1
|
Song B, Yang P, Zhang S. Cell fate regulation governed by p53: Friends or reversible foes in cancer therapy. Cancer Commun (Lond) 2024; 44:297-360. [PMID: 38311377 PMCID: PMC10958678 DOI: 10.1002/cac2.12520] [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: 07/26/2023] [Revised: 01/03/2024] [Accepted: 01/11/2024] [Indexed: 02/10/2024] Open
Abstract
Cancer is a leading cause of death worldwide. Targeted therapies aimed at key oncogenic driver mutations in combination with chemotherapy and radiotherapy as well as immunotherapy have benefited cancer patients considerably. Tumor protein p53 (TP53), a crucial tumor suppressor gene encoding p53, regulates numerous downstream genes and cellular phenotypes in response to various stressors. The affected genes are involved in diverse processes, including cell cycle arrest, DNA repair, cellular senescence, metabolic homeostasis, apoptosis, and autophagy. However, accumulating recent studies have continued to reveal novel and unexpected functions of p53 in governing the fate of tumors, for example, functions in ferroptosis, immunity, the tumor microenvironment and microbiome metabolism. Among the possibilities, the evolutionary plasticity of p53 is the most controversial, partially due to the dizzying array of biological functions that have been attributed to different regulatory mechanisms of p53 signaling. Nearly 40 years after its discovery, this key tumor suppressor remains somewhat enigmatic. The intricate and diverse functions of p53 in regulating cell fate during cancer treatment are only the tip of the iceberg with respect to its equally complicated structural biology, which has been painstakingly revealed. Additionally, TP53 mutation is one of the most significant genetic alterations in cancer, contributing to rapid cancer cell growth and tumor progression. Here, we summarized recent advances that implicate altered p53 in modulating the response to various cancer therapies, including chemotherapy, radiotherapy, and immunotherapy. Furthermore, we also discussed potential strategies for targeting p53 as a therapeutic option for cancer.
Collapse
Affiliation(s)
- Bin Song
- Laboratory of Radiation MedicineWest China Second University HospitalSichuan UniversityChengduSichuanP. R. China
| | - Ping Yang
- Laboratory of Radiation MedicineWest China Second University HospitalSichuan UniversityChengduSichuanP. R. China
| | - Shuyu Zhang
- Laboratory of Radiation MedicineWest China Second University HospitalSichuan UniversityChengduSichuanP. R. China
- The Second Affiliated Hospital of Chengdu Medical CollegeChina National Nuclear Corporation 416 HospitalChengduSichuanP. R. China
- Laboratory of Radiation MedicineNHC Key Laboratory of Nuclear Technology Medical TransformationWest China School of Basic Medical Sciences & Forensic MedicineSichuan UniversityChengduSichuanP. R. China
| |
Collapse
|
2
|
Liao H, Yang S, Liang Z, Xiao L, Xie S, Lin P, Xia F, Fang C, Chen Q, Ling D, Li F. A Cancer Cell Selective Replication Stress Nano Amplifier Promotes Replication Fork Catastrophe to Overcome Radioresistance. ACS NANO 2023; 17:18548-18561. [PMID: 37706454 DOI: 10.1021/acsnano.3c06774] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
Replication stress (RS) induced by DNA damage plays a significant role in conferring the anticancer effects of radiotherapy and is tightly associated with radioresistance of cancer cells. Amplification of RS represents an effective approach to improving the efficacy of radiotherapy, although the development of selective RS amplifiers remains an unexplored frontier. We herein present an RS nano amplifier (RSNA) consisting of a catalytic FePt nanoparticle loaded with the chemotherapeutic doxorubicin (DOX), which selectively exacerbates RS in cancer cells by promoting replication fork (RF) catastrophe. RSNA converts the excessive reactive oxygen species (ROS) in cancer cells into oxygen, enhancing the DNA-damaging effects of radiotherapy to create more template lesions that impede RF progression in coalition with DOX. After radiation, ROS scavenging by RSNA accelerates RF progression through damaged template strands, increasing the frequency of RF collapse into double-strand breaks. Moreover, pretreatment with RSNA accumulates cancer cells in the S phase, exposing more RFs to radiation-induced RS. These effects of RSNA convergently maximize RS in cancer cells, effectively overcoming the radioresistance of cancer cells without affecting normal cells. Our study demonstrates the feasibility of selectively amplifying RS to boost radiotherapy.
Collapse
Affiliation(s)
- Hongwei Liao
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shengfei Yang
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zeyu Liang
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Lin Xiao
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shangzhi Xie
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Peihua Lin
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fan Xia
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chunyan Fang
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qian Chen
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Daishun Ling
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
- WLA Laboratories, Shanghai 201203, China
| | - Fangyuan Li
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- WLA Laboratories, Shanghai 201203, China
| |
Collapse
|
3
|
Shadfar S, Parakh S, Jamali MS, Atkin JD. Redox dysregulation as a driver for DNA damage and its relationship to neurodegenerative diseases. Transl Neurodegener 2023; 12:18. [PMID: 37055865 PMCID: PMC10103468 DOI: 10.1186/s40035-023-00350-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 03/16/2023] [Indexed: 04/15/2023] Open
Abstract
Redox homeostasis refers to the balance between the production of reactive oxygen species (ROS) as well as reactive nitrogen species (RNS), and their elimination by antioxidants. It is linked to all important cellular activities and oxidative stress is a result of imbalance between pro-oxidants and antioxidant species. Oxidative stress perturbs many cellular activities, including processes that maintain the integrity of DNA. Nucleic acids are highly reactive and therefore particularly susceptible to damage. The DNA damage response detects and repairs these DNA lesions. Efficient DNA repair processes are therefore essential for maintaining cellular viability, but they decline considerably during aging. DNA damage and deficiencies in DNA repair are increasingly described in age-related neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and Huntington's disease. Furthermore, oxidative stress has long been associated with these conditions. Moreover, both redox dysregulation and DNA damage increase significantly during aging, which is the biggest risk factor for neurodegenerative diseases. However, the links between redox dysfunction and DNA damage, and their joint contributions to pathophysiology in these conditions, are only just emerging. This review will discuss these associations and address the increasing evidence for redox dysregulation as an important and major source of DNA damage in neurodegenerative disorders. Understanding these connections may facilitate a better understanding of disease mechanisms, and ultimately lead to the design of better therapeutic strategies based on preventing both redox dysregulation and DNA damage.
Collapse
Affiliation(s)
- Sina Shadfar
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Macquarie University, Sydney, NSW, 2109, Australia.
| | - Sonam Parakh
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Macquarie University, Sydney, NSW, 2109, Australia
| | - Md Shafi Jamali
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Macquarie University, Sydney, NSW, 2109, Australia
| | - Julie D Atkin
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Macquarie University, Sydney, NSW, 2109, Australia.
- La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Melbourne, VIC, 3086, Australia.
