 10.1016/j.tox.2022.153397] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022]
Abstract
Ataxia telangiectasia mutated (ATM) is a pivotal sensor during the DNA damage response that slows cell passage through the cell cycle checkpoints to facilitate DNA repair, and liver fibrosis is an irreversible pathological consequence of the sustained wound-healing process, However, the effects of ATM on the development of liver fibrosis are still not fully understood. Therefore, the aim of the study was to investigate the effects and potential mechanisms of ATM on the progression of liver fibrosis. Wild-type and ATM-deficient were administered with carbon tetrachloride (CCl4, 5 ml/kg, i.p.) for 8 weeks to induce liver fibrosis, and the liver tissues and serum were collected for analysis. KU-55933 (10 μM) was used to investigate the effects of ATM blockage on CCl4-induced hepatocyte injury in vitro. The results showed that ATM deficiency aggravated the increased serum transaminase levels and liver MDA, HYP, and 8-OHdG contents compared with the model group (p < 0.05). Sirius red staining showed that ATM deficiency exacerbated liver collagen deposition in vivo, which was associated with the activation of TGF-β1/Smad2 signaling. Furthermore, blocking ATM with KU-55933 exacerbated the production of ROS and DNA damage caused by CCl4 exposure in HepG2 cells, and KU-55933 treatment also reversed the downregulated expression of CDK1 and CDK2 after CCl4 exposure in vitro. Moreover, the loss of ATM perturbed the regulation of the hepatic cell ChK2-CDC25A/C-CDK1/2 cascade and apoptosis in vivo, which was accompanied by increased Ki67-positive and TUNEL-positive cells after chronic CCl4 treatment. In conclusion, our results indicated that ATM might be a critical regulator of liver fibrosis progression, and the underlying mechanisms of exacerbated liver fibrosis development in ATM-deficient mice might be associated with the dysregulation of hepatic cell proliferation and apoptosis.
Collapse
|
49
|
Sensitization of cervical cancer cells to radiation by the cyclin-dependent kinase inhibitor dinaciclib. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 40:68. [PMID: 36586018 DOI: 10.1007/s12032-022-01890-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 11/08/2022] [Indexed: 01/01/2023]
Abstract
Dinaciclib is a selective cyclin-dependent kinase inhibitor, but its radiosensitizing effect remains unclear. The aim of this study is to investigate the radiosensitizing effect of Dinaciclib on cervical cancer cells. Two cervical cancer cell lines, Hela and Siha, were selected, and the IC50 was determined by CCK8. The radiosensitizing effect of Dinaciclib was verified by plate cloning assay, and the G2/M phase arrest and apoptosis of IR cells were verified by flow cytometry. Immunofluorescence assay was used to verify the formation of γH2AX foci following DNA damage. Western blot was performed to detect cell cycle, apoptosis, autophagy, and DNA damage-related pathways. Dinaciclib increased the cell sensitivity to IR. IR induced G2/M phase arrest and apoptosis, and Dinaciclib enhanced this effect. Further, Dinaciclib delayed DNA repair, including non-homologous end joining repair and homologous recombination repair, and reduced the expression of DNA repair proteins Ku80 (SiHa cells), Ku70, and RAD51, as well as the expression of apoptotic marker Bcl-2. The expression of autophagy marker Beclin1 induced tumor cell death and increased the formation of DNA damage marker γH2AX foci. Dinaciclib improves the sensitivity of cervical cancer cells to IR by inducing cell cycle arrest, delaying DNA repair, and increasing apoptosis. However, further research is needed to unravel the complexity of DNA repair pathways.
