1
|
Crisol BM, Rocha MB, Franco B, Morelli AP, Katashima CK, Junior SJA, Carneiro FS, Braga RR, Brícola RS, de Azambuja G, Costa RG, Esteves AM, Mori MA, Oliveira MCG, Cintra DE, Pauli JR, Larsen FJ, da Silva ASR, Ropelle ER. Excessive exercise elicits poly (ADP-ribose) Polymerase-1 activation and global protein PARylation driving muscle dysfunction and performance impairment. Mol Metab 2025; 96:102135. [PMID: 40187729 PMCID: PMC12018203 DOI: 10.1016/j.molmet.2025.102135] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/27/2025] [Accepted: 03/29/2025] [Indexed: 04/07/2025] Open
Abstract
Excessive exercise combined with inadequate recovery time may trigger fatigue, performance impairment, and ultimately the overtraining syndrome. The intramyocellular mechanisms involved in the overtraining syndrome remain only partially known. Here, we combined multi-omics analyses from isogenic BXD mouse strains with a mouse model of overtraining and excessive exercise protocol in mice and humans to evaluate the molecular mechanism involved in the performance impairment induced by excessive exercise. We identified that BXD mouse strains with elevated levels of Parp1 gene expression in the skeletal muscle displayed features like overtraining syndrome and abnormal muscle genetic signature. High PARP1 protein content and aberrant PARylation of proteins were detected in the skeletal muscle of overtrained, but not in trained mice. Overtraining syndrome reduced mitochondrial function promoted by exercise training, induced muscle hyperalgesia, reduced muscle fiber size and promoted a similar gene signature of myopathy and atrophy models. Short periods of excessive exercise also increased PARylation in the skeletal muscle of mice and healthy subjects. The pharmacological inhibition of PARP1, using Olaparib, and genetic Parp1 ablation, preserved muscle fiber morphology and protected against physical performance impairment and other symptoms of the overtraining syndrome in mice. In conclusion, PARP1 excessive activation is related to muscle abnormalities led by long or short periods of excessive exercise, and here we suggest that PARP1 is a potential target in the treatment and prevention of overtraining syndrome.
Collapse
Affiliation(s)
- Barbara M Crisol
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), Limeira, SP, 13484-350, Brazil
| | - Matheus B Rocha
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), Limeira, SP, 13484-350, Brazil
| | - Beatriz Franco
- School of Applied Sciences, University of Campinas, Limeira, SP, Brazil
| | - Ana Paula Morelli
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), Limeira, SP, 13484-350, Brazil
| | - Carlos K Katashima
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), Limeira, SP, 13484-350, Brazil
| | - Scylas J A Junior
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), Limeira, SP, 13484-350, Brazil
| | - Fernanda S Carneiro
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), Limeira, SP, 13484-350, Brazil
| | - Renata R Braga
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), Limeira, SP, 13484-350, Brazil
| | - Rafael S Brícola
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), Limeira, SP, 13484-350, Brazil
| | | | - Raul Gobato Costa
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Brazil
| | - Andrea M Esteves
- School of Applied Sciences, University of Campinas, Limeira, SP, Brazil
| | - Marcelo A Mori
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Brazil
| | | | - Dennys E Cintra
- Laboratory of Nutritional Genomic, School of Applied Sciences, University of Campinas (UNICAMP), Limeira, SP, 13484-350, Brazil; Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Brazil
| | - José R Pauli
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), Limeira, SP, 13484-350, Brazil; School of Applied Sciences, University of Campinas, Limeira, SP, Brazil; Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Brazil
| | - Filip J Larsen
- The Swedish School of Sport and Health Sciences, GIH, Åstrand Laboratory, Department of Physiology, Nutrition and Biomechanics, Stockholm, 114 33, Sweden
| | - Adelino S R da Silva
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil; School of Physical Education and Sport of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Eduardo R Ropelle
- Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Sciences, University of Campinas (UNICAMP), Limeira, SP, 13484-350, Brazil; School of Applied Sciences, University of Campinas, Limeira, SP, Brazil; Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Brazil.
| |
Collapse
|
2
|
Hu Y, Meng Y, Zhuang Z, Li Y, Nan J, Xu N, Ye Z, Jing J. Prospects for PARG inhibitors in cancer therapy. J Mol Cell Biol 2025; 16:mjae050. [PMID: 39668635 PMCID: PMC12123320 DOI: 10.1093/jmcb/mjae050] [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: 08/27/2024] [Revised: 11/22/2024] [Accepted: 12/11/2024] [Indexed: 12/14/2024] Open
Abstract
Poly(ADP-ribose) glycosylhydrolase (PARG) is an enzyme involved in hydrolyzing the ribose-ribose bonds present in poly(ADP-ribose) (PAR), which are primarily found in the nucleus. Along with poly(ADP-ribose) polymerase, PARG regulates the level of PAR in cells, playing a crucial role in DNA maintenance and repair processes. Recent studies have revealed elevated levels of PARG in various cancers, such as breast, liver, prostate, and esophageal cancers, indicating a link to unfavorable cancer outcomes. PARG is a significant molecular target for treating PAR-related cancers. This review provides a comprehensive overview of the physiological role of PARG and the development of its inhibitors, highlighting its potential as an innovative target for cancer treatment.
Collapse
Affiliation(s)
- Yangchan Hu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Yuxin Meng
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Zirui Zhuang
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences (UCAS), Hangzhou 310024, China
| | - Yuancong Li
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Junjun Nan
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Ning Xu
- Institute of Drug Development & Chemical Biology, College of Pharmaceutical Science, Zhejiang University of Technology, Huzhou 313200, China
| | - Zu Ye
- Gastric Cancer Center, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HlM), Chinese Academy of Sciences, Hangzhou 310022, China
- Zhejiang Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Hangzhou 310022, China
| | - Ji Jing
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
- Zhejiang Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer, Hangzhou 310022, China
| |
Collapse
|
3
|
Chen J, Yue L, Pan Y, Jiang B, Wan J, Lin H, Guo F, Li H, Li Y, Zhao Q. Novel Cyano-Artemisinin Dimer ZQJ29 Targets PARP1 to Induce Ferroptosis in Pancreatic Cancer Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e01935. [PMID: 40387570 DOI: 10.1002/advs.202501935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/29/2025] [Indexed: 05/20/2025]
Abstract
Pancreatic cancer remains one of the most lethal malignancies in the digestive system, with limited available drugs and a need for improved efficacy. This unmet clinical need highlights the urgency to discover novel, highly efficient small-molecule compounds. Herein, a novel cyano-containing artemisinin dimer derivative, ZQJ29, is synthesized through structural modifications of artemisinin. Biological evaluation demonstrated that ZQJ29 effectively inhibits the proliferation of pancreatic cancer cells both in vitro and in vivo. ZQJ29 selectively targets PARP1 and has distinct structural features comparable to established PARP1 inhibitors such as Olaparib. Notably, ZQJ29 is the first reported artemisinin derivative to inhibit PARP1. Furthermore, the inhibition of PARP1 by ZQJ29 enhances the expression of TP53 and inhibits the SLC7A11/GPX4 pathway. The work first demonstrates that targeting PARP1 can induce ferroptosis in pancreatic cancer. These findings not only identify promising artemisinin derivatives for the development of therapies targeting pancreatic cancer but also provide scientific evidence supporting therapeutic strategies aimed at inducing ferroptosis in pancreatic cancer. This research lays a robust foundation for subsequent preclinical studies.
Collapse
Affiliation(s)
- Jianping Chen
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Shanghai Frontiers Science Center for TCM Chemical Biology, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lingyun Yue
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Shanghai Frontiers Science Center for TCM Chemical Biology, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Chemistry, College of Sciences, Shanghai University, Shanghai, 200444, China
| | - Yanna Pan
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Shanghai Frontiers Science Center for TCM Chemical Biology, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Bingying Jiang
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Shanghai Frontiers Science Center for TCM Chemical Biology, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Junfeng Wan
- College of Mathematics and Physics, Shanghai University of Electric Power, Shanghai, 201306, China
| | - Haixia Lin
- Department of Chemistry, College of Sciences, Shanghai University, Shanghai, 200444, China
| | - Fujiang Guo
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Huiyu Li
- College of Mathematics and Physics, Shanghai University of Electric Power, Shanghai, 201306, China
| | - Yajuan Li
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Qingjie Zhao
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Shanghai Frontiers Science Center for TCM Chemical Biology, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
4
|
Yang Y, Li Z, Yu X, Zheng Y, Yu Y, Yang M, Zhang N. WDR11-DT enhances radiosensitivity via promoting PARP1 degradation and homologous recombination deficiency. Cancer Lett 2025; 625:217757. [PMID: 40320037 DOI: 10.1016/j.canlet.2025.217757] [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: 12/25/2024] [Revised: 04/08/2025] [Accepted: 04/28/2025] [Indexed: 05/07/2025]
Abstract
Radiotherapy is an important management for non-small cell lung cancer (NSCLC). Although long non-coding RNAs (lncRNAs) have been reported to be involved in modulating radiosensitivity, the underlying mechanisms are still largely unclear. Here, we found that tumor suppressor WDR11-DT is a novel radiation-induced lncRNA, which is transcriptionally regulated by SPDEF, in NSCLC. In contrast to normal tissues, WDR11-DT is down-regulated in NSCLC specimens and its low expression was associated with poor prognosis of patient receiving radiotherapy. Importantly, WDR11-DT can markedly enhance NSCLC cells' radiosensitivity in vitro and in vivo. WDR11-DT functions through distinct mechanisms via binding different proteins. WDR11-DT facilitates interactions between PARP1 and its E3 ligase TRIP12, promotes PARP1 protein degradation and suppresses PARP1-controlled Single-strand breaks (SSBs) repair. Additionally, WDR11-DT binds RNA-bind protein HNRNPK, represses its functions in improving RNA stability of homologous recombination (HR) genes, decreases expression of BRCA1, ATM, BLM and RAD50, and suppresses radiotherapy-triggered HR repair. WDR11-DT-induced dual restraints of PARP1 and the HR pathway lead to the accumulation of double-strand breaks as well as synthetic lethal effects of malignant cells, which, thereby, enhances radiosensitivity and inhibits progression of lung cancer. These results extend our current knowledge of radio-biology and elucidate that WDR11-DT may be a new target for boosting cancer radiotherapy.
Collapse
Affiliation(s)
- Yanting Yang
- Shandong Provincial Key Laboratory of Precision Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China
| | - Zongcheng Li
- Shandong Provincial Key Laboratory of Precision Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China
| | - Xinyuan Yu
- Shandong Provincial Key Laboratory of Precision Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China
| | - Yanxiu Zheng
- Shandong Provincial Key Laboratory of Precision Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China
| | - Yongquan Yu
- Department of Radiology, Weihai Central Hospital, Qingdao University, Weihai, Shandong, China
| | - Ming Yang
- Shandong Provincial Key Laboratory of Precision Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China; School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, Shandong Province, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Nasha Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
5
|
Kulkarni S, Seneviratne N, Tosun Ç, Madhusudan S. PARP inhibitors in ovarian cancer: Mechanisms of resistance and implications to therapy. DNA Repair (Amst) 2025; 149:103830. [PMID: 40203475 DOI: 10.1016/j.dnarep.2025.103830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 04/11/2025]
Abstract
Advanced epithelial ovarian cancer of the high-grade serous subtype (HGSOC) remains a significant clinical challenge due to the development of resistance to current platinum-based chemotherapies. PARP1/2 inhibitors (PARPi) exploit the well-characterised homologous recombination repair deficiency (HRD) in HGSOC and offer an effective targeted approach to treatment. Several clinical trials demonstrated that PARPi (olaparib, rucaparib, niraparib) significantly improved progression-free survival (PFS) in HGSOC in the recurrent maintenance setting. However, 40-70 % of patients develop Resistance to PARPi presenting an ongoing challenge in the clinic. Therefore, there is an unmet need for novel targeted therapies and biomarkers to identify intrinsic or acquired resistance to PARPi in ovarian cancer. Understanding the mechanisms of resistance to PARPi is crucial for identifying molecular vulnerabilities, developing effective biomarkers for patient stratification and guiding treatment decisions. Here, we summarise the current landscape of mechanisms associated with PARPi resistance such as restored homologous recombination repair functionality, replication fork stability and alterations to PARP1 and PARP2 and the DNA damage response. We highlight the role of circulating tumour DNA (ctDNA) in identifying acquired resistance biomarkers and its potential in guiding 'real-time' treatment decisions. Moreover, we explore other innovative treatment strategies aimed at overcoming specific resistance mechanisms, including the inhibition of ATR, WEE1 and POLQ. We also examine the role of PARPi rechallenge in patients with acquired resistance.
Collapse
Affiliation(s)
- Sanat Kulkarni
- Medical Sciences Division, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | | | - Çağla Tosun
- Naaz-Coker Ovarian Cancer Research Centre, Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham NG7 3RD, UK
| | - Srinivasan Madhusudan
- Naaz-Coker Ovarian Cancer Research Centre, Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham NG7 3RD, UK; Department of Oncology, Nottingham University Hospitals, Nottingham NG51PB, UK.
| |
Collapse
|
6
|
Li H, Chatla S, Liu X, Tian Z, Vekariya U, Wang P, Kim D, Octaviani S, Lian Z, Morton G, Feng Z, Yang D, Sullivan-Reed K, Childers W, Yu X, Chitrala KN, Madzo J, Skorski T, Huang J. ZNF251 haploinsufficiency confers PARP inhibitors resistance in BRCA1-mutated cancer cells through activation of homologous recombination. Cancer Lett 2025; 613:217505. [PMID: 39892701 DOI: 10.1016/j.canlet.2025.217505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 01/21/2025] [Accepted: 01/24/2025] [Indexed: 02/04/2025]
Abstract
Poly (ADP-ribose) polymerase inhibitors (PARPis) represent a promising new class of agents that have demonstrated efficacy in treating various cancers, particularly those with BRCA1/2 mutations. Cancer-associated BRCA1/2 mutations disrupt DNA double-strand break (DSB) repair via homologous recombination (HR). PARP inhibitors (PARPis) have been used to trigger synthetic lethality in BRCA1/2-mutated cancer cells by promoting the accumulation of toxic DSBs. Unfortunately, resistance to PARPis is common and can occur through multiple mechanisms, including the restoration of HR and/or stabilization of replication forks. To gain a better understanding of the mechanisms underlying PARPis resistance, we conducted an unbiased CRISPR-pooled genome-wide library screen to identify new genes whose deficiency confers resistance to the PARPi olaparib. Our research revealed that haploinsufficiency of the ZNF251 gene, which encodes zinc finger protein 251, is associated with resistance to PARPis in various breast and ovarian cancer cell lines carrying BRCA1 mutations. Mechanistically, we discovered that ZNF251 haploinsufficiency leads to stimulation of RAD51-mediated HR repair of DSBs in olaparib-treated BRCA1-mutated cancer cells. Moreover, we demonstrated that a RAD51 inhibitor reversed PARPi resistance in ZNF251 haploinsufficient cancer cells harboring BRCA1 mutations. Our findings provide important insights into the mechanisms underlying PARPis resistance by highlighting the role of RAD51 in this phenomenon.