| |
Collapse
|
4
|
Callaghan CM, Abukhiran IM, Masaadeh A, Van Rheeden RV, Kalen AL, Rodman SN, Petronek MS, Mapuskar KA, George BN, Coleman MC, Goswami PC, Allen BG, Spitz DR, Caster JM. Manipulation of Redox Metabolism Using Pharmacologic Ascorbate Opens a Therapeutic Window for Radio-Sensitization by ATM Inhibitors in Colorectal Cancer. Int J Radiat Oncol Biol Phys 2023; 115:933-944. [PMID: 36228747 PMCID: PMC9974877 DOI: 10.1016/j.ijrobp.2022.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/30/2022] [Accepted: 10/05/2022] [Indexed: 11/11/2022]
Abstract
PURPOSE Ataxia telangiectasia mutated kinase (ATM) inhibitors are potent radiosensitizers that regulate DNA damage responses and redox metabolism, but they have not been translated clinically because of the potential for excess normal tissue toxicity. Pharmacologic ascorbate (P-AscH-; intravenous administration achieving mM plasma concentrations) selectively enhances H2O2-induced oxidative stress and radiosensitization in tumors while acting as an antioxidant and mitigating radiation damage in normal tissues including the bowel. We hypothesized that P-AscH- could enhance the therapeutic index of ATM inhibitor-based chemoradiation by simultaneously enhancing the intended effects of ATM inhibitors in tumors and mitigating off-target effects in adjacent normal tissues. METHODS AND MATERIALS Clonogenic survival was assessed in human (human colon tumor [HCT]116, SW480, HT29) and murine (CT26, MC38) colorectal tumor lines and normal cells (human umbilical vein endothelial cell, FHs74) after radiation ± DNA repair inhibitors ± P-AscH-. Tumor growth delay was assessed in mice with HCT116 or MC38 tumors after fractionated radiation (5 Gy × 3) ± the ATM inhibitor KU60019 ± P-AscH-. Intestinal injury, oxidative damage, and transforming growth factor β immunoreactivity were quantified using immunohistochemistry after whole abdominal radiation (10 Gy) ± KU60019 ± P-AscH-. Cell cycle distribution and ATM subcellular localization were assessed using flow cytometry and immunohistochemistry. The role of intracellular H2O2 fluxes was assessed using a stably expressed doxycycline-inducible catalase transgene. RESULTS KU60019 with P-AscH- enhanced radiosensitization in colorectal cancer models in vitro and in vivo by H2O2-dependent oxidative damage to proteins and enhanced DNA damage, abrogation of the postradiation G2 cell cycle checkpoint, and inhibition of ATM nuclear localization. In contrast, concurrent P-AscH- markedly reduced intestinal toxicity and oxidative damage with KU60019. CONCLUSIONS We provide evidence that redox modulating drugs, such as P-AscH-, may facilitate the clinical translation of ATM inhibitors by enhancing tumor radiosensitization while simultaneously protecting normal tissues.
Collapse
Affiliation(s)
- Cameron M Callaghan
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa
| | - Ibrahim M Abukhiran
- Department of Pathology, University of Iowa Hospitals and Clinics and Carver College of Medicine, Iowa City, Iowa
| | - Amr Masaadeh
- Department of Pathology, University of Iowa Hospitals and Clinics and Carver College of Medicine, Iowa City, Iowa
| | | | - Amanda L Kalen
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa; Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
| | - Samuel N Rodman
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa; Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
| | - Michael S Petronek
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa; Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
| | - Kranti A Mapuskar
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa; Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
| | - Benjamin N George
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa
| | - Mitchell C Coleman
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa; Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
| | - Prabhat C Goswami
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa; Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
| | - Bryan G Allen
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa; Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
| | - Douglas R Spitz
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa; Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
| | - Joseph M Caster
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa; Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa.
| |
Collapse
|
5
|
Yamaguchi K, Horie C, Takane K, Ikenoue T, Nakagawa S, Isobe Y, Ota Y, Ushiku T, Tanaka M, Fujishiro J, Hoshino N, Arisue A, Nishizuka S, Aikou S, Shida D, Furukawa Y. Identification of odontogenic ameloblast associated as a novel target gene of the Wnt/β-catenin signaling pathway. Cancer Sci 2023; 114:948-960. [PMID: 36382598 PMCID: PMC9986071 DOI: 10.1111/cas.15657] [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: 06/28/2022] [Revised: 10/31/2022] [Accepted: 11/09/2022] [Indexed: 11/17/2022] Open
Abstract
The Wnt/β-catenin signaling pathway plays a key role in development and carcinogenesis. Although some target genes of this signaling have been identified in various tissues and neoplasms, the comprehensive understanding of the target genes and their roles in the development of human cancer, including hepatoma and colorectal cancer remain to be fully elucidated. In this study, we searched for genes regulated by the Wnt signaling in liver cancer using HuH-7 hepatoma cells. A comparison of the expression profiles between cells expressing an active form of mutant β-catenin and cells expressing enhanced green fluorescent protein (EGFP) identified seven genes upregulated by the mutant β-catenin gene (CTNNB1). Among the seven genes, we focused in this study on ODAM, odontogenic, ameloblast associated, as a novel target gene. Interestingly, its expression was frequently upregulated in hepatocellular carcinoma, colorectal adenocarcinoma, and hepatoblastoma. We additionally identified a distant enhancer region that was associated with the β-catenin/TCF7L2 complex. Further analyses revealed that ODAM plays an important role in the regulation of the cell cycle, DNA synthesis, and cell proliferation. These data may be useful for clarification of the main molecular mechanism(s) underlying these cancers.
Collapse
Affiliation(s)
- Kiyoshi Yamaguchi
- Division of Clinical Genome Research, The Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Chiaki Horie
- Division of Clinical Genome Research, The Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Kiyoko Takane
- Division of Clinical Genome Research, The Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Tsuneo Ikenoue
- Division of Clinical Genome Research, The Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Saya Nakagawa
- Division of Clinical Genome Research, The Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Yumiko Isobe
- Division of Clinical Genome Research, The Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Yasunori Ota
- Department of Pathology, Research Hospital, The Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Tetsuo Ushiku
- Department of Pathology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Mariko Tanaka
- Department of Pathology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Jun Fujishiro
- Department of Pediatric Surgery, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Noriko Hoshino
- Department of Pediatric Surgery, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Atsuhiro Arisue
- Department of SurgeryIwate Medical University School of MedicineYahabaJapan
| | - Satoshi Nishizuka
- Division of Biomedical Research and DevelopmentIwate Medical University Institute for Biomedical SciencesYahabaJapan
| | - Susumu Aikou
- Division of Frontier Surgery, The Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Dai Shida
- Division of Frontier Surgery, The Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Yoichi Furukawa
- Division of Clinical Genome Research, The Institute of Medical ScienceThe University of TokyoTokyoJapan
| |
Collapse
|
6
|
Barbagallo C, Stella M, Broggi G, Russo A, Caltabiano R, Ragusa M. Genetics and RNA Regulation of Uveal Melanoma. Cancers (Basel) 2023; 15:775. [PMID: 36765733 PMCID: PMC9913768 DOI: 10.3390/cancers15030775] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/20/2023] [Accepted: 01/23/2023] [Indexed: 01/28/2023] Open
Abstract
Uveal melanoma (UM) is the most common intraocular malignant tumor and the most frequent melanoma not affecting the skin. While the rate of UM occurrence is relatively low, about 50% of patients develop metastasis, primarily to the liver, with lethal outcome despite medical treatment. Notwithstanding that UM etiopathogenesis is still under investigation, a set of known mutations and chromosomal aberrations are associated with its pathogenesis and have a relevant prognostic value. The most frequently mutated genes are BAP1, EIF1AX, GNA11, GNAQ, and SF3B1, with mutually exclusive mutations occurring in GNAQ and GNA11, and almost mutually exclusive ones in BAP1 and SF3B1, and BAP1 and EIF1AX. Among chromosomal aberrations, monosomy of chromosome 3 is the most frequent, followed by gain of chromosome 8q, and full or partial loss of chromosomes 1 and 6. In addition, epigenetic mechanisms regulated by non-coding RNAs (ncRNA), namely microRNAs and long non-coding RNAs, have also been investigated. Several papers investigating the role of ncRNAs in UM have reported that their dysregulated expression affects cancer-related processes in both in vitro and in vivo models. This review will summarize current findings about genetic mutations, chromosomal aberrations, and ncRNA dysregulation establishing UM biology.
