1
|
Sharma R, Mishra A, Bhardwaj M, Singh G, Indira Harahap LV, Vanjani S, Pan CH, Nepali K. Medicinal chemistry breakthroughs on ATM, ATR, and DNA-PK inhibitors as prospective cancer therapeutics. J Enzyme Inhib Med Chem 2025; 40:2489720. [PMID: 40256842 PMCID: PMC12013171 DOI: 10.1080/14756366.2025.2489720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 04/22/2025] Open
Abstract
This review discusses the critical roles of Ataxia Telangiectasia Mutated Kinase (ATM), ATM and Rad3-related Kinase (ATR), and DNA-dependent protein kinase (DNA-PK) in the DNA damage response (DDR) and their implications in cancer. Emphasis is placed on the intricate interplay between these kinases, highlighting their collaborative and distinct roles in maintaining genomic integrity and promoting tumour development under dysregulated conditions. Furthermore, the review covers ongoing clinical trials, patent literature, and medicinal chemistry campaigns on ATM/ATR/DNA-PK inhibitors as antitumor agents. Notably, the medicinal chemistry campaigns employed robust drug design strategies and aimed at assembling new structural templates with amplified DDR kinase inhibitory ability, as well as outwitting the pharmacokinetic liabilities of the existing DDR kinase inhibitors. Given the success attained through such endeavours, the clinical pipeline of DNA repair kinase inhibitors is anticipated to be supplemented by a reasonable number of tractable entries (DDR kinase inhibitors) soon.
Collapse
Affiliation(s)
- Ram Sharma
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Anshul Mishra
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Monika Bhardwaj
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Gurpreet Singh
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, India
| | | | - Sakshi Vanjani
- Molecular Medicine, University of South Florida, Tampa, FL, USA
| | - Chun Hsu Pan
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Kunal Nepali
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
2
|
Peng X, Feng J, Yang H, Xia P, Pu F. Nrf2: A key regulator in chemoradiotherapy resistance of osteosarcoma. Genes Dis 2025; 12:101335. [PMID: 40242036 PMCID: PMC12000747 DOI: 10.1016/j.gendis.2024.101335] [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: 11/13/2023] [Revised: 01/24/2024] [Accepted: 04/03/2024] [Indexed: 04/18/2025] Open
Abstract
Osteosarcoma (OS), frequently observed in children and adolescents, is one of the most common primary malignant tumors of the bone known to be associated with a high capacity for invasion and metastasis. The incidence of osteosarcoma in children and adolescents is growing annually, although improvements in survival remain limited. With the clinical application of neoadjuvant chemotherapy, chemotherapy combined with limb-preserving surgery has gained momentum as a major intervention. However, certain patients with OS experience treatment failure owing to chemoradiotherapy resistance or metastasis. Nuclear factor E2-related factor 2 (Nrf2), a key antioxidant factor in organisms, plays a crucial role in maintaining cellular physiological homeostasis; however, its overactivation in cancer cells restricts reactive oxygen species production, promotes DNA repair and drug efflux, and ultimately leads to chemoradiotherapy resistance. Recent studies have also identified the functions of Nrf2 beyond its antioxidative function, including the promotion of proliferation, metastasis, and regulation of metabolism. The current review describes the multiple mechanisms of chemoradiotherapy resistance in OS and the substantial role of Nrf2 in the signaling regulatory network to elucidate the function of Nrf2 in promoting OS chemoradiotherapy resistance and formulating relevant therapeutic strategies.
Collapse
Affiliation(s)
- Xianglin Peng
- Department of Orthopedics, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Department of Orthopedics, Wuhan No.1 Hospital, Wuhan 430022, China
| | - Jing Feng
- Department of Orthopedics, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Department of Orthopedics, Wuhan No.1 Hospital, Wuhan 430022, China
| | - Han Yang
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi 563000, China
- Department of Immunology, Zunyi Medical University, Zunyi 563000, China
| | - Ping Xia
- Department of Orthopedics, Wuhan Fourth Hospital, Wuhan 430030, China
| | - Feifei Pu
- Department of Orthopedics, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Department of Orthopedics, Wuhan No.1 Hospital, Wuhan 430022, China
| |
Collapse
|
3
|
Yadav V, Ahuja T, Kharbanda H, Kumar D, Siddhanta S. Shining Light on DNA Mutations through Machine Learning-Augmented Vibrational Spectroscopy. Anal Chem 2025; 97:12080-12089. [PMID: 40462527 DOI: 10.1021/acs.analchem.5c00183] [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: 06/18/2025]
Abstract
A method to directly predict the number of nucleic acid bases in a single-stranded DNA (ssDNA) or a genomic DNA has been proposed with a combination of Raman spectroscopy and an Artificial Neural Network (ANN) algorithm. In this work, the algorithm was trained by using the Raman spectroscopic signatures from a cohort of 32 ssDNAs. The algorithm could predict the number of bases in an unknown sequence with an R2 value of more than 0.83. Chemical mutation using the hydroxylamine method was performed on a ssDNA and also a genomic herring sperm DNA, and the extent was monitored using optical absorbance measurements. The mutation of bases, such as cytosine, can introduce subtle alterations in the DNA structure, potentially leading to significant biological consequences, including neurodegenerative and epigenetic disorders. Also, during the mutation process, the unstable intermediate can undergo further transformation, converting bases such as cytosine to uracil, thus significantly altering the base-pairing properties of the DNA. A one-to-one correspondence was observed between the experimentally and computationally predicted mutated bases in both the single- and double-stranded DNA (dsDNA), thus opening up avenues for the detection of mutations in a diagnostic setup.
Collapse
Affiliation(s)
- Vikas Yadav
- Department of Chemistry, Indian Institute of Technology Delhi, Hauz Khas New Delhi 110016, India
| | - Tripti Ahuja
- Environmental Monitoring and Intervention Hub, CSIR-IITR, Lucknow 226001, India
| | - Himanshi Kharbanda
- Department of Chemistry, Indian Institute of Technology Delhi, Hauz Khas New Delhi 110016, India
| | - Dinesh Kumar
- Department of Chemistry, Indian Institute of Technology Delhi, Hauz Khas New Delhi 110016, India
| | - Soumik Siddhanta
- Department of Chemistry, Indian Institute of Technology Delhi, Hauz Khas New Delhi 110016, India
| |
Collapse
|
4
|
Cassera E, Ferrari E, Vignati DAL, Capucciati A. The interaction between metals and catecholamines: oxidative stress, DNA damage, and implications for human health. Brain Res Bull 2025; 226:111366. [PMID: 40306586 DOI: 10.1016/j.brainresbull.2025.111366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/07/2025] [Accepted: 04/27/2025] [Indexed: 05/02/2025]
Abstract
The interaction between metals and catecholamines plays a pivotal role in the generation of reactive oxygen species (ROS), leading to oxidative stress and DNA damage. ROS are linked to several diseases, including neurodegenerative disorders such as Parkinson's and Alzheimer's diseases. This review examines how essential metals (iron, copper, zinc, manganese) and a few non-essential metal(loid)s (mercury, chromium, arsenic, aluminum, cadmium, and nickel) contribute to oxidative stress in the presence of catecholamines. In the presence of metals, catecholamines can cause oxidative DNA modification, possibly resulting in cell apoptosis, by taking part in redox reactions and oxidizing to the corresponding aminochrome with simultaneous ROS production. Essential metals are vital for physiological functions, but imbalances in their homeostasis can be harmful. Furthermore, non-essential metals, commonly encountered through environmental or occupational exposure, can exhibit significant toxicity. Previous studies on catecholamine-induced oxidative stress focused on copper and iron, but this review emphasizes the need to investigate other neurotoxic metals and expand existing knowledge on the interactions between metals, catecholamines, and DNA damage. Results from such research could help prioritizing the development of new assessment methods associated with adverse outcome pathways, to reliably predict harmful effects on human health, aiding in the development of therapeutical strategies. The present work will help to shed light on the interplay of metals, catecholamines, and DNA damage in different diseases hopefully fostering new research in this still understudied topic. Future research should investigate the molecular mechanisms through which these metals affect neuronal health and contribute to disease pathogenesis.
Collapse
Affiliation(s)
- Elena Cassera
- Department of Chemistry, University of Pavia, Viale Taramelli 12, Pavia 27100, Italy
| | - Emanuele Ferrari
- National Research Council of Italy, Water Research Institute (CNR-IRSA) Molecular Ecology Group (MEG), Largo Tonolli 50, Verbania 28922, Italy.
| | | | - Andrea Capucciati
- Department of Chemistry, University of Pavia, Viale Taramelli 12, Pavia 27100, Italy; Fondazione Grigioni per il Morbo di Parkinson, Via Gianfranco Zuretti 35, Milano 20125, Italy
| |
Collapse
|
5
|
Liu J, Lu Q, Fan Z, Lin J, He N, Zhang X, Han Z, Zhu T, Wu Z, Xu Y, Wang Y. IncRNA-ZFAS1, an Emerging Gate-Keeper in DNA Damage-Dependent Transcriptional Regulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e12385. [PMID: 40411394 DOI: 10.1002/advs.202412385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 04/15/2025] [Indexed: 05/26/2025]
Abstract
Numerous long noncoding RNAs (lncRNAs) are generated in response to external stimuli, but the scope and functions of such activity are not known. Here, this study provides insight into how the transcription of lncRNAs is connected to DNA damage response by identifying the lncRNA ZFAS1, which is required for cell cycle arrest, transcription regulation, and DNA repair. Mechanistically, ZFAS1 facilitates dynamic changes in hyperphosphorylated forms of the large subunit of RNA polymerase II (RNAPII) around transcription initiation sites by directly targeting the regulated genes. It is shown that extensive transcription shutdown and concomitant stimulated engagement of RNAPII-Ser2P are crucial for repair and cell survival upon genotoxic stress. Finally, ZFAS1 knockout in mice dampened nucleotide excision repair (NER) and led to kidney dysplasia. Overall, the findings extend the understanding of lncRNAs in DNA damage repair (DDR) and imply a protective role of lncRNA against DDR-deficient developmental disorders.
Collapse
Affiliation(s)
- Jiena Liu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, P. R. China
| | - Qing Lu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, P. R. China
| | - Zixuan Fan
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, P. R. China
| | - Jiahui Lin
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, P. R. China
| | - Nan He
- School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Xin Zhang
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, P. R. China
| | - Zhaoya Han
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, P. R. China
| | - Tingting Zhu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, P. R. China
| | - Zhenzhen Wu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, P. R. China
| | - Yingying Xu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, P. R. China
| | - Yuming Wang
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 511436, P. R. China
| |
Collapse
|
6
|
Yang C, Zhong M, Jiao X, Cao H, Qin Y, Xu H, Guo F, Tang Z, Lv T, Guan L, Wang Y, Gao Y, Zhang K. Polysaccharides from Dicliptera chinensis (L.) Juss. Attenuates drug-induced liver injury by phosphorylating AMPK and thus facilitating the entry of FOXO3 into the cell nucleus. Int Immunopharmacol 2025; 155:114633. [PMID: 40239335 DOI: 10.1016/j.intimp.2025.114633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND Isoniazid and rifampicin, frontline tuberculosis drugs, frequently induce drug-induced liver injury (DILI), marked by hepatitis and hepatocyte necrosis. Polysaccharides from Dicliptera chinensis (L.) Juss. (DCP) exhibit anti-inflammatory, antioxidant, and hepatoprotective properties, but their effects on DILI remain unexplored OBJECTIVE: This study investigated DCP therapeutic potential against DILI and elucidated its molecular mechanisms METHODS: In vivo (using C57BL/6 mice) and in vitro (using HepG2 cells) DILI models were established and treated with DCP. Transcriptomics, qRT-PCR, and Western blotting were employed to analyze pathway regulation RESULTS: DCP significantly attenuated hepatocyte apoptosis, inflammation, and oxidative stress in DILI mice. Transcriptomic analysis linked DCP'S effects to the modulation of AMPK-FOXO3, p53, and NF-κB pathways, alongside regulation of antioxidant and cell cycle genes. In HepG2 cells, DCP similarly protected against DILI by enhancing AMPK phosphorylation, which facilitated the FOXO3 nuclear translocation. Both models demonstrated DCP'S suppression of p53 and NF-κB activation, restoration of antioxidant defenses, and correction of cell cycle dysregulation CONCLUSION: DCP mitigates DILI by reducing apoptosis, oxidative stress, and inflammation through activation of the AMPK-FOXO3 pathway, inhibition of p53/NF-κB signaling, and stabilization of the cell cycle. These findings highlight DCP'S potential as a therapeutic agent for DILI prevention and treatment.
Collapse
Affiliation(s)
- Chaoyue Yang
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, China
| | - Mingli Zhong
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, China; Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Key Laboratory of Diabetic Systems Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin 541100, China
| | - Xuefei Jiao
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, China
| | - Houkang Cao
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, China; Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Key Laboratory of Diabetic Systems Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin 541100, China
| | - Yandan Qin
- Guangxi Vocational University of Agriculture,Nanning 530009, China
| | - Hengjie Xu
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, China
| | - Fengyue Guo
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, China
| | - Zixuan Tang
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, China
| | - Tiansong Lv
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, China
| | - Lilin Guan
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, China
| | - Yongwang Wang
- Department of Anesthesiology, Affiliated Hospital of Guilin Medical University, Guilin 541001, China.
| | - Ya Gao
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, China; Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Key Laboratory of Diabetic Systems Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin 541100, China.
| | - Kefeng Zhang
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, China; Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Key Laboratory of Diabetic Systems Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin 541100, China.
| |
Collapse
|
7
|
Xing Z, Cai X, He T, Li P, He J, Qiu Y, Li N, Mi L, Li R, Zhu J, Li Z, Su A, Ye H, Wu W. VCP's nuclear journey: Initiated by interacting with KPNB1 to repair DNA damage. Proc Natl Acad Sci U S A 2025; 122:e2416045122. [PMID: 40339118 DOI: 10.1073/pnas.2416045122] [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: 08/08/2024] [Accepted: 03/27/2025] [Indexed: 05/10/2025] Open
Abstract
DNA damage repair (DDR) is essential for cancer cell survival and treatment resistance, making it a critical target for tumor therapy. The eukaryotic AAA+ adenosine triphosphatase valosin-containing protein (VCP), which is transported from the cytoplasm into the nucleus, plays a critical role in the DDR process. However, the nuclear translocation and molecular mechanism of VCP for DDR remain elusive. Here, we define VCP as a KPNB1 interacting protein through a combination of chemical and immunoprecipitation mass spectrometry approaches. Further biochemical studies elucidate that KPNB1 directly transports VCP into the nucleus. We also identify withaferin A (WA) as a small molecule that can retard VCP nuclear localization via covalent binding to CYS 158 of KPNB1. Further studies verify WA as an effective antitumor drug candidate via blocking VCP nuclear localization to impact on the DDR pathway in vivo. Our findings underly the unclear VCP's role in DDR in a KPNB1-dependent manner and provide an important theoretical basis for developing small-molecule inhibitors targeting this process.