Collapse
Affiliation(s)
- Huan Li
- Coriell Institute for Medical Research, Camden, NJ, United States
| | - Srinivas Chatla
- Fels Cancer Institute for Personalized Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Xiaolei Liu
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Zhen Tian
- Coriell Institute for Medical Research, Camden, NJ, United States
| | - Umeshkumar Vekariya
- Fels Cancer Institute for Personalized Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Peng Wang
- Coriell Institute for Medical Research, Camden, NJ, United States
| | - Dongwook Kim
- Coriell Institute for Medical Research, Camden, NJ, United States
| | - Stacia Octaviani
- Coriell Institute for Medical Research, Camden, NJ, United States
| | - Zhaorui Lian
- Coriell Institute for Medical Research, Camden, NJ, United States
| | - George Morton
- Moulder Center for Drug Discovery Research, School of Pharmacy, Department of Pharmaceutical Sciences, Temple University, Philadelphia, PA, United States
| | - Zijie Feng
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Institute of Diabetes, Obesity, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Dan Yang
- Coriell Institute for Medical Research, Camden, NJ, United States
| | - Katherine Sullivan-Reed
- Fels Cancer Institute for Personalized Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Wayne Childers
- Moulder Center for Drug Discovery Research, School of Pharmacy, Department of Pharmaceutical Sciences, Temple University, Philadelphia, PA, United States
| | - Xiang Yu
- Shanghai Jiao Tong University, School of Life Science and Biotechnology, Shanghai, PR China
| | - Kumaraswamy Naidu Chitrala
- Fels Cancer Institute for Personalized Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Jozef Madzo
- Coriell Institute for Medical Research, Camden, NJ, United States
| | - Tomasz Skorski
- Fels Cancer Institute for Personalized Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States; Department of Cancer and Cellular Biology, Temple University Lewis Katz School of Medicine, Philadelphia, PA, United States; Nuclear Dynamics Cancer Program, Fox Chase Cancer Center, Philadelphia, PA, United States.
| | - Jian Huang
- Coriell Institute for Medical Research, Camden, NJ, United States; Temple University Lewis Katz School of Medicine, Center for Metabolic Disease Research, Philadelphia, PA, United States; Cooper Medical School of Rowan University, Camden, NJ, United States.
| |
Collapse
|
7
|
Chen YH, Hsieh CY, Chiou CT, Caro EJGV, Tayo LL, Tsai PW. In Vitro and In Silico Studies on the Anti-H1N1 Activity of Bioactive Compounds from Marine-Derived Streptomyces ardesiacus. Mar Drugs 2025; 23:149. [PMID: 40278270 PMCID: PMC12028705 DOI: 10.3390/md23040149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/26/2025] Open
Abstract
This study explores the potential anti-H1N1 Influenza A activity of bioactive compounds extracted from Streptomyces ardesiacus, a marine-derived microorganism known for producing diverse secondary metabolites. Four major compounds-1-acetyl-β-carboline, 1H-indole-3-carbaldehyde, anthranilic acid, and indole-3-carboxylic acid-were isolated and characterized through NMR. Among these, the identified structure of 1-acetyl-β-carboline showed the highest IC50 effect, with a dose of 9.71 μg/mL in anti-influenza assays. Using network pharmacology and molecular docking analyses, the interactions of these compounds with key proteins involved in H1N1 pathogenesis were examined. Protein-protein interaction (PPI) networks and Gene Ontology enrichment analysis revealed CDC25B, PARP1, and PTGS2 as key targets, associating these compounds with pathways related to catalytic activity, inflammation, and cell cycle regulation. The molecular docking results demonstrated that 1-acetyl-β-carboline exhibited binding affinities comparable to Tamiflu, the positive control drug, with LibDock scores of 81.89, 77.49, and 89.21 for CDC25B, PARP1, and PTGS2, respectively, compared to Tamiflu's scores of 84.34, 86.13, and 91.29. These findings highlight the potential of the active compound 1-acetyl-β-carboline from S. ardesiacus as a novel anti-influenza agent, offering insights into their molecular mechanisms of action. The results support further in vitro and in vivo studies to validate the observed inhibitory mechanisms and therapeutic applications against H1N1 Influenza A.
Collapse
Affiliation(s)
- Yung-Husan Chen
- Xiamen Key Laboratory of Natural Products Resources of Marine Medicine, Xiamen Medical College, Xiamen 361023, China;
- Fujian Provincial University Marine Biomedical Resources Engineering Research Center, Xiamen Medical College, Xiamen 361023, China
| | - Cheng-Yang Hsieh
- Department of Food Science, National Taiwan Ocean University, Keelung 202, Taiwan;
- Department of Chemical and Materials Engineering, National I-Lan University, Yilan 260, Taiwan
| | - Chun-Tang Chiou
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei 112, Taiwan;
| | - Engelo John Gabriel V. Caro
- School of Chemical, Biological, and Materials Engineering and Sciences, Mapúa University, Manila 1002, Philippines;
| | - Lemmuel L. Tayo
- Department of Biology, School of Health Sciences, Mapúa University, Makati 1200, Philippines
| | - Po-Wei Tsai
- Department of Food Science, National Taiwan Ocean University, Keelung 202, Taiwan;
| |
Collapse
|
8
|
Zhu T, Tan JZA, Zhang L, Huang H, Das SS, Cheng F, Padmanabhan P, Jones MJK, Lee M, Lee A, Widagdo J, Anggono V. FTO suppresses DNA repair by inhibiting PARP1. Nat Commun 2025; 16:2925. [PMID: 40133293 PMCID: PMC11937437 DOI: 10.1038/s41467-025-58309-0] [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: 07/06/2023] [Accepted: 03/17/2025] [Indexed: 03/27/2025] Open
Abstract
Maintaining genomic integrity and faithful transmission of genetic information is essential for the survival and proliferation of cells and organisms. DNA damage, which threatens the integrity of the genome, is rapidly sensed and repaired by mechanisms collectively known as the DNA damage response. The RNA demethylase FTO has been implicated in this process; however, the underlying mechanism by which FTO regulates DNA repair remains unclear. Here, we use an unbiased quantitative proteomic approach to identify the proximal interactome of endogenous FTO protein. Our results demonstrate a direct interaction with the DNA damage sensor protein PARP1, which dissociates upon ultraviolet stimulation. FTO inhibits PARP1 catalytic activity and controls its clustering in the nucleolus. Loss of FTO enhances PARP1 enzymatic activity and the rate of PARP1 recruitment to DNA damage sites, accelerating DNA repair and promoting cell survival. Interestingly, FTO regulates PARP1 function and DNA damage response independent of its catalytic activity. We conclude that FTO is an endogenous negative regulator of PARP1 and the DNA damage response in cells beyond its role as an RNA demethylase.
Collapse
Affiliation(s)
- Tianyi Zhu
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, Brisbane, Australia
| | - Jing Zhi Anson Tan
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, Brisbane, Australia
| | - Lingrui Zhang
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, Brisbane, Australia
| | - He Huang
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, Brisbane, Australia
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Sooraj S Das
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, Brisbane, Australia
| | - Flora Cheng
- Motor Neuron Disease Research Centre, Macquarie Medical School, Faculty of Medicine Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Pranesh Padmanabhan
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, Brisbane, Australia
- School of Biomedical Sciences, Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, Brisbane, Australia
- NHMRC Centre for Research Excellence in Mechanisms in NeuroDegeneration - Alzheimer's Disease (MIND-AD CRE), Brisbane, Australia
| | - Mathew J K Jones
- Frazer Institute, Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, Brisbane, Australia
- School of Chemistry & Molecular Biosciences, Faculty of Science, The University of Queensland, Brisbane, Australia
| | - Mihwa Lee
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
- School of Chemistry, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Australia
| | - Albert Lee
- Motor Neuron Disease Research Centre, Macquarie Medical School, Faculty of Medicine Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Jocelyn Widagdo
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, Brisbane, Australia.
| | - Victor Anggono
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, Faculty of Health, Medicine and Behavioural Sciences, The University of Queensland, Brisbane, Australia.
- NHMRC Centre for Research Excellence in Mechanisms in NeuroDegeneration - Alzheimer's Disease (MIND-AD CRE), Brisbane, Australia.
| |
Collapse
|
9
|
Zhu Z, Shi Y. Poly (ADP-ribose) polymerase inhibitors in cancer therapy. Chin Med J (Engl) 2025; 138:634-650. [PMID: 39932206 PMCID: PMC11925422 DOI: 10.1097/cm9.0000000000003471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Indexed: 03/17/2025] Open
Abstract
ABSTRACT Poly(ADP-ribose) polymerase (PARP) inhibitors (PARPis) have emerged as critical agents for cancer therapy. By inhibiting the catalytic activity of PARP enzymes and trapping them in the DNA, PARPis disrupt DNA repair, ultimately leading to cell death, particularly in cancer cells with homologous recombination repair deficiencies, such as those harboring BRCA mutations. This review delves into the mechanisms of action of PARPis in anticancer treatments, including the inhibition of DNA repair, synthetic lethality, and replication stress. Furthermore, the clinical applications of PARPis in various cancers and their adverse effects as well as their combinations with other therapies and the mechanisms underlying resistance are summarized. This review provides comprehensive insights into the role and mechanisms of PARP and PARPis in DNA repair, with a particular focus on the potential of PARPi-based therapies in precision medicine for cancer treatment.
Collapse
Affiliation(s)
- Ziqi Zhu
- Department of Pathology & Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yujun Shi
- Department of Pathology & Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
10
|
Chiolo I, Altmeyer M, Legube G, Mekhail K. Nuclear and genome dynamics underlying DNA double-strand break repair. Nat Rev Mol Cell Biol 2025:10.1038/s41580-025-00828-1. [PMID: 40097581 DOI: 10.1038/s41580-025-00828-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2025] [Indexed: 03/19/2025]
Abstract
Changes in nuclear shape and in the spatial organization of chromosomes in the nucleus commonly occur in cancer, ageing and other clinical contexts that are characterized by increased DNA damage. However, the relationship between nuclear architecture, genome organization, chromosome stability and health remains poorly defined. Studies exploring the connections between the positioning and mobility of damaged DNA relative to various nuclear structures and genomic loci have revealed nuclear and cytoplasmic processes that affect chromosome stability. In this Review, we discuss the dynamic mechanisms that regulate nuclear and genome organization to promote DNA double-strand break (DSB) repair, genome stability and cell survival. Genome dynamics that support DSB repair rely on chromatin states, repair-protein condensates, nuclear or cytoplasmic microtubules and actin filaments, kinesin or myosin motor proteins, the nuclear envelope, various nuclear compartments, chromosome topology, chromatin loop extrusion and diverse signalling cues. These processes are commonly altered in cancer and during natural or premature ageing. Indeed, the reshaping of the genome in nuclear space during DSB repair points to new avenues for therapeutic interventions that may take advantage of new cancer cell vulnerabilities or aim to reverse age-associated defects.
Collapse
Affiliation(s)
- Irene Chiolo
- Department of Molecular and Computational Biology, University of Southern California, Los Angeles, CA, USA.
| | - Matthias Altmeyer
- Department of Molecular Mechanisms of Disease, University of Zurich (UZH), Zurich, Switzerland.
| | - Gaëlle Legube
- MCD, Centre de Biologie Intégrative (CBI), CNRS, Université de Toulouse, UT3, Toulouse, France.
| | - Karim Mekhail
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
11
|
Matsumoto K, Matsumoto Y, Wada J. PARylation-mediated post-transcriptional modifications in cancer immunity and immunotherapy. Front Immunol 2025; 16:1537615. [PMID: 40134437 PMCID: PMC11933034 DOI: 10.3389/fimmu.2025.1537615] [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: 12/01/2024] [Accepted: 02/17/2025] [Indexed: 03/27/2025] Open
Abstract
Poly-ADP-ribosylation (PARylation) is a post-translational modification in which ADP-ribose is added to substrate proteins. PARylation is mediated by a superfamily of ADP-ribosyl transferases known as PARPs and influences a wide range of cellular functions, including genome integrity maintenance, and the regulation of proliferation and differentiation. We and others have recently reported that PARylation of SH3 domain-binding protein 2 (3BP2) plays a role in bone metabolism, immune system regulation, and cytokine production. Additionally, PARylation has recently gained attention as a target for cancer treatment. In this review, we provide an overview of PARylation, its involvement in several signaling pathways related to cancer immunity, and the potential of combination therapies with PARP inhibitors and immune checkpoint inhibitors.
Collapse
Affiliation(s)
| | - Yoshinori Matsumoto
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Faculty of
Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | | |
Collapse
|
12
|
Chen K, Wang S, Fu S, Kim J, Park P, Liu R, Lei K. 4(3 H)-Quinazolinone: A Natural Scaffold for Drug and Agrochemical Discovery. Int J Mol Sci 2025; 26:2473. [PMID: 40141117 PMCID: PMC11941892 DOI: 10.3390/ijms26062473] [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: 02/13/2025] [Revised: 03/09/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
4(3H)-quinazolinone is a functional scaffold that exists widely both in natural products and synthetic organic compounds. Its drug-like derivatives have been extensively synthesized with interesting biological features including anticancer, anti-inflammatory, antiviral, antimalarial, antibacterial, antifungal, and herbicidal, etc. In this review, we highlight the medicinal and agrochemical versatility of the 4(3H)-quinazolinone scaffold according to the studies published in the past six years (2019-2024), and comprehensively give a summary of the target recognition, structure-activity relationship, and mechanism of its analogs. The present review is expected to provide valuable guidance for discovering novel lead compounds containing 4(3H)-quinazolinone moiety in both drug and agrochemical research.
Collapse
Affiliation(s)
- Ke Chen
- Department of Biotechnology, The University of Suwon, Hwaseong-si 18323, Gyeonggi-do, Republic of Korea; (K.C.); (J.K.); (P.P.)
| | - Shumin Wang
- School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng 252059, China; (S.W.); (S.F.)
| | - Shuyue Fu
- School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng 252059, China; (S.W.); (S.F.)
| | - Junehyun Kim
- Department of Biotechnology, The University of Suwon, Hwaseong-si 18323, Gyeonggi-do, Republic of Korea; (K.C.); (J.K.); (P.P.)
| | - Phumbum Park
- Department of Biotechnology, The University of Suwon, Hwaseong-si 18323, Gyeonggi-do, Republic of Korea; (K.C.); (J.K.); (P.P.)
| | - Rui Liu
- Department of Biotechnology, The University of Suwon, Hwaseong-si 18323, Gyeonggi-do, Republic of Korea; (K.C.); (J.K.); (P.P.)
| | - Kang Lei
- School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng 252059, China; (S.W.); (S.F.)
| |
Collapse
|
13
|
Mei B, Li J, Wang D, Feng L, Huang J, Zhang G. All-trans Retinoic Acid Sensitizes Epithelial Ovarian Cancer to PARP Inhibition after Exposure to Cisplatin. Mol Cancer Ther 2025; 24:453-463. [PMID: 39676352 PMCID: PMC11876957 DOI: 10.1158/1535-7163.mct-24-0140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/27/2024] [Accepted: 12/12/2024] [Indexed: 12/17/2024]
Abstract
Epithelial ovarian cancer (EOC) is the most lethal of gynecologic malignancies. The standard-of-care treatment for EOC is platinum-based chemotherapy such as cisplatin (CDDP). Notably, platinum-based chemotherapy induces resistance of EOC to PARP inhibition. However, therapeutic approaches targeting PARP inhibitor (PARPi) resistance remain to be explored. In this study, we show that all-trans retinoic acid (ATRA) reduces PARPi resistance-associated EOC cells induced by CDDP treatment. Clinically applicable ATRA suppressed the outgrowth of CDDP-treated EOC cells both in vitro and in vivo. Moreover, a CDDP treatment followed by niraparib maintenance therapy in combination with ATRA improved the survival of EOC-bearing mice. These phenotypes correlated with the PARPi-resistant EOC signature, which consists of elevated expression of aldehyde dehydrogenase 1 family member A1, nicotinamide phosphoribosyltransferase, PARP1, and checkpoint kinase 1, as well as elevated NAD+ level-mediated high activity of aldehyde dehydrogenase 1 family member A1 and PARP1. Mechanistically, ATRA downregulates the expression of these genes and level of intracellular NAD+. Our results suggest that ATRA in conjunction with PARPi represents a promising maintenance therapeutic strategy for EOC.