Collapse
Affiliation(s)
- Cristina Barbagallo
- Department of Biomedical and Biotechnological Sciences—Section of Biology and Genetics, University of Catania, 95123 Catania, Italy
| | - Michele Stella
- Department of Biomedical and Biotechnological Sciences—Section of Biology and Genetics, University of Catania, 95123 Catania, Italy
| | - Giuseppe Broggi
- Department of Medical, Surgical Sciences and Advanced Technologies G.F. Ingrassia—Section of Anatomic Pathology, University of Catania, 95123 Catania, Italy
| | - Andrea Russo
- Department of Ophthalmology, University of Catania, 95123 Catania, Italy
| | - Rosario Caltabiano
- Department of Medical, Surgical Sciences and Advanced Technologies G.F. Ingrassia—Section of Anatomic Pathology, University of Catania, 95123 Catania, Italy
| | - Marco Ragusa
- Department of Biomedical and Biotechnological Sciences—Section of Biology and Genetics, University of Catania, 95123 Catania, Italy
| |
Collapse
|
7
|
Minor Kinases with Major Roles in Cytokinesis Regulation. Cells 2022; 11:cells11223639. [PMID: 36429067 PMCID: PMC9688779 DOI: 10.3390/cells11223639] [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: 10/07/2022] [Revised: 11/07/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022] Open
Abstract
Cytokinesis, the conclusive act of cell division, allows cytoplasmic organelles and chromosomes to be faithfully partitioned between two daughter cells. In animal organisms, its accurate regulation is a fundamental task for normal development and for preventing aneuploidy. Cytokinesis failures produce genetically unstable tetraploid cells and ultimately result in chromosome instability, a hallmark of cancer cells. In animal cells, the assembly and constriction of an actomyosin ring drive cleavage furrow ingression, resulting in the formation of a cytoplasmic intercellular bridge, which is severed during abscission, the final event of cytokinesis. Kinase-mediated phosphorylation is a crucial process to orchestrate the spatio-temporal regulation of the different stages of cytokinesis. Several kinases have been described in the literature, such as cyclin-dependent kinase, polo-like kinase 1, and Aurora B, regulating both furrow ingression and/or abscission. However, others exist, with well-established roles in cell-cycle progression but whose specific role in cytokinesis has been poorly investigated, leading to considering these kinases as "minor" actors in this process. Yet, they deserve additional attention, as they might disclose unexpected routes of cell division regulation. Here, we summarize the role of multifunctional kinases in cytokinesis with a special focus on those with a still scarcely defined function during cell cleavage. Moreover, we discuss their implication in cancer.
Collapse
|
8
|
Soni A, Duan X, Stuschke M, Iliakis G. ATR Contributes More Than ATM in Intra-S-Phase Checkpoint Activation after IR, and DNA-PKcs Facilitates Recovery: Evidence for Modular Integration of ATM/ATR/DNA-PKcs Functions. Int J Mol Sci 2022; 23:7506. [PMID: 35886852 PMCID: PMC9316047 DOI: 10.3390/ijms23147506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/24/2022] [Accepted: 07/04/2022] [Indexed: 11/25/2022] Open
Abstract
The intra-S-phase checkpoint was among the first reported cell cycle checkpoints in mammalian cells. It transiently slows down the rate of DNA replication after DNA damage to facilitate repair and thus prevents genomic instability. The ionizing radiation (IR)-induced intra-S-phase checkpoint in mammalian cells is thought to be mainly dependent upon the kinase activity of ATM. Defects in the intra-S-phase checkpoint result in radio-resistant DNA synthesis (RDS), which promotes genomic instability. ATM belongs to the PI3K kinase family along with ATR and DNA-PKcs. ATR has been shown to be the key kinase for intra-S-phase checkpoint signaling in yeast and has also been implicated in this checkpoint in higher eukaryotes. Recently, contributions of DNA-PKcs to IR-induced G2-checkpoint could also be established. Whether and how ATR and DNA-PKcs are involved in the IR-induced intra-S-phase checkpoint in mammalian cells is incompletely characterized. Here, we investigated the contributions of ATM, ATR, and DNA-PKcs to intra-S-phase checkpoint activation after exposure to IR of human and hamster cells. The results suggest that the activities of both ATM and ATR are essential for efficient intra-S-phase checkpoint activation. Indeed, in a wild-type genetic background, ATR inhibition generates stronger checkpoint defects than ATM inhibition. Similar to G2 checkpoint, DNA-PKcs contributes to the recovery from the intra-S-phase checkpoint. DNA-PKcs-deficient cells show persistent, mainly ATR-dependent intra-S-phase checkpoints. A correlation between the degree of DSB end resection and the strength of the intra-S-phase checkpoint is observed, which again compares well to the G2 checkpoint response. We conclude that the organization of the intra-S-phase checkpoint has a similar mechanistic organization to that of the G2 checkpoint in cells irradiated in the G2 phase.
Collapse
Affiliation(s)
- Aashish Soni
- Division of Experimental Radiation Biology, Department of Radiation Therapy, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (A.S.); (M.S.)
- Institute of Medical Radiation Biology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany;
| | - Xiaolu Duan
- Institute of Medical Radiation Biology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany;
| | - Martin Stuschke
- Division of Experimental Radiation Biology, Department of Radiation Therapy, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (A.S.); (M.S.)
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, and German Cancer Research Center (DKFZ), 45147 Essen, Germany
| | - George Iliakis
- Division of Experimental Radiation Biology, Department of Radiation Therapy, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (A.S.); (M.S.)
- Institute of Medical Radiation Biology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany;
| |
Collapse
|
9
|
Daks A, Fedorova O, Parfenyev S, Nevzorov I, Shuvalov O, Barlev NA. The Role of E3 Ligase Pirh2 in Disease. Cells 2022; 11:1515. [PMID: 35563824 PMCID: PMC9101203 DOI: 10.3390/cells11091515] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 02/04/2023] Open
Abstract
The p53-dependent ubiquitin ligase Pirh2 regulates a number of proteins involved in different cancer-associated processes. Targeting the p53 family proteins, Chk2, p27Kip1, Twist1 and others, Pirh2 participates in such cellular processes as proliferation, cell cycle regulation, apoptosis and cellular migration. Thus, it is not surprising that Pirh2 takes part in the initiation and progression of different diseases and pathologies including but not limited to cancer. In this review, we aimed to summarize the available data on Pirh2 regulation, its protein targets and its role in various diseases and pathological processes, thus making the Pirh2 protein a promising therapeutic target.
Collapse
Affiliation(s)
- Alexandra Daks
- Institute of Cytology RAS, 194064 St. Petersburg, Russia; (O.F.); (S.P.); (I.N.); (O.S.)
| | | | | | | | | | - Nickolai A. Barlev
- Institute of Cytology RAS, 194064 St. Petersburg, Russia; (O.F.); (S.P.); (I.N.); (O.S.)
| |
Collapse
|
10
|
Chen SY, Chao CN, Huang HY, Fang CY. Auranofin induces urothelial carcinoma cell death via reactive oxygen species production and synergy with cisplatin. Oncol Lett 2022; 23:61. [PMID: 35069870 PMCID: PMC8756563 DOI: 10.3892/ol.2021.13179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/30/2021] [Indexed: 11/13/2022] Open
Abstract
Urothelial carcinoma (UC) is one of the most common cancer types of the urinary tract. UC is associated with poor 5-year survival rate, and resistance to cisplatin-based therapy remains a challenge for invasive bladder cancer treatment. Therefore, there is an urgent need to develop new drugs for advanced UC therapy. Auranofin (AF) was developed over 30 years ago for the treatment of rheumatoid arthritis and has been reported to exert an antitumor effect by increasing the level of reactive oxygen species (ROS) in cancer cells. The aim of the present study was to examine the effects of AF on cancer cell proliferation, cell cycle and apoptosis, either alone or in combination with cisplatin. AF induced cell death in two separate cell lines, HT 1376 and BFTC 909, in a concentration- and time-dependent manner by inducing cell cycle arrest. However, the distribution of cells in different phases of the cell cycle differed between the two cell lines, with G0/G1 cell cycle arrest in HT 1376 cells and S phase arrest in BFTC 909 cells. In addition, AF induced apoptosis in HT 1376, as well as redox imbalance in both HT 1376 and BFTC 909 cells. Cell viability was rescued following treatment with N-acetyl-L-cysteine, a ROS scavenger. Furthermore, AF treatment synergistically increased the cytotoxicity of HT 1376 and BFTC 909 cells when combined with cisplatin treatment. These findings suggest that AF may represent a potential candidate drug against UC and increase the therapeutic effect of cisplatin.