Collapse
Affiliation(s)
- Zhichao Xing
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoying Cai
- Department of Biotherapy, Cancer Center and State Key laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ting He
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Peiheng Li
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jun He
- Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu 610041, China
| | - Yuxuan Qiu
- Department of Ultrasound, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310030, China
| | - Na Li
- Department of Biotherapy, Cancer Center and State Key laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Li Mi
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ruixi Li
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jingqiang Zhu
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhihui Li
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Anping Su
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Haoyu Ye
- Department of Biotherapy, Cancer Center and State Key laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wenshuang Wu
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
8
|
Toprani SM, Mordukhovich I, McNeely E, Nagel ZD. Suppressed DNA repair capacity in flight attendants after air travel. Sci Rep 2025; 15:16513. [PMID: 40360675 PMCID: PMC12075667 DOI: 10.1038/s41598-025-98934-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
Elevated cancer risk and compromised reproductive health have been well documented in flight attendants (FA), but the etiology remains unknown. Many studies using cell and animal models suggest that air travel related exposures might plausibly explain the adverse health outcomes observed in flight crew, but our understanding of the underlying biological mechanisms is incomplete. During air travel, FA are constantly exposed to complex mixtures of mutagens in the flight cabin that may contribute to genomic instability by inducing DNA damage and interfering with DNA repair. Defects in DNA repair capacity (DRC) have been associated with risk of cancer and other diseases. To explore our hypothesis that alterations in DNA damage and repair in FA are related to flight travel, we conducted a pilot study of FA's DNA damage and assess global DNA repair efficiency pre and post flight. We collected venous blood samples from nine FA before and after flight. Differential blood cell counts were carried out to assess immune responses and functional assays were performed to assess the DNA damage response. The CometChip assay was employed to quantify baseline DNA damage and repair kinetics for DNA damage induced by X-rays. Fluorescence multiplex based host cell reactivation (FM-HCR) assays were utilized to assess DRC in five major DNA repair pathways. Our findings revealed a significant increase in lymphocyte counts as well as diminished repair of ionizing radiation induced DNA damage and excision of 8oxoG:C lesions in after flight samples. Our results illustrate the potential for using biological samples to identify molecular mechanisms that may implicate impaired genomic stability and altered immune responses in the etiology of excess cancer in FAs.
Collapse
Affiliation(s)
- Sneh M Toprani
- John B. Little Center of Radiation Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Irina Mordukhovich
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- SHINE, Human Flourishing Program Institute for Quantitative Science, Harvard University, Cambridge, MA, USA
| | - Eileen McNeely
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- SHINE, Human Flourishing Program Institute for Quantitative Science, Harvard University, Cambridge, MA, USA
| | - Zachary D Nagel
- John B. Little Center of Radiation Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
9
|
Kasetthamrongrat P, Phumsankhot R, Duangya A, Watcharawipha A, Nobnop W, Autsavapromporn N. Comparison of Tumor Cell Responses to Different Radiotherapy Techniques: Three-Dimensional Conformal Radiotherapy (3D-CRT), Volumetric Modulated Arc Therapy (VMAT), and Helical Tomotherapy (HT). BIOLOGY 2025; 14:529. [PMID: 40427718 PMCID: PMC12109413 DOI: 10.3390/biology14050529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2025] [Revised: 05/05/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025]
Abstract
Currently, advanced RT techniques such as VMAT and HT are being developed to optimize tumor coverage while minimizing radiation exposure to the surrounding organs that are at risk. Despite their growing clinical use, comparative studies evaluating the dosimetric and radiobiological effects of these modalities remain limited. In this study, A549, HeLa, and HepG2 cells were exposed to a single 2 Gy dose, using three RT techniques (3D-CRT, dual arc VMAT, and HT). Treatment plans were generated using a water phantom to ensure consistent target coverage and comparable dosimetric parameters across the techniques. Multiple radiobiological endpoints were assessed to evaluate the cellular responses. Although all three techniques yielded similar dosimetric parameters without statistically significant differences, the biological responses varied among the cell lines. Notably, VMAT and HT demonstrated superior tumor cell suppression compared to 3D-CRT. This was likely due to their enhanced dose conformity and modulation precision, which potentially led to improved tumor cell killing. These findings highlight the importance of integrating radiobiological assessments with physical dose metrics to inform the clinical application of advanced RT technologies. However, this study had several limitations. The use of a single radiation dose limited its clinical relevance, and the immediate post-irradiation assessments may not have captured delayed biological responses. Additionally, the small number of replicates may have reduced the study's statistical power. Future studies incorporating dose fractionation schemes, time course analyses, and larger sample sizes are warranted to better simulate clinical conditions and further elucidate the radiobiological effects of advanced RT techniques.
Collapse
Affiliation(s)
- Phanwadee Kasetthamrongrat
- Medical Physics Program, Department of Radiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (P.K.); (R.P.)
| | - Rinwarat Phumsankhot
- Medical Physics Program, Department of Radiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (P.K.); (R.P.)
| | - Aphidet Duangya
- Division of Radiation Oncology, Department of Radiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (A.D.); (A.W.); (W.N.)
| | - Anirut Watcharawipha
- Division of Radiation Oncology, Department of Radiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (A.D.); (A.W.); (W.N.)
| | - Wannapha Nobnop
- Division of Radiation Oncology, Department of Radiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (A.D.); (A.W.); (W.N.)
| | - Narongchai Autsavapromporn
- Division of Radiation Oncology, Department of Radiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (A.D.); (A.W.); (W.N.)
| |
Collapse
|
10
|
Bai D, Cao Z, Attada N, Song J, Zhu C. Single-cell parallel analysis of DNA damage and transcriptome reveals selective genome vulnerability. Nat Methods 2025; 22:962-972. [PMID: 40128288 DOI: 10.1038/s41592-025-02632-3] [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: 06/20/2024] [Accepted: 02/18/2025] [Indexed: 03/26/2025]
Abstract
Maintenance of genome integrity is paramount to molecular programs in multicellular organisms. Throughout the lifespan, various endogenous and environmental factors pose persistent threats to the genome, which can result in DNA damage. Understanding the functional consequences of DNA damage requires investigating their preferred genomic distributions and influences on gene regulatory programs. However, such analysis is hindered by both the complex cell-type compositions within organs and the high background levels due to the stochasticity of damage formation. To address these challenges, we developed Paired-Damage-seq for joint analysis of oxidative and single-stranded DNA damage with gene expression in single cells. We applied this approach to cultured HeLa cells and the mouse brain as a proof of concept. Our results indicated the associations between damage formation and epigenetic changes. The distribution of oxidative DNA damage hotspots exhibits cell-type-specific patterns; this selective genome vulnerability, in turn, can predict cell types and dysregulated molecular programs that contribute to disease risks.
Collapse
Affiliation(s)
| | - Zhenkun Cao
- Physiology, Biophysics and Systems Biology Graduate Program, Weill Cornell Medicine, New York, NY, USA
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Jinghui Song
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Chenxu Zhu
- New York Genome Center, New York, NY, USA.
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
11
|
Ghediri S, Sarma PAP, Saravanan V, Abbadie C, Blossey R, Cleri F. Mechanisms of DNA Damage Recognition by UDG and PARP1 in the Nucleosome. Biomolecules 2025; 15:649. [PMID: 40427542 PMCID: PMC12108792 DOI: 10.3390/biom15050649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Revised: 04/23/2025] [Accepted: 04/26/2025] [Indexed: 05/29/2025] Open
Abstract
The DNA base-excision repair (BER) pathway shares the second part of its enzymatic chain with the single-strand break (SSB) repair pathway. BER is initiated by a glycosylase, such as UDG, while SSBR is initiated by the multifunctional enzyme PARP1. The very early steps in the identification of the DNA damage are crucial to the correct initiation of the repair chains, and become even more complex when considering the realistic environment of damage to the DNA in the nucleosome. We performed molecular dynamics computer simulations of the interaction between the glycosylase UDG and a mutated uracil (as could result from oxidative deamination of cytosine), and between the Zn1-Zn2 fragment of PARP1 and a simulated SSB. The model system is a whole nucleosome in which DNA damage is inserted at various typical positions along the 145-bp sequence. It is shown that damage recognition by the enzymes requires very strict conditions, unlikely to be matched by pure random search along the DNA. We propose that mechanical deformation of the DNA around the defective sites may help signaling the presence of the defect, accelerating the search process.
Collapse
Affiliation(s)
- Safwen Ghediri
- Université de Lille, Institut d’Electronique Microelectronique et Nanotechnologie (IEMN CNRS, UMR8520) and Département de Physique, F59652 Villeneuve d’Ascq, France; (S.G.); (P.A.P.S.)
- Université de Lille, Unité de Glycobiologie Structurale et Fonctionnelle (UGSF CNRS UMR8576), F59000 Lille, France; (V.S.); (R.B.)
| | - Parvathy A. P. Sarma
- Université de Lille, Institut d’Electronique Microelectronique et Nanotechnologie (IEMN CNRS, UMR8520) and Département de Physique, F59652 Villeneuve d’Ascq, France; (S.G.); (P.A.P.S.)
- Université de Lille, CNRS UMR9020 and Inserm U1277-CANTHER-Cancer Heterogeneity, Plasticity and Resistance to Therapies, F59000 Lille, France;
| | - Vinnarasi Saravanan
- Université de Lille, Unité de Glycobiologie Structurale et Fonctionnelle (UGSF CNRS UMR8576), F59000 Lille, France; (V.S.); (R.B.)
| | - Corinne Abbadie
- Université de Lille, CNRS UMR9020 and Inserm U1277-CANTHER-Cancer Heterogeneity, Plasticity and Resistance to Therapies, F59000 Lille, France;
| | - Ralf Blossey
- Université de Lille, Unité de Glycobiologie Structurale et Fonctionnelle (UGSF CNRS UMR8576), F59000 Lille, France; (V.S.); (R.B.)
| | - Fabrizio Cleri
- Université de Lille, Institut d’Electronique Microelectronique et Nanotechnologie (IEMN CNRS, UMR8520) and Département de Physique, F59652 Villeneuve d’Ascq, France; (S.G.); (P.A.P.S.)
- Laboratory for Integrated Micro Mechatronics, LIMMS CNRS IRL2820 and University of Tokyo, Komaba, Meguro-ku, Tokyo 153-8505, Japan
| |
Collapse
|
12
|
Ma YT, Li C, Shen Y, You WH, Han MX, Mu YF, Han FJ. Mechanisms of the JNK/p38 MAPK signaling pathway in drug resistance in ovarian cancer. Front Oncol 2025; 15:1533352. [PMID: 40352594 PMCID: PMC12063130 DOI: 10.3389/fonc.2025.1533352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 04/02/2025] [Indexed: 05/14/2025] Open
Abstract
Ovarian cancer (OC) is the most lethal malignancy in the female reproductive system, and chemotherapy drug resistance is the main cause of treatment failure. The Mitogen-Activated Protein Kinases (MAPK) pathway plays a pivotal role in regulating cell proliferation, migration, and invasive capacity in response to extracellular stimuli. This review focuses on the mechanisms and therapeutic strategies related to the JNK/p38 MAPK signaling pathway in OC resistance. The JNK/p38 MAPK pathway plays a dual role in OC chemoresistance. This review examines its role in mediating OC treatment resistance by exploring the mechanisms of action of the JNK/p38 MAPK signaling pathway, particularly its involvement in several key biological processes, including apoptosis, autophagy, DNA damage response, the tumor microenvironment (TME), and drug efflux. Additionally, the review investigates the timing of activation of this pathway and its crosstalk with other signaling pathways such as PI3K/AKT and NF-κB. Targeting JNK/p38 MAPK signaling has shown promise in reversing chemoresistance, with several inhibitors and natural compounds demonstrating potential in preclinical studies. Regulating JNK/p38 MAPK may transform what was once a terminal obstacle into a manageable challenge for OC patients with chemotherapy resistance, ultimately improving survival and quality of life.
Collapse
Affiliation(s)
- Yu-Ting Ma
- Department of Obstetrics and Gynecology, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Chan Li
- Department of Obstetrics and Gynecology, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Ying Shen
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Wan-Hui You
- Department of Obstetrics and Gynecology, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Ming-Xuan Han
- Department of Obstetrics and Gynecology, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Yi-Fan Mu
- Department of Obstetrics and Gynecology, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Feng-Juan Han
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| |
Collapse
|
13
|
Sobkowiak K, Kohzaki M, Böhm R, Mailler J, Huber F, Emamzadah S, Tropia L, Hiller S, Halazonetis TD. REV7 functions with REV3 as a checkpoint protein delaying mitotic entry until DNA replication is completed. Cell Rep 2025; 44:115431. [PMID: 40106439 DOI: 10.1016/j.celrep.2025.115431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 12/18/2024] [Accepted: 02/24/2025] [Indexed: 03/22/2025] Open
Abstract
REV7, also named MAD2B or MAD2L2, is a subunit of the DNA translesion polymerase zeta and also part of the 53BP1-shieldin complex, which is present at sites of DNA double-strand breaks. REV7 has high sequence similarity to the MAD2 spindle assembly checkpoint protein, prompting us to examine whether REV7 has a checkpoint function. We observed that, in chicken and human cells exposed to agents that induce DNA replication stress, REV7 inhibits mitotic entry; this effect is most evident when the canonical DNA replication stress checkpoint, mediated by ATR, is inhibited. Similar to MAD2, REV7 undergoes conformational changes upon ligand binding, and its checkpoint function depends on its ability to homodimerize and bind its ligands. Notably, even in unchallenged cells, deletion of the REV7 gene leads to premature mitotic entry, raising the possibility that the REV7 checkpoint monitors ongoing DNA replication.
Collapse
Affiliation(s)
- Katarzyna Sobkowiak
- Department of Molecular and Cellular Biology, University of Geneva, 1205 Geneva, Switzerland
| | - Masaoki Kohzaki
- Department of Molecular and Cellular Biology, University of Geneva, 1205 Geneva, Switzerland.
| | - Raphael Böhm
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Jonathan Mailler
- Department of Molecular and Cellular Biology, University of Geneva, 1205 Geneva, Switzerland
| | - Florian Huber
- Department of Molecular and Cellular Biology, University of Geneva, 1205 Geneva, Switzerland
| | - Soheila Emamzadah
- Department of Molecular and Cellular Biology, University of Geneva, 1205 Geneva, Switzerland
| | - Laurence Tropia
- Department of Molecular and Cellular Biology, University of Geneva, 1205 Geneva, Switzerland
| | | | - Thanos D Halazonetis
- Department of Molecular and Cellular Biology, University of Geneva, 1205 Geneva, Switzerland.
| |
Collapse
|
14
|
Liu X, Wang S, Lv H, Chen E, Yan L, Yu J. Advances in the relationship of immune checkpoint inhibitors and DNA damage repair. Curr Res Transl Med 2025; 73:103494. [PMID: 39824061 DOI: 10.1016/j.retram.2025.103494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 01/08/2025] [Accepted: 01/11/2025] [Indexed: 01/20/2025]
Abstract
Cancer immunotherapy, alongside surgery, radiation therapy, and chemotherapy, has emerged as a key treatment modality. Immune checkpoint inhibitors (ICIs) represent a promising immunotherapy that plays a critical role in the management of various solid tumors. However, the limited efficacy of ICI monotherapy and the development of primary or secondary resistance to combination therapy remain a challenge. Consequently, identifying molecular markers for predicting ICI efficacy has become an area of active clinical research. Notably, the correlation between DNA damage repair (DDR) mechanisms and the effectiveness of ICI treatment has been established. This review outlines the two primary pathways of DDR, namely, the homologous recombination repair pathway and the mismatch repair pathway. The relationship between these key genes and ICIs has been discussed and the potential of these genes as molecular markers for predicting ICI efficacy summarized.