Collapse
Affiliation(s)
- Bingjie Mei
- Department of Gynecological Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Junyang Li
- Department of Gynecological Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Dengfeng Wang
- Department of Gynecological Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Lu Feng
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jianming Huang
- Biochemistry and Molecular Biology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Guonan Zhang
- Department of Gynecological Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
14
|
Merkuryev AV, Egorov VV. Role of PARP-1 structural and functional features in PARP-1 inhibitors development. Bioorg Chem 2025; 156:108188. [PMID: 39855113 DOI: 10.1016/j.bioorg.2025.108188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 12/18/2024] [Accepted: 01/17/2025] [Indexed: 01/27/2025]
Abstract
Poly(ADP-ribose) polymerase-1 (PARP-1) is the key enzyme among other PARPs for post-translational modification of DNA repair proteins. It has four functional domains for DNA-binding, automodification and enzymatic activity. PARP-1 participates in poly-ADP-ribosylation of itself or other proteins during DNA damage response. It recruits reparation machinery proteins that restore native DNA sequence. PARP-1 participates in chromatin structure organization and gene expression regulation. It was shown that PARP-1-dependent regulation mechanisms affect on possible risk of carcinogenesis. Therefore, PARP-1 was proposed as a novel target for cancer treatment. Three generations of PARP-1 inhibitors had been developed depending on pharmacophore structure. To date, four PARP-1 inhibitors have been approved for cancer treatment as a chemotherapy potentiators or as a stand-alone therapy. However, different cytotoxicity effects of specific PARP-1 inhibitors were observed due to diverse PARP-1 activity in cellular processes. Moreover, cancer cells can develop resistance to PARP-1 inhibitors and decrease chemotherapy efficacy. There are promising strategies how to avoid these disadvantages including dual-targeted inhibitors and combination therapy.
Collapse
|
15
|
Wu Y, Zeng Y, Wu Y, Ha X, Feng Z, Liu C, Liu Z, Wang J, Ju X, Huang S, Liang L, Zheng B, Yang L, Wang J, Wu X, Li S, Wen H. HIF-1α-induced long noncoding RNA LINC02776 promotes drug resistance of ovarian cancer by increasing polyADP-ribosylation. Clin Transl Med 2025; 15:e70244. [PMID: 40118782 PMCID: PMC11928293 DOI: 10.1002/ctm2.70244] [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: 09/19/2024] [Revised: 01/06/2025] [Accepted: 02/12/2025] [Indexed: 03/23/2025] Open
Abstract
BACKGROUND Chemoresistance remains a major hurdle in ovarian cancer (OC) treatment, as many patients eventually develop resistance to platinum-based chemotherapy and/or PARP inhibitors (PARPi). METHODS We performed transcriptome-wide analysis by RNA sequencing (RNA-seq) data of platinum-resistant and -sensitive OC tissues. We demonstrated the role of LINC02776 in platinum resistance in OC cells, mice models and patient-derived organoid (PDO) models. RESULTS We identify the long noncoding RNA LINC02776 as a critical factor of platinum resistance. Elevated expression of LINC02776 is observed in platinum-resistant OC and serves as an independent prognostic factor for OC patients. Functionally, silencing LINC02776 reduces proliferation and DNA damage repair in OC cells, thereby enhancing sensitivity to platinum and PARPi in both xenograft mouse models and patient-derived organoid (PDO) models with acquired chemoresistance. Mechanistically, LINC02776 binds to the catalytic domain of poly (ADP-ribose) polymerase 1 (PARP1), promoting PARP1-dependent polyADP-ribosylation (PARylation) and facilitating homologous recombination (HR) restoration. Additionally, high HIF-1α expression in platinum-resistant tissues further stimulates LINC02776 transcription. CONCLUSIONS Our findings suggest that targeting LINC02776 represents a promising therapeutic strategy for OC patients who have developed resistance to platinum or PARPi. KEY POINTS LINC02776 promotes OC cell proliferation by regulating DNA damage and apoptosis signaling pathways. LINC02776 binds PARP1 to promote DNA damage-triggered PARylation in OC cells. LINC02776 mediates cisplatin and olaparib resistance in OC cells by enhancing PARP1-mediated PARylation activity and regulating the PARP1-mediated HR pathway. The high expression of LINC02776 is induced by HIF-1α in platinum-resistant OC cells and tissues.
Collapse
Affiliation(s)
- Yangjun Wu
- Department of Gynecologic OncologyFudan University Shanghai Cancer Center, Fudan UniversityShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
| | - Yu Zeng
- Precision Research Center for Refractory Diseases and Shanghai Key Laboratory of Pancreatic DiseasesShanghai General Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yong Wu
- Department of Gynecologic OncologyFudan University Shanghai Cancer Center, Fudan UniversityShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
| | - Xinyu Ha
- Department of Gynecologic OncologyFudan University Shanghai Cancer Center, Fudan UniversityShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
| | - Zheng Feng
- Department of Gynecologic OncologyFudan University Shanghai Cancer Center, Fudan UniversityShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
| | - Chaohua Liu
- Department of Gynecologic OncologyFudan University Shanghai Cancer Center, Fudan UniversityShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
| | - Ziqi Liu
- Department of Gynecologic OncologyFudan University Shanghai Cancer Center, Fudan UniversityShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
| | - Jiajia Wang
- Department of Gynecologic OncologyFudan University Shanghai Cancer Center, Fudan UniversityShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
| | - Xingzhu Ju
- Department of Gynecologic OncologyFudan University Shanghai Cancer Center, Fudan UniversityShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
| | - Shenglin Huang
- Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical SciencesFudan University Shanghai Cancer Center, Fudan UniversityShanghaiChina
| | - Linhui Liang
- Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical SciencesFudan University Shanghai Cancer Center, Fudan UniversityShanghaiChina
| | - Bin Zheng
- Accurate International Biotechnology Co. Ltd.GuangzhouChina
| | - Lulu Yang
- Wuhan Benagen Technology Co., LtdWuhanChina
| | - Jun Wang
- Wuhan Benagen Technology Co., LtdWuhanChina
| | - Xiaohua Wu
- Department of Gynecologic OncologyFudan University Shanghai Cancer Center, Fudan UniversityShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
| | - Shengli Li
- Precision Research Center for Refractory Diseases and Shanghai Key Laboratory of Pancreatic DiseasesShanghai General Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Hao Wen
- Department of Gynecologic OncologyFudan University Shanghai Cancer Center, Fudan UniversityShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
| |
Collapse
|
16
|
Shanmugam N, Chatterjee S, Cisneros GA. Impact of a Cancer-Associated Mutation on Poly(ADP-ribose) Polymerase1 Inhibition. J Phys Chem B 2025; 129:2175-2186. [PMID: 39962867 PMCID: PMC12005076 DOI: 10.1021/acs.jpcb.4c07960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
Poly(ADP-ribose) polymerase1 (PARP1) plays a vital role in DNA repair, and its inhibition in cancer cells may cause cell apoptosis. In this study, we investigated the effects of a PARP1 variant, V762A, which is strongly associated with several cancers in humans, on the inhibition of PARP1 by three FDA-approved inhibitors: niraparib, rucaparib, and talazoparib. Specifically, we compared the inhibition of the mutant to that of wild-type (WT) PARP1. Additionally, we investigated how the mutation influences the binding of these inhibitors to PARP1. Our work suggests that while mutant PARP1 exhibits only minor differences in residual fluctuations, backbone deviations, and residue motion correlations compared to the WT under niraparib and rucaparib inhibitions, it shows significant and distinct differences in these features when inhibited by talazoparib. Among the three inhibitions, talazoparib inhibition uniquely lowers the average residue fluctuations in the mutant than the WT including lower fluctuations of mutant's N- and C-terminal residues in the catalytic domain, conserved H-Y-E traid residues, and donor loop (D-loop) residues which are important for catalysis more effectively than other inhibitions. However, talazoparib also significantly enhances destabilizing interactions between the mutation site in the HD domain in the mutant than WT. Further, among the three inhibitions, talazoparib inhibition uniquely and significantly disrupts the functional fluctuations of terminal regions in the mutant, which are otherwise present in the WT. The mutation and inhibition do not significantly affect PARP1's essential dynamics. Lastly, these inhibitors bind to the V762A mutant more effectively than to the WT, with similar binding free energies between them.
Collapse
Affiliation(s)
- Neel Shanmugam
- Department of Chemistry, University of North Texas, Denton, Texas 76201, United States; Present Address: Department of Computer Science, Columbia University, New York, New York 10027, United States
| | - Shubham Chatterjee
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - G. Andrés Cisneros
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States; Department of Physics, University of Texas at Dallas, Richardson, Texas 75080, United States
| |
Collapse
|
17
|
Breunig K, Lei X, Montalbano M, Guardia GDA, Ostadrahimi S, Alers V, Kosti A, Chiou J, Klein N, Vinarov C, Wang L, Li M, Song W, Kraus WL, Libich DS, Tiziani S, Weintraub ST, Galante PAF, Penalva LO. SERBP1 interacts with PARP1 and is present in PARylation-dependent protein complexes regulating splicing, cell division, and ribosome biogenesis. eLife 2025; 13:RP98152. [PMID: 39937575 PMCID: PMC11820137 DOI: 10.7554/elife.98152] [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] [Indexed: 02/13/2025] Open
Abstract
RNA binding proteins (RBPs) containing intrinsically disordered regions (IDRs) are present in diverse molecular complexes where they function as dynamic regulators. Their characteristics promote liquid-liquid phase separation (LLPS) and the formation of membraneless organelles such as stress granules and nucleoli. IDR-RBPs are particularly relevant in the nervous system and their dysfunction is associated with neurodegenerative diseases and brain tumor development. Serpine1 mRNA-binding protein 1 (SERBP1) is a unique member of this group, being mostly disordered and lacking canonical RNA-binding domains. We defined SERBP1's interactome, uncovered novel roles in splicing, cell division and ribosomal biogenesis, and showed its participation in pathological stress granules and Tau aggregates in Alzheimer's brains. SERBP1 preferentially interacts with other G-quadruplex (G4) binders, implicated in different stages of gene expression, suggesting that G4 binding is a critical component of SERBP1 function in different settings. Similarly, we identified important associations between SERBP1 and PARP1/polyADP-ribosylation (PARylation). SERBP1 interacts with PARP1 and its associated factors and influences PARylation. Moreover, protein complexes in which SERBP1 participates contain mostly PARylated proteins and PAR binders. Based on these results, we propose a feedback regulatory model in which SERBP1 influences PARP1 function and PARylation, while PARylation modulates SERBP1 functions and participation in regulatory complexes.
Collapse
Affiliation(s)
- Kira Breunig
- Children’s Cancer Research Institute, UT Health San AntonioSan AntonioUnited States
| | - Xuifen Lei
- Children’s Cancer Research Institute, UT Health San AntonioSan AntonioUnited States
| | - Mauro Montalbano
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical BranchGalvestonUnited States
- Department of Neurology, University of Texas Medical BranchGalvestonUnited States
| | | | - Shiva Ostadrahimi
- Children’s Cancer Research Institute, UT Health San AntonioSan AntonioUnited States
- Department of Cell Systems and Anatomy, UT Health San AntonioSan AntonioUnited States
| | - Victoria Alers
- Children’s Cancer Research Institute, UT Health San AntonioSan AntonioUnited States
- Department of Cell Systems and Anatomy, UT Health San AntonioSan AntonioUnited States
- Department of Biochemistry and Structural Biology, UT Health San AntonioSan AntonioUnited States
| | - Adam Kosti
- Children’s Cancer Research Institute, UT Health San AntonioSan AntonioUnited States
- Department of Cell Systems and Anatomy, UT Health San AntonioSan AntonioUnited States
| | - Jennifer Chiou
- Department of Nutritional Sciences, College of Natural Sciences, University of Texas at AustinAustinUnited States
| | - Nicole Klein
- Children’s Cancer Research Institute, UT Health San AntonioSan AntonioUnited States
| | - Corina Vinarov
- Children’s Cancer Research Institute, UT Health San AntonioSan AntonioUnited States
| | - Lily Wang
- Children’s Cancer Research Institute, UT Health San AntonioSan AntonioUnited States
| | - Mujia Li
- Children’s Cancer Research Institute, UT Health San AntonioSan AntonioUnited States
| | - Weidan Song
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences,The University of Texas Southwestern Medical CenterDallasUnited States
| | - W Lee Kraus
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences,The University of Texas Southwestern Medical CenterDallasUnited States
| | - David S Libich
- Children’s Cancer Research Institute, UT Health San AntonioSan AntonioUnited States
- Department of Biochemistry and Structural Biology, UT Health San AntonioSan AntonioUnited States
| | - Stefano Tiziani
- Department of Nutritional Sciences, College of Natural Sciences, University of Texas at AustinAustinUnited States
- Department of Pediatrics, Dell Medical School, University of Texas at AustinAustinUnited States
- Department of Oncology, Dell Medical School, University of Texas at AustinAustinUnited States
| | - Susan T Weintraub
- Department of Biochemistry and Structural Biology, UT Health San AntonioSan AntonioUnited States
| | - Pedro AF Galante
- Centro de Oncologia Molecular, Hospital Sírio-LibanêsSão PauloBrazil
| | - Luiz O Penalva
- Children’s Cancer Research Institute, UT Health San AntonioSan AntonioUnited States
- Department of Cell Systems and Anatomy, UT Health San AntonioSan AntonioUnited States
| |
Collapse
|
18
|
Buckley-Benbow L, Agnarelli A, Bellelli R. 'Where is my gap': mechanisms underpinning PARP inhibitor sensitivity in cancer. Biochem Soc Trans 2025; 53:BST20241633. [PMID: 39927794 DOI: 10.1042/bst20241633] [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: 12/08/2024] [Revised: 01/20/2025] [Accepted: 01/23/2025] [Indexed: 02/11/2025]
Abstract
The introduction of poly-ADP ribose polymerase (PARP) inhibitors (PARPi) has completely changed the treatment landscape of breast cancer susceptibility 1-2 (BRCA1-BRCA2)-mutant cancers and generated a new avenue of research in the fields of DNA damage response and cancer therapy. Despite this, primary and secondary resistances to PARPi have become a challenge in the clinic, and novel therapies are urgently needed to address this problem. After two decades of research, a unifying model explaining sensitivity of cancer cells to PARPi is still missing. Here, we review the current knowledge in the field and the increasing evidence pointing to a crucial role for replicative gaps in mediating sensitization to PARPi in BRCA-mutant and 'wild-type' cancer cells. Finally, we discuss the challenges to be addressed to further improve the utilization of PARPi and tackle the emergence of resistance in the clinical context.