Collapse
|
50
|
Srikanth P, Chowdhury AR, Low GKM, Saraswathy R, Fujimori A, Banerjee B, Martinez-Lopez W, Hande MP. Oxidative Damage Induced Telomere Mediated Genomic Instability in Cells from Ataxia Telangiectasia Patients. Genome Integr 2022; 13:e20220002. [PMID: 38021281 PMCID: PMC10557037 DOI: 10.14293/genint.13.1.003] [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] [Indexed: 12/01/2023] Open
Abstract
Our cellular genome is susceptible to cytotoxic lesions which include single strand breaks and double strand breaks among other lesions. Ataxia telangiectasia mutated (ATM) protein was one of the first DNA damage sensor proteins to be discovered as being involved in DNA repair and as well as in telomere maintenance. Telomeres help maintain the stability of our chromosomes by protecting the ends from degradation. Cells from ataxia telangiectasia (AT) patients lack ATM and accumulate chromosomal alterations. AT patients display heightened susceptibility to cancer. In this study, cells from AT patients (called as AT -/- and AT +/- cells) were characterized for genome stability status and it was observed that AT -/- cells show considerable telomere attrition. Furthermore, DNA damage and genomic instability were compared between normal (AT +/+ cells) and AT -/- cells exhibiting increased frequencies of spontaneous DNA damage and genomic instability markers. Both AT -/- and AT +/- cells were sensitive to sodium arsenite (1.5 and 3.0 μg/ml) and ionizing radiation-induced (2 Gy, gamma rays) oxidative stress. Interestingly, telomeric fragments were detected in the comet tails as revealed by comet-fluorescence in situ hybridization analysis, suggestive of telomeric instability in AT -/- cells upon exposure to sodium arsenite or radiation. Besides, there was an increase in the number of chromosome alterations in AT -/- cells following arsenite treatment or irradiation. In addition, complex chromosome aberrations were detected by multicolor fluorescence in situ hybridization in AT -/- cells in comparison to AT +/- and normal cells. Telomere attrition and chromosome alterations were detected even at lower doses of sodium arsenite. Peptide nucleic acid - FISH analysis revealed defective chromosome segregation in cells lacking ATM proteins. The data obtained in this study substantiates the role of ATM in telomere stability under oxidative stress.
Collapse
Affiliation(s)
- Prarthana Srikanth
- Department of Physiology, Yong Loo Lin School of Medicine, National
University of Singapore, Singapore
- Department of Biomedical Sciences, School of Biosciences and
Technology, Vellore Institute of Technology, Vellore, India
| | - Amit Roy Chowdhury
- inDNA Center for Research and Innovation in Molecular Diagnostics,
inDNA Life Sciences Private Limited, Bhubaneswar, India
| | - Grace Kah Mun Low
- Department of Physiology, Yong Loo Lin School of Medicine, National
University of Singapore, Singapore
| | - Radha Saraswathy
- Department of Biomedical Sciences, School of Biosciences and
Technology, Vellore Institute of Technology, Vellore, India
| | - Akira Fujimori
- Molecular and Cellular Radiation Biology Group, Department of
Charged Particle Therapy Research Institute for Quantum Medical Science
Chiba, Japan
| | - Birendranath Banerjee
- Department of Physiology, Yong Loo Lin School of Medicine, National
University of Singapore, Singapore
- inDNA Center for Research and Innovation in Molecular Diagnostics,
inDNA Life Sciences Private Limited, Bhubaneswar, India
| | - Wilner Martinez-Lopez
- Department of Biomedical Sciences, School of Biosciences and
Technology, Vellore Institute of Technology, Vellore, India
- Genetics Department and Biodosimetry Services, Instituto de
Investigaciones Biológicas Clemente Estable, Montevideo,
Uruguay
- Associate Unit on Genomic Stability, Faculty of Medicine,
University of the Republic (UdelaR), Montevideo, Uruguay
| | - M. Prakash Hande
- Department of Physiology, Yong Loo Lin School of Medicine, National
University of Singapore, Singapore
- Department of Biomedical Sciences, School of Biosciences and
Technology, Vellore Institute of Technology, Vellore, India
- Department of Applied Zoology, Mangalore University, Mangalore,
India
| |
Collapse
|