Collapse
Affiliation(s)
- Xiaolin Liu
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Shan Wang
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Hongwei Lv
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Enli Chen
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Li Yan
- School of Humanities, Beijing University of Chinese Medicine, Beijing, PR China
| | - Jing Yu
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China.
| |
Collapse
|
15
|
Santoro A, Angelico G, Travaglino A, Inzani F, Spadola S, Pettinato A, Mazzucchelli M, Bragantini E, Maccio L, Zannoni GF. The multiple facets of ovarian high grade serous carcinoma: A review on morphological, immunohistochemical and molecular features. Crit Rev Oncol Hematol 2025; 208:104603. [PMID: 39732305 DOI: 10.1016/j.critrevonc.2024.104603] [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: 09/15/2024] [Revised: 12/06/2024] [Accepted: 12/20/2024] [Indexed: 12/30/2024] Open
Abstract
High-grade serous ovarian carcinoma (HGSOC) is the most aggressive subtype of epithelial ovarian cancer and a leading cause of mortality among gynecologic malignancies. This review aims to comprehensively analyze the morphological, immunohistochemical, and molecular features of HGSOC, highlighting its pathogenesis and identifying biomarkers with diagnostic, prognostic, and therapeutic significance. Special emphasis is placed on the role of tumor microenvironment (TME) and genomic instability in shaping the tumor's behavior and therapeutic vulnerabilities. Key advancements, such as the identification of TP53 and BRCA mutations, the classification of homologous recombination repair (HRR) deficiencies, and the clinical implications of biomarkers like folate receptor alpha (FRα) and PD-L1 are discussed. These findings reveal actionable insights into targeted therapies, including immune checkpoint inhibitors and PARP inhibitors, which hold promise for improving outcomes in HGSOC. This synthesis of knowledge aims to bridge gaps in understanding HGSOC's multifaceted biology, enhance clinical decision-making, and foster the development of precision therapies.
Collapse
MESH Headings
- Humans
- Female
- Ovarian Neoplasms/pathology
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/metabolism
- Ovarian Neoplasms/diagnosis
- Ovarian Neoplasms/therapy
- Cystadenocarcinoma, Serous/pathology
- Cystadenocarcinoma, Serous/genetics
- Cystadenocarcinoma, Serous/metabolism
- Cystadenocarcinoma, Serous/diagnosis
- Cystadenocarcinoma, Serous/therapy
- Biomarkers, Tumor/metabolism
- Biomarkers, Tumor/genetics
- Tumor Microenvironment
- Neoplasm Grading
- Immunohistochemistry
- Prognosis
- Carcinoma, Ovarian Epithelial/pathology
- Carcinoma, Ovarian Epithelial/genetics
- Carcinoma, Ovarian Epithelial/metabolism
- Mutation
Collapse
Affiliation(s)
- Angela Santoro
- Pathology Institute, Catholic University of Sacred Heart, Rome 00168, Italy; Pathology Unit, Department of Woman and Child's Health and Public Health Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome 00168, Italy
| | - Giuseppe Angelico
- Department of Medicine and Surgery, Kore University of Enna, Enna 94100, Italy
| | - Antonio Travaglino
- Pathology Unit, Department of Medicine and Technological Innovation, University of Insubria, Varese, Italy
| | - Frediano Inzani
- Anatomic Pathology Unit, Department of Molecular Medicine, University of Pavia and Fondazione IRCCS San Matteo Hospital, Pavia 27100, Italy
| | - Saveria Spadola
- Department of Medicine and Surgery, Kore University of Enna, Enna 94100, Italy
| | - Angela Pettinato
- Department of Pathological Anatomy, A.O.E. Cannizzaro, Via Messina, 829, Catania 95126, Italy
| | - Manuel Mazzucchelli
- Department of Medical and Surgical Sciences and Advanced Technologies "G.F. Ingrassia", Anatomic Pathology, University of Catania, Catania, Italy
| | - Emma Bragantini
- Unit of Surgical Pathology, Santa Chiara Hospital, APSS, Trento, Italy
| | - Livia Maccio
- Unit of Surgical Pathology, Santa Chiara Hospital, APSS, Trento, Italy
| | - Gian Franco Zannoni
- Pathology Institute, Catholic University of Sacred Heart, Rome 00168, Italy; Pathology Unit, Department of Woman and Child's Health and Public Health Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome 00168, Italy.
| |
Collapse
|
16
|
Yang Y, Wang P, Zhou K, Zhang W, Liu S, Ouyang J, Bai M, Ding G, Huang S, Jia Z, Zhang A. HUWE1-Mediated Degradation of MUTYH Facilitates DNA Damage and Mitochondrial Dysfunction to Promote Acute Kidney Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412250. [PMID: 39921445 PMCID: PMC11967787 DOI: 10.1002/advs.202412250] [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: 10/02/2024] [Revised: 01/26/2025] [Indexed: 02/10/2025]
Abstract
The role of MUTYH, a DNA repair glycosylase in the pathogenesis of acute kidney injury (AKI) is unclear. In this study, it is found that MUTYH protein levels are significantly decreased in the kidneys of cisplatin- or folic acid (FA)-induced mouse AKI models and patients with AKI. MUTYH deficiency aggravates renal dysfunction and tubular injury following cisplatin and FA treatment, along with the accumulation of 7, 8-dihydro-8-oxoguanine (8-oxoG) and impairs mitochondrial function. Importantly, the overexpression of type 2 MUTYH (nuclear) significantly ameliorates cisplatin-induced apoptosis, oxidative stress, mitochondrial dysfunction, and DNA damage in vivo and in vitro. In contrast, overexpression of type 1 MUTYH (mitochondrial) shows a marginal effect against cisplatin-induced injury, indicating the chief role of type 2 MUTYH in antagonizing AKI. Interestingly, the results also indicate that the upregulation of the E3 ligase HUWE1 causes the ubiquitination and degradation of MUTYH in tubular epithelial cells. HUWE1 knockout or treatment with the HUWE1 inhibitor BI8622 significantly protect against cisplatin-induced AKI. Taken together, these results suggest that the ubiquitin E3 ligase HUWE1-mediates ubiquitination and degradation of MUTYH can aggravate DNA damage in the nucleus and mitochondria and promote AKI. Targeting the HUWE1/MUTYH pathway may be a potential strategy for AKI treatment.
Collapse
Affiliation(s)
- Yunwen Yang
- Department of NephrologyChildren's Hospital of Nanjing Medical University72 Guangzhou RoadNanjing210008P. R. China
- Nanjing Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing210008P. R. China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029P. R. China
| | - Peipei Wang
- Department of NephrologyChildren's Hospital of Nanjing Medical University72 Guangzhou RoadNanjing210008P. R. China
- Nanjing Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing210008P. R. China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029P. R. China
| | - Kaiqian Zhou
- Department of NephrologyChildren's Hospital of Nanjing Medical University72 Guangzhou RoadNanjing210008P. R. China
- Nanjing Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing210008P. R. China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029P. R. China
| | - Wen Zhang
- Department of NephrologyAffiliated Hospital of Integrated Traditional Chinese and Western MedicineNanjing University of Chinese MedicineNanjing210028P. R. China
| | - Suwen Liu
- Department of PediatricsShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinan250021P. R. China
| | - Jing Ouyang
- Department of NephrologyChildren's Hospital of Nanjing Medical University72 Guangzhou RoadNanjing210008P. R. China
- Nanjing Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing210008P. R. China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029P. R. China
| | - Mi Bai
- Department of NephrologyChildren's Hospital of Nanjing Medical University72 Guangzhou RoadNanjing210008P. R. China
- Nanjing Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing210008P. R. China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029P. R. China
| | - Guixia Ding
- Department of NephrologyChildren's Hospital of Nanjing Medical University72 Guangzhou RoadNanjing210008P. R. China
- Nanjing Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing210008P. R. China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029P. R. China
| | - Songming Huang
- Department of NephrologyChildren's Hospital of Nanjing Medical University72 Guangzhou RoadNanjing210008P. R. China
- Nanjing Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing210008P. R. China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029P. R. China
| | - Zhanjun Jia
- Department of NephrologyChildren's Hospital of Nanjing Medical University72 Guangzhou RoadNanjing210008P. R. China
- Nanjing Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing210008P. R. China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029P. R. China
| | - Aihua Zhang
- Department of NephrologyChildren's Hospital of Nanjing Medical University72 Guangzhou RoadNanjing210008P. R. China
- Nanjing Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjing210008P. R. China
- Jiangsu Key Laboratory of Early Development and Chronic Diseases Prevention in ChildrenNanjing Medical UniversityNanjing210029P. R. China
| |
Collapse
|
17
|
Zhang XL, Yue HW, Liu YJ, Wang JY, Duan HT, Liu YH, Jiang LL, Hu HY. Designer polyQ fusion proteins sequester USP7/HDM2 for modulating P53 functionality. iScience 2025; 28:112025. [PMID: 40104064 PMCID: PMC11914518 DOI: 10.1016/j.isci.2025.112025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 11/05/2024] [Accepted: 02/11/2025] [Indexed: 03/20/2025] Open
Abstract
Overexpression of USP7 and HDM2 inactivates P53 signaling in tumor cells and facilitates their progression, but suppression of these targets by conventional strategies to reactivate P53 function remains a challenge. We applied polyQ sequences and target-interacting peptides to engineer polyQ fusion proteins that specifically sequester the targets, hence depleting their availabilities and modulating the P53 functionality. We have revealed that the designer fusion Atx793Q-N172-IRF (IRF sequence: SPGEGPSGTG) sequesters USP7 and/or HDM2 into aggregates and thereby increases the P53 level, but it depends on the IRF repeats fused, suggesting that depletion of the USP7 availability plays a dual role in controlling P53 stability. Direct sequestration of HDM2 by Atx793Q-N172-PMI (PMI: TSFAEYWNLLSP) remarkably reduces the protein level of soluble HDM2 and hence increases the P53 level, which consequently up-regulates expression of the downstream genes. The polyQ-fusion strategy is feasible to modulate the P53 stability and functionality, furnishing a therapeutic potential for cancers.
Collapse
Affiliation(s)
- Xiang-Le Zhang
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Hong-Wei Yue
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Ya-Jun Liu
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Jian-Yang Wang
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Heng-Tong Duan
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Yin-Hu Liu
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Lei-Lei Jiang
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, P.R. China
| | - Hong-Yu Hu
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, P.R. China
| |
Collapse
|
18
|
Jin Y, Xue B, Zhou X. Protein Biomarkers of DNA Damage in Yeast Cells for Genotoxicity Screening. ENVIRONMENT & HEALTH (WASHINGTON, D.C.) 2025; 3:250-258. [PMID: 40144325 PMCID: PMC11934195 DOI: 10.1021/envhealth.4c00160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/27/2024] [Accepted: 11/01/2024] [Indexed: 03/28/2025]
Abstract
Providing an unbiased and comprehensive view of the DNA damage response in cells is critical in genotoxicity screening to identify substances that cause diverse types of DNA damage. Considering that S. cerevisiae is one of the most well-characterized model organisms in molecular and cellular biology, we created a map of the DNA damage response network containing the reported signaling pathways in yeast cells programmed to constitutively respond to DNA damage. A collection of GFP-fused S. cerevisiae yeast strains treated with typical genotoxic agents illuminated the cellular response to DNA damage, thereby identifying 15 protein biomarkers encompassing all eight documented DNA damage response pathways. Three statistical and one deep learning models were proposed to interpret the quantitative molecular toxicity end point, i.e. protein effect level index (PELI), by introducing weights of 15 biomarkers in genotoxicity assessment. The method based on standard deviation exhibited the best performance, with an R 2 of 0.916 compared to the SOS/umu test and an R 2 of 0.989 compared to the comet assay. The GFP-fused yeast-based proteomic assay has minute-level resolution of pathway activation data. It provides a concise alternative for fast, efficient, and mechanistic genotoxicity screening for various environmental and health applications.
Collapse
Affiliation(s)
- Yushi Jin
- Center for Sensor Technology of Environment
and Health, School of Environment, Tsinghua
University, Beijing 100084, China
| | - Boyuan Xue
- Center for Sensor Technology of Environment
and Health, School of Environment, Tsinghua
University, Beijing 100084, China
| | - Xiaohong Zhou
- Center for Sensor Technology of Environment
and Health, School of Environment, Tsinghua
University, Beijing 100084, China
| |
Collapse
|
19
|
Solomon AD, Gouttia OG, Wang L, Zhu S, Wang F, Li Y, Paydar M, Bessho T, Kwok BH, Peng A. γ-tubulin mediates DNA double-strand break repair. J Cell Sci 2025; 138:jcs262255. [PMID: 40135584 PMCID: PMC12050090 DOI: 10.1242/jcs.262255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 12/20/2024] [Indexed: 03/27/2025] Open
Abstract
Double-strand breaks (DSBs) in DNA pose a critical threat to genomic integrity, potentially leading to the onset and progression of various diseases, including cancer. Cellular responses to such lesions entail sophisticated repair mechanisms primarily mediated by non-homologous end joining (NHEJ) and homologous recombination (HR). Interestingly, the efficient recruitment of repair proteins and completion of DSB repair likely involve complex, inter-organelle communication and coordination of cellular components. In this study, we report a role of γ-tubulin in DSB repair. γ-tubulin is a major microtubule nucleation factor governing microtubule dynamics. We show that γ-tubulin is recruited to the site of DNA damage and is required for efficient DSB repair via both NHEJ and HR. Suppression of γ-tubulin impedes DNA repair and exacerbates DNA damage accumulation. Furthermore, γ-tubulin mediates the mobilization and formation of DNA damage foci, which serve as repair centers, thereby facilitating the recruitment of HR and NHEJ repair proteins on damaged chromatin. Finally, pharmacological inhibition of γ-tubulin enhances the cytotoxic effect of DNA-damaging agents, consistent with the DNA repair function of γ-tubulin, and underscoring the potential of its therapeutic intervention in cancer therapy.
Collapse
Affiliation(s)
- Abhishikt David Solomon
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Odjo G. Gouttia
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ling Wang
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Songli Zhu
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE 68583, USA
| | - Feifei Wang
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE 68583, USA
| | - Yanqui Li
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE 68583, USA
| | - Mohammadjavad Paydar
- Institute for Research in Immunology and Cancer (IRIC), Département de médecine, Université de Montréal, Montréal H3C 3J7, Canada
| | - Tadayoshi Bessho
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Benjamin H. Kwok
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE 68583, USA
| | - Aimin Peng
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
20
|
Zhou Y, Geng S, Tang RC, Yu H, Zhang A, Bai Y, Zhang J. Clinical and functional significance of SPATA2 in cancer particularly in LIHC. Sci Rep 2025; 15:8392. [PMID: 40069269 PMCID: PMC11897323 DOI: 10.1038/s41598-025-91386-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 02/20/2025] [Indexed: 03/15/2025] Open
Abstract
Spermatogenesis-associated protein 2 (SPATA2) is primarily named for its important role in spermatogenesis. Its function in tumorigenesis remains elusive. Here, we used various bioinformatic tools to systematically analyze the expression patterns of SPATA2 in cancers, the correlation of SPATA2 expression with clinical parameters, genetic variation, methylation, phosphorylation, immune infiltration and immune therapy. SPATA2 is significantly upregulated in multiple cancers and its expression was associated with tumor stage, grade and serve as a potential prognostic marker in LIHC. Notably, SPATA2 was also linked to immune suppression, exhibiting positive correlations with immune checkpoint genes and immune suppressive cells such as regulatory T cells and MDSCs. Furthermore, SPATA2 interacted and co-expressed with proteins involved in DNA repair mechanisms, indicating its potential role in maintaining genomic stability. Finally, we conducted biological experiments to investigate the role of SPATA2 in LIHC. SPATA2 knockdown enhances the migration and proliferation capabilities of Hep-G2 and HuH7 cell lines. These findings underscore the significance of SPATA2 in cancer biology, suggests its role in both the tumor microenvironment and the tumor cell level and its potential as a prognostic marker and therapeutic target in oncology, particularly in LIHC.