Collapse
Affiliation(s)
- Lauryn Buckley-Benbow
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, Barbican, London EC1M 6BQ, U.K
| | - Alessandro Agnarelli
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, Barbican, London EC1M 6BQ, U.K
| | - Roberto Bellelli
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, Barbican, London EC1M 6BQ, U.K
| |
Collapse
|
19
|
Bilkis R, Lake RJ, Fan HY. ATP-Dependent Chromatin Remodeler CSB Couples DNA Repair Pathways to Transcription with Implications for Cockayne Syndrome and Cancer Therapy. Cells 2025; 14:239. [PMID: 39996712 PMCID: PMC11852979 DOI: 10.3390/cells14040239] [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: 01/10/2025] [Revised: 02/01/2025] [Accepted: 02/04/2025] [Indexed: 02/26/2025] Open
Abstract
Efficient DNA lesion repair is crucial for cell survival, especially within actively transcribed DNA regions that contain essential genetic information. Additionally, DNA breaks in regions of active transcription are prone to generating insertions and deletions, which are hallmark features of cancer genomes. Cockayne syndrome protein B (CSB) is the sole ATP-dependent chromatin remodeler that is essential for coupling DNA repair pathways with transcription, leading to more efficient DNA repair in regions of active transcription. CSB is best known for its essential function in transcription-coupled nucleotide excision repair (TC-NER), a process that rapidly removes helix-distorting DNA lesions that stall RNA polymerase II, such as those created by chemotherapeutic platinum compounds and UV irradiation. In addition to NER, CSB has also been reported to couple homologous recombination to transcription. Most recently, CSB has also been shown to couple single-strand DNA break repair to transcription. In this review, we will discuss the overlapping and distinct mechanisms by which CSB couples these different DNA repair pathways to transcription. We will also discuss how these CSB functions may account for Cockayne syndrome and the emerging roles of CSB as an innovative target for cancer therapy.
Collapse
Affiliation(s)
- Rabeya Bilkis
- Biomedical Sciences Graduate Program, University of New Mexico Health Science Center, Albuquerque, NM 87131, USA;
| | - Robert J. Lake
- Program in Cell and Molecular Oncology, University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA;
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM 87131, USA
| | - Hua-Ying Fan
- Program in Cell and Molecular Oncology, University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA;
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM 87131, USA
| |
Collapse
|
20
|
Liu YJ, Kimura M, Li X, Sulc J, Wang Q, Rodríguez-López S, Scantlebery AML, Strotjohann K, Gallart-Ayala H, Vijayakumar A, Myers RP, Ivanisevic J, Houtkooper RH, Subramanian GM, Takebe T, Auwerx J. ACMSD inhibition corrects fibrosis, inflammation, and DNA damage in MASLD/MASH. J Hepatol 2025; 82:174-188. [PMID: 39181211 PMCID: PMC11741923 DOI: 10.1016/j.jhep.2024.08.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 07/01/2024] [Accepted: 08/01/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND & AIMS Recent findings reveal the importance of tryptophan-initiated de novo nicotinamide adenine dinucleotide (NAD+) synthesis in the liver, a process previously considered secondary to biosynthesis from nicotinamide. The enzyme α-amino-β-carboxymuconate-ε-semialdehyde decarboxylase (ACMSD), primarily expressed in the liver and kidney, acts as a modulator of de novo NAD+ synthesis. Boosting NAD+ levels has previously demonstrated remarkable metabolic benefits in mouse models. In this study, we aimed to investigate the therapeutic implications of ACMSD inhibition in the treatment of metabolic dysfunction-associated steatotic liver disease/steatohepatitis (MASLD/MASH). METHODS In vitro experiments were conducted in primary rodent hepatocytes, Huh7 human liver carcinoma cells and induced pluripotent stem cell-derived human liver organoids (HLOs). C57BL/6J male mice were fed a western-style diet and housed at thermoneutrality to recapitulate key aspects of MASLD/MASH. Pharmacological ACMSD inhibition was given therapeutically, following disease onset. HLO models of steatohepatitis were used to assess the DNA damage responses to ACMSD inhibition in human contexts. RESULTS Inhibiting ACMSD with a novel specific pharmacological inhibitor promotes de novo NAD+ synthesis and reduces DNA damage ex vivo, in vivo, and in HLO models. In mouse models of MASLD/MASH, de novo NAD+ biosynthesis is suppressed, and transcriptomic DNA damage signatures correlate with disease severity; in humans, Mendelian randomization-based genetic analysis suggests a notable impact of genomic stress on liver disease susceptibility. Therapeutic inhibition of ACMSD in mice increases liver NAD+ and reverses MASLD/MASH, mitigating fibrosis, inflammation, and DNA damage, as observed in HLO models of steatohepatitis. CONCLUSIONS Our findings highlight the benefits of ACMSD inhibition in enhancing hepatic NAD+ levels and enabling genomic protection, underscoring its therapeutic potential in MASLD/MASH. IMPACT AND IMPLICATIONS Enhancing NAD+ levels has been shown to induce remarkable health benefits in mouse models of metabolic dysfunction-associated steatotic liver disease/steatohepatitis (MASLD/MASH), yet liver-specific NAD+ boosting strategies remain underexplored. Here, we present a novel pharmacological approach to enhance de novo synthesis of NAD+ in the liver by inhibiting α-amino-β-carboxymuconate-ε-semialdehyde decarboxylase (ACMSD), an enzyme highly expressed in the liver. Inhibiting ACMSD increases NAD+ levels, enhances mitochondrial respiration, and maintains genomic stability in hepatocytes ex vivo and in vivo. These molecular benefits prevent disease progression in both mouse and human liver organoid models of steatohepatitis. Our preclinical study identifies ACMSD as a promising target for MASLD/MASH management and lays the groundwork for developing ACMSD inhibitors as a clinical treatment.
Collapse
Affiliation(s)
- Yasmine J Liu
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Masaki Kimura
- Division of Gastroenterology, Hepatology and Nutrition & Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Xiaoxu Li
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Jonathan Sulc
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Qi Wang
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Sandra Rodríguez-López
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | | | - Keno Strotjohann
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Hector Gallart-Ayala
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | | | | | - Julijana Ivanisevic
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Riekelt H Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, Amsterdam, The Netherlands; Amsterdam Gastroenterology, Endocrinology and Metabolism institute, Amsterdam UMC, Amsterdam, The Netherlands; Amsterdam Cardiovascular Sciences institute, Amsterdam UMC, Amsterdam, The Netherlands
| | | | - Takanori Takebe
- Division of Gastroenterology, Hepatology and Nutrition & Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), and Division of Stem Cell and Organoid Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland.
| |
Collapse
|
21
|
Roggero CM, Ghosh AB, Devineni A, Ma S, Blatt E, Raj GV, Yin Y. CDK4/6 inhibitors promote PARP1 degradation and synergize with PARP inhibitors in non-small cell lung cancer. Transl Oncol 2025; 52:102231. [PMID: 39662449 PMCID: PMC11683282 DOI: 10.1016/j.tranon.2024.102231] [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: 06/24/2024] [Revised: 10/02/2024] [Accepted: 11/28/2024] [Indexed: 12/13/2024] Open
Abstract
Despite widespread deregulation of CDK4/6 activity in non-small cell lung cancer (NSCLC), clinical trials with CDK4/6 inhibitor (CDK4/6i) as a monotherapy have shown poor antitumor activity. Preclinical studies indicate that CDK4/6i may collaborate by influencing DNA damage repair pathways during radiotherapy. Since PARP1 expression was also significantly upregulated in NSCLC, we analyzed the efficacy of combining PARP1 and CDK4/6 inhibition in NSCLC models. We found that CDK4/6is synergize with PARP1 inhibitors (PARPis) to inhibit the clonogenic growth of RB-proficient NSCLC models. This synergy correlates with increased accumulation of DNA damage, interrupted cell-cycle checkpoints, and enhanced apoptotic cell death. Mechanistically, we showed that CDK4/6is promote PARP1 protein degradation, which lead to decreased availability of DNA repair factors involved in homologous recombination and suppression of DNA repair competency. Furthermore, we showed that PARP trapping is engaged in this synergy. We then confirmed that combining PARPi and CDK4/6i blocked the growth of NSCLC xenografts in vivo and patient-derived explant models ex vivo. Our data reveal a previously uncharacterized impact of CDK4/6i on PARP1 levels in RB-proficient NSCLC models and the engagement of PARP trapping in the synergy between CDK4/6i and PARPi. Our findings suggest combining CDK4/6i with PARPi could be a viable therapeutic strategy for patients with RB-proficient NSCLC.
Collapse
Affiliation(s)
- Carlos M Roggero
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States; Instituto de Histología y Embriología de Mendoza (IHEM)-CONICET-Universidad Nacional de Cuyo, Argentina
| | - Anwesha B Ghosh
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Anvita Devineni
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Shihong Ma
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Eliot Blatt
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Ganesh V Raj
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States; Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Yi Yin
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States; Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States.
| |
Collapse
|
22
|
Brosey CA, Shen R, Tainer JA. NADH-bound AIF activates the mitochondrial CHCHD4/MIA40 chaperone by a substrate-mimicry mechanism. EMBO J 2025; 44:1220-1248. [PMID: 39806100 PMCID: PMC11832770 DOI: 10.1038/s44318-024-00360-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 01/16/2025] Open
Abstract
Mitochondrial metabolism requires the chaperoned import of disulfide-stabilized proteins via CHCHD4/MIA40 and its enigmatic interaction with oxidoreductase Apoptosis-inducing factor (AIF). By crystallizing human CHCHD4's AIF-interaction domain with an activated AIF dimer, we uncover how NADH allosterically configures AIF to anchor CHCHD4's β-hairpin and histidine-helix motifs to the inner mitochondrial membrane. The structure further reveals a similarity between the AIF-interaction domain and recognition sequences of CHCHD4 substrates. NMR and X-ray scattering (SAXS) solution measurements, mutational analyses, and biochemistry show that the substrate-mimicking AIF-interaction domain shields CHCHD4's redox-sensitive active site. Disrupting this shield critically activates CHCHD4 substrate affinity and chaperone activity. Regulatory-domain sequestration by NADH-activated AIF directly stimulates chaperone binding and folding, revealing how AIF mediates CHCHD4 mitochondrial import. These results establish AIF as an integral component of the metazoan disulfide relay and point to NADH-activated dimeric AIF as an organizational import center for CHCHD4 and its substrates. Importantly, AIF regulation of CHCHD4 directly links AIF's cellular NAD(H) sensing to CHCHD4 chaperone function, suggesting a mechanism to balance tissue-specific oxidative phosphorylation (OXPHOS) capacity with NADH availability.
Collapse
Affiliation(s)
- Chris A Brosey
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Runze Shen
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - John A Tainer
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, 77030, USA.
- Department of Cancer Biology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, 77030, USA.
- MBIB Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA.
| |
Collapse
|
23
|
Wang J, Gao Y, Xiong X, Yan Y, Lou J, Guo M, Noman M, Li D, Song F. Poly(ADP-ribose) polymerase FonPARP1-catalyzed PARylation of protein disulfide isomerase FonPdi1 regulates pathogenicity of Fusarium oxysporum f. sp. niveum on watermelon. Int J Biol Macromol 2025; 291:139046. [PMID: 39708869 DOI: 10.1016/j.ijbiomac.2024.139046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 11/25/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
Poly(ADP-ribosyl)ation (PARylation), catalyzed by poly(ADP-ribose) polymerases (PARPs) and hydrolyzed by poly(ADP-ribose) glycohydrolase (PARG), is an important reversible post-translational protein modification in all eukaryotes, including plant pathogenic fungi. Previously, we revealed that FonPARP1, an active PARP, is crucial for the pathogenicity of Fusarium oxysporum f. sp. niveum (Fon), the causative agent of watermelon Fusarium wilt. This study explores the enzymatic activity and substrates of FonPARP1 in regulating Fon pathogenicity. FonPARP1 is localized in nuclei of Fon macroconidia and hyphae. Essential conserved domains and a key glutamic acid residue at position 729 are critical for FonPARP1 enzyme activity and pathogenicity function in Fon. FonPARP1 interacts with protein disulfide isomerase FonPdi1 and PARylates it at 13 glutamic acid residues, affecting the interaction ability, PDI activity, ER homeostasis, and pathogenicity function. FonPARG1, interacting with both FonPARP1 and FonPdi1, hydrolyzes poly(ADP-ribose) chains from auto-PARylated FonPARP1 and FonPARP1-PARylated FonPdi1. These findings underscore the role of FonPARP1-catalyzed PARylation in regulating Fon pathogenicity and its significance in plant pathogenic fungi.
Collapse
Affiliation(s)
- Jiajing Wang
- Key Laboratory of Crop Diseases and Insect Pests of Ministry of Agriculture and Rural Affairs, Institute of Biotechnology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, People's Republic of China; Zhejiang Provincial Key Laboratory of Biology of Crop Pathogens and Insects, Institute of Biotechnology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, People's Republic of China; State Key Laboratory of Rice Biology and Breeding, Institute of Biotechnology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Yizhou Gao
- Key Laboratory of Crop Diseases and Insect Pests of Ministry of Agriculture and Rural Affairs, Institute of Biotechnology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, People's Republic of China; Zhejiang Provincial Key Laboratory of Biology of Crop Pathogens and Insects, Institute of Biotechnology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, People's Republic of China; State Key Laboratory of Rice Biology and Breeding, Institute of Biotechnology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, People's Republic of China; School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou 310023, People's Republic of China
| | - Xiaohui Xiong
- Key Laboratory of Crop Diseases and Insect Pests of Ministry of Agriculture and Rural Affairs, Institute of Biotechnology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, People's Republic of China; Zhejiang Provincial Key Laboratory of Biology of Crop Pathogens and Insects, Institute of Biotechnology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, People's Republic of China; State Key Laboratory of Rice Biology and Breeding, Institute of Biotechnology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Yuqing Yan
- Key Laboratory of Crop Diseases and Insect Pests of Ministry of Agriculture and Rural Affairs, Institute of Biotechnology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, People's Republic of China; Zhejiang Provincial Key Laboratory of Biology of Crop Pathogens and Insects, Institute of Biotechnology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, People's Republic of China; State Key Laboratory of Rice Biology and Breeding, Institute of Biotechnology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Jiajun Lou
- Key Laboratory of Crop Diseases and Insect Pests of Ministry of Agriculture and Rural Affairs, Institute of Biotechnology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, People's Republic of China; Zhejiang Provincial Key Laboratory of Biology of Crop Pathogens and Insects, Institute of Biotechnology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, People's Republic of China; State Key Laboratory of Rice Biology and Breeding, Institute of Biotechnology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Mengmeng Guo
- Key Laboratory of Crop Diseases and Insect Pests of Ministry of Agriculture and Rural Affairs, Institute of Biotechnology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, People's Republic of China; Zhejiang Provincial Key Laboratory of Biology of Crop Pathogens and Insects, Institute of Biotechnology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, People's Republic of China; State Key Laboratory of Rice Biology and Breeding, Institute of Biotechnology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Muhammad Noman
- Key Laboratory of Crop Diseases and Insect Pests of Ministry of Agriculture and Rural Affairs, Institute of Biotechnology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, People's Republic of China; Zhejiang Provincial Key Laboratory of Biology of Crop Pathogens and Insects, Institute of Biotechnology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, People's Republic of China; State Key Laboratory of Rice Biology and Breeding, Institute of Biotechnology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, People's Republic of China; State Key Laboratory for Managing Biotic and Chemical Treats to the Quality and Safety of Agro-products, Institute of Plant Protection and Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou 310022, People's Republic of China
| | - Dayong Li
- Key Laboratory of Crop Diseases and Insect Pests of Ministry of Agriculture and Rural Affairs, Institute of Biotechnology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, People's Republic of China; Zhejiang Provincial Key Laboratory of Biology of Crop Pathogens and Insects, Institute of Biotechnology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, People's Republic of China; State Key Laboratory of Rice Biology and Breeding, Institute of Biotechnology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Fengming Song
- Key Laboratory of Crop Diseases and Insect Pests of Ministry of Agriculture and Rural Affairs, Institute of Biotechnology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, People's Republic of China; Zhejiang Provincial Key Laboratory of Biology of Crop Pathogens and Insects, Institute of Biotechnology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, People's Republic of China; State Key Laboratory of Rice Biology and Breeding, Institute of Biotechnology, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, People's Republic of China.
| |
Collapse
|
24
|
Wang Y, Zhang J, Wu X, Huang L, Xiao W, Guo C. The Potential of PARP Inhibitors as Antitumor Drugs and the Perspective of Molecular Design. J Med Chem 2025; 68:18-48. [PMID: 39723587 DOI: 10.1021/acs.jmedchem.4c02642] [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: 12/28/2024]
Abstract
PARP (poly-ADP ribose polymerase) has received widespread attention in cancer treatment. Research has shown that PARP plays a crucial role in DNA damage repair and has become a popular target for drug design. Based on the mechanism of "synthetic lethality", multiple PARPis (PARP inhibitors) have been launched for the treatment of BRCA deficient tumors. For example, the approved PARPis have shown significant potential in cancer treatment, particularly in breast cancer and cancers associated with BRCA1/BRCA2 deficiencies. However, the clinical efficacy and safety of PARP inhibitors in different cancers remain issues that cannot be overlooked. The design of PARPis aims to eliminate their resistance and broaden their application scope. Designing selective PARP-1 inhibitors is also a potential strategy. PROTACs (Proteolysis Targeting Chimeras) to degrade PARP have become a potential novel cancer treatment strategy.