Collapse
Affiliation(s)
- San-Yuan Chen
- Department of Chinese Medicine, Ditmanson Medical Foundation, Chiayi Christian Hospital, Chiayi 60002, Taiwan, R.O.C
- Department of Sports Management, Chia Nan University of Pharmacy and Science, Tainan 71710, Taiwan, R.O.C
| | - Chun-Nun Chao
- Department of Pediatrics, Ditmanson Medical Foundation, Chiayi Christian Hospital, Chiayi 60002, Taiwan, R.O.C
- Department of Biotechnology, Asia University, Taichung 41354, Taiwan, R.O.C
| | - Hsin-Yi Huang
- Department of Medical Research, Ditmanson Medical Foundation, Chiayi Christian Hospital, Chiayi 60002, Taiwan, R.O.C
| | - Chiung-Yao Fang
- Department of Medical Research, Ditmanson Medical Foundation, Chiayi Christian Hospital, Chiayi 60002, Taiwan, R.O.C
| |
Collapse
|
11
|
Boice AG, Lopez KE, Pandita RK, Parsons MJ, Charendoff CI, Charaka V, Carisey AF, Pandita TK, Bouchier-Hayes L. Caspase-2 regulates S-phase cell cycle events to protect from DNA damage accumulation independent of apoptosis. Oncogene 2022; 41:204-219. [PMID: 34718349 PMCID: PMC8738157 DOI: 10.1038/s41388-021-02085-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 10/13/2021] [Accepted: 10/18/2021] [Indexed: 11/09/2022]
Abstract
In addition to its classical role in apoptosis, accumulating evidence suggests that caspase-2 has non-apoptotic functions, including regulation of cell division. Loss of caspase-2 is known to increase proliferation rates but how caspase-2 is regulating this process is currently unclear. We show that caspase-2 is activated in dividing cells in G1-phase of the cell cycle. In the absence of caspase-2, cells exhibit numerous S-phase defects including delayed exit from S-phase, defects in repair of chromosomal aberrations during S-phase, and increased DNA damage following S-phase arrest. In addition, caspase-2-deficient cells have a higher frequency of stalled replication forks, decreased DNA fiber length, and impeded progression of DNA replication tracts. This indicates that caspase-2 protects from replication stress and promotes replication fork protection to maintain genomic stability. These functions are independent of the pro-apoptotic function of caspase-2 because blocking caspase-2-induced cell death had no effect on cell division, DNA damage-induced cell cycle arrest, or DNA damage. Thus, our data supports a model where caspase-2 regulates cell cycle and DNA repair events to protect from the accumulation of DNA damage independently of its pro-apoptotic function.
Collapse
Affiliation(s)
- Ashley G Boice
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, TX, 77030, USA
- Texas Children's Hospital William T. Shearer Center for Human Immunobiology, Houston, TX, 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Karla E Lopez
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, TX, 77030, USA
- Texas Children's Hospital William T. Shearer Center for Human Immunobiology, Houston, TX, 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Raj K Pandita
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
- Texas A&M Institute of Biosciences and Technology, Houston, TX, 77030, USA
| | - Melissa J Parsons
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Chloe I Charendoff
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, TX, 77030, USA
- Texas Children's Hospital William T. Shearer Center for Human Immunobiology, Houston, TX, 77030, USA
| | - Vijay Charaka
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Alexandre F Carisey
- Texas Children's Hospital William T. Shearer Center for Human Immunobiology, Houston, TX, 77030, USA
- Department of Pediatrics, Section of Allergy and Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Tej K Pandita
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
- Texas A&M Institute of Biosciences and Technology, Houston, TX, 77030, USA
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Lisa Bouchier-Hayes
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, TX, 77030, USA.
- Texas Children's Hospital William T. Shearer Center for Human Immunobiology, Houston, TX, 77030, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
12
|
Khan FH, Dervan E, Bhattacharyya DD, McAuliffe JD, Miranda KM, Glynn SA. The Role of Nitric Oxide in Cancer: Master Regulator or NOt? Int J Mol Sci 2020; 21:ijms21249393. [PMID: 33321789 PMCID: PMC7763974 DOI: 10.3390/ijms21249393] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/06/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023] Open
Abstract
Nitric oxide (NO) is a key player in both the development and suppression of tumourigenesis depending on the source and concentration of NO. In this review, we discuss the mechanisms by which NO induces DNA damage, influences the DNA damage repair response, and subsequently modulates cell cycle arrest. In some circumstances, NO induces cell cycle arrest and apoptosis protecting against tumourigenesis. NO in other scenarios can cause a delay in cell cycle progression, allowing for aberrant DNA repair that promotes the accumulation of mutations and tumour heterogeneity. Within the tumour microenvironment, low to moderate levels of NO derived from tumour and endothelial cells can activate angiogenesis and epithelial-to-mesenchymal transition, promoting an aggressive phenotype. In contrast, high levels of NO derived from inducible nitric oxide synthase (iNOS) expressing M1 and Th1 polarised macrophages and lymphocytes may exert an anti-tumour effect protecting against cancer. It is important to note that the existing evidence on immunomodulation is mainly based on murine iNOS studies which produce higher fluxes of NO than human iNOS. Finally, we discuss different strategies to target NO related pathways therapeutically. Collectively, we present a picture of NO as a master regulator of cancer development and progression.
Collapse
Affiliation(s)
- Faizan H. Khan
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, National University of Ireland Galway (NUIG), H91 YR71 Galway, Ireland; (F.H.K.); (E.D.); (D.D.B.); (J.D.M.)
| | - Eoin Dervan
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, National University of Ireland Galway (NUIG), H91 YR71 Galway, Ireland; (F.H.K.); (E.D.); (D.D.B.); (J.D.M.)
| | - Dibyangana D. Bhattacharyya
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, National University of Ireland Galway (NUIG), H91 YR71 Galway, Ireland; (F.H.K.); (E.D.); (D.D.B.); (J.D.M.)
| | - Jake D. McAuliffe
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, National University of Ireland Galway (NUIG), H91 YR71 Galway, Ireland; (F.H.K.); (E.D.); (D.D.B.); (J.D.M.)
| | - Katrina M. Miranda
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA;
| | - Sharon A. Glynn
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, National University of Ireland Galway (NUIG), H91 YR71 Galway, Ireland; (F.H.K.); (E.D.); (D.D.B.); (J.D.M.)
- Correspondence:
| |
Collapse
|
13
|
Wang L, Xu X, Teng M, Zhao G, Lei A. Coping with DNA Double-Strand Breaks via ATM Signaling Pathway in Bovine Oocytes. Int J Mol Sci 2020; 21:ijms21238892. [PMID: 33255251 PMCID: PMC7727702 DOI: 10.3390/ijms21238892] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/10/2020] [Accepted: 11/20/2020] [Indexed: 12/11/2022] Open
Abstract
As a common injury almost all cells face, DNA damage in oocytes—especially double-strand breaks (DSBs), which occur naturally during the first meiosis phase (meiosis I) due to synaptic complex separation—affects the fertilization ability of oocytes, instead of causing cancer (as in somatic cells). The mechanism of oocytes to effectively repair DSB damage has not yet been clearly studied, especially considering medically induced DSBs superimposed on naturally occurring DSBs in meiosis I. It was found that maturation rates decreased or increased, respectively corresponding with overexpression or interference of p21 in bovine oocytes. At the same time, the maturation rate of bovine oocytes decreased with a gradual increase in Zeocin dose, and the p21 expression in those immature oocytes changed significantly with the gradual increase in Zeocin dose (same as increased DSB intensity). Same as p21, the variation trend of ATM expression was consistent with the gradual increase in Zeocin dose. Furthermore, the oocytes demonstrated tolerance to DSBs during meiosis I, while the maturation rates decreased when the damage exceeded a certain threshold; according to which, it may be that ATM regulates the p53–p21 pathway to affect the completion of meiosis. In addition, nonhomologous recombination and cumulus cells are potentially involved in the process by which oocytes respond to DSB damage.