Collapse
Affiliation(s)
- Yunxuan Zhou
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-Related Diseases, Peking University, Beijing, China
| | - Shijin Geng
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-Related Diseases, Peking University, Beijing, China
| | - Rong-Chun Tang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-Related Diseases, Peking University, Beijing, China
| | - Hengxiang Yu
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-Related Diseases, Peking University, Beijing, China
| | - Ao Zhang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-Related Diseases, Peking University, Beijing, China
| | - Yuekui Bai
- General Surgery, Haidian Hospital, Beijing, China.
| | - Jun Zhang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-Related Diseases, Peking University, Beijing, China.
| |
Collapse
|
21
|
Gong P, Guo Z, Wang S, Gao S, Cao Q. Histone Phosphorylation in DNA Damage Response. Int J Mol Sci 2025; 26:2405. [PMID: 40141048 PMCID: PMC11941871 DOI: 10.3390/ijms26062405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/01/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
The DNA damage response (DDR) is crucial for maintaining genomic stability and preventing the accumulation of mutations that can lead to various diseases, including cancer. The DDR is a complex cellular regulatory network that involves DNA damage sensing, signal transduction, repair, and cell cycle arrest. Modifications in histone phosphorylation play important roles in these processes, facilitating DNA repair factor recruitment, damage signal transduction, chromatin remodeling, and cell cycle regulation. The precise regulation of histone phosphorylation is critical for the effective repair of DNA damage, genomic integrity maintenance, and the prevention of diseases such as cancer, where DNA repair mechanisms are often compromised. Thus, understanding histone phosphorylation in the DDR provides insights into DDR mechanisms and offers potential therapeutic targets for diseases associated with genomic instability, including cancers.
Collapse
Affiliation(s)
- Ping Gong
- Hunan Institute of Microbiology, Changsha 410009, China; (Z.G.); (S.W.); (S.G.)
| | - Zhaohui Guo
- Hunan Institute of Microbiology, Changsha 410009, China; (Z.G.); (S.W.); (S.G.)
| | - Shengping Wang
- Hunan Institute of Microbiology, Changsha 410009, China; (Z.G.); (S.W.); (S.G.)
| | - Shufeng Gao
- Hunan Institute of Microbiology, Changsha 410009, China; (Z.G.); (S.W.); (S.G.)
| | - Qinhong Cao
- College of Biological Sciences, China Agricultural University, No.2 Yuan-Ming-Yuan West Road, Beijing 100193, China
| |
Collapse
|
22
|
Carbone FP, Ancona P, Volinia S, Terrazzan A, Bianchi N. Druggable Molecular Networks in BRCA1/BRCA2-Mutated Breast Cancer. BIOLOGY 2025; 14:253. [PMID: 40136510 PMCID: PMC11940086 DOI: 10.3390/biology14030253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 03/27/2025]
Abstract
Mutations in the tumor suppressor genes BRCA1 and BRCA2 are associated with the triple-negative breast cancer phenotype, particularly aggressive and hard-to-treat tumors lacking estrogen, progesterone, and human epidermal growth factor receptor 2. This research aimed to understand the metabolic and genetic links behind BRCA1 and BRCA2 mutations and investigate their relationship with effective therapies. Using the Cytoscape software, two networks were generated through a bibliographic analysis of articles retrieved from the PubMed-NCBI database. We identified 98 genes deregulated by BRCA mutations, and 24 were modulated by therapies. In particular, BIRC5, SIRT1, MYC, EZH2, and CSN2 are influenced by BRCA1, while BCL2, BAX, and BRIP1 are influenced by BRCA2 mutation. Moreover, the study evaluated the efficacy of several promising therapies, targeting only BRCA1/BRCA2-mutated cells. In this context, CDDO-Imidazolide was shown to increase ROS levels and induce DNA damage. Similarly, resveratrol decreased the expression of the anti-apoptotic gene BIRC5 while it increased SIRT1 both in vitro and in vivo. Other specific drugs were found to induce apoptosis selectively in BRCA-mutated cells or block cell growth when the mutation occurs, i.e., 3-deazaneplanocin A, genistein or daidzein, and PARP inhibitors. Finally, over-representation analysis on the genes highlights ferroptosis and proteoglycan pathways as potential drug targets for more effective treatments.
Collapse
Affiliation(s)
- Francesca Pia Carbone
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.P.C.); (P.A.); (S.V.); (N.B.)
| | - Pietro Ancona
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.P.C.); (P.A.); (S.V.); (N.B.)
| | - Stefano Volinia
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.P.C.); (P.A.); (S.V.); (N.B.)
- Genomics Core Facility, Centre of New Technologies, University of Warsaw, 02-097 Warsaw, Poland
- Laboratory for Technologies of Advanced Therapies (LTTA), 44121 Ferrara, Italy
| | - Anna Terrazzan
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.P.C.); (P.A.); (S.V.); (N.B.)
- Genomics Core Facility, Centre of New Technologies, University of Warsaw, 02-097 Warsaw, Poland
- Laboratory for Technologies of Advanced Therapies (LTTA), 44121 Ferrara, Italy
| | - Nicoletta Bianchi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (F.P.C.); (P.A.); (S.V.); (N.B.)
| |
Collapse
|
23
|
Stewart J, Krastev DB, Brough R, Zatreanu D, Song F, Baxter JS, Sridhar S, Frankum J, Konde A, Yang W, Haider S, Alexander J, Betteridge K, Gulati A, Attygalle AD, Vroobel K, Natrajan R, Khalique S, Roumeliotis TI, Choudhary JS, Yeung J, Wicks AJ, Marlow R, Banerjee S, Pettitt SJ, Tutt ANJ, Lord CJ. PPP2R1A mutations cause ATR inhibitor sensitivity in ovarian clear cell carcinoma. Oncogene 2025; 44:618-629. [PMID: 39939726 PMCID: PMC11850283 DOI: 10.1038/s41388-024-03265-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 12/04/2024] [Accepted: 12/12/2024] [Indexed: 02/14/2025]
Abstract
Identification of ARID1A/ATR synthetic lethality led to ATR inhibitor phase II trials in ovarian clear cell carcinoma (OCCC), a cancer of unmet need. Using multiple CRISPR-Cas9 mutagenesis and interference screens, we show that inactivation of protein phosphatase 2A (PP2A) subunits, including PPP2R1A, enhance ATRi sensitivity in ARID1A mutant OCCC. Analysis of a new OCCC cohort indicates that 52% possess oncogenic PPP2R1A p.R183 mutations and of these, one half possessed both ARID1A as well as PPP2R1A mutations. Using CRISPR-prime editing to generate new isogenic models of PPP2R1A mutant OCCC, we found that PPP2R1A p.R183W and p.R183P mutations cause ATRi-induced S phase stress, premature mitotic entry, genomic instability and ATRi sensitivity in OCCC tumour cells. p.R183 mutation also enhanced both in vitro and in vivo ATRi sensitivity in preclinical models of ARID1A mutant OCCC. These results argue for the assessment of PPP2R1A mutations as a biomarker of ATRi sensitivity.
Collapse
Affiliation(s)
- James Stewart
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, SW3 6JB, UK
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London, UK and Division of Clinical Studies, Institute of Cancer Research, London, UK
| | - Dragomir B Krastev
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, SW3 6JB, UK
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Rachel Brough
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, SW3 6JB, UK
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Diana Zatreanu
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, SW3 6JB, UK
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Feifei Song
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, SW3 6JB, UK
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Joseph S Baxter
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, SW3 6JB, UK
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Sandhya Sridhar
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, SW3 6JB, UK
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Jessica Frankum
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, SW3 6JB, UK
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Asha Konde
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, SW3 6JB, UK
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - William Yang
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, SW3 6JB, UK
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Syed Haider
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - John Alexander
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Kai Betteridge
- Light microscopy Facility, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Aditi Gulati
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Ayoma D Attygalle
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London, UK and Division of Clinical Studies, Institute of Cancer Research, London, UK
| | - Katherine Vroobel
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London, UK and Division of Clinical Studies, Institute of Cancer Research, London, UK
| | - Rachael Natrajan
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Saira Khalique
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London, UK and Division of Clinical Studies, Institute of Cancer Research, London, UK
| | | | - Jyoti S Choudhary
- Functional Proteomics Laboratory, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Jason Yeung
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, SW3 6JB, UK
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Andrew J Wicks
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, SW3 6JB, UK
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Rebecca Marlow
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Susana Banerjee
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London, UK and Division of Clinical Studies, Institute of Cancer Research, London, UK
| | - Stephen J Pettitt
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, SW3 6JB, UK
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Andrew N J Tutt
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK
| | - Christopher J Lord
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, SW3 6JB, UK.
- Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, SW3 6JB, UK.
| |
Collapse
|
24
|
Chen F, Xu W, Tang M, Tian Y, Shu Y, He X, Zhou L, Liu Q, Zhu Q, Lu X, Zhang J, Zhu WG. hnRNPA2B1 deacetylation by SIRT6 restrains local transcription and safeguards genome stability. Cell Death Differ 2025; 32:382-396. [PMID: 39511404 PMCID: PMC11893882 DOI: 10.1038/s41418-024-01412-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/15/2024] Open
Abstract
Repair of double strand breaks (DSBs) by RNA-binding proteins (RBPs) is vital for ensuring genome integrity. DSB repair is accompanied by local transcriptional repression in the vicinity of transcriptionally active genes, but the mechanism by which RBPs regulate transcriptional regulation is unclear. Here, we demonstrated that RBP hnRNPA2B1 functions as a RNA polymerase-associated factor that stabilizes the transcription complex under physiological conditions. Following a DSB, hnRNPA2B1 is released from damaged chromatin, reducing the efficiency of RNAPII complex assembly, leading to local transcriptional repression. Mechanistically, SIRT6 deacetylates hnRNPA2B1 at K113/173 residues, enforcing its rapid detachment from DSBs. This process disrupts the integrity of the RNAPII complex on active chromatin, which is a pre-requisite for transient but complete repression of local transcription. Functionally, the overexpression of an acetylation mimic stabilizes the transcription complex and facilitates the functioning of the transcription machinery. hnRNPA2B1 acetylation status was negatively correlated with SIRT6 expression, and acetylation mimic enhanced radio-sensitivity in vivo. Our findings demonstrate that hnRNPA2B1 is crucial for transcriptional repression. We have uncovered the missing link between DSB repair and transcriptional regulation in genome stability maintenance, highlighting the potential of hnRNPA2B1 as a therapeutic target.
Collapse
Affiliation(s)
- Feng Chen
- Department of Biochemistry and Molecular Biology, International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
| | - Wenchao Xu
- Department of Biochemistry and Molecular Biology, International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
| | - Ming Tang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuan Tian
- Department of Biochemistry and Molecular Biology, International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
| | - Yuxin Shu
- Department of Biochemistry and Molecular Biology, International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
- School of Basic Medical Sciences, Wannan Medical College, Wuhu, Anhui, China
| | - Xingkai He
- Department of Biochemistry and Molecular Biology, International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
| | - Linmin Zhou
- Department of Biochemistry and Molecular Biology, International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
| | - Qi Liu
- Department of Biochemistry and Molecular Biology, International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
| | - Qian Zhu
- Department of Biochemistry and Molecular Biology, International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
| | - Xiaopeng Lu
- Department of Biochemistry and Molecular Biology, International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
| | - Jun Zhang
- Department of Biochemistry and Molecular Biology, International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China.
| | - Wei-Guo Zhu
- Department of Biochemistry and Molecular Biology, International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China.
- School of Basic Medical Sciences, Wannan Medical College, Wuhu, Anhui, China.
| |
Collapse
|
25
|
Liu Y, Tang Q, Tao Q, Dong H, Shi Z, Zhou L. Low-frequency magnetic field therapy for glioblastoma: Current advances, mechanisms, challenges and future perspectives. J Adv Res 2025; 69:531-543. [PMID: 38565404 PMCID: PMC11954840 DOI: 10.1016/j.jare.2024.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/10/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most common malignant tumour of the central nervous system. Despite recent advances in multimodal GBM therapy incorporating surgery, radiotherapy, systemic therapy (chemotherapy, targeted therapy), and supportive care, the overall survival (OS) remains poor, and long-term survival is rare. Currently, the primary obstacles hindering the effectiveness of GBM treatment are still the blood-brain barrier and tumor heterogeneity. In light of its substantial advantages over conventional therapies, such as strong penetrative ability and minimal side effects, low-frequency magnetic fields (LF-MFs) therapy has gradually caught the attention of scientists. AIM OF REVIEW In this review, we shed the light on the current status of applying LF-MFs in the treatment of GBM. We specifically emphasize our current understanding of the mechanisms by which LF-MFs mediate anticancer effects and the challenges faced by LF-MFs in treating GBM cells. Furthermore, we discuss the prospective applications of magnetic field therapy in the future treatment of GBM. Key scientific concepts of review: The review explores the current progress on the use of LF-MFs in the treatment of GBM with a special focus on the potential underlying mechanisms of LF-MFs in anticancer effects. Additionally, we also discussed the complex magnetic field features and biological characteristics related to magnetic bioeffects. Finally, we proposed a promising magnetic field treatment strategy for future applications in GBM therapy.
Collapse
Affiliation(s)
- Yinlong Liu
- Department of Neurosurgery, Huashan Hospital, Fudan University, China
| | - Qisheng Tang
- Department of Neurosurgery, Huashan Hospital, Fudan University, China; National Center for Neurological Disorders, China; Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, China; Neurosurgical Institute of Fudan University, Shanghai, China; Shanghai Clinical Medical Center of Neurosurgery, China
| | - Quan Tao
- Shanghai Institute of Microsystem and Information Technology, China
| | - Hui Dong
- Shanghai Institute of Microsystem and Information Technology, China
| | - Zhifeng Shi
- Department of Neurosurgery, Huashan Hospital, Fudan University, China; National Center for Neurological Disorders, China; Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, China; Neurosurgical Institute of Fudan University, Shanghai, China; Shanghai Clinical Medical Center of Neurosurgery, China.
| | - Liangfu Zhou
- Department of Neurosurgery, Huashan Hospital, Fudan University, China; National Center for Neurological Disorders, China; Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, China; Neurosurgical Institute of Fudan University, Shanghai, China; Shanghai Clinical Medical Center of Neurosurgery, China.
| |
Collapse
|
26
|
Qiu H, Ye C. Phospholipid Biosynthesis: An Unforeseen Modulator of Nuclear Metabolism. Biol Cell 2025; 117:e70002. [PMID: 40123381 DOI: 10.1111/boc.70002] [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: 08/17/2024] [Revised: 02/27/2025] [Accepted: 03/05/2025] [Indexed: 03/25/2025]
Abstract
Glycerophospholipid biosynthesis is crucial not only for providing structural components required for membrane biogenesis during cell proliferation but also for facilitating membrane remodeling under stress conditions. The biosynthetic pathways for glycerophospholipid tails, glycerol backbones, and diverse head group classes intersect with various other metabolic processes, sharing intermediary metabolites. Recent studies have revealed intricate connections between glycerophospholipid synthesis and nuclear metabolism, including metabolite-mediated crosstalk with the epigenome, signaling pathways that govern genome integrity, and CTP-involved regulation of nucleotide and antioxidant biosynthesis. This review highlights recent advances in understanding the functional roles of glycerophospholipid biosynthesis beyond their structural functions in budding yeast and mammalian cells. We propose that glycerophospholipid biosynthesis plays an integrative role in metabolic regulation, providing a new perspective on lipid biology.