Collapse
Affiliation(s)
- Yinghan Wang
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Jingtao Zhang
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Xiaochen Wu
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Longjiang Huang
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Wenjing Xiao
- Department of Radiation Therapy, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Chuanlong Guo
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| |
Collapse
|
25
|
Frey Y, Haj M, Ziv Y, Elkon R, Shiloh Y. Broad repression of DNA repair genes in senescent cells identified by integration of transcriptomic data. Nucleic Acids Res 2025; 53:gkae1257. [PMID: 39739833 PMCID: PMC11724277 DOI: 10.1093/nar/gkae1257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 11/19/2024] [Accepted: 12/06/2024] [Indexed: 01/02/2025] Open
Abstract
Cellular senescence plays a significant role in tissue aging. Senescent cells, which resist apoptosis while remaining metabolically active, generate endogenous DNA-damaging agents, primarily reactive oxygen species. Efficient DNA repair is therefore crucial in these cells, especially when they undergo senescence escape, resuming DNA replication and cellular proliferation. To investigate whether senescent cell transcriptomes reflect adequate DNA repair capacity, we conducted a comprehensive meta-analysis of 60 transcriptomic datasets comparing senescent to proliferating cells. Our analysis revealed a striking downregulation of genes encoding essential components across DNA repair pathways in senescent cells. This includes pathways active in different cell cycle phases such as nucleotide excision repair, base excision repair, nonhomologous end joining and homologous recombination repair of double-strand breaks, mismatch repair and interstrand crosslink repair. The downregulation observed suggests a significant accumulation of DNA lesions. Experimental monitoring of DNA repair readouts in cells that underwent radiation-induced senescence supported this conclusion. This phenomenon was consistent across various senescence triggers and was also observed in primary cell lines from aging individuals. These findings highlight the potential of senescent cells as 'ticking bombs' in aging-related diseases and tumors recurring following therapy-induced senescence.
Collapse
Affiliation(s)
- Yann Frey
- The David and Inez Myers Laboratory for Cancer Research, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Human Molecular Genetics and Biochemistry, School of Medicine, Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Majd Haj
- The David and Inez Myers Laboratory for Cancer Research, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Human Molecular Genetics and Biochemistry, School of Medicine, Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Yael Ziv
- The David and Inez Myers Laboratory for Cancer Research, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Human Molecular Genetics and Biochemistry, School of Medicine, Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ran Elkon
- Department of Human Molecular Genetics and Biochemistry, School of Medicine, Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Yosef Shiloh
- The David and Inez Myers Laboratory for Cancer Research, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Human Molecular Genetics and Biochemistry, School of Medicine, Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
26
|
Cai SX, Ma N, Wang X, Guo M, Jiang Y, Tian YE. The Discovery of a Potent PARP1 Inhibitor Senaparib. Mol Cancer Ther 2025; 24:47-55. [PMID: 38920409 DOI: 10.1158/1535-7163.mct-23-0625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 04/02/2024] [Accepted: 06/24/2024] [Indexed: 06/27/2024]
Abstract
PARP1 is a critical enzyme involved in DNA damage repair. It belongs to a superfamily of proteins and catalyzes poly(ADP-ribosyl)ation (PARylation). PARP1 inhibitors are effective to treat tumors that have homologous recombination deficiency such as those with BRCA1/2 mutations. The PARP1 inhibitors that have been approved by FDA inhibit both PARP1 and PARP2. PARP2 has also been suggested to play a similar function in DNA repair as PARP1. In addition to inhibiting PARP1 enzymatic activities, PARP1 inhibitors cause the PARP1 enzyme to be "trapped" on DNA, stalling the DNA replication fork and eventually causing double-strand DNA breaks and cell death. Here, we report a PARP1 inhibitor, Senaparib, which has a novel chemical structure and high potency inhibiting PARP1/2 enzymes. Senaparib was highly potent in cell viability tests against tumor cells with BRCA1/2 mutations. It was efficacious in cell line-derived and patient-derived xenograft models in tumors harboring BRCA1/2 mutations. In combination studies, Senaparib used with temozolomide had shown strong synergistic cytotoxicity in both in vitro and in vivo experiments. Senaparib represents a novel class of PARP1 inhibitors that can be used for the treatment of cancer. A phase III clinical study of Senaparib for maintenance treatment following first-line chemotherapy in patients with advanced ovarian cancer has met its primary endpoint, and a new drug application of Senaparib has been accepted by the National Medical Products Administration of China for review.
Collapse
Affiliation(s)
- Sui X Cai
- IMPACT Therapeutics Inc., Shanghai, China
| | - Ning Ma
- IMPACT Therapeutics Inc., Shanghai, China
| | | | | | | | - Ye E Tian
- IMPACT Therapeutics Inc., Shanghai, China
| |
Collapse
|
27
|
Sakari M, Bhadane R, Kumar S, Azevedo R, Malakoutikhah M, Masoumi A, Littler DR, Härmä H, Kopra K, Pulliainen AT. ADP-ribosyltransferase-based biocatalysis of nonhydrolyzable NAD+ analogs. J Biol Chem 2025; 301:108106. [PMID: 39706271 PMCID: PMC11786771 DOI: 10.1016/j.jbc.2024.108106] [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: 11/22/2024] [Accepted: 12/11/2024] [Indexed: 12/23/2024] Open
Abstract
Enzyme promiscuity is the ability of an enzyme to catalyze an unexpected side reaction in addition to its main reaction. Here, we describe a biocatalytic process to produce nonhydrolyzable NAD+ analogs based on the ADP-ribosyltransferase activity of pertussis toxin PtxS1 subunit. First, in identical manner to normal catalysis, PtxS1 activates NAD+ to form the reactive oxocarbenium cation. Subsequently, the electrophilic ribose 1' carbon of the oxocarbenium cation is subject of an attack by the nitrogen atom of an amino group coupled to nicotinamide mimicking compounds. The nitrogen atom acts as the nucleophile instead of the natural sulfur atom substrate of the human Gαi protein. The invention builds on structural data indicating the presence of an NAD+ analog, benzamide amino adenine dinucleotide, at the NAD+ binding site of PtxS1. This was witnessed upon cocrystallization of PtxS1 with NAD+ and 3-aminobenzamide (3-AB). A pharmacophore-based screening on 3-AB followed by quantum mechanical simulations identified analogs of 3-AB with capacity to react with the oxocarbenium cation. Based on HPLC and mass spectrometry, we confirmed the formation of benzamide amino adenine dinucleotide by PtxS1, and also identified two new chemical entities. We name the new entities as isoindolone amine adenine dinucleotide, and isoquinolinone amine adenine dinucleotide, the latter being a highly fluorescent compound. The new NAD+ analogs emerge as valuable tools to study the structural biology and enzymology of NAD+ binding and consuming enzymes, such as human poly(ADP-ribose) polymerases and bacterial ADP-ribosyltransferase exotoxins, and to advance the ongoing drug development efforts.
Collapse
Affiliation(s)
- Moona Sakari
- Institute of Biomedicine, University of Turku, Turku, Finland
| | | | - Sujit Kumar
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Rita Azevedo
- Institute of Biomedicine, University of Turku, Turku, Finland
| | | | | | - Dene R Littler
- Infection and Immunity Program & Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Harri Härmä
- Department of Chemistry, University of Turku, Turku, Finland
| | - Kari Kopra
- Department of Chemistry, University of Turku, Turku, Finland
| | | |
Collapse
|
28
|
Caggiano R, Prokhorova E, Duma L, Schützenhofer K, Lauro R, Catara G, Melillo RM, Celetti A, Smith R, Weroha SJ, Kaufmann SH, Ahel I, Palazzo L. Suppression of ADP-ribosylation reversal triggers cell vulnerability to alkylating agents. Neoplasia 2025; 59:101092. [PMID: 39615107 PMCID: PMC11648251 DOI: 10.1016/j.neo.2024.101092] [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: 10/14/2024] [Revised: 11/06/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024]
Abstract
The ADP-ribosyl hydrolases PARG and ARH3 counteract PARP enzymatic activity by removing ADP-ribosylation. PARG and ARH3 activities have a synthetic lethal effect; however, the specific molecular mechanisms underlying this response remain unknown. Here, we show that the PARG and ARH3 synthetic lethality is enhanced further in the presence of DNA alkylating agents, suggesting that the inability to revert ADP-ribosylation primarily affects the repair of alkylated DNA bases. ARH3 knockout cells, treated with PARG inhibitor and alkylating genotoxins, accumulated single-stranded DNA and DNA damage, resulting in G2/M cell cycle arrest and apoptosis. Furthermore, we reveal a reduction in PARP1/PARP2 levels in ARH3-deficient cells treated with PARG inhibitor due to excessive ADP-ribosylation, which may contribute to alkylating agents' vulnerability. Collectively, these results uncover the potential of targeting ADP-ribosyl hydrolases in combination with alkylating agents for cancer therapy and provide insights into the mechanisms underlying the synthetic lethal effect.
Collapse
Affiliation(s)
- Rocco Caggiano
- Institute of Experimental Endocrinology and Oncology, National Research Council of Italy, Naples, Italy
| | - Evgeniia Prokhorova
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Lena Duma
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Kira Schützenhofer
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Raffaella Lauro
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Giuliana Catara
- Institute of Biochemistry and Cell Biology, National Research Council of Italy, Naples, Italy
| | - Rosa Marina Melillo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Angela Celetti
- Institute of Experimental Endocrinology and Oncology, National Research Council of Italy, Naples, Italy
| | - Rebecca Smith
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - S John Weroha
- Department of Oncology, Mayo Clinic, Rochester, Minnesota, United States
| | - Scott H Kaufmann
- Department of Oncology, Mayo Clinic, Rochester, Minnesota, United States
| | - Ivan Ahel
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom.
| | - Luca Palazzo
- Institute of Experimental Endocrinology and Oncology, National Research Council of Italy, Naples, Italy; Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy.
| |
Collapse
|
29
|
Kang C, Kim J, Jeong Y, Yoo JW, Jung Y. Colon-Targeted Poly(ADP-ribose) Polymerase Inhibitors Synergize Therapeutic Effects of Mesalazine Against Rat Colitis Induced by 2,4-Dinitrobenzenesulfonic Acid. Pharmaceutics 2024; 16:1546. [PMID: 39771525 PMCID: PMC11728683 DOI: 10.3390/pharmaceutics16121546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/14/2024] [Accepted: 11/25/2024] [Indexed: 01/16/2025] Open
Abstract
Background/Objectives: In addition to oncological applications, poly(ADP-ribose) polymerase (PARP) inhibitors have potential as anti-inflammatory agents. Colon-targeted delivery of PARP inhibitors has been evaluated as a pharmaceutical strategy to enhance their safety and therapeutic efficacy against gut inflammation. Methods: Colon-targeted PARP inhibitors 5-aminoisoquinoline (5-AIQ) and 3-aminobenzamide (3-AB) were designed and synthesized by azo coupling with salicylic acid (SA), yielding 5-AIQ azo-linked with SA (AQSA) and 3-AB azo-linked with SA (ABSA). Additional conjugation of AQSA with acidic amino acids yielded glutamic acid-conjugated AQSA (AQSA-Glu) and aspartic acid-conjugated AQSA, which further increased the hydrophilicity of AQSA. Results: The distribution coefficients of PARP inhibitors were lowered by chemical modifications, which correlated well with drug permeability via the Caco-2 cell monolayer. All derivatives were effectively converted to their corresponding PARP inhibitors in the cecal contents. Compared with observations in the oral administration of PARP inhibitors, AQSA-Glu and ABSA resulted in the accumulation of much greater amounts of each PARP inhibitor in the cecum. ABSA accumulated mesalazine (5-ASA) in the cecum to a similar extent as sulfasalazine (SSZ), a colon-targeted 5-ASA prodrug. In the DNBS-induced rat colitis model, AQSA-Glu enhanced the anticolitic potency of 5-AIQ. Furthermore, ABSA was more effective against rat colitis than SSZ or AQSA-Glu, and the anticolitic effects of AQSA-Glu were augmented by combined treatment with a colon-targeted 5-ASA prodrug. In addition, the colon-targeted delivery of PARP inhibitors substantially reduced their systemic absorption. Conclusions: Colon-targeted PARP inhibitors may improve the therapeutic and toxicological properties of inhibitors and synergize the anticolitic effects of 5-ASA.
Collapse
Affiliation(s)
| | | | | | | | - Yunjin Jung
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea; (C.K.); (J.K.); (Y.J.); (J.-W.Y.)
| |
Collapse
|
30
|
Feng S, Xie N, Liu Y, Qin C, Savas AC, Wang TY, Li S, Rao Y, Shambayate A, Chou TF, Brenner C, Huang C, Feng P. Cryptic phosphoribosylase activity of NAMPT restricts the virion incorporation of viral proteins. Nat Metab 2024; 6:2300-2318. [PMID: 39572750 DOI: 10.1038/s42255-024-01162-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 10/04/2024] [Indexed: 12/21/2024]
Abstract
As obligate intracellular pathogens, viruses activate host metabolic enzymes to supply intermediates that support progeny production. Nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme of salvage nicotinamide adenine dinucleotide (NAD+) synthesis, is an interferon-inducible protein that inhibits the replication of several RNA and DNA viruses through unknown mechanisms. Here, we show that NAMPT restricts herpes simplex virus type 1 (HSV-1) replication by impeding the virion incorporation of viral proteins owing to its phosphoribosyl-hydrolase (phosphoribosylase) activity, which is independent of the role of NAMPT in NAD+ synthesis. Proteomics analysis of HSV-1-infected cells identifies phosphoribosylated viral structural proteins, particularly glycoproteins and tegument proteins, which are de-phosphoribosylated by NAMPT in vitro and in cells. Chimeric and recombinant HSV-1 carrying phosphoribosylation-resistant mutations show that phosphoribosylation promotes the incorporation of structural proteins into HSV-1 virions and subsequent virus entry. Loss of NAMPT renders mice highly susceptible to HSV-1 infection. Our work describes an additional enzymatic activity of a metabolic enzyme in viral infection and host defence, offering a system to interrogate the roles of protein phosphoribosylation in metazoans.