Collapse
Affiliation(s)
- Lili Wang
- Shaanxi Stem Cell Engineering and Technology Research Center, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (L.W.); (X.X.); (M.T.)
| | - Xiaolei Xu
- Shaanxi Stem Cell Engineering and Technology Research Center, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (L.W.); (X.X.); (M.T.)
| | - Mingming Teng
- Shaanxi Stem Cell Engineering and Technology Research Center, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (L.W.); (X.X.); (M.T.)
| | - Guimin Zhao
- Key Laboratory of Infection and Immunity of Shandong Province, Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, China;
| | - Anmin Lei
- Shaanxi Stem Cell Engineering and Technology Research Center, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; (L.W.); (X.X.); (M.T.)
- Correspondence: ; Tel./Fax: +86-029-87080068
| |
Collapse
|
14
|
Puts G, Jarrett S, Leonard M, Matsangos N, Snyder D, Wang Y, Vincent R, Portney B, Abbotts R, McLaughlin L, Zalzman M, Rassool F, Kaetzel D. Metastasis Suppressor NME1 Modulates Choice of Double-Strand Break Repair Pathways in Melanoma Cells by Enhancing Alternative NHEJ while Inhibiting NHEJ and HR. Int J Mol Sci 2020; 21:ijms21165896. [PMID: 32824412 PMCID: PMC7460576 DOI: 10.3390/ijms21165896] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/09/2020] [Accepted: 08/13/2020] [Indexed: 01/15/2023] Open
Abstract
Reduced NME1 expression in melanoma cell lines, mouse models of melanoma, and melanoma specimens in human patients is associated with increased metastatic activity. Herein, we investigate the role of NME1 in repair of double-stranded breaks (DSBs) and choice of double-strand break repair (DSBR) pathways in melanoma cells. Using chromatin immunoprecipitation, NME1 was shown to be recruited rapidly and directly to DSBs generated by the homing endonuclease I-PpoI. NME1 was recruited to DSBs within 30 min, in concert with recruitment of ataxia-telangiectasia mutated (ATM) protein, an early step in DSBR complex formation, as well as loss of histone 2B. NME1 was detected up to 5 kb from the break site after DSB induction, suggesting a role in extending chromatin reorganization away from the repair site. shRNA-mediated silencing of NME1 expression led to increases in the homologous recombination (HR) and non-homologous end-joining (NHEJ) pathways of double-strand break repair (DSBR), and reduction in the low fidelity, alternative-NHEJ (A-NHEJ) pathway. These findings suggest low expression of NME1 drives DSBR towards higher fidelity pathways, conferring enhanced genomic stability necessary for rapid and error-free proliferation in invasive and metastatic cells. The novel mechanism highlighted in the current study appears likely to impact metastatic potential and therapy-resistance in advanced melanoma and other cancers.
Collapse
Affiliation(s)
- Gemma Puts
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (G.P.); (M.L.); (N.M.); (D.S.); (Y.W.); (R.V.); (B.P.); (M.Z.)
| | - Stuart Jarrett
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA;
| | - Mary Leonard
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (G.P.); (M.L.); (N.M.); (D.S.); (Y.W.); (R.V.); (B.P.); (M.Z.)
| | - Nicolette Matsangos
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (G.P.); (M.L.); (N.M.); (D.S.); (Y.W.); (R.V.); (B.P.); (M.Z.)
| | - Devin Snyder
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (G.P.); (M.L.); (N.M.); (D.S.); (Y.W.); (R.V.); (B.P.); (M.Z.)
| | - Ying Wang
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (G.P.); (M.L.); (N.M.); (D.S.); (Y.W.); (R.V.); (B.P.); (M.Z.)
| | - Richard Vincent
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (G.P.); (M.L.); (N.M.); (D.S.); (Y.W.); (R.V.); (B.P.); (M.Z.)
| | - Benjamin Portney
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (G.P.); (M.L.); (N.M.); (D.S.); (Y.W.); (R.V.); (B.P.); (M.Z.)
| | - Rachel Abbotts
- Department of Radiation Oncology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (R.A.); (L.M.); (F.R.)
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
| | - Lena McLaughlin
- Department of Radiation Oncology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (R.A.); (L.M.); (F.R.)
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
| | - Michal Zalzman
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (G.P.); (M.L.); (N.M.); (D.S.); (Y.W.); (R.V.); (B.P.); (M.Z.)
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
- Department of Otorhinolaryngology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Feyruz Rassool
- Department of Radiation Oncology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (R.A.); (L.M.); (F.R.)
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
| | - David Kaetzel
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA; (G.P.); (M.L.); (N.M.); (D.S.); (Y.W.); (R.V.); (B.P.); (M.Z.)
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
- Correspondence: ; Tel.: +1-410-706-5080; Fax: +1-410-706-8297
| |
Collapse
|
15
|
Evangelisti G, Barra F, Moioli M, Sala P, Stigliani S, Gustavino C, Costantini S, Ferrero S. Prexasertib: an investigational checkpoint kinase inhibitor for the treatment of high-grade serous ovarian cancer. Expert Opin Investig Drugs 2020; 29:779-792. [PMID: 32539469 DOI: 10.1080/13543784.2020.1783238] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Introduction Patients with high-grade serous ovarian cancer (HGSOC) have a poor prognosis, and current chemotherapy regimens for treating advanced disease are far from satisfactory. Prexasertib (LY2606368) is a novel checkpoint kinase inhibitor (CHK) under investigation for the treatment of HGSOC. Data from a recent phase II trial showed promising efficacy and safety results for treating wild-type BRCA HGSOC. Areas covered This article reviews the available data on the pharmacokinetics, pharmacodynamics, clinical efficacy, and safety of prexasertib in the treatment of HGSOC. Expert opinion Until now, prexasertib demonstrated clinical activity in phase I and II clinical trial for treating wild-type BRCA HGSOC, whereas its promising efficacy as monotherapy and combined with olaparib in BRCA-mutated HGSOC has been preliminary evidenced only in phase I studies. Compared to other drugs of the same class, prexasertib showed a better tolerability profile, causing moderate hematological toxicity. Further studies are needed to confirm efficacy and safety profiles of prexasertib in combined regimens. New early clinical trials may investigate prexasertib administered with programmed cell death ligand 1 (PD-L1) and PI3 K inhibitors due to the preclinical evidence of a synergic action.
Collapse
Affiliation(s)
- Giulio Evangelisti
- Academic Unit of Obstetrics and Gynecology, IRCCS Ospedale Policlinico San Martino , Genoa, Italy.,Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child, Health (Dinogmi), University of Genoa , Italy
| | - Fabio Barra
- Academic Unit of Obstetrics and Gynecology, IRCCS Ospedale Policlinico San Martino , Genoa, Italy.,Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child, Health (Dinogmi), University of Genoa , Italy
| | - Melita Moioli
- Academic Unit of Obstetrics and Gynecology, IRCCS Ospedale Policlinico San Martino , Genoa, Italy.,Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child, Health (Dinogmi), University of Genoa , Italy
| | - Paolo Sala
- Unit of Obstetrics and Gynecology, IRCCS Ospedale Policlinico San Martino , Genoa, Italy.,LILT - Lega Italiana per la Lotta contro i Tumori, Rome, Italy
| | - Sara Stigliani
- Academic Unit of Obstetrics and Gynecology, IRCCS Ospedale Policlinico San Martino , Genoa, Italy.,Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child, Health (Dinogmi), University of Genoa , Italy
| | - Claudio Gustavino
- Unit of Obstetrics and Gynecology, IRCCS Ospedale Policlinico San Martino , Genoa, Italy
| | - Sergio Costantini
- Academic Unit of Obstetrics and Gynecology, IRCCS Ospedale Policlinico San Martino , Genoa, Italy.,Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child, Health (Dinogmi), University of Genoa , Italy
| | - Simone Ferrero
- Academic Unit of Obstetrics and Gynecology, IRCCS Ospedale Policlinico San Martino , Genoa, Italy.,Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child, Health (Dinogmi), University of Genoa , Italy
| |
Collapse
|
16
|
Dhyani A, Favaro P, Olalla Saad ST. ANKHD1 is an S phase protein required for histone synthesis and DNA repair in multiple myeloma cells. Blood Cells Mol Dis 2020; 84:102460. [PMID: 32562952 DOI: 10.1016/j.bcmd.2020.102460] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/04/2020] [Accepted: 06/07/2020] [Indexed: 11/17/2022]
Abstract
ANKHD1 is highly expressed in various cancers such as leukemia and multiple myeloma. Silencing of ANKHD1 expression leads to decreased cell proliferation and accumulation of cells at the S phase. In this study we found ANKHD1 expression to be higher at the S phase, suggesting it to be an S phase protein. We observed that ANKHD1 interacts with histone promoter regions and its inhibition downregulates expression of all core histones, implying a role in histone synthesis. Since histone synthesis occurs in parallel with DNA replication at S phase, we evaluated PCNA (Proliferating Cell Nuclear Antigen) expression, a protein involved in DNA replication and repair. PCNA expression was found to be significantly decreased in ANKHD1 silenced cells. We further observed accumulation γH2AX, a marker for DNA double stranded breaks and an early sign of DNA damage induced by replication stress, upon ANKHD1 silencing. The expressions of several genes implicated in DNA repair were also modulated in ANKHD1 silenced cells, confirming the role of ANKHD1 in DNA repair. Based on this study we speculate that ANKHD1 is an S phase protein required for histone synthesis and DNA repair. These results however, are preliminary and require thorough investigation.