Collapse
Affiliation(s)
- Hong Qiu
- Zhejiang Key Laboratory of Molecular Cancer Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Cunqi Ye
- Zhejiang Key Laboratory of Molecular Cancer Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Hainan Institute of Zhejiang University, Zhejiang University, Sanya, China
| |
Collapse
|
27
|
Sun L, Fan G, Zhang Z, Chang D, Zhang X, Zhang T, Geng J, Zhang X, Lin M, Hu C, Zhou J, Wang M, Cao L, Zhang M, He B, Zhang S, Wang C. Phosphorylation of SIRT7 by ATM causes DNA mismatch repair downregulation and adaptive mutability during chemotherapy. Cell Rep 2025; 44:115269. [PMID: 39908142 DOI: 10.1016/j.celrep.2025.115269] [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: 07/09/2024] [Revised: 12/03/2024] [Accepted: 01/15/2025] [Indexed: 02/07/2025] Open
Abstract
Drug resistance significantly limits the efficacy of chemotherapy. The DNA mismatch repair (MMR) system maintains genomic stability by correcting DNA errors. During DNA-damaging treatments, cancer cells transiently increase their adaptive mutability, also known as microsatellite instability (MSI), to evade therapeutic pressure through MMR downregulation, conferring drug resistance. However, an understanding of the underlying mechanisms of MMR protein downregulation under DNA-damaging drugs remains limited. Our study reveals a negative correlation between SIRT7 protein levels and MMR core protein MSH2 levels in cervical and lung cancer tissues. SIRT7 destabilizes MSH2, promoting MSI and mutagenesis. Molecularly, DNA damage triggers ATM kinase-dependent phosphorylation and subcellular redistribution of SIRT7. Phosphorylated SIRT7 interacts with and deacetylates MSH2, impairing MMR, and inducing MSI and drug resistance. Our findings suggest that SIRT7 drives MMR downregulation under therapeutic stress and that ATM-dependent phosphorylation of SIRT7 may serve as a predictive biomarker for chemotherapeutic efficacy and a target for cancer treatment.
Collapse
Affiliation(s)
- Lianhui Sun
- Biomedical Translational Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China
| | - Guangjian Fan
- Biomedical Translational Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China
| | - Zhuqing Zhang
- Biomedical Translational Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China
| | - Dong Chang
- Biomedical Translational Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China
| | - Xiaoyu Zhang
- Biomedical Translational Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China
| | - Tongqing Zhang
- Biomedical Translational Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China
| | - Jichuan Geng
- Biomedical Translational Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China
| | - Xiaoxia Zhang
- Biomedical Translational Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China
| | - Menghan Lin
- Biomedical Translational Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China
| | - Chen Hu
- Biomedical Translational Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China
| | - Jiaqi Zhou
- Biomedical Translational Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China
| | - Mengxue Wang
- Biomedical Translational Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China
| | - Liu Cao
- Health Sciences Institute, College of Basic Medical Sciences, China Medical University, Shenyang 110122, China
| | - Mary Zhang
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, 4100 John R., Detroit, MI 48201, USA
| | - Baokun He
- Institute of Chinese Materia Medica, The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, China
| | - Shengping Zhang
- Biomedical Translational Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China.
| | - Chuangui Wang
- Biomedical Translational Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255049, China.
| |
Collapse
|
28
|
Zhang S, Xie X, Zhang H, Zhao Z, Xia K, Song H, Li Q, Li M, Ge Z. Visualizing Reactive Oxygen Species-Induced DNA Damage Process in Higher-Ordered Origami Nanostructures. JACS AU 2025; 5:965-974. [PMID: 40017784 PMCID: PMC11863157 DOI: 10.1021/jacsau.4c01203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/13/2025] [Accepted: 01/15/2025] [Indexed: 03/01/2025]
Abstract
The genetic information on organisms is stored in the cell nucleus in the form of higher-ordered DNA structures. Here, we use DNA framework nanostructures (DFNs) to simulate the compaction and stacking density of nucleosome DNA for precise conformational and structure determination, particularly the dynamic structural changes, preferential reaction regions, and sites of DFNs during the reactive oxygen species (ROS) reaction process. By developing an atomic force microscopy-based single-particle analysis (SPA) data reconstruction method to collect and reanalyze imaging information, we demonstrate that the geometric morphology of DFNs constrains their reaction kinetics with ROS, where local mechanical stress and regional base distribution are two key factors affecting their kinetics. Furthermore, we plot the reaction process diagram for ROS and DFNs, showing the reaction process and intermediate products with individual activation energies. This SPA method offers new research tools and insights for studying the dynamic changes of highly folded and organized DNA structural domains within the nucleus and helps to reveal the key mechanisms behind their functional differences in topologically associating domains.
Collapse
Affiliation(s)
- Shuangye Zhang
- School
of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory,
Frontiers Science Center for Transformative Molecules, Zhangjiang
Institute for Advanced Study and National Center for Translational
Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiaodong Xie
- School
of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory,
Frontiers Science Center for Transformative Molecules, Zhangjiang
Institute for Advanced Study and National Center for Translational
Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hairuo Zhang
- School
of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory,
Frontiers Science Center for Transformative Molecules, Zhangjiang
Institute for Advanced Study and National Center for Translational
Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ziwei Zhao
- Shanghai
Artificial Intelligence Research Institute, Shanghai 200240, China
| | - Kai Xia
- Shanghai
Artificial Intelligence Research Institute, Shanghai 200240, China
| | - Haitao Song
- Shanghai
Artificial Intelligence Research Institute, Shanghai 200240, China
| | - Qian Li
- School
of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory,
Frontiers Science Center for Transformative Molecules, Zhangjiang
Institute for Advanced Study and National Center for Translational
Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Mingqiang Li
- School
of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory,
Frontiers Science Center for Transformative Molecules, Zhangjiang
Institute for Advanced Study and National Center for Translational
Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhilei Ge
- School
of Chemistry and Chemical Engineering, New Cornerstone Science Laboratory,
Frontiers Science Center for Transformative Molecules, Zhangjiang
Institute for Advanced Study and National Center for Translational
Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
29
|
Chen X, Wu S, He H, Tang J, Zhong Y, Fang H, Huang Q, Hong L, Shao L, Wu J. G2M-checkpoint related immune barrier structure and signature for prognosis and immunotherapy response in hepatocellular carcinoma: insights from spatial transcriptome and machine learning. J Transl Med 2025; 23:202. [PMID: 39966861 PMCID: PMC11837653 DOI: 10.1186/s12967-024-06051-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 12/24/2024] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) treatment remains challenging, particularly for immune checkpoint inhibitors (ICIs) non-response patients. Spatial transcriptome (ST) data and machine learning algorithms offer new insights into understanding HCC heterogeneity and ICIs resistance mechanisms. METHODS Utilizing ST data from HCC patients on ICIs treatment, we analyzed pathway activity and immune infiltration. We combined 167 machine learning models to develop a G2M-checkpoint related signature (G2MRS) based on differential gene expression. The four cohorts and in-house cohort was used to validate G2MRS, and KPNA2's role was further examined through in vitro experiments in two different liver cancer cell lines. RESULTS Our analysis revealed a distinct suppressive immune barrier structure (SIBS) in ICIs non-response patients, associated with upregulated G2M-checkpoint levels. G2MRS, consisting of KPNA2, CENPA, and UCK2, accurately predicted HCC prognosis and ICIs response. Further in vitro experiments demonstrated KPNA2's role in regulating migration, proliferation, and apoptosis in liver cancer. CONCLUSIONS This study highlights the importance of spatial heterogeneity and machine learning in refining HCC prognosis and ICIs response prediction. G2MRS and KPNA2 emerge as promising biomarkers for personalized HCC management.
Collapse
Affiliation(s)
- Xingte Chen
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Shiji Wu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Hongxin He
- Department of Gastrointestinal Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Jingjing Tang
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Yaqi Zhong
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Huipeng Fang
- Department of Hepatopancreatobiliary Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Qizhen Huang
- Department of Radiation Oncology, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Liang Hong
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Lingdong Shao
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China.
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, 420 Fuma Rd, Jin'an District, Fuzhou, Fujian, China.
| | - Junxin Wu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China.
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, 420 Fuma Rd, Jin'an District, Fuzhou, Fujian, China.
| |
Collapse
|
30
|
Yang Z, Zhou M, Yin T, Wang CY, Zhu GY, Bai LP, Jiang ZH, Zhang W. Functionalized Carbon Dots With Intrinsic Wnt/ β-Catenin Inhibition to Synergistically Promote 5-Fluorouracil Chemotherapy. Int J Nanomedicine 2025; 20:1951-1964. [PMID: 39963418 PMCID: PMC11831914 DOI: 10.2147/ijn.s503540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 01/22/2025] [Indexed: 02/20/2025] Open
Abstract
Background 5-fluorouracil (5-FU) is the most widely used anti-pyrimidine drug that exerts its cytotoxic effect by causing DNA damage. The Wnt/β-catenin pathway has been considered a promising strategy to improve chemosensitivity by enhancing the DNA damage response of chemotherapy drugs. Combination therapies against cancers could inevitably affect endogenous levels of ribonucleotides (RNs) and deoxyribonucleotides (dRNs) which are critical for DNA synthesis and repair. However, exploring satisfactory Wnt/β-catenin inhibitors for synergistic therapy through regulating RNs and dRNs remains challenging. Methods and Results Here, aloe vera-derived carbon dots (A-CDs) with good bioactivity were synthesized via a one-step hydrothermal process, demonstrating both intrinsic Wnt/β-catenin inhibition and bioimaging capabilities to overcome the limitations of conventional Wnt/β-catenin inhibitors. The as-prepared A-CDs were further served as the transport vehicle for 5-FU, facilitating synergistic combination therapy by inhibiting the Wnt/β-catenin pathway, which could possibly accelerate nucleotide imbalance of dATP, ATP, TMP, and dUMP, ultimately leading to enhanced 5-FU efficiency. Additionally, the tumor-targeted material (HA-CDs@5-FU) was synthesized by modifying hyaluronic acid (HA) onto CDs@5-FU and exhibited superior antitumor efficacy in vivo with negligible side effects. Conclusion Overall, this study provided a novel strategy for Wnt/β-catenin inhibition and synergistic therapy, providing insights into the application of nano-agents in cancer therapy.
Collapse
Affiliation(s)
- Ziwei Yang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, People’s Republic of China
| | - Mingyue Zhou
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, People’s Republic of China
| | - Tianpeng Yin
- Biomedical Research and Development Center, Zunyi Medical University, Zhuhai, People’s Republic of China
| | - Cai-Yun Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, People’s Republic of China
| | - Guo-Yuan Zhu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, People’s Republic of China
| | - Li-Ping Bai
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, People’s Republic of China
| | - Zhi-Hong Jiang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, People’s Republic of China
| | - Wei Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, People’s Republic of China
| |
Collapse
|
31
|
Guo Y, Li P, Zhang J, Hao S, Zhou X, Di C, Long F, Zhang H, Si J. Carbon ion irradiation conquers the radioresistance by inducing complex DNA damage and apoptosis in U251 human glioblastomas cells. Med Oncol 2025; 42:64. [PMID: 39903402 DOI: 10.1007/s12032-025-02616-5] [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: 11/01/2024] [Accepted: 01/22/2025] [Indexed: 02/06/2025]
Abstract
Glioblastoma multiforme (GBM) is the most malignant brain tumor, with radiotherapy frequently employed following surgical resection. However, conventional radiation therapies often yield suboptimal results. This study investigated the effects of X-ray and carbon ion irradiation on the glioblastoma cell line U251 to assess the distinctive advantages of carbon ion treatment and explore mechanisms for overcoming radiation resistance. The findings indicated that carbon ion irradiation more effectively inhibited colony formation and induced more severe apoptosis and cell cycle disorder in U251 cells. Immunofluorescence assays revealed larger and more abundant ϒ-H2AX and 53BP1 foci in the carbon ion irradiation group. Western blot analysis demonstrated that carbon ion-induced DNA damage repair involved a complex array of pathways, with the RAD51-mediated homologous recombination (HR) pathway being predominant, while the Rad23B-mediated nucleotide excision repair (NER) pathway and XRCC1-mediated base excision repair (BER) were more relevant in response to X-ray irradiation. These results suggest that carbon ion irradiation may overcome radioresistance by inducing more complex DNA damage and apoptosis, thus providing insights for targeting new strategies in combining gene therapy with radiotherapy.
Collapse
Affiliation(s)
- Yulu Guo
- Department of Medical Physics, Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou, 730000, Gansu, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Pingping Li
- Department of Medical Physics, Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou, 730000, Gansu, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Jinhua Zhang
- Department of Medical Physics, Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou, 730000, Gansu, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Sijia Hao
- Department of Medical Physics, Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou, 730000, Gansu, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Xuan Zhou
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Cuixia Di
- Department of Medical Physics, Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou, 730000, Gansu, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
| | - Feng Long
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Hong Zhang
- Department of Medical Physics, Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou, 730000, Gansu, China.
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China.
| | - Jing Si
- Department of Medical Physics, Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou, 730000, Gansu, China.
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China.
| |
Collapse
|
32
|
Paganelli F, Poli A, Truocchio S, Martelli AM, Palumbo C, Lattanzi G, Chiarini F. At the nucleus of cancer: how the nuclear envelope controls tumor progression. MedComm (Beijing) 2025; 6:e70073. [PMID: 39866838 PMCID: PMC11758262 DOI: 10.1002/mco2.70073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 12/09/2024] [Accepted: 12/12/2024] [Indexed: 01/28/2025] Open
Abstract
Historically considered downstream effects of tumorigenesis-arising from changes in DNA content or chromatin organization-nuclear alterations have long been seen as mere prognostic markers within a genome-centric model of cancer. However, recent findings have placed the nuclear envelope (NE) at the forefront of tumor progression, highlighting its active role in mediating cellular responses to mechanical forces. Despite significant progress, the precise interplay between NE components and cancer progression remains under debate. In this review, we provide a comprehensive and up-to-date overview of how changes in NE composition affect nuclear mechanics and facilitate malignant transformation, grounded in the latest molecular and functional studies. We also review recent research that uses advanced technologies, including artificial intelligence, to predict malignancy risk and treatment outcomes by analyzing nuclear morphology. Finally, we discuss how progress in understanding nuclear mechanics has paved the way for mechanotherapy-a promising cancer treatment approach that exploits the mechanical differences between cancerous and healthy cells. Shifting the perspective on NE alterations from mere diagnostic markers to potential therapeutic targets, this review calls for further investigation into the evolving role of the NE in cancer, highlighting the potential for innovative strategies to transform conventional cancer therapies.
Collapse
Affiliation(s)
- Francesca Paganelli
- Department of Biomedical and Neuromotor SciencesAlma Mater StudiorumUniversity of BolognaBolognaItaly
| | - Alessandro Poli
- IFOM ETS ‐ The AIRC Institute of Molecular OncologyMilanItaly
| | - Serena Truocchio
- Department of Biomedical and Neuromotor SciencesAlma Mater StudiorumUniversity of BolognaBolognaItaly
| | - Alberto M. Martelli
- Department of Biomedical and Neuromotor SciencesAlma Mater StudiorumUniversity of BolognaBolognaItaly
| | - Carla Palumbo
- Department of BiomedicalMetabolic and Neural SciencesUniversity of Modena and Reggio EmiliaModenaItaly
| | - Giovanna Lattanzi
- CNR Institute of Molecular Genetics “Luigi Luca Cavalli‐Sforza”Unit of BolognaBolognaItaly
- IRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Francesca Chiarini
- Department of BiomedicalMetabolic and Neural SciencesUniversity of Modena and Reggio EmiliaModenaItaly
| |
Collapse
|
33
|
Kalashnikova AA, Toibazarova AB, Artyushin OI, Anikina LV, Globa AA, Klemenkova ZS, Andreev MV, Radchenko EV, Palyulin VA, Aleksandrova YR, Syzdykbayev MI, Appazov NO, Chubarev VN, Neganova ME, Brel VK. Design of New Daunorubicin Derivatives with High Cytotoxic Potential. Int J Mol Sci 2025; 26:1270. [PMID: 39941040 PMCID: PMC11818560 DOI: 10.3390/ijms26031270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/16/2025] [Accepted: 01/30/2025] [Indexed: 02/16/2025] Open
Abstract
Chemotherapy with anthracycline antibiotics is a common method of treating tumors of various etiologies. To create more highly effective cytostatics based on daunorubicin, we used the method of reductive amination using polyalkoxybenzaldehydes. The obtained derivatives of the anthracycline structure have much greater cytotoxicity compared to daunorubicin due to increased affinity for DNA, the ability to disrupt the cell cycle, and their inhibition of the glycolysis process, which is confirmed by data from extensive biological studies and the results of molecular modeling.