Collapse
Affiliation(s)
- Shu Feng
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Na Xie
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and West China, School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, PR China
| | - Yongzhen Liu
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Chao Qin
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Ali Can Savas
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Ting-Yu Wang
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
- Proteome Exploration Laboratory, Beckman Institute, California Institute of Technology, Pasadena, CA, USA
| | - Shutong Li
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Youliang Rao
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Alexandra Shambayate
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Tsui-Fen Chou
- Proteome Exploration Laboratory, Beckman Institute, California Institute of Technology, Pasadena, CA, USA
| | - Charles Brenner
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital and West China, School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, PR China
| | - Pinghui Feng
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
31
|
Huang D, Su Z, Mei Y, Shao Z. The complex universe of inactive PARP1. Trends Genet 2024; 40:1074-1085. [PMID: 39306519 DOI: 10.1016/j.tig.2024.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 12/06/2024]
Abstract
Poly(ADP-ribose) polymerase 1 (PARP1) is a crucial member of the PARP family, which modifies targets through ADP-ribosylation and plays key roles in a variety of biological processes. PARP inhibitors (PARPis) hinder ADP-ribosylation and lead to the retention of PARP1 at the DNA lesion (also known as trapping), which underlies their toxicity. However, inhibitors and mutations that make PARP1 inactive do not necessarily correlate with trapping potency, challenging the current understanding of inactivation-caused trapping. Recent studies on mouse models indicate that both trapping and non-trapping inactivating mutations of PARP1 lead to embryonic lethality, suggesting the unexpected toxicity of the current inhibition strategy. The allosteric model, complicated automodification, and various biological functions of PARP1 all contribute to the complexity of PARP1 inactivation.
Collapse
Affiliation(s)
- Doudou Huang
- Department of Pathology and Pathophysiology, Institute of Colorectal Surgery and Oncology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ziyi Su
- Department of Pathology and Pathophysiology, Institute of Colorectal Surgery and Oncology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yanxia Mei
- Department of Colorectal Surgery and Oncology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhengping Shao
- Department of Pathology and Pathophysiology, Institute of Colorectal Surgery and Oncology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang University Cancer Center, Hangzhou, China.
| |
Collapse
|
32
|
Lei XY, He KY, Li QT, Zhang L, Wu DH, Yang JY, Guo JR, Liu MJ, Zhao ZL, Li JQ, Liu H, Zhao Y, Li YJ, Sun QH, Wu CG, Wang YF, Cao GS, Wang G, Jian YP, Xu ZX. PARylation of HMGA1 desensitizes esophageal squamous cell carcinoma to olaparib. Clin Transl Med 2024; 14:e70111. [PMID: 39690136 DOI: 10.1002/ctm2.70111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 11/01/2024] [Accepted: 11/15/2024] [Indexed: 12/19/2024] Open
Abstract
As a chromatin remodelling factor, high mobility group A1 (HMGA1) plays various roles in both physiological and pathological conditions. However, its role in DNA damage response and DNA damage-based chemotherapy remains largely unexplored. In this study, we report the poly ADP-ribosylation (PARylation) of HMGA1 during DNA damage, leading to desensitization of esophageal squamous cell carcinoma (ESCC) cells to the poly(ADP-ribose) polymerase 1 (PARP1) inhibitor, olaparib. We found that HMGA1 accumulates at sites of DNA damage, where it interacts with PARP1 and undergoes PARylation at residues E47 and E50 in its conserved AT-hook domain. This modification enhances the accumulation of Ku70/Ku80 at the site of DNA damage and activates the DNA-dependent protein kinase catalytic subunit, facilitating nonhomologous end-joining repair. In both subcutaneous tumour models and genetically engineered mouse models of in situ esophageal cancer, HMGA1 interference increased tumour sensitivity to olaparib. Moreover, HMGA1 was highly expressed in ESCC tissues and positively correlated with PARP1 levels as well as poor prognosis in ESCC patients. Taken together, these findings reveal a mechanistic link between HMGA1 and PARP1 in regulating cell responses to DNA damage and suggest that targeting HMGA1 could be a promising strategy to increase cancer cell sensitivity to olaparib.
Collapse
Affiliation(s)
- Xin-Yuan Lei
- School of Life Sciences, Henan University, Kaifeng, China
| | - Kai-Yue He
- School of Life Sciences, Henan University, Kaifeng, China
| | - Qiu-Tong Li
- School of Life Sciences, Henan University, Kaifeng, China
| | - Lei Zhang
- School of Life Sciences, Henan University, Kaifeng, China
| | - Dan-Hui Wu
- School of Life Sciences, Henan University, Kaifeng, China
| | - Jing-Yu Yang
- School of Life Sciences, Henan University, Kaifeng, China
| | - Jin-Rong Guo
- School of Life Sciences, Henan University, Kaifeng, China
| | - Meng-Jie Liu
- School of Life Sciences, Henan University, Kaifeng, China
| | - Zi-Long Zhao
- School of Life Sciences, Henan University, Kaifeng, China
| | - Jun-Qi Li
- School of Life Sciences, Henan University, Kaifeng, China
| | - Huai Liu
- School of Life Sciences, Henan University, Kaifeng, China
| | - Yuan Zhao
- School of Life Sciences, Henan University, Kaifeng, China
| | - Yu-Jia Li
- School of Life Sciences, Henan University, Kaifeng, China
| | - Qian-Hui Sun
- School of Life Sciences, Henan University, Kaifeng, China
| | - Chen-Guang Wu
- School of Life Sciences, Henan University, Kaifeng, China
| | - Yun-Fan Wang
- School of Life Sciences, Henan University, Kaifeng, China
| | - Geng-Sheng Cao
- School of Life Sciences, Henan University, Kaifeng, China
| | - Gang Wang
- School of Life Sciences, Henan University, Kaifeng, China
| | - Yong-Ping Jian
- School of Life Sciences, Henan University, Kaifeng, China
| | - Zhi-Xiang Xu
- School of Life Sciences, Henan University, Kaifeng, China
| |
Collapse
|
33
|
Naumenko KN, Berezhnev EA, Kurgina TA, Sukhanova MV, Lavrik OI. Effects of the RNA-Binding Protein Sam68 on Poly(ADP-Ribose)polymerase 1 Activity. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:2143-2154. [PMID: 39865028 DOI: 10.1134/s0006297924120046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/10/2024] [Accepted: 10/15/2024] [Indexed: 01/28/2025]
Abstract
Taking into account involvement of the RNA-binding proteins in regulation of activity of poly(ADP-ribose) polymerase 1 (PARP1), a key factor of DNA repair, the effect of the intrinsically disordered protein Sam68 (Src-associated substrate during mitosis of 68 kDa) on catalytic activity of this enzyme was studied. Plasmid containing coding sequence of the Sam68 protein was obtained. Using the obtained construct, conditions for the Sam68 expression in Escherichia coli cells were optimized and procedure for protein purification was developed. It was found that Sam68 is able to regulate catalytic activity of PARP1, stimulating auto-poly(ADP-ribosyl)ation of PARP1, interacting with the damaged DNA and purified poly(ADP-ribose) (PAR). Based on the experimental data, a hypothesis on the mechanism of PARP1 activity stimulation by the Sam68 protein was proposed, which involves formation of a complex of Sam68 with poly(ADP-ribosyl)ated PARP1. Sam68 interacts with PAR, shielding its negative charge, which increases the time of PARP1 in the complex with damaged DNA and the overall yield of PAR synthesized by this enzyme.
Collapse
Affiliation(s)
- Konstantin N Naumenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| | - Egor A Berezhnev
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| | - Tatyana A Kurgina
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| | - Maria V Sukhanova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia
| | - Olga I Lavrik
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090, Russia.
| |
Collapse
|
34
|
Chin Sang C, Moore G, Tereshchenko M, Zhang H, Nosella ML, Dasovich M, Alderson TR, Leung AKL, Finkelstein IJ, Forman-Kay JD, Lee HO. PARP1 condensates differentially partition DNA repair proteins and enhance DNA ligation. EMBO Rep 2024; 25:5635-5666. [PMID: 39496836 PMCID: PMC11624282 DOI: 10.1038/s44319-024-00285-5] [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: 07/26/2024] [Revised: 09/21/2024] [Accepted: 10/01/2024] [Indexed: 11/06/2024] Open
Abstract
Poly(ADP-ribose) polymerase 1 (PARP1) is one of the first responders to DNA damage and plays crucial roles in recruiting DNA repair proteins through its activity - poly(ADP-ribosyl)ation (PARylation). The enrichment of DNA repair proteins at sites of DNA damage has been described as the formation of a biomolecular condensate. However, it remains unclear how exactly PARP1 and PARylation contribute to the formation and organization of DNA repair condensates. Using recombinant human single-strand repair proteins in vitro, we find that PARP1 readily forms viscous biomolecular condensates in a DNA-dependent manner and that this depends on its three zinc finger (ZnF) domains. PARylation enhances PARP1 condensation in a PAR chain length-dependent manner and increases the internal dynamics of PARP1 condensates. DNA and single-strand break repair proteins XRCC1, LigIII, Polβ, and FUS partition in PARP1 condensates, although in different patterns. While Polβ and FUS are both homogeneously mixed within PARP1 condensates, FUS enrichment is greatly enhanced upon PARylation whereas Polβ partitioning is not. XRCC1 and LigIII display an inhomogeneous organization within PARP1 condensates; their enrichment in these multiphase condensates is enhanced by PARylation. Functionally, PARP1 condensates concentrate short DNA fragments, which correlates with PARP1 clusters compacting long DNA and bridging DNA ends. Furthermore, the presence of PARP1 condensates significantly promotes DNA ligation upon PARylation. These findings provide insight into how PARP1 condensation and PARylation regulate the assembly and biochemical activities of DNA repair factors, which may inform on how PARPs function in DNA repair foci and other PAR-driven condensates in cells.
Collapse
Affiliation(s)
| | - Gaelen Moore
- Department of Biochemistry, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Maria Tereshchenko
- Department of Biochemistry, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Hongshan Zhang
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA
- Center for Systems and Synthetic Biology, University of Texas at Austin, Austin, TX, USA
| | - Michael L Nosella
- Department of Biochemistry, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
| | - Morgan Dasovich
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
- Green Centre for Reproductive Biology Sciences, University of Texas Southwestern Medical Centre, Dallas, TX, USA
| | - T Reid Alderson
- Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Institute of Structural Biology, Helmholtz Zentrum München, Munich, Bavaria, Germany
| | - Anthony K L Leung
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
- Department of Molecular Biology and Genetics, Department of Oncology, and Department of Genetic Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Ilya J Finkelstein
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA
- Center for Systems and Synthetic Biology, University of Texas at Austin, Austin, TX, USA
| | - Julie D Forman-Kay
- Department of Biochemistry, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Molecular Medicine Program, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
| | - Hyun O Lee
- Department of Biochemistry, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
35
|
Nickerson JA, Momen-Heravi F. Long non-coding RNAs: roles in cellular stress responses and epigenetic mechanisms regulating chromatin. Nucleus 2024; 15:2350180. [PMID: 38773934 PMCID: PMC11123517 DOI: 10.1080/19491034.2024.2350180] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 04/22/2024] [Indexed: 05/24/2024] Open
Abstract
Most of the genome is transcribed into RNA but only 2% of the sequence codes for proteins. Non-coding RNA transcripts include a very large number of long noncoding RNAs (lncRNAs). A growing number of identified lncRNAs operate in cellular stress responses, for example in response to hypoxia, genotoxic stress, and oxidative stress. Additionally, lncRNA plays important roles in epigenetic mechanisms operating at chromatin and in maintaining chromatin architecture. Here, we address three lncRNA topics that have had significant recent advances. The first is an emerging role for many lncRNAs in cellular stress responses. The second is the development of high throughput screening assays to develop causal relationships between lncRNAs across the genome with cellular functions. Finally, we turn to recent advances in understanding the role of lncRNAs in regulating chromatin architecture and epigenetics, advances that build on some of the earliest work linking RNA to chromatin architecture.
Collapse
Affiliation(s)
- Jeffrey A Nickerson
- Division of Genes & Development, Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Fatemeh Momen-Heravi
- College of Dental Medicine, Columbia University Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| |
Collapse
|
36
|
Liu J, Geng Y, Jiang S, Guan L, Gao J, Niu MM, Li J. Discovery of novel PARP1/NRP1 dual-targeting inhibitors with strong antitumor potency. Front Pharmacol 2024; 15:1454957. [PMID: 39679370 PMCID: PMC11637875 DOI: 10.3389/fphar.2024.1454957] [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: 06/26/2024] [Accepted: 11/15/2024] [Indexed: 12/17/2024] Open
Abstract
Given that overexpression of Poly (ADP-ribose) polymerase-1 (PARP1) and Neuropilin-1 (NRP1) is implicated in the pathogenesis of human breast cancer, the design of dual PARP1/NRP1 inhibitors has wide therapeutic prospect. However, there have been no reports of such inhibitors so far. Herein, we discovered novel small molecule inhibitors that simultaneously target PARP1 and NRP1 using structure-based virtual screening for the treatment of breast cancer. Notably, PPNR-4 was the most potent inhibitor targeting PARP1 (IC50 = 7.71 ± 0.39 nM) and NRP1 (IC50 = 24.48 ± 2.16 nM). PPNR-4 showed high affinity and binding stability to PARP1 and NRP1. The cytotoxicity assays showed that PPNR-4 demonstrated significant antiproliferative activity on MDA-MB-231 cells (IC50 = 0.21 μM) without effect on normal human cells. In vivo experiments exhibited that PPNR-4 showed more effective than the positive controls in inhibiting the growth of tumors. Overall, these data suggest that PPNR-4 is an effective antitumor candidate and deserves further research.
Collapse
Affiliation(s)
- Juanjuan Liu
- Department of Pharmacy, Taizhou School of Clinical Medicine, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou, China
| | - Yifei Geng
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China
| | - Su Jiang
- Department of Pharmacy, Taizhou School of Clinical Medicine, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou, China
| | - Lixia Guan
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China
| | - Junyi Gao
- Department of Pharmacy, Taizhou School of Clinical Medicine, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou, China
| | - Miao-Miao Niu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing, China
| | - Jindong Li
- Department of Pharmacy, Taizhou School of Clinical Medicine, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou, China
| |
Collapse
|
37
|
Sukhanova MV, Anarbaev RO, Naumenko KN, Hamon L, Singatulina AS, Pastré D, Lavrik OI. Phase Separation of FUS with Poly(ADP-ribosyl)ated PARP1 Is Controlled by Polyamines, Divalent Metal Cations, and Poly(ADP-ribose) Structure. Int J Mol Sci 2024; 25:12445. [PMID: 39596510 PMCID: PMC11594298 DOI: 10.3390/ijms252212445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/29/2024] [Accepted: 11/01/2024] [Indexed: 11/28/2024] Open
Abstract
Fused in sarcoma (FUS) is involved in the formation of nuclear biomolecular condensates associated with poly(ADP-ribose) [PAR] synthesis catalyzed by a DNA damage sensor such as PARP1. Here, we studied FUS microphase separation induced by poly(ADP-ribosyl)ated PARP1WT [PAR-PARP1WT] or its catalytic variants PARP1Y986S and PARP1Y986H, respectively, synthesizing (short PAR)-PARP1Y986S or (short hyperbranched PAR)-PARP1Y986H using dynamic light scattering, fluorescence microscopy, turbidity assays, and atomic force microscopy. We observed that biologically relevant cations such as Mg2+, Ca2+, or Mn2+ or polyamines (spermine4+ or spermidine3+) were essential for the assembly of FUS with PAR-PARP1WT and FUS with PAR-PARP1Y986S in vitro. We estimated the range of the FUS-to-PAR-PARP1 molar ratio and the cation concentration that are favorable for the stability of the protein's microphase-separated state. We also found that FUS microphase separation induced by PAR-PARP1Y986H (i.e., a PARP1 variant attaching short hyperbranched PAR to itself) can occur in the absence of cations. The dependence of PAR-PARP1-induced FUS microphase separation on cations and on the branching of the PAR structure points to a potential role of the latter in the regulation of the formation of FUS-related biological condensates and requires further investigation.