Collapse
Affiliation(s)
- Anamika Dhyani
- Hematology and Hemotherapy Center-University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas, Brazil.
| | - Patricia Favaro
- Department of Biological Sciences, Federal University of Sao Paulo, Diadema, São Paulo, Brazil
| | - Sara T Olalla Saad
- Hematology and Hemotherapy Center-University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas, Brazil
| |
Collapse
|
17
|
Huang RX, Zhou PK. DNA damage response signaling pathways and targets for radiotherapy sensitization in cancer. Signal Transduct Target Ther 2020; 5:60. [PMID: 32355263 PMCID: PMC7192953 DOI: 10.1038/s41392-020-0150-x] [Citation(s) in RCA: 531] [Impact Index Per Article: 132.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/20/2020] [Accepted: 03/16/2020] [Indexed: 12/19/2022] Open
Abstract
Radiotherapy is one of the most common countermeasures for treating a wide range of tumors. However, the radioresistance of cancer cells is still a major limitation for radiotherapy applications. Efforts are continuously ongoing to explore sensitizing targets and develop radiosensitizers for improving the outcomes of radiotherapy. DNA double-strand breaks are the most lethal lesions induced by ionizing radiation and can trigger a series of cellular DNA damage responses (DDRs), including those helping cells recover from radiation injuries, such as the activation of DNA damage sensing and early transduction pathways, cell cycle arrest, and DNA repair. Obviously, these protective DDRs confer tumor radioresistance. Targeting DDR signaling pathways has become an attractive strategy for overcoming tumor radioresistance, and some important advances and breakthroughs have already been achieved in recent years. On the basis of comprehensively reviewing the DDR signal pathways, we provide an update on the novel and promising druggable targets emerging from DDR pathways that can be exploited for radiosensitization. We further discuss recent advances identified from preclinical studies, current clinical trials, and clinical application of chemical inhibitors targeting key DDR proteins, including DNA-PKcs (DNA-dependent protein kinase, catalytic subunit), ATM/ATR (ataxia-telangiectasia mutated and Rad3-related), the MRN (MRE11-RAD50-NBS1) complex, the PARP (poly[ADP-ribose] polymerase) family, MDC1, Wee1, LIG4 (ligase IV), CDK1, BRCA1 (BRCA1 C terminal), CHK1, and HIF-1 (hypoxia-inducible factor-1). Challenges for ionizing radiation-induced signal transduction and targeted therapy are also discussed based on recent achievements in the biological field of radiotherapy.
Collapse
Affiliation(s)
- Rui-Xue Huang
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, 410078, Changsha, People's Republic of China
| | - Ping-Kun Zhou
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, AMMS, 100850, Beijing, People's Republic of China.
- Institute for Chemical Carcinogenesis, State Key Laboratory of Respiratory, Guangzhou Medical University, 511436, Guangzhou, People's Republic of China.
| |
Collapse
|
18
|
Fiévet A, Bellanger D, Zahed L, Burglen L, Derrien AC, Dubois d'Enghien C, Lespinasse J, Parfait B, Pedespan JM, Rieunier G, Stoppa-Lyonnet D, Stern MH. DNA repair functional analyses of NBN hypomorphic variants associated with NBN-related infertility. Hum Mutat 2019; 41:608-618. [PMID: 31729086 DOI: 10.1002/humu.23955] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 10/17/2019] [Accepted: 11/03/2019] [Indexed: 01/17/2023]
Abstract
Nijmegen breakage syndrome caused by biallelic pathogenic variants of the DNA-damage response gene NBN, is characterized by severe microcephaly, cancer proneness, infertility, and karyotype abnormalities. We previously reported NBN variants in siblings suffering from fertility defects. Here, we identify a new founder NBN variant (c.442A>G, p.(Thr148Ala)) in Lebanese patients associated with isolated infertility. Functional analyses explored preserved or altered functions correlated with their remarkably mild phenotype. Transcript and protein analyses supported the use of an alternative transcript with in-frame skipping of exons 4-5, leading to p84-NBN protein with a preserved forkhead-associated (FHA) domain. The level of NBN was dramatically reduced and the MRN complex delocalized to the cytoplasm. Interestingly, ataxia-elangiectasia mutated (ATM) also shifted from the nucleus to the cytoplasm, suggesting some interaction between ATM and the MRN complex at a steady state. The ATM pathway activation, attenuated in typical patients with NBS, appeared normal under camptothecin treatment in these new NBN-related infertile patients. Cell cycle checkpoint defect was present in these atypical patients, although to a lesser extent than in typical patients with NBS. In conclusion, we report three new NBN-related infertile patients and we suggest that preserved FHA domain could be responsible for the mild phenotype and intermediate DNA-damage response defects.