Collapse
Affiliation(s)
- Aleksandra A. Kalashnikova
- Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, Vavilova St. 28, Bld. 1, Moscow 119991, Russia; (A.A.K.); (O.I.A.); (Z.S.K.); (M.V.A.); (Y.R.A.)
| | - Altynkul B. Toibazarova
- Laboratory of Engineering Profile, Korkyt Ata Kyzylorda University, Ayteke bi Str., 29A, Kyzylorda 120014, Kazakhstan;
| | - Oleg I. Artyushin
- Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, Vavilova St. 28, Bld. 1, Moscow 119991, Russia; (A.A.K.); (O.I.A.); (Z.S.K.); (M.V.A.); (Y.R.A.)
| | - Lada V. Anikina
- Institute of Physiologically Active Compounds of the FSBIS of the Federal Research Center for Problems of Chemical Physics and Medicinal Chemistry of the RAS, 1 Severnyi Proezd, Chernogolovka 142432, Russia; (L.V.A.); (A.A.G.)
| | - Anastasiya A. Globa
- Institute of Physiologically Active Compounds of the FSBIS of the Federal Research Center for Problems of Chemical Physics and Medicinal Chemistry of the RAS, 1 Severnyi Proezd, Chernogolovka 142432, Russia; (L.V.A.); (A.A.G.)
| | - Zinaida S. Klemenkova
- Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, Vavilova St. 28, Bld. 1, Moscow 119991, Russia; (A.A.K.); (O.I.A.); (Z.S.K.); (M.V.A.); (Y.R.A.)
| | - Maxim V. Andreev
- Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, Vavilova St. 28, Bld. 1, Moscow 119991, Russia; (A.A.K.); (O.I.A.); (Z.S.K.); (M.V.A.); (Y.R.A.)
| | - Eugene V. Radchenko
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory, Moscow 119991, Russia; (E.V.R.); (V.A.P.)
| | - Vladimir A. Palyulin
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory, Moscow 119991, Russia; (E.V.R.); (V.A.P.)
| | - Yulia R. Aleksandrova
- Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, Vavilova St. 28, Bld. 1, Moscow 119991, Russia; (A.A.K.); (O.I.A.); (Z.S.K.); (M.V.A.); (Y.R.A.)
| | - Marat I. Syzdykbayev
- Department of Biology, Geography and Chemistry, Laboratory of Engineering Profile, Korkyt Ata Kyzylorda University, Ayteke bi Str., 29A, Kyzylorda 120014, Kazakhstan;
| | - Nurbol O. Appazov
- Laboratory of Engineering Profile, Department of Engineering Technology, Korkyt Ata Kyzylorda University, Ayteke bi Str., 29A, Kyzylorda 120014, Kazakhstan;
- “CNEC” LLP, Dariger Ali Lane, 2, Kyzylorda 120001, Kazakhstan
| | - Vladimir N. Chubarev
- Department of Pharmacology, The Institute of Pharmacy Named after A.P. Nelyubin, Sechenov University, Trubetskaya St., 8-2, Moscow 119991, Russia;
| | - Margarita E. Neganova
- Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, Vavilova St. 28, Bld. 1, Moscow 119991, Russia; (A.A.K.); (O.I.A.); (Z.S.K.); (M.V.A.); (Y.R.A.)
| | - Valery K. Brel
- Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, Vavilova St. 28, Bld. 1, Moscow 119991, Russia; (A.A.K.); (O.I.A.); (Z.S.K.); (M.V.A.); (Y.R.A.)
| |
Collapse
|
34
|
Fan X, Zhang X, Zhang Y, Jiang S, Song W, Song D. IR-Driven Multisignal Conditioning for Multiplex Detection: Thermal-Responsive Triple DNA-Mediated Reconfigurable Photoelectrochemical/Photothermal Dual-Mode Strategy. ACS Sens 2025; 10:292-300. [PMID: 39752297 DOI: 10.1021/acssensors.4c02504] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2025]
Abstract
Superior to traditional multiplex photoelectrochemical (PEC) sensors, integrated multitarget assay on a single reconstructive electrode interface is promising in real-time detection through eliminating the need of specialized instrumentation and cumbersome interfacial modifications. Current interface reconstruction approaches including pH modulation and bioenzyme cleavage involve biohazardous and time-consuming operations, which cannot meet the demand for rapid, eco-friendly, and portable detection, which are detrimental to the development of multiplex PEC sensors toward portability. Herein, we report a pioneer work on IR-driven "four-to-one" multisignal conditioning to facile reconfigure electrode interface for multitarget detection via photoelectrochemical/photothermal dual mode. The copper sulfide quantum dot (CuS QD) with excellent photoelectrochemical properties and a photothermal effect is first labeled on DNA S2. Once the CuS QD-S2 complementarily pairs with the DNA S3 on the photocathode surface, thermal-responsive triple DNA is formed, and the photocurrent and photothermal dual-mode signals for one target assay are produced. Upon the dissociation of the triple DNA by IR irradiation, the electrode interface is reconfigured for the self-calibrating dual-mode detection of another target. The feasibility of the IR-driven multisignal conditioning sensor is confirmed by detecting coexistent antibiotics kanamycin (KANA) and neomycin (NEO) in complex real samples. The low-loss interface reconfiguration and rapid "four-to-one" multisignal modulation highlight a broad prospect for self-calibrating multiplex assay in the fields of environment, medicine, and food safety.
Collapse
Affiliation(s)
- Xue Fan
- College of Chemistry, Jilin Province Research Center for Engineering and Technology of Spectral Analytical Instruments, Jilin University, Changchun 130012, China
| | - Xuechen Zhang
- College of Chemistry, Jilin Province Research Center for Engineering and Technology of Spectral Analytical Instruments, Jilin University, Changchun 130012, China
| | - Yanru Zhang
- College of Chemistry, Jilin Province Research Center for Engineering and Technology of Spectral Analytical Instruments, Jilin University, Changchun 130012, China
| | - Shan Jiang
- College of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, China
| | - Wenbo Song
- College of Chemistry, Jilin Province Research Center for Engineering and Technology of Spectral Analytical Instruments, Jilin University, Changchun 130012, China
| | - Daqian Song
- College of Chemistry, Jilin Province Research Center for Engineering and Technology of Spectral Analytical Instruments, Jilin University, Changchun 130012, China
| |
Collapse
|
35
|
Rimal P, Paul SK, Panday SK, Alexov E. Further Development of SAMPDI-3D: A Machine Learning Method for Predicting Binding Free Energy Changes Caused by Mutations in Either Protein or DNA. Genes (Basel) 2025; 16:101. [PMID: 39858648 PMCID: PMC11764785 DOI: 10.3390/genes16010101] [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: 12/27/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Predicting the effects of protein and DNA mutations on the binding free energy of protein-DNA complexes is crucial for understanding how DNA variants impact wild-type cellular function. As many cellular interactions involve protein-DNA binding, accurately predicting changes in binding free energy (ΔΔG) is valuable for distinguishing pathogenic mutations from benign ones. METHODS This study describes the development and optimization of the SAMPDI-3Dv2 machine learning method, which is trained on an expanded database of experimentally measured ΔΔGs. This enhanced model incorporates new features, including the 3D structure of the mutant protein, features of the mutant structure, and a position-specific scoring matrix (PSSM). Benchmarking was conducted using 5-fold cross-validation. RESULTS The updated SAMPDI-3D model (SAMPDI-3Dv2) achieved Pearson correlation coefficients (PCCs) of 0.68 for protein and 0.80 for DNA mutations. These results represent significant improvements over existing tools. Additionally, the method's rapid execution time enables genome-scale predictions. CONCLUSIONS The improved SAMPDI-3Dv2 shows enhanced predictive performance for analyzing mutations in protein-DNA complexes. By leveraging structural information and an expanded training dataset, SAMPDI-3Dv2 provides researchers with a more accurate and efficient tool for mutation analysis, contributing to identifying pathogenic variants and improving our understanding of cellular function.
Collapse
Affiliation(s)
| | | | | | - Emil Alexov
- Department of Physics and Astronomy, College of Science, Clemson University, Clemson, SC 29634, USA; (P.R.); (S.K.P.); (S.K.P.)
| |
Collapse
|
36
|
Zhao B, Nepovimova E, Wu Q. The role of circadian rhythm regulator PERs in oxidative stress, immunity, and cancer development. Cell Commun Signal 2025; 23:30. [PMID: 39825442 PMCID: PMC11740368 DOI: 10.1186/s12964-025-02040-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/11/2025] [Indexed: 01/20/2025] Open
Abstract
The complex interaction between circadian rhythms and physiological functions is essential for maintaining human health. At the heart of this interaction lies the PERIOD proteins (PERs), pivotal to the circadian clock, influencing the timing of physiological and behavioral processes and impacting oxidative stress, immune functionality, and tumorigenesis. PER1 orchestrates the cooperation of the enzyme GPX1, modulating mitochondrial dynamics in sync with daily rhythms and oxidative stress, thus regulating the mechanisms managing energy substrates. PERs in innate immune cells modulate the temporal patterns of NF-κB and TNF-α activities, as well as the response to LPS-induced toxic shock, initiating inflammatory responses that escalate into chronic inflammatory conditions. Crucially, PERs modulate cancer cell behaviors including proliferation, apoptosis, and migration by influencing the levels of cell cycle proteins and stimulating the expression of oncogenes c-Myc and MDM2. PER2/3, as antagonists in cancer stem cell biology, play important roles in differentiating cancer stem cells and in maintaining their stemness. Importantly, the expression of Pers serve as a significant factor for early cancer diagnosis and prognosis. This review delves into the link between circadian rhythm regulator PERs, disruptions in circadian rhythm, and oncogenesis. We examine the evidence that highlights how dysfunctions in PERs activities initiate cancer development, aid tumor growth, and modify cancer cell metabolism through pathways involved in oxidative stress and immune system. Comprehending these connections opens new pathways for the development of circadian rhythm-based therapeutic strategies, with the aims of boosting immune responses and enhancing cancer treatments.
Collapse
Affiliation(s)
- Baimei Zhao
- College of Life Science, Yangtze University, Jingzhou, 434025, China
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové , 500 03, Czech Republic
| | - Qinghua Wu
- College of Life Science, Yangtze University, Jingzhou, 434025, China.
| |
Collapse
|
37
|
Li Z, Zhang Z. A tale of two strands: Decoding chromatin replication through strand-specific sequencing. Mol Cell 2025; 85:238-261. [PMID: 39824166 PMCID: PMC11750172 DOI: 10.1016/j.molcel.2024.10.035] [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: 08/15/2024] [Revised: 10/03/2024] [Accepted: 10/25/2024] [Indexed: 01/20/2025]
Abstract
DNA replication, a fundamental process in all living organisms, proceeds with continuous synthesis of the leading strand by DNA polymerase ε (Pol ε) and discontinuous synthesis of the lagging strand by polymerase δ (Pol δ). This inherent asymmetry at each replication fork necessitates the development of methods to distinguish between these two nascent strands in vivo. Over the past decade, strand-specific sequencing strategies, such as enrichment and sequencing of protein-associated nascent DNA (eSPAN) and Okazaki fragment sequencing (OK-seq), have become essential tools for studying chromatin replication in eukaryotic cells. In this review, we outline the foundational principles underlying these methodologies and summarize key mechanistic insights into DNA replication, parental histone transfer, epigenetic inheritance, and beyond, gained through their applications. Finally, we discuss the limitations and challenges of current techniques, highlighting the need for further technological innovations to better understand the dynamics and regulation of chromatin replication in eukaryotic cells.
Collapse
Affiliation(s)
- Zhiming Li
- Institute for Cancer Genetics and Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA; West China School of Public Health and West China Fourth Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Zhiguo Zhang
- Institute for Cancer Genetics and Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Pediatrics and Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
38
|
Sarmiento-Mañús R, Fontcuberta-Cervera S, Kawade K, Oikawa A, Tsukaya H, Quesada V, Micol JL, Ponce MR. Functional conservation and divergence of arabidopsis VENOSA4 and human SAMHD1 in DNA repair. Heliyon 2025; 11:e41019. [PMID: 39801971 PMCID: PMC11720913 DOI: 10.1016/j.heliyon.2024.e41019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 11/28/2024] [Accepted: 12/05/2024] [Indexed: 01/16/2025] Open
Abstract
The human deoxyribonucleoside triphosphatase (dNTPase) Sterile alpha motif and histidine-aspartate domain containing protein 1 (SAMHD1) has a dNTPase-independent role in repairing DNA double-strand breaks (DSBs) by homologous recombination (HR). Here, we show that VENOSA4 (VEN4), the probable Arabidopsis thaliana ortholog of SAMHD1, also functions in DSB repair by HR. The ven4 loss-of-function mutants showed increased DNA ploidy and deregulated DNA repair genes, suggesting DNA damage accumulation. Hydroxyurea, which blocks DNA replication and generates DSBs, induced VEN4 expression. The ven4 mutants were hypersensitive to hydroxyurea, with decreased DSB repair by HR. Metabolomic analysis of the strong ven4-0 mutant revealed depletion of metabolites associated with DNA damage responses. In contrast to SAMHD1, VEN4 showed no evident involvement in preventing R-loop accumulation. Our study thus reveals functional conservation in DNA repair by VEN4 and SAMHD1.