Collapse
Affiliation(s)
- Maria V. Sukhanova
- Institute of Chemical Biology and Fundamental Medicine (ICBFM), Siberian Branch of the Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia; (M.V.S.); (R.O.A.); (K.N.N.); (A.S.S.)
| | - Rashid O. Anarbaev
- Institute of Chemical Biology and Fundamental Medicine (ICBFM), Siberian Branch of the Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia; (M.V.S.); (R.O.A.); (K.N.N.); (A.S.S.)
| | - Konstantin N. Naumenko
- Institute of Chemical Biology and Fundamental Medicine (ICBFM), Siberian Branch of the Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia; (M.V.S.); (R.O.A.); (K.N.N.); (A.S.S.)
| | - Loic Hamon
- INSERM U1204, Univ-Evry, University Paris Saclay, 91025 Evry, France;
| | - Anastasia S. Singatulina
- Institute of Chemical Biology and Fundamental Medicine (ICBFM), Siberian Branch of the Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia; (M.V.S.); (R.O.A.); (K.N.N.); (A.S.S.)
| | - David Pastré
- INSERM U1204, Univ-Evry, University Paris Saclay, 91025 Evry, France;
| | - Olga I. Lavrik
- Institute of Chemical Biology and Fundamental Medicine (ICBFM), Siberian Branch of the Russian Academy of Sciences (SB RAS), Novosibirsk 630090, Russia; (M.V.S.); (R.O.A.); (K.N.N.); (A.S.S.)
| |
Collapse
|
38
|
Lodovichi S, Nepomuceno TC, Woods NT, Rix U, Koomen JM, Pellicioli A, Galli A, Monteiro ANA. SART1 modulates poly-(ADP-ribose) chain accumulation and PARP1 chromatin localization. iScience 2024; 27:111252. [PMID: 39569366 PMCID: PMC11576398 DOI: 10.1016/j.isci.2024.111252] [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: 03/15/2023] [Revised: 02/23/2024] [Accepted: 10/22/2024] [Indexed: 11/22/2024] Open
Abstract
PARP1 inhibitors (PARPis) are used for treatment of cancers with mutations in BRCA1 or BRCA2 that are deficient in homologous recombination. The identification of modulators of PARP1 activity is critical to understand and overcome resistance to PARPis. We integrated data from three omics-scale screens to discover new regulators of PARP1 activity. We identified SART1 and show that its silencing leads to an increase in poly-ADP ribosylation and chromatin-bound PARP1. SART1 is recruited to chromatin following DNA damage and limits PARP1 chromatin retention and activity. The SART1 N-terminus is sufficient to regulate the accumulation of PAR chains and PARP1 on chromatin, an activity dependent on the RGG/RG box. Silencing of SART1 leads to an increased sensitivity of cells to DNA damage induced by IR, irrespective of BRCA1 status and to PARPis only in absence of BRCA1. These results suggest that SART1 could be clinically utilized to improve PARPi efficacy.
Collapse
Affiliation(s)
- Samuele Lodovichi
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
- Yeast Genetics and Genomics, Laboratory of Functional Genetics and Genomics, Institute of Clinical Physiology, CNR, 56125 Pisa, Italy
- Dipartimento di Bioscienze, Università degli Studi di Milano, 20131 Milano, Italy
| | - Thales C Nepomuceno
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Nicholas T Woods
- Gastrointestinal Cancer Program, Eppley Institute for Research in Cancer, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Uwe Rix
- Department of Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - John M Koomen
- Molecular Oncology and Molecular Medicine Program, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Achille Pellicioli
- Dipartimento di Bioscienze, Università degli Studi di Milano, 20131 Milano, Italy
| | - Alvaro Galli
- Yeast Genetics and Genomics, Laboratory of Functional Genetics and Genomics, Institute of Clinical Physiology, CNR, 56125 Pisa, Italy
| | - Alvaro N A Monteiro
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| |
Collapse
|
39
|
Shanmugam N, Chatterjee S, Cisneros GA. Impact of a Cancer-Associated Mutation on Poly(ADP-ribose) Polymerase1 Inhibition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.13.623412. [PMID: 39605557 PMCID: PMC11601374 DOI: 10.1101/2024.11.13.623412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Poly(ADP-ribose) polymerase1 (PARP1) plays a vital role in DNA repair and its inhibition in cancer cells may cause cell apoptosis. In this study, we investigated the effects of a PARP1 variant, V762A, which is strongly associated with several cancers in humans, on the inhibition of PARP1 by three FDA approved inhibitors: niraparib, rucaparib and talazoparib. Our work suggests that these inhibitors bind to the V762A mutant more effectively than to the wild-type (WT), with similar binding free energies between them. Talazoparib inhibition uniquely lowers the average residue fluctuations in the mutant than the WT including lower fluctuations of mutant's N- and C-terminal residues, conserved H-Y-E traid residues and donor loop (D-loop) residues which important for catalysis more effectively than other inhibitions. However, talazoparib also enhances destabilizing interactions between the mutation site in the HD domain in the mutant than WT. Further, talazoparib inhibition significantly disrupts the functional fluctuations of terminal regions in the mutant, which are otherwise present in the WT. Lastly, the mutation and inhibition do not significantly affect PARP1's essential dynamics.
Collapse
Affiliation(s)
- Neel Shanmugam
- Department of Chemistry, University of North Texas, Denton, TX 76201, USA
| | - Shubham Chatterjee
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - G. Andrés Cisneros
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, TX, 75080, USA
- Department of Physics, University of Texas at Dallas, Richardson, TX, 75080, USA
| |
Collapse
|
40
|
Elfar G, Aning O, Ngai T, Yeo P, Chan J, Sim S, Goh L, Yuan J, Phua C, Yeo J, Mak S, Goh B, Chow PH, Tam W, Ho Y, Cheok C. p53-dependent crosstalk between DNA replication integrity and redox metabolism mediated through a NRF2-PARP1 axis. Nucleic Acids Res 2024; 52:12351-12377. [PMID: 39315696 PMCID: PMC11551750 DOI: 10.1093/nar/gkae811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 08/24/2024] [Accepted: 09/10/2024] [Indexed: 09/25/2024] Open
Abstract
Mechanisms underlying p53-mediated protection of the replicating genome remain elusive, despite the quintessential role of p53 in maintaining genomic stability. Here, we uncover an unexpected function of p53 in curbing replication stress by limiting PARP1 activity and preventing the unscheduled degradation of deprotected stalled forks. We searched for p53-dependent factors and elucidated RRM2B as a prime factor. Deficiency in p53/RRM2B results in the activation of an NRF2 antioxidant transcriptional program, with a concomitant elevation in basal PARylation in cells. Dissecting the consequences of p53/RRM2B loss revealed a crosstalk between redox metabolism and genome integrity that is negotiated through a hitherto undescribed NRF2-PARP1 axis, and pinpoint G6PD as a primary oxidative stress-induced NRF2 target and activator of basal PARylation. This study elucidates how loss of p53 could be destabilizing for the replicating genome and, importantly, describes an unanticipated crosstalk between redox metabolism, PARP1 and p53 tumor suppressor pathway that is broadly relevant in cancers and can be leveraged therapeutically.
Collapse
Affiliation(s)
- Gamal Ahmed Elfar
- NUS Department of Pathology, National University of Singapore, Yong Loo Lin School of Medicine, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Obed Aning
- NUS Department of Pathology, National University of Singapore, Yong Loo Lin School of Medicine, Singapore
| | - Tsz Wai Ngai
- NUS Department of Pathology, National University of Singapore, Yong Loo Lin School of Medicine, Singapore
| | - Pearlyn Yeo
- NUS Department of Pathology, National University of Singapore, Yong Loo Lin School of Medicine, Singapore
| | - Joel Wai Kit Chan
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Shang Hong Sim
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Leonard Goh
- NUS Department of Pathology, National University of Singapore, Yong Loo Lin School of Medicine, Singapore
| | - Ju Yuan
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Cheryl Zi Jin Phua
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Joanna Zhen Zhen Yeo
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Shi Ya Mak
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Brian Kim Poh Goh
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore and National Cancer Centre Singapore, Singapore
| | - Pierce Kah-Hoe Chow
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore and National Cancer Centre Singapore, Singapore
- Surgery Academic ClinicalProgramme, Duke-NUS Medical School, National University of Singapore, Singapore
| | - Wai Leong Tam
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University Singapore, Singapore
| | - Ying Swan Ho
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Chit Fang Cheok
- NUS Department of Pathology, National University of Singapore, Yong Loo Lin School of Medicine, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University Singapore, Singapore
| |
Collapse
|
41
|
Luo S, Yue M, Wang D, Lu Y, Wu Q, Jiang J. Breaking the barrier: Epigenetic strategies to combat platinum resistance in colorectal cancer. Drug Resist Updat 2024; 77:101152. [PMID: 39369466 DOI: 10.1016/j.drup.2024.101152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/22/2024] [Accepted: 09/20/2024] [Indexed: 10/08/2024]
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related mortality worldwide. Platinum-based drugs, such as cisplatin and oxaliplatin, are frontline chemotherapy for CRC, effective in both monotherapy and combination regimens. However, the clinical efficacy of these treatments is often undermined by the development of drug resistance, a significant obstacle in cancer therapy. In recent years, epigenetic alterations have been recognized as key players in the acquisition of resistance to platinum drugs. Targeting these dysregulated epigenetic mechanisms with small molecules represents a promising therapeutic strategy. This review explores the complex relationship between epigenetic changes and platinum resistance in CRC, highlighting current epigenetic therapies and their effectiveness in countering resistance mechanisms. By elucidating the epigenetic underpinnings of platinum resistance, this review aims to contribute to ongoing efforts to improve treatment outcomes for CRC patients.
Collapse
Affiliation(s)
- Shiwen Luo
- Institute of Infection, Immunology and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Ming Yue
- Department of Pharmacy, the Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Dequan Wang
- Institute of Infection, Immunology and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yukang Lu
- Institute of Infection, Immunology and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Qingming Wu
- Institute of Infection, Immunology and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China; Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan 430065, China.
| | - Jue Jiang
- Institute of Infection, Immunology and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan 430065, China.
| |
Collapse
|
42
|
Rack JGM, Voorneveld J, Longarini EJ, Wijngaarden S, Zhu K, Peters A, Sia JJ, Prokhorova E, Ahel D, Matić I, Filippov DV, Ahel I. Reversal of tyrosine-linked ADP-ribosylation by ARH3 and PARG. J Biol Chem 2024; 300:107838. [PMID: 39342999 PMCID: PMC11541775 DOI: 10.1016/j.jbc.2024.107838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 10/01/2024] Open
Abstract
ADP-ribosylation is an ancient posttranslational modification with exceptional versatility in terms of breadth of modification targets including at least seven different amino acid side chains, various moieties on nucleic acids, and a variety of small chemical compounds. The spatiotemporal signaling dynamic of the different modification variations is tightly regulated and depends on the writers, erases, and readers of each type. Among these, tyrosine ADP-ribosylation (Tyr-ADPr) has been consistently detected as a novel modification type, but systematic analysis of its potential physiological role, modification establishment, and reversal are still lacking. Here we present a re-analysis of recent ADP-ribosylome data and show that Tyr-ADPr sites are conserved and enriched among ribosome biogenesis and mRNA processing proteins and that these sites are affected by the status of the (ADP-ribosyl)hydrolase ARH3. To facilitate the study of Tyr-ADPr, we establish methodologies for the synthesis of well-defined Tyr-ADPr peptides and with these could show that Tyr-ADPr is reversed both by ARH3 and PARG enzymes. Together, our work lays the foundation for the future exploration of the Tyr-ADPr.
Collapse
Affiliation(s)
| | - Jim Voorneveld
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Edoardo José Longarini
- Research Group of Proteomics and ADP-ribosylation Signalling, Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Sven Wijngaarden
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Kang Zhu
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Alessandra Peters
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Jia Jhing Sia
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | | | - Dragana Ahel
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Ivan Matić
- Research Group of Proteomics and ADP-ribosylation Signalling, Max Planck Institute for Biology of Ageing, Cologne, Germany; Cologne Excellence Cluster for Stress Responses in Ageing-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Dmitri V Filippov
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands.
| | - Ivan Ahel
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK.
| |
Collapse
|
43
|
Huang YZ, Sang MY, Xi PW, Xu RX, Cai MY, Wang ZW, Zhao JY, Li YH, Wei JF, Ding Q. FANCI Inhibition Induces PARP1 Redistribution to Enhance the Efficacy of PARP Inhibitors in Breast Cancer. Cancer Res 2024; 84:3447-3463. [PMID: 39037758 DOI: 10.1158/0008-5472.can-23-2738] [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: 09/08/2023] [Revised: 03/27/2024] [Accepted: 07/19/2024] [Indexed: 07/23/2024]
Abstract
Breast cancer is a global public health concern with high mortality rates, necessitating the development of innovative treatment strategies. PARP inhibitors have shown efficacy in certain patient populations, but their application is largely limited to cancers with homologous recombination deficiency. Here, we identified the suppression of FANCI as a therapeutic strategy to enhance the efficacy of PARP inhibitors in breast cancer. Elevated FANCI expression in breast cancer was associated with poor prognosis and increased cell proliferation and migration. FANCI interacted with PARP1, and suppressing FANCI limited the nuclear localization and functionality of PARP1. Importantly, FANCI inhibition sensitized breast cancer cells to the PARP inhibitor talazoparib in the absence of BRCA mutations. Additionally, the CDK4/6 inhibitor palbociclib enhanced the sensitivity of breast cancer cells to talazoparib through FANCI inhibition. These findings highlight the potential of targeting FANCI to enhance the efficacy of PARP inhibitors in treating breast cancer. Significance: Targeting FANCI is a promising therapeutic strategy for enhancing PARP inhibitor sensitivity in breast cancer that holds potential for broader therapeutic applications beyond cancers harboring BRCA mutations.