Collapse
Affiliation(s)
- Alice Fiévet
- INSERM U830, Institut Curie, PSL Research University, Paris, France.,D.R.U.M. Team, INSERM U830, Institut Curie, Paris, France.,Service de Génétique, Institut Curie Hôpital, Paris, France.,Service Génétique des Tumeurs, Gustave Roussy, Villejuif, France
| | - Dorine Bellanger
- INSERM U830, Institut Curie, PSL Research University, Paris, France.,D.R.U.M. Team, INSERM U830, Institut Curie, Paris, France
| | - Laila Zahed
- Department of Clinical Laboratory, Saint George Hospital University Medical Center, Beirut, Lebanon
| | - Lydie Burglen
- Centre de Référence des, Malformations et Maladies Congénitales du Cervelet, Paris, France.,GRC n°19, Pathologies Congénitales du Cervelet-LeucoDystrophies, Hôpital Armand Trousseau (APHP), Sorbonne Université, Paris, France.,INSERM U1141, Université Paris Diderot, Paris, France.,Département de Génétique Médicale (GHUEP), Hôpital Armand Trousseau (APHP), Paris, France
| | - Anne-Céline Derrien
- INSERM U830, Institut Curie, PSL Research University, Paris, France.,D.R.U.M. Team, INSERM U830, Institut Curie, Paris, France
| | | | - James Lespinasse
- Génétique Chromosomique, Centre Hospitalier Metropole Savoie, Chambéry-Hôtel-Dieu, Chambéry, France
| | - Béatrice Parfait
- Centre de ressources Biologiques, Hôpital Cochin, Assistance Publique - Hôpitaux de Paris, Paris, France
| | | | - Guillaume Rieunier
- INSERM U830, Institut Curie, PSL Research University, Paris, France.,D.R.U.M. Team, INSERM U830, Institut Curie, Paris, France
| | - Dominique Stoppa-Lyonnet
- D.R.U.M. Team, INSERM U830, Institut Curie, Paris, France.,Service de Génétique, Institut Curie Hôpital, Paris, France.,Université Paris Descartes, Sorbonne-Paris-Cité, Paris, France
| | - Marc-Henri Stern
- INSERM U830, Institut Curie, PSL Research University, Paris, France.,D.R.U.M. Team, INSERM U830, Institut Curie, Paris, France.,Service de Génétique, Institut Curie Hôpital, Paris, France
| |
Collapse
|
19
|
Fiévet A, Bellanger D, Rieunier G, Dubois d'Enghien C, Sophie J, Calvas P, Carriere JP, Anheim M, Castrioto A, Flabeau O, Degos B, Ewenczyk C, Mahlaoui N, Touzot F, Suarez F, Hully M, Roubertie A, Aladjidi N, Tison F, Antoine-Poirel H, Dahan K, Doummar D, Nougues MC, Ioos C, Rougeot C, Masurel A, Bourjault C, Ginglinger E, Prieur F, Siri A, Bordigoni P, Nguyen K, Philippe N, Bellesme C, Demeocq F, Altuzarra C, Mathieu-Dramard M, Couderc F, Dörk T, Auger N, Parfait B, Abidallah K, Moncoutier V, Collet A, Stoppa-Lyonnet D, Stern MH. Functional classification of ATM variants in ataxia-telangiectasia patients. Hum Mutat 2019; 40:1713-1730. [PMID: 31050087 DOI: 10.1002/humu.23778] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 04/24/2019] [Accepted: 04/29/2019] [Indexed: 12/11/2022]
Abstract
Ataxia-telangiectasia (A-T) is a recessive disorder caused by biallelic pathogenic variants of ataxia-telangiectasia mutated (ATM). This disease is characterized by progressive ataxia, telangiectasia, immune deficiency, predisposition to malignancies, and radiosensitivity. However, hypomorphic variants may be discovered associated with very atypical phenotypes, raising the importance of evaluating their pathogenic effects. In this study, multiple functional analyses were performed on lymphoblastoid cell lines from 36 patients, comprising 49 ATM variants, 24 being of uncertain significance. Thirteen patients with atypical phenotype and presumably hypomorphic variants were of particular interest to test strength of functional analyses and to highlight discrepancies with typical patients. Western-blot combined with transcript analyses allowed the identification of one missing variant, confirmed suspected splice defects and revealed unsuspected minor transcripts. Subcellular localization analyses confirmed the low level and abnormal cytoplasmic localization of ATM for most A-T cell lines. Interestingly, atypical patients had lower kinase defect and less altered cell-cycle distribution after genotoxic stress than typical patients. In conclusion, this study demonstrated the pathogenic effects of the 49 variants, highlighted the strength of KAP1 phosphorylation test for pathogenicity assessment and allowed the establishment of the Ataxia-TeLangiectasia Atypical Score to predict atypical phenotype. Altogether, we propose strategies for ATM variant detection and classification.
Collapse
Affiliation(s)
- Alice Fiévet
- Institut Curie, PSL Research University, INSERM U830, Paris, France.,Institut Curie, Hôpital, Service de Génétique, Paris, France
| | - Dorine Bellanger
- Institut Curie, PSL Research University, INSERM U830, Paris, France
| | | | | | - Julia Sophie
- CHU de Toulouse, Service de Génétique Médicale, Toulouse, France
| | - Patrick Calvas
- CHU de Toulouse, Service de Génétique Médicale, Toulouse, France
| | - Jean-Paul Carriere
- Hopital des enfants de Toulouse, Unité de Neuropédiatrie, Toulouse, France
| | - Mathieu Anheim
- CHU de Strasbourg, Service de Neurologie, Strasbourg, France
| | - Anna Castrioto
- CHU de Grenoble, Pole de Psychiatrie et de Neurologie, Grenoble, France
| | - Olivier Flabeau
- CH de la côte Basque, Service de Neurologie, Bayonne, France
| | - Bertrand Degos
- Département des Maladies du Système Nerveux, Hôpitaux Universitaires Pitié Salpêtrière - Charles Foix, Paris, France
| | - Claire Ewenczyk
- Hôpitaux universitaires Pitié Salpêtrière - Charles Foix, Service de Génétique, Paris, France
| | - Nizar Mahlaoui
- Hôpital Necker Enfants Malades, Service d'Immunologie, d'Hématologie et de Rhumatologie Pédiatriques, Paris, France
| | - Fabien Touzot
- Hôpital Necker Enfants Malades, Service d'Immunologie, d'Hématologie et de Rhumatologie Pédiatriques, Paris, France
| | - Felipe Suarez
- Hôpital Necker Enfants Malades, Service d'Hématologie Adulte, Paris, France
| | - Marie Hully
- Hôpital Necker Enfants Malades, Service de Neurologie Pédiatrique, Paris, France
| | - Agathe Roubertie
- CHU de Montpellier, Service de Neuropédiatrie, Montpellier, France
| | | | - François Tison
- CHU de Bordeaux, Département de Neurologie, Bordeaux, France
| | - Hélène Antoine-Poirel
- Centre de Génétique Humaine, Cliniques Universitaires Saint-Luc & Université Catholique de Louvain, Brussels, Belgium
| | - Karine Dahan
- Centre de Génétique Humaine, Cliniques Universitaires Saint-Luc & Université Catholique de Louvain, Brussels, Belgium
| | - Diane Doummar
- Hopital Armand Trousseau, Service de Neurologie Pédiatrique, Paris, France
| | | | - Christine Ioos
- Hôpital Raymond Poincaré, Pôle de Pédiatrie, Garches, France
| | | | - Alice Masurel
- Hopital d'Enfants de Dijon, Service de Génétique, Dijon, France
| | - Caroline Bourjault
- CH de Bretagne sud, Site du Scorff, Service de Pédiatrie, Lorient, France
| | | | - Fabienne Prieur
- CHU de St Etienne, Hôpital Nord, Service de Génétique Médicale, Saint Etienne, France
| | - Aurélie Siri
- CHU de Nancy, Service de Neurologie, Nancy, France
| | - Pierre Bordigoni
- CHU Nancy, Hôpitaux de Brabois, Service de Pédiatrie II, Vandoeuvre, France
| | - Karine Nguyen
- Département de Génétique Médicale, Hopital de la Timone, Marseille, France
| | - Noel Philippe
- Hopital Debrousse, Service d'Hématologie Pédiatrique, Lyon, France
| | - Céline Bellesme
- GH Cochin-saint-Vincent de Paul, Service d'Endocrinologie et de Neurologie Pédiatrique, Paris, France
| | - François Demeocq
- CHU de Clermont-Ferrand, Hôtel Dieu, Service de Pédiatrie B, Clermont-Ferrand, France
| | | | | | - Fanny Couderc
- CH d'Aix en Provence - du Pays d'Aix, Service de Pédiatrie, Aix en Provence, France
| | - Thilo Dörk
- Gynecology Research Unit, Hannover Medical School, Hannover, Germany
| | - Nathalie Auger
- Gustave Roussy, Service Génétique des Tumeurs, Villejuif, France
| | - Béatrice Parfait
- Centre de ressources Biologiques, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris, France
| | | | | | - Agnès Collet
- Institut Curie, Hôpital, Service de Génétique, Paris, France
| | - Dominique Stoppa-Lyonnet
- Institut Curie, PSL Research University, INSERM U830, Paris, France.,Institut Curie, Hôpital, Service de Génétique, Paris, France.,University Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Marc-Henri Stern
- Institut Curie, PSL Research University, INSERM U830, Paris, France.,Institut Curie, Hôpital, Service de Génétique, Paris, France
| |
Collapse
|
20
|
Fiévet A, Bellanger D, Valence S, Mobuchon L, Afenjar A, Giuliano F, Dubois d'Enghien C, Parfait B, Pedespan JM, Auger N, Rieunier G, Collet A, Burglen L, Stoppa-Lyonnet D, Stern MH. Three new cases of ataxia-telangiectasia-like disorder: No impairment of the ATM pathway, but S-phase checkpoint defect. Hum Mutat 2019; 40:1690-1699. [PMID: 31033087 DOI: 10.1002/humu.23773] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 04/18/2019] [Accepted: 04/24/2019] [Indexed: 11/09/2022]
Abstract
Ataxia-telangiectasia-like disorder (ATLD) is a rare genomic instability syndrome caused by biallelic variants of MRE11 (meiotic recombination 11) characterized by progressive cerebellar ataxia and typical karyotype abnormalities. These symptoms are common to those of ataxia-telangiectasia, which is consistent with the key role of MRE11 in ataxia-telangiectasia mutated (ATM) activation after DNA double-strand breaks. Three unrelated French patients were referred with ataxia. Only one had typical karyotype abnormalities. Unreported biallelic MRE11 variants were found in these three cases. Interestingly, one variant (c.424G>A) was present in two cases and haplotype analysis strongly suggested a French founder variant. Variants c.544G>A and c.314+4_314+7del lead to splice defects. The level of MRE11 in lymphoblastoid cell lines was consistently and dramatically reduced. Functional consequences were evaluated on activation of the ATM pathway via phosphorylation of ATM targets (KAP1 and CHK2), but no consistent defect was observed. However, an S-phase checkpoint activation defect after camptothecin was observed in these patients with ATLD. In conclusion, we report the first three French ATLD patients and a French founder variant, and propose an S-phase checkpoint activation study to evaluate the pathogenicity of MRE11 variants.