Collapse
Affiliation(s)
- Raquel Sarmiento-Mañús
- Instituto de Bioingeniería, Universidad Miguel Hernández, Campus de Elche, 03202, Elche, Spain
| | | | - Kensuke Kawade
- Graduate School of Science and Engineering, Saitama University, Saitama City, 338-8570, Saitama, Japan
- Center for Sustainable Resource Science, RIKEN, Yokohama, 230-0045, Kanagawa, Japan
- Exploratory Research Center on Life and Living Systems, Okazaki, 444-8787, Aichi, Japan
| | - Akira Oikawa
- Center for Sustainable Resource Science, RIKEN, Yokohama, 230-0045, Kanagawa, Japan
- Graduate School of Agriculture, Kyoto University, 606-8502, Kyoto, Japan
| | - Hirokazu Tsukaya
- Exploratory Research Center on Life and Living Systems, Okazaki, 444-8787, Aichi, Japan
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Bunkyo-ku, 113-0033, Tokyo, Japan
| | - Víctor Quesada
- Instituto de Bioingeniería, Universidad Miguel Hernández, Campus de Elche, 03202, Elche, Spain
| | - José Luis Micol
- Instituto de Bioingeniería, Universidad Miguel Hernández, Campus de Elche, 03202, Elche, Spain
| | - María Rosa Ponce
- Instituto de Bioingeniería, Universidad Miguel Hernández, Campus de Elche, 03202, Elche, Spain
| |
Collapse
|
39
|
Lu PS, Sun SC. Mycotoxin toxicity and its alleviation strategy on female mammalian reproduction and fertility. J Adv Res 2025:S2090-1232(25)00041-4. [PMID: 39814223 DOI: 10.1016/j.jare.2025.01.022] [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: 07/10/2024] [Revised: 12/23/2024] [Accepted: 01/12/2025] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Mycotoxin, a secondary metabolite of fungus, found worldwide and concerning in crops and food, causes multiple acute and chronic toxicities. Its toxic profile includes hepatotoxicity, carcinogenicity, teratogenicity, estrogenicity, immunotoxicity, and neurotoxicity, leading to deleterious impact on human and animal health. Emerging evidence suggests that it adversely affects perinatal health and progeny by its ability to cross placental barriers. AIM OF REVIEW Due to its wide occurrence and potential toxicity on reproductive health, it is essential to understand the mechanisms of mycotoxin-related reproductive toxicity. This review summarizes the toxicities and mechanisms of mycotoxin on maternal and offspring reproduction among mammalian species. Approaches for effective mycotoxin alleviation are also discussed, providing strategies against mycotoxin contamination. KEY SCIENTIFIC CONCEPTS OF REVIEW The profound mycotoxin toxicities in female mammalian reproduction affect follicle assembly, embryo development, and fetus growth, thereby decreasing offspring fertility. Factors from endocrine system such as hypothalamic-pituitary-gonadal axis and gut-ovarian axis, placenta ABC transporters, organelle and cytoskeleton dynamics, cell cycle control, genomic stability, and redox homeostasis are found to be closely related to mycotoxin toxicities. Approaches from physical, chemical, biological, and supplementation of natural antioxidants are discussed for the mycotoxin elimination, while their applications are not widespread. Available ways for mycotoxin and its toxicities alleviation need further study. Since a species-, time-, and dose-specific response might exist in mycotoxin toxicities, more consideration should be given to the protocols for mycotoxin toxicity studies, such as experimental animal models, exposure duration, and dosage. Specific mechanism for mycotoxin, especially form a molecular biology perspective, could be investigated with multi-omics technologies and advanced imaging techniques. Mass spectrometry with algorithms may provide more accurate exposure assessments, and it may be further helpful to identify the high-risk individuals in the future.
Collapse
Affiliation(s)
- Ping-Shuang Lu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Shao-Chen Sun
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China; Key Laboratory of Research On Clinical Molecular Diagnosis for High Incidence Diseases in Western Guangxi of Guangxi Higher Education Institutions, Reproductive Medicine of Guangxi Medical and Health Key Discipline Construction Project, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China.
| |
Collapse
|
40
|
Compton ZT, Mellon W, Harris VK, Rupp S, Mallo D, Kapsetaki SE, Wilmot M, Kennington R, Noble K, Baciu C, Ramirez LN, Peraza A, Martins B, Sudhakar S, Aksoy S, Furukawa G, Vincze O, Giraudeau M, Duke EG, Spiro S, Flach E, Davidson H, Li CI, Zehnder A, Graham TA, Troan BV, Harrison TM, Tollis M, Schiffman JD, Aktipis CA, Abegglen LM, Maley CC, Boddy AM. Cancer Prevalence across Vertebrates. Cancer Discov 2025; 15:227-244. [PMID: 39445720 PMCID: PMC11726020 DOI: 10.1158/2159-8290.cd-24-0573] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/17/2024] [Accepted: 09/24/2024] [Indexed: 10/25/2024]
Abstract
Cancer is pervasive across multicellular species, but what explains the differences in cancer prevalence across species? Using 16,049 necropsy records for 292 species spanning three clades of tetrapods (amphibians, sauropsids, and mammals), we found that neoplasia and malignancy prevalence increases with adult mass (contrary to Peto's paradox) and somatic mutation rate but decreases with gestation time. The relationship between adult mass and malignancy prevalence was only apparent when we controlled for gestation time. Evolution of cancer susceptibility appears to have undergone sudden shifts followed by stabilizing selection. Outliers for neoplasia prevalence include the common porpoise (<1.3%), the Rodrigues fruit bat (<1.6%), the black-footed penguin (<0.4%), ferrets (63%), and opossums (35%). Discovering why some species have particularly high or low levels of cancer may lead to a better understanding of cancer syndromes and novel strategies for the management and prevention of cancer. Significance: Evolution has discovered mechanisms for suppressing cancer in a wide variety of species. By analyzing veterinary necropsy records, we can identify species with exceptionally high or low cancer prevalence. Discovering the mechanisms of cancer susceptibility and resistance may help improve cancer prevention and explain cancer syndromes. See related commentary by Metzger, p. 14.
Collapse
Affiliation(s)
- Zachary T. Compton
- Arizona Cancer Evolution Center, The Biodesign Institute, Arizona State University, Tempe, Arizona
- University of Arizona Cancer Center, Tucson, Arizona
- University of Arizona College of Medicine, Tucson, Arizona
| | - Walker Mellon
- Arizona Cancer Evolution Center, The Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Valerie K. Harris
- Arizona Cancer Evolution Center, The Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Shawn Rupp
- Arizona Cancer Evolution Center, The Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Diego Mallo
- Arizona Cancer Evolution Center, The Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Stefania E. Kapsetaki
- Arizona Cancer Evolution Center, The Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Mallory Wilmot
- Department of Pediatrics and Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Ryan Kennington
- Department of Pediatrics and Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Kathleen Noble
- Department of Pediatrics and Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Cristina Baciu
- Arizona Cancer Evolution Center, The Biodesign Institute, Arizona State University, Tempe, Arizona
- Department of Psychology, Arizona State University, Tempe, Arizona
| | - Lucia N. Ramirez
- Genomic Sciences Graduate Program, North Carolina State University, Raleigh, North Carolina
| | - Ashley Peraza
- Arizona Cancer Evolution Center, The Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Brian Martins
- Arizona Cancer Evolution Center, The Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Sushil Sudhakar
- Arizona Cancer Evolution Center, The Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Selin Aksoy
- Arizona Cancer Evolution Center, The Biodesign Institute, Arizona State University, Tempe, Arizona
| | - Gabriela Furukawa
- Department of Pediatrics and Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Orsolya Vincze
- Institute of Aquatic Ecology, Centre for Ecological Research, Debrecen, Hungary
- Evolutionary Ecology Group, Hungarian Department of Biology and Ecology, Babeş-Bolyai University, Cluj-Napoca, Romania
| | | | - Elizabeth G. Duke
- North Carolina State College of Veterinary Medicine, Raleigh, North Carolina
- Exotic Species Cancer Research Alliance, North Carolina State College of Veterinary Medicine, Raleigh, North Carolina
| | - Simon Spiro
- Wildlife Health Services, Zoological Society of London, London, United Kingdom
| | - Edmund Flach
- Wildlife Health Services, Zoological Society of London, London, United Kingdom
| | - Hannah Davidson
- North Carolina State College of Veterinary Medicine, Raleigh, North Carolina
- Exotic Species Cancer Research Alliance, North Carolina State College of Veterinary Medicine, Raleigh, North Carolina
| | - Christopher I. Li
- Translational Research Program and Epidemiology Program, Fred Hutchinson Cancer Center, Seattle, Washington
- Department of Epidemiology, University of Washington, Seattle, Washington
| | - Ashley Zehnder
- Exotic Species Cancer Research Alliance, North Carolina State College of Veterinary Medicine, Raleigh, North Carolina
| | - Trevor A. Graham
- Centre for Evolution and Cancer, Institute of Cancer Research, London, United Kingdom
| | - Brigid V. Troan
- North Carolina State College of Veterinary Medicine, Raleigh, North Carolina
- Exotic Species Cancer Research Alliance, North Carolina State College of Veterinary Medicine, Raleigh, North Carolina
- The North Carolina Zoo, Asheboro, North Carolina
| | - Tara M. Harrison
- North Carolina State College of Veterinary Medicine, Raleigh, North Carolina
- Exotic Species Cancer Research Alliance, North Carolina State College of Veterinary Medicine, Raleigh, North Carolina
| | - Marc Tollis
- School of Informatics, Computing and Cyber Systems, Northern Arizona University, Flagstaff, Arizona
| | - Joshua D. Schiffman
- Arizona Cancer Evolution Center, The Biodesign Institute, Arizona State University, Tempe, Arizona
- Department of Pediatrics and Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
- Peel Therapeutics, Inc., Salt Lake City, Utah
| | - C. Athena Aktipis
- Arizona Cancer Evolution Center, The Biodesign Institute, Arizona State University, Tempe, Arizona
- School of Life Sciences, Arizona State University, Tempe, Arizona
| | - Lisa M. Abegglen
- Arizona Cancer Evolution Center, The Biodesign Institute, Arizona State University, Tempe, Arizona
- Department of Pediatrics and Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
- Peel Therapeutics, Inc., Salt Lake City, Utah
| | - Carlo C. Maley
- Arizona Cancer Evolution Center, The Biodesign Institute, Arizona State University, Tempe, Arizona
- School of Life Sciences, Arizona State University, Tempe, Arizona
- Biodesign Center for Biocomputing, Security and Society, Arizona State University, Tempe, Arizona
| | - Amy M. Boddy
- Arizona Cancer Evolution Center, The Biodesign Institute, Arizona State University, Tempe, Arizona
- University of California Santa Barbara, Santa Barbara, California
| |
Collapse
|
41
|
Lange M, Wölk M, Doubravsky CE, Hendricks JM, Kato S, Otoki Y, Styler B, Nakagawa K, Fedorova M, Olzmann JA. FSP1-mediated lipid droplet quality control prevents neutral lipid peroxidation and ferroptosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.06.631537. [PMID: 39829838 PMCID: PMC11741373 DOI: 10.1101/2025.01.06.631537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Lipid droplets (LDs) are organelles that store and supply lipids based on cellular needs. While mechanisms preventing oxidative damage to membrane phospholipids are established, the vulnerability of LD neutral lipids to peroxidation and protective mechanisms are unknown. Here, we identify LD-localized Ferroptosis Suppressor Protein 1 (FSP1) as a critical regulator that prevents neutral lipid peroxidation by recycling coenzyme Q10 (CoQ10) to its lipophilic antioxidant form. Lipidomics reveal that FSP1 loss leads to the accumulation of oxidized triacylglycerols and cholesteryl esters, and biochemical reconstitution of FSP1 with CoQ10 and NADH suppresses triacylglycerol peroxidation in vitro. Notably, polyunsaturated fatty acid (PUFA)-rich triacylglycerols enhance cancer cell sensitivity to FSP1 loss and inducing PUFA-rich LDs triggers triacylglycerol peroxidation and LD-initiated ferroptosis when FSP1 activity is impaired. These findings uncover the first LD lipid quality control pathway, wherein LD-localized FSP1 maintains neutral lipid integrity to prevent the buildup of oxidized lipids and induction of ferroptosis.
Collapse
Affiliation(s)
- Mike Lange
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Michele Wölk
- Center of Membrane Biochemistry and Lipid Research, University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden Dresden, Germany
| | - Cody E. Doubravsky
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Joseph M. Hendricks
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Shunji Kato
- Laboratory of Food Function Analysis, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Yurika Otoki
- Laboratory of Food Function Analysis, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Benjamin Styler
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kiyotaka Nakagawa
- Laboratory of Food Function Analysis, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Maria Fedorova
- Center of Membrane Biochemistry and Lipid Research, University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden Dresden, Germany
| | - James A. Olzmann
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
42
|
Zhang S, Wang X, Gao X, Chen X, Li L, Li G, Liu C, Miao Y, Wang R, Hu K. Radiopharmaceuticals and their applications in medicine. Signal Transduct Target Ther 2025; 10:1. [PMID: 39747850 PMCID: PMC11697352 DOI: 10.1038/s41392-024-02041-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/30/2024] [Accepted: 10/28/2024] [Indexed: 01/04/2025] Open
Abstract
Radiopharmaceuticals involve the local delivery of radionuclides to targeted lesions for the diagnosis and treatment of multiple diseases. Radiopharmaceutical therapy, which directly causes systematic and irreparable damage to targeted cells, has attracted increasing attention in the treatment of refractory diseases that are not sensitive to current therapies. As the Food and Drug Administration (FDA) approvals of [177Lu]Lu-DOTA-TATE, [177Lu]Lu-PSMA-617 and their complementary diagnostic agents, namely, [68Ga]Ga-DOTA-TATE and [68Ga]Ga-PSMA-11, targeted radiopharmaceutical-based theranostics (radiotheranostics) are being increasingly implemented in clinical practice in oncology, which lead to a new era of radiopharmaceuticals. The new generation of radiopharmaceuticals utilizes a targeting vector to achieve the accurate delivery of radionuclides to lesions and avoid off-target deposition, making it possible to improve the efficiency and biosafety of tumour diagnosis and therapy. Numerous studies have focused on developing novel radiopharmaceuticals targeting a broader range of disease targets, demonstrating remarkable in vivo performance. These include high tumor uptake, prolonged retention time, and favorable pharmacokinetic properties that align with clinical standards. While radiotheranostics have been widely applied in tumor diagnosis and therapy, their applications are now expanding to neurodegenerative diseases, cardiovascular diseases, and inflammation. Furthermore, radiotheranostic-empowered precision medicine is revolutionizing the cancer treatment paradigm. Diagnostic radiopharmaceuticals play a pivotal role in patient stratification and treatment planning, leading to improved therapeutic outcomes in targeted radionuclide therapy. This review offers a comprehensive overview of the evolution of radiopharmaceuticals, including both FDA-approved and clinically investigated agents, and explores the mechanisms of cell death induced by radiopharmaceuticals. It emphasizes the significance and future prospects of theranostic-based radiopharmaceuticals in advancing precision medicine.
Collapse
Grants
- 82372002 National Natural Science Foundation of China (National Science Foundation of China)
- 0104002 Beijing Nova Program
- L248087; L234044 Natural Science Foundation of Beijing Municipality (Beijing Natural Science Foundation)
- Nonprofit Central Research Institute Fund of the Chinese Academy of Medical Sciences (No. 2022-RC350-04), the CAMS Innovation Fund for Medical Sciences (Nos. 2021-I2M-1-026, 2022-I2M-2-002-2, and 2021-I2M-3-001), the National Key Research and Development Program of China (No. 2022YFE0111700),the Fundamental Research Funds for the Central Universities (Nos. 3332023044 and 3332023151), the CIRP Open Fund of Radiation Protection Laboratories (No. ZHYLYB2021005), and the China National Nuclear Corporation Young Talent Program.
- Fundamental Research Funds for the Central Universities,Nos. 3332023044
- Fundamental Research Funds for the Central Universities,Nos. 3332023151
- he Nonprofit Central Research Institute Fund of Chinese Academy of Medical Sciences,No. 2022-RC350-04;the CAMS Innovation Fund for Medical Sciences,Nos. 2021-I2M-1-026, 2022-I2M-2-002-2, and 2021-I2M-3-001;the National Key Research and Development Program of China,No. 2022YFE0111700
Collapse
Affiliation(s)
- Siqi Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Xingkai Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Xin Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Xueyao Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Linger Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Guoqing Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Can Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Yuan Miao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Rui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China.