Collapse
Affiliation(s)
- Yu-Zhou Huang
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, PR China
| | - Ming-Yi Sang
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, PR China
| | - Pei-Wen Xi
- Health Management Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, PR China
| | - Ruo-Xi Xu
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, PR China
| | - Meng-Yuan Cai
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, PR China
| | - Zi-Wen Wang
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, PR China
| | - Jian-Yi Zhao
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, PR China
| | - Yi-Han Li
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, PR China
| | - Ji-Fu Wei
- Department of Pharmacy, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, PR China
| | - Qiang Ding
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, PR China
| |
Collapse
|
44
|
Yang LL, Zhang XK, Cao Y, Shi LY, Xie SY, Yang YJ, Wu SJ, Sun HZ, Tang XJ, Yuan DL, Zhang D, Xu XF, Li Q, Ying XY. PARP1 acetylation at K119 is essential in regulating the progression and proliferation of cervical cancer cells. Med Oncol 2024; 41:273. [PMID: 39400626 DOI: 10.1007/s12032-024-02315-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/27/2024] [Indexed: 10/15/2024]
Abstract
Cervical cancer, CC, is one of the malignant cancers in women worldwide. Many studies about the genesis and progression of CC have been done at genomic, transcriptional, translational, and epigenetic levels. However, much less is done at post-translational modification (PTM) level. We first used pan-PTM antibodies to compare the pan PTM levels between clinical normal cervical tissues and CC tissues; we then sent the selected samples for label-free identification of acetylation sites. Next, we employed WT or K119A mutant PARP1-EGFP-STREPII plasmid transfection in Hela cells and examined various indexes including colony formation, wound healing, ROS generation, early apoptosis, and immunofluorescence and quantification of proliferation markers (Ki67, PCNA, and p-P53). Last, we examined the levels of multiple important kinases regulating cervical cancer progression. We found that pan-acetylation was the most downregulated in clinical CC samples, whereas the acetylation of PARP1, Poly(ADP-ribose) polymerase-1, was upregulated at K119. Next, we showed that PARP1-WT overexpression significantly suppressed the proliferation and progression in CC cell line Hela, while K119A overexpression didn't show any impact. Finally, PARP1-WT overexpression significantly decreased p-ERK1/2 while didn't affect the phosphorylation levels of other important kinases such as AKT, MTOR, and RPS6. This study discovered a new type of PTM of PARP1 in CC, and showed that PARP1 acetylation at K119 is essential in regulating the proliferation and progression of CC through ERK1/2. Further studies are required to investigate how PARP1 acetylation impact its function.
Collapse
Affiliation(s)
- Li-Li Yang
- The Second Affiliated Hospital of Nanjing Medical University, 121 Jiangjia Garden, Nanjing, 210029, China
- Taizhou People's Hospital Affiliated to Nanjing Medical University, 366 Taihu Road, Taizhou, 225300, China
| | - Xue-Ke Zhang
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China
| | - Ying Cao
- The Second Affiliated Hospital of Nanjing Medical University, 121 Jiangjia Garden, Nanjing, 210029, China
| | - Li-Ya Shi
- Reproductive Medicine Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200082, China
- Obstetrics and Gynecology, Shanghai East Hospital Ji'an Hospital, Ji'an, 343006, China
| | - Shi-Ya Xie
- State Key Lab of Reproductive Medicine and Offspring Health, Nanjing Medical University, 101 Longmian Ave., Nanjing, 211166, China
| | - Yan-Jie Yang
- State Key Lab of Reproductive Medicine and Offspring Health, Nanjing Medical University, 101 Longmian Ave., Nanjing, 211166, China
| | - Shao-Jun Wu
- Taizhou People's Hospital Affiliated to Nanjing Medical University, 366 Taihu Road, Taizhou, 225300, China
| | - Hong-Zhan Sun
- Taizhou People's Hospital Affiliated to Nanjing Medical University, 366 Taihu Road, Taizhou, 225300, China
| | - Xue-Jun Tang
- The Second Affiliated Hospital of Nanjing Medical University, 121 Jiangjia Garden, Nanjing, 210029, China
| | - Dong-Lan Yuan
- Taizhou People's Hospital Affiliated to Nanjing Medical University, 366 Taihu Road, Taizhou, 225300, China.
| | - Dong Zhang
- State Key Lab of Reproductive Medicine and Offspring Health, Nanjing Medical University, 101 Longmian Ave., Nanjing, 211166, China.
| | - Xiao-Feng Xu
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China.
| | - Qian Li
- Department of Gynecology, Nanjing Women and Children's Healthcare Hospital, 123 Tianfei Lane, Mochou Road, Nanjing, 210018, China.
| | - Xiao-Yan Ying
- The Second Affiliated Hospital of Nanjing Medical University, 121 Jiangjia Garden, Nanjing, 210029, China.
| |
Collapse
|
45
|
Breunig K, Lei X, Montalbano M, Guardia GDA, Ostadrahimi S, Alers V, Kosti A, Chiou J, Klein N, Vinarov C, Wang L, Li M, Song W, Kraus WL, Libich DS, Tiziani S, Weintraub ST, Galante PAF, Penalva LOF. SERBP1 interacts with PARP1 and is present in PARylation-dependent protein complexes regulating splicing, cell division, and ribosome biogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.22.586270. [PMID: 38585848 PMCID: PMC10996453 DOI: 10.1101/2024.03.22.586270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
RNA binding proteins (RBPs) containing intrinsically disordered regions (IDRs) are present in diverse molecular complexes where they function as dynamic regulators. Their characteristics promote liquid-liquid phase separation (LLPS) and the formation of membraneless organelles such as stress granules and nucleoli. IDR-RBPs are particularly relevant in the nervous system and their dysfunction is associated with neurodegenerative diseases and brain tumor development. Serpine1 mRNA-binding protein 1 (SERBP1) is a unique member of this group, being mostly disordered and lacking canonical RNA-binding domains. We defined SERBP1's interactome, uncovered novel roles in splicing, cell division and ribosomal biogenesis, and showed its participation in pathological stress granules and Tau aggregates in Alzheimer's brains. SERBP1 preferentially interacts with other G-quadruplex (G4) binders, implicated in different stages of gene expression, suggesting that G4 binding is a critical component of SERBP1 function in different settings. Similarly, we identified important associations between SERBP1 and PARP1/polyADP-ribosylation (PARylation). SERBP1 interacts with PARP1 and its associated factors and influences PARylation. Moreover, protein complexes in which SERBP1 participates contain mostly PARylated proteins and PAR binders. Based on these results, we propose a feedback regulatory model in which SERBP1 influences PARP1 function and PARylation, while PARylation modulates SERBP1 functions and participation in regulatory complexes.
Collapse
|
46
|
Jessop M, Broadway BJ, Miller K, Guettler S. Regulation of PARP1/2 and the tankyrases: emerging parallels. Biochem J 2024; 481:1097-1123. [PMID: 39178157 DOI: 10.1042/bcj20230230] [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: 06/10/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 08/25/2024]
Abstract
ADP-ribosylation is a prominent and versatile post-translational modification, which regulates a diverse set of cellular processes. Poly-ADP-ribose (PAR) is synthesised by the poly-ADP-ribosyltransferases PARP1, PARP2, tankyrase (TNKS), and tankyrase 2 (TNKS2), all of which are linked to human disease. PARP1/2 inhibitors have entered the clinic to target cancers with deficiencies in DNA damage repair. Conversely, tankyrase inhibitors have continued to face obstacles on their way to clinical use, largely owing to our limited knowledge of their molecular impacts on tankyrase and effector pathways, and linked concerns around their tolerability. Whilst detailed structure-function studies have revealed a comprehensive picture of PARP1/2 regulation, our mechanistic understanding of the tankyrases lags behind, and thereby our appreciation of the molecular consequences of tankyrase inhibition. Despite large differences in their architecture and cellular contexts, recent structure-function work has revealed striking parallels in the regulatory principles that govern these enzymes. This includes low basal activity, activation by intra- or inter-molecular assembly, negative feedback regulation by auto-PARylation, and allosteric communication. Here we compare these poly-ADP-ribosyltransferases and point towards emerging parallels and open questions, whose pursuit will inform future drug development efforts.
Collapse
Affiliation(s)
- Matthew Jessop
- Division of Structural Biology, The Institute of Cancer Research (ICR), London, U.K
- Division of Cancer Biology, The Institute of Cancer Research (ICR), London, U.K
| | - Benjamin J Broadway
- Division of Structural Biology, The Institute of Cancer Research (ICR), London, U.K
- Division of Cancer Biology, The Institute of Cancer Research (ICR), London, U.K
| | - Katy Miller
- Division of Structural Biology, The Institute of Cancer Research (ICR), London, U.K
- Division of Cancer Biology, The Institute of Cancer Research (ICR), London, U.K
| | - Sebastian Guettler
- Division of Structural Biology, The Institute of Cancer Research (ICR), London, U.K
- Division of Cancer Biology, The Institute of Cancer Research (ICR), London, U.K
| |
Collapse
|
47
|
Chiu SP, Camacho CV, Kraus WL. Development and characterization of recombinant ADP-ribose binding reagents that allow simultaneous detection of mono and poly ADP-ribose. J Biol Chem 2024; 300:107609. [PMID: 39074634 PMCID: PMC11388009 DOI: 10.1016/j.jbc.2024.107609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/29/2024] [Accepted: 07/05/2024] [Indexed: 07/31/2024] Open
Abstract
ADP-ribosylation (ADPRylation) is a post-translational modification (PTM) of proteins mediated by the activity of a variety of ADP-ribosyltransferase (ART) enzymes, such as the Poly (ADP-ribose) Polymerase (PARP) family of proteins. This PTM is diverse in both form and biological functions, which makes it a highly interesting modification, but difficult to study due to limitations in reagents available to detect the diversity of ADPRylation. Recently we developed a set of recombinant antibody-like ADP-ribose (ADPR) binding proteins using naturally occurring ADPR binding domains (ARBDs), including macrodomains and WWE domains, functionalized by fusion to the constant "Fc" region of rabbit immunoglobulin. Herein, we present an expansion of this biological toolkit, where we have replaced the rabbit Fc sequence with the sequence from two other species, mouse and goat. These new reagents are based on a previously characterized set of naturally occurring ARBDs with known specificity. Characterization of the new reagents demonstrates that they can be detected in a species-dependent manner by secondary immunological tools, recognize specific ADPR moieties, and can be used for simultaneous detection of mono ADPR and poly ADPR at single-cell resolution in various antibody-based assays. The expansion of this toolkit will allow for more multiplexed assessments of the complexity of ADPRylation biology in many biological systems.
Collapse
Affiliation(s)
- Shu-Ping Chiu
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Section of Laboratory Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Cristel V Camacho
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Section of Laboratory Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| | - W Lee Kraus
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Section of Laboratory Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| |
Collapse
|
48
|
Dibitetto D, Widmer CA, Rottenberg S. PARPi, BRCA, and gaps: controversies and future research. Trends Cancer 2024; 10:857-869. [PMID: 39004561 DOI: 10.1016/j.trecan.2024.06.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/17/2024] [Accepted: 06/21/2024] [Indexed: 07/16/2024]
Abstract
In recent years, various poly(ADP-ribose) polymerase (PARP) inhibitors (PARPis) have been approved for the treatment of several cancers to target the vulnerability of homologous recombination (HR) deficiency (e.g., due to BRCA1/2 dysfunction). In this review we analyze the ongoing debates and recent breakthroughs in the use of PARPis for BRCA1/2-deficient cancers, juxtaposing the 'double-strand break (DSB)' and 'single-stranded DNA (ssDNA) gap' models of synthetic lethality induced by PARPis. We spotlight the complexity of this interaction, highlighting emerging research on the role of DNA polymerase theta (POLθ) and ssDNA gaps in shaping therapy responses. We scrutinize the clinical ramifications of these findings, especially concerning PARPi efficacy and resistance mechanisms, underscoring the heterogeneity of BRCA-mutated tumors and the urgent need for advanced research to bridge the gap between laboratory models and patient outcomes.
Collapse
Affiliation(s)
- Diego Dibitetto
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Länggassstrasse 122, 3012 Bern, Switzerland; Bern Center for Precision Medicine and Cancer Therapy Research Cluster, Department for Biomedical Research, University of Bern, 3012 Bern, Switzerland; Molecular Oncology and DNA Damage Response Laboratory, Department of Experimental Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy.
| | - Carmen A Widmer
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Länggassstrasse 122, 3012 Bern, Switzerland; Bern Center for Precision Medicine and Cancer Therapy Research Cluster, Department for Biomedical Research, University of Bern, 3012 Bern, Switzerland
| | - Sven Rottenberg
- Institute of Animal Pathology, Vetsuisse Faculty, University of Bern, Länggassstrasse 122, 3012 Bern, Switzerland; Bern Center for Precision Medicine and Cancer Therapy Research Cluster, Department for Biomedical Research, University of Bern, 3012 Bern, Switzerland; Division of Molecular Pathology, The Netherlands Cancer Institute, 1066CX Amsterdam, The Netherlands.
| |
Collapse
|
49
|
Huang M, Zhu X, Wang C, He L, Li L, Wang H, Fan G, Wang Y. PINX1 loss confers susceptibility to PARP inhibition in pan-cancer cells. Cell Death Dis 2024; 15:610. [PMID: 39174499 PMCID: PMC11341912 DOI: 10.1038/s41419-024-07009-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 08/12/2024] [Accepted: 08/16/2024] [Indexed: 08/24/2024]
Abstract
PARP1 is crucial in DNA damage repair, chromatin remodeling, and transcriptional regulation. The principle of synthetic lethality has effectively guided the application of PARP inhibitors in treating tumors carrying BRCA1/2 mutations. Meanwhile, PARP inhibitors have exhibited efficacy in BRCA-proficient patients, further highlighting the necessity for a deeper understanding of PARP1 function and its inhibition in cancer therapy. Here, we unveil PIN2/TRF1-interacting telomerase inhibitor 1 (PINX1) as an uncharacterized PARP1-interacting protein that synergizes with PARP inhibitors upon its depletion across various cancer cell lines. Loss of PINX1 compromises DNA damage repair capacity upon etoposide treatment. The vulnerability of PINX1-deficient cells to etoposide and PARP inhibitors could be effectively restored by introducing either a full-length or a mutant form of PINX1 lacking telomerase inhibitory activity. Mechanistically, PINX1 is recruited to DNA lesions through binding to the ZnF3-BRCT domain of PARP1, facilitating the downstream recruitment of the DNA repair factor XRCC1. In the absence of DNA damage, PINX1 constitutively binds to PARP1, promoting PARP1-chromatin association and transcription of specific DNA damage repair proteins, including XRCC1, and transcriptional regulators, including GLIS3. Collectively, our findings identify PINX1 as a multifaceted partner of PARP1, crucial for safeguarding cells against genotoxic stress and emerging as a potential candidate for targeted tumor therapy.
Collapse
Affiliation(s)
- Mei Huang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Xiaotong Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Chen Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Liying He
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Lei Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Haopeng Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Shanghai Clinical Research and Trial Center, Shanghai, 201210, China
| | - Gaofeng Fan
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
- Shanghai Clinical Research and Trial Center, Shanghai, 201210, China.
| | - Yu Wang
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
50
|
Otarbayev D, Myung K. Exploring factors influencing choice of DNA double-strand break repair pathways. DNA Repair (Amst) 2024; 140:103696. [PMID: 38820807 DOI: 10.1016/j.dnarep.2024.103696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/20/2024] [Accepted: 05/20/2024] [Indexed: 06/02/2024]
Abstract
DNA double-strand breaks (DSBs) represent one of the most severe threats to genomic integrity, demanding intricate repair mechanisms within eukaryotic cells. A diverse array of factors orchestrates the complex choreography of DSB signaling and repair, encompassing repair pathways, such as non-homologous end-joining, homologous recombination, and polymerase-θ-mediated end-joining. This review looks into the intricate decision-making processes guiding eukaryotic cells towards a particular repair pathway, particularly emphasizing the processing of two-ended DSBs. Furthermore, we elucidate the transformative role of Cas9, a site-specific endonuclease, in revolutionizing our comprehension of DNA DSB repair dynamics. Additionally, we explore the burgeoning potential of Cas9's remarkable ability to induce sequence-specific DSBs, offering a promising avenue for precise targeting of tumor cells. Through this comprehensive exploration, we unravel the intricate molecular mechanisms of cellular responses to DSBs, shedding light on both fundamental repair processes and cutting-edge therapeutic strategies.
Collapse
Affiliation(s)
- Daniyar Otarbayev
- Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, South Korea; Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, South Korea
| | - Kyungjae Myung
- Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, South Korea; Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, South Korea.
| |
Collapse
|