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 2019RU066, 730000, Lanzhou, China.
| | - Kuan Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China.
| |
Collapse
|
43
|
Yang TT, Zhou LH, Gu LF, Qian LL, Bao YL, Jing P, Sun JT, Du C, Shan TK, Wang SB, Wang WJ, Chen JY, Wang ZM, Wang H, Wang QM, Wang RX, Wang LS. CHK1 attenuates cardiac dysfunction via suppressing SIRT1-ubiquitination. Metabolism 2025; 162:156048. [PMID: 39454820 DOI: 10.1016/j.metabol.2024.156048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/21/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024]
Abstract
BACKGROUND Mitochondrial dysfunction is linked to myocardial ischemia-reperfusion (I/R) injury. Checkpoint kinase 1 (CHK1) could facilitate cardiomyocyte proliferation, however, its role on mitochondrial function in I/R injury remains unknown. METHODS To investigate the role of CHK1 on mitochondrial function following I/R injury, cardiomyocyte-specific knockout/overexpression mouse models were generated. Adult mouse cardiomyocytes (AMCMs) were isolated for in vitro study. Mass spectrometry-proteomics analysis and protein co-immunoprecipitation assays were conducted to dissect the molecular mechanism. RESULTS CHK1 was downregulated in myocardium post I/R and AMCMs post oxygen-glucose deprivation/re‑oxygenation (OGD/R). In vivo, CHK1 overexpression protected against I/R induced cardiac dysfunction, while heterogenous CHK1 knockout exacerbated cardiomyopathy. In vitro, CHK1 inhibited OGD/R-induced cardiomyocyte apoptosis and bolstered cardiomyocyte survival. Mechanistically, CHK1 attenuated oxidative stress and preserved mitochondrial metabolism in cardiomyocytes under I/R. Moreover, disrupted mitochondrial homeostasis in I/R myocardium was restored by CHK1 through the promotion of mitochondrial biogenesis and mitophagy. Through mass spectrometry analysis following co-immunoprecipitation, SIRT1 was identified as a direct target of CHK1. The 266-390 domain of CHK1 interacted with the 160-583 domain of SIRT1. Importantly, CHK1 phosphorylated SIRT1 at Thr530 residue, thereby inhibiting SMURF2-mediated degradation of SIRT1. The role of CHK1 in maintaining mitochondrial dynamics control and myocardial protection is abolished by SIRT1 inhibition, while inactivated mutation of SIRT1 Thr530 fails to reverse the impaired mitochondrial dynamics following CHK1 knockdown. CHK1 Δ390 amino acids (aa) mutant functioned similarly to full-length CHK1 in scavenging ROS and maintaining mitochondrial dynamics. Consistently, cardiac-specific SIRT1 knockdown attenuated the protective role of CHK1 in I/R injury. CONCLUSIONS Our findings revealed that CHK1 mitigates I/R injury and restores mitochondrial dynamics in cardiomyocytes through a SIRT1-dependent mechanism.
Collapse
Affiliation(s)
- Tong-Tong Yang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Liu-Hua Zhou
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Ling-Feng Gu
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Ling-Ling Qian
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical center, Nanjing Medical University, Wuxi 214023, China
| | - Yu-Lin Bao
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Peng Jing
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jia-Teng Sun
- Department of Cardiology, Drum Tower Hospital, Medical School of Nanjing University, No. 321 Zhongshan Road, Nanjing, China
| | - Chong Du
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Tian-Kai Shan
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Si-Bo Wang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Wen-Jing Wang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jia-Yi Chen
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Ze-Mu Wang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Hao Wang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Qi-Ming Wang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Ru-Xing Wang
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical center, Nanjing Medical University, Wuxi 214023, China.
| | - Lian-Sheng Wang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
44
|
Washif M, Kawasumi R, Hirota K. PrimPol-mediated repriming elicits gap-filling by template switching and promotes cellular tolerance to cidofovir. DNA Repair (Amst) 2025; 145:103787. [PMID: 39577201 DOI: 10.1016/j.dnarep.2024.103787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/27/2024] [Accepted: 11/12/2024] [Indexed: 11/24/2024]
Abstract
A nucleoside analog, Cidofovir (CDV), is used for the treatment of viral diseases such as cytomegalovirus retinitis and herpes virus infection. CDV converts to its active diphosphate metabolite (CDVpp) through cellular kinases and acts as a competitive inhibitor for viral polymerase thereby interfering with viral replication. However, the effect of this drug on the replication of healthy host cells and the mechanisms involved in the cellular tolerance to CDV are yet to be fully understood. In this study, we explored the mechanisms underlying cellular tolerance to CDV by screening mutant cell lines exhibiting hypersensitivity to CDV from a collection of DT40 mutants deficient in various genome maintenance systems. We identified Rad17 and PrimPol as critical factors for CDV tolerance. We found that Rad17 plays a pivotal role in activating intra-S phase checkpoint by the phosphorylation of Chk1, a vital checkpoint mediator. We showed that PrimPol, a factor involved in the release of stalled replication, plays critical roles in CDV tolerance in tandem with Rad17. We found that PrimPol deficient cells showed slower replication on the CDV-incorporated template strand than did wild-type cells, indicating a critical role of PrimPol in the continuous replication fork progression on the CDV-incorporated damaged template. PrimPol releases replication arrest with its DNA-damage bypass function and its repriming function, we thus investigated which PrimPol function is involved in CDV tolerance using the separation of function mutant genes of PRIMPOL. The CDV hypersensitive phenotype of PrimPol deficient cells was restored by PRIMPOLY89D (primase active / reduced polymerase activity), indicating that the repriming function of PrimPol is required for maintaining replication on the CDV-damaged template. Moreover, we found that the number of sister chromatid exchange (SCE) was reduced in PrimPol-deficient cells. These data indicate that gaps generated by PrimPol-mediated repriming on CDV-damaged templates promote post-replicative gap-filing by template switching.
Collapse
Affiliation(s)
- Mubasshir Washif
- Department of Chemistry, Graduate School of Science, Tokyo Metropolitan University, Minamiosawa 1-1, Hachioji-shi, Tokyo 192-0397, Japan
| | - Ryotaro Kawasumi
- Department of Chemistry, Graduate School of Science, Tokyo Metropolitan University, Minamiosawa 1-1, Hachioji-shi, Tokyo 192-0397, Japan
| | - Kouji Hirota
- Department of Chemistry, Graduate School of Science, Tokyo Metropolitan University, Minamiosawa 1-1, Hachioji-shi, Tokyo 192-0397, Japan.
| |
Collapse
|
45
|
DeWitt JT, Raghunathan M, Haricharan S. Nonrepair functions of DNA mismatch repair proteins: new avenues for precision oncology. Trends Cancer 2025; 11:49-61. [PMID: 39490324 PMCID: PMC12077842 DOI: 10.1016/j.trecan.2024.10.001] [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: 05/29/2024] [Revised: 10/01/2024] [Accepted: 10/07/2024] [Indexed: 11/05/2024]
Abstract
DNA damage repair (DDR) proteins are well recognized as guardians of the genome that are frequently lost during malignant transformation of normal cells across cancer types. To date, their tumor suppressor functions have been generally regarded as a consequence of their roles in maintaining genomic stability: more genomic instability increases the risk of oncogenic transformation events. However, recent discoveries centering around DNA mismatch repair (MMR) proteins suggest a broader impact of the loss of DDR proteins on cellular processes beyond genomic instability. Here, we explore the clinical implications of nonrepair roles for DDR proteins, using the growing evidence supporting roles for DNA MMR proteins in cell cycle and apoptosis regulation, metabolic function, the cellular secretome, and immunomodulation.
Collapse
Affiliation(s)
- Jerry Tyler DeWitt
- Department of Biology, San Diego State University, San Diego, CA, USA; Cancer Biology and Signaling Program, UCSD Moores Cancer Center, San Diego, CA, USA
| | - Megha Raghunathan
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Svasti Haricharan
- Department of Biology, San Diego State University, San Diego, CA, USA; Cancer Biology and Signaling Program, UCSD Moores Cancer Center, San Diego, CA, USA.
| |
Collapse
|
46
|
Fu Z, Wang W, Gao Y. Understanding the impact of ER stress on lung physiology. Front Cell Dev Biol 2024; 12:1466997. [PMID: 39744015 PMCID: PMC11688383 DOI: 10.3389/fcell.2024.1466997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 11/22/2024] [Indexed: 01/04/2025] Open
Abstract
Human lungs consist of a distinctive array of cell types, which are subjected to persistent challenges from chemical, mechanical, biological, immunological, and xenobiotic stress throughout life. The disruption of endoplasmic reticulum (ER) homeostatic function, triggered by various factors, can induce ER stress. To overcome the elevated ER stress, an adaptive mechanism known as the unfolded protein response (UPR) is activated in cells. However, persistent ER stress and maladaptive UPR can lead to defects in proteostasis at the cellular level and are typical features of the lung aging. The aging lung and associated lung diseases exhibit signs of ER stress-related disruption in cellular homeostasis. Dysfunction resulting from ER stress and maladaptive UPR can compromise various cellular and molecular processes associated with aging. Hence, comprehending the mechanisms of ER stress and UPR components implicated in aging and associated lung diseases could enable to develop appropriate therapeutic strategies for the vulnerable population.
Collapse
Affiliation(s)
- Zhiling Fu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Wei Wang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yuan Gao
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
47
|
Chauhan A, Kamal R, Bhatia R, Singh TG, Awasthi A. From Bench to Bedside: ROS-Responsive Nanocarriers in Cancer Therapy. AAPS PharmSciTech 2024; 26:10. [PMID: 39668268 DOI: 10.1208/s12249-024-03011-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 11/25/2024] [Indexed: 12/14/2024] Open
Abstract
Reactive oxygen species (ROS) play a dual role in cancer, acting as both signaling molecules that promote tumour growth and as agents that can inhibit tumour progression through cytotoxic effects. In cancer therapy, ROS-responsive drug delivery systems take advantage of the elevated ROS levels found in tumors compared to healthy tissues. These systems are engineered to release drugs precisely in response to increased ROS levels in tumour cells, allowing targeted and controlled treatment, minimizing side effects, and enhancing therapeutic outcomes. ROS generation in cancer cells is linked to metabolic changes, mitochondrial dysfunction, and oncogenic signaling, leading to increased oxidative stress. Tumour cells manage this by upregulating antioxidant defenses to prevent ROS from reaching harmful levels. This balance between ROS production and neutralization is critical for cancer cell survival, making ROS both a challenge and an opportunity for targeted therapies. ROS also connect inflammation and cancer. Chronic inflammation leads to elevated ROS, which can damage DNA and proteins, promoting mutations and cancer development. Additionally, ROS contribute to protein degradation, affecting essential cellular functions. Therapeutic strategies targeting ROS aim to either increase ROS beyond tolerable levels for cancer cells or inhibit their antioxidant defenses. Nanocarriers responsive to ROS show great potential in improving the precision of cancer treatments by releasing drugs specifically in high ROS environments, like tumors. This review discusses the mechanisms of ROS in cancer, its role in inflammation and protein degradation, and the advances in ROS-targeted nanocarrier therapies across different cancer types.
Collapse
Affiliation(s)
- Abhishek Chauhan
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Raj Kamal
- School of Pharmacy, Desh Bhagat University, 147301, Punjab, India, Mandi Gobindgarh
| | - Rohit Bhatia
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | | | - Ankit Awasthi
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| |
Collapse
|
48
|
Lim HE, Lim HJ, Yoo HY. Interaction of DDB1 with NBS1 in a DNA Damage Checkpoint Pathway. Int J Mol Sci 2024; 25:13097. [PMID: 39684807 DOI: 10.3390/ijms252313097] [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: 10/10/2024] [Revised: 11/19/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
Various DNA damage checkpoint control mechanisms in eukaryotic cells help maintain genomic integrity. Among these, NBS1, a key component of the MRE11-RAD50-NBS1 (MRN) complex, is an essential protein involved in the DNA damage response (DDR). In this study, we discovered that DNA damage-binding protein 1 (DDB1) interacts with NBS1. DDB1 is a DDR sensor protein found in UV-induced DNA replication blocks. Through pull-down and immunoprecipitation assays conducted in Xenopus egg extracts and human cell lines, we demonstrated a specific interaction between NBS1 and DDB1. DDB1 was also found to associate with several proteins that interact with NBS1, including DNA topoisomerase 2-binding protein 1 (TopBP1) and Mediator of DNA damage checkpoint protein 1 (MDC1). Notably, the interaction between DDB1 and NBS1 is disrupted in MDC1-depleted egg extracts, indicating that MDC1 is necessary for this interaction. Furthermore, the depletion of DDB1 leads to increased Chk1 activation upon DNA damage. These novel findings regarding the interaction between NBS1 and DDB1 provide new insights into how DDB1 regulates DNA damage pathways.
Collapse
Affiliation(s)
- Hoe Eun Lim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, Republic of Korea
- Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Hee Jung Lim
- Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Hae Yong Yoo
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, Republic of Korea
- Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Republic of Korea
| |
Collapse
|
49
|
Kitaoka M, Yamashita YM. Running the gauntlet: challenges to genome integrity in spermiogenesis. Nucleus 2024; 15:2339220. [PMID: 38594652 PMCID: PMC11005813 DOI: 10.1080/19491034.2024.2339220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 04/02/2024] [Indexed: 04/11/2024] Open
Abstract
Species' continuity depends on gametogenesis to produce the only cell types that can transmit genetic information across generations. Spermiogenesis, which encompasses post-meiotic, haploid stages of male gametogenesis, is a process that leads to the formation of sperm cells well-known for their motility. Spermiogenesis faces three major challenges. First, after two rounds of meiotic divisions, the genome lacks repair templates (no sister chromatids, no homologous chromosomes), making it incredibly vulnerable to any genomic insults over an extended time (typically days-weeks). Second, the sperm genome becomes transcriptionally silent, making it difficult to respond to new perturbations as spermiogenesis progresses. Third, the histone-to-protamine transition, which is essential to package the sperm genome, counterintuitively involves DNA break formation. How spermiogenesis handles these challenges remains poorly understood. In this review, we discuss each challenge and their intersection with the biology of protamines. Finally, we discuss the implication of protamines in the process of evolution.
Collapse
Affiliation(s)
- Maiko Kitaoka
- Whitehead Institute for Biomedical Research and Howard Hughes Medical Institute, Cambridge, MA, USA
| | - Yukiko M. Yamashita
- Whitehead Institute for Biomedical Research and Howard Hughes Medical Institute, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
50
|
Mackova V, Raudenska M, Polanska HH, Jakubek M, Masarik M. Navigating the redox landscape: reactive oxygen species in regulation of cell cycle. Redox Rep 2024; 29:2371173. [PMID: 38972297 PMCID: PMC11637001 DOI: 10.1080/13510002.2024.2371173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2024] Open
Abstract
Objectives: To advance our knowledge of disease mechanisms and therapeutic options, understanding cell cycle regulation is critical. Recent research has highlighted the importance of reactive oxygen species (ROS) in cell cycle regulation. Although excessive ROS levels can lead to age-related pathologies, ROS also play an essential role in normal cellular functions. Many cell cycle regulatory proteins are affected by their redox status, but the precise mechanisms and conditions under which ROS promote or inhibit cell proliferation are not fully understood.Methods: This review presents data from the scientific literature and publicly available databases on changes in redox state during the cell cycle and their effects on key regulatory proteins.Results: We identified redox-sensitive targets within the cell cycle machinery and analysed different effects of ROS (type, concentration, duration of exposure) on cell cycle phases. For example, moderate levels of ROS can promote cell proliferation by activating signalling pathways involved in cell cycle progression, whereas excessive ROS levels can induce DNA damage and trigger cell cycle arrest or cell death.Discussion: Our findings encourage future research focused on identifying redox-sensitive targets in the cell cycle machinery, potentially leading to new treatments for diseases with dysregulated cell proliferation.
Collapse
Affiliation(s)
- Viktoria Mackova
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Martina Raudenska
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Hana Holcova Polanska
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Milan Jakubek
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Michal Masarik
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic
- Institute of Pathophysiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|