Collapse
Affiliation(s)
- Alice Fiévet
- Institut Curie, PSL Research University, Paris, France.,INSERM U830, D.R.U.M. team, Paris, France.,Institut Curie, Hôpital, Service de Génétique, Paris, France
| | - Dorine Bellanger
- Institut Curie, PSL Research University, Paris, France.,INSERM U830, D.R.U.M. team, Paris, France
| | - Stéphanie Valence
- APHP, GHUEP, Hôpital Armand Trousseau, Service de Neurologie Pédiatrique, Paris, France.,Centre de Référence Maladies Rares "Malformations et Maladies Congénitales du Cervelet", Paris-Lyon-Lille, France.,Sorbonne Université, GRC n°19, Pathologies Congénitales du Cervelet-LeucoDystrophies, APHP, Hôpital Armand Trousseau, Paris, France.,INSERM U1141, Université Paris Diderot, Paris, France
| | - Lenha Mobuchon
- Institut Curie, PSL Research University, Paris, France.,INSERM U830, D.R.U.M. team, Paris, France
| | - Alexandra Afenjar
- Centre de Référence Maladies Rares "Malformations et Maladies Congénitales du Cervelet", APHP, Hôpital Armand Trousseau, Paris, France
| | - Fabienne Giuliano
- Service de Génétique Médicale, CHU de Nice, Hôpital l'Archet 2, Nice, France
| | | | - Béatrice Parfait
- Centre de Ressources Biologiques, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris, France
| | | | - Nathalie Auger
- Department of Biopathology, Gustave Roussy, Villejuif, France
| | - Guillaume Rieunier
- Institut Curie, PSL Research University, Paris, France.,INSERM U830, D.R.U.M. team, Paris, France
| | - Agnès Collet
- Institut Curie, Hôpital, Service de Génétique, Paris, France
| | - Lydie Burglen
- Centre de Référence Maladies Rares "Malformations et Maladies Congénitales du Cervelet", Paris-Lyon-Lille, France.,Sorbonne Université, GRC n°19, Pathologies Congénitales du Cervelet-LeucoDystrophies, APHP, Hôpital Armand Trousseau, Paris, France.,INSERM U1141, Université Paris Diderot, Paris, France.,Département de Génétique Médicale, APHP, GHUEP, Hôpital Armand Trousseau, Paris, France
| | - Dominique Stoppa-Lyonnet
- INSERM U830, D.R.U.M. team, Paris, France.,Institut Curie, Hôpital, Service de Génétique, Paris, France.,Faculté de Médecine, Université Paris-Descartes, Paris, France
| | - Marc-Henri Stern
- Institut Curie, PSL Research University, Paris, France.,INSERM U830, D.R.U.M. team, Paris, France.,Institut Curie, Hôpital, Service de Génétique, Paris, France
| |
Collapse
|
21
|
Chung S, Vail P, Witkiewicz AK, Knudsen ES. Coordinately Targeting Cell-Cycle Checkpoint Functions in Integrated Models of Pancreatic Cancer. Clin Cancer Res 2018; 25:2290-2304. [PMID: 30538111 DOI: 10.1158/1078-0432.ccr-18-1620] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 10/19/2018] [Accepted: 12/07/2018] [Indexed: 01/02/2023]
Abstract
PURPOSE Cancer cells often have deficiencies in cell-cycle control mechanisms and could be dependent on specific cell-cycle checkpoints to maintain viability. Because of the documented role of KRAS in driving replication stress, we targeted the checkpoint governing DNA replication using CHK1 kinase inhibitors in pancreatic ductal adenocarcinoma (PDAC) models and examined mechanisms of resistance. EXPERIMENTAL DESIGN Single-agent efficacy of CHK1 inhibition was investigated in established and primary PDAC lines. Drug screening was performed to identify cooperative agents. In vitro and in vivo studies were employed to interrogate combination treatment efficacy and mechanisms of resistance. RESULTS Many PDAC models evade single-agent inhibition through mechanisms that allow S-phase progression with CHK1 inhibited. Gene expression analysis revealed FOXM1 as a potential marker of CHK1 sensitivity and defined a form of pancreatic cancer with poor prognosis. Drug screen analysis identified WEE1 as a cooperative agent with CHK1 and was effective in cell culture. In vivo experiments validated the combination efficacy; however, resistance could evolve. Resistance was due to selection of a stable subclone from the original PDX tumor, which harbored high baseline replication stress. In vitro analysis revealed that gemcitabine could eliminate viability in the resistant models. The triplet regimen of gemcitabine, CHK1, and WEE1 inhibition provided strong disease control in all xenograft models interrogated. CONCLUSIONS These results demonstrate the therapeutic resiliency of pancreatic cancer and indicate that coordinately targeting cell-cycle checkpoints in concert with chemotherapy could be particularly efficacious.
Collapse
Affiliation(s)
- Sejin Chung
- Center for Personalized Medicine, Roswell Park Cancer Institute, Buffalo, New York.,Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, New York
| | - Paris Vail
- The University of Arizona Cancer Center, Tucson, Arizona
| | - Agnieszka K Witkiewicz
- Center for Personalized Medicine, Roswell Park Cancer Institute, Buffalo, New York. .,Department of Pathology, Roswell Park Cancer Institute, Buffalo, New York
| | - Erik S Knudsen
- Center for Personalized Medicine, Roswell Park Cancer Institute, Buffalo, New York. .,Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, New York
| |
Collapse
|
22
|
Qiao S, Koyuturk M, Ozsoyoglu MZ. Querying of Disparate Association and Interaction Data in Biomedical Applications. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2018; 15:1052-1065. [PMID: 27959818 DOI: 10.1109/tcbb.2016.2637344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
In biomedical applications, network models are commonly used to represent interactions and higher-level associations among biological entities. Integrated analyses of these interaction and association data has proven useful in extracting knowledge, and generating novel hypotheses for biomedical research. However, since most datasets provide their own schema and query interface, opportunities for exploratory and integrative querying of disparate data are currently limited. In this study, we utilize RDF-based representations of biomedical interaction and association data to develop a querying framework that enables flexible specification and efficient processing of graph template matching queries. The proposed framework enables integrative querying of biomedical databases to discover complex patterns of associations among a diverse range of biological entities, including biomolecules, biological processes, organisms, and phenotypes. Our experimental results on the UniProt dataset show that the proposed framework can be used to efficiently process complex queries, and identify biologically relevant patterns of associations that cannot be readily obtained by querying each dataset independently.
Collapse
|
23
|
Matsuya Y, McMahon SJ, Tsutsumi K, Sasaki K, Okuyama G, Yoshii Y, Mori R, Oikawa J, Prise KM, Date H. Investigation of dose-rate effects and cell-cycle distribution under protracted exposure to ionizing radiation for various dose-rates. Sci Rep 2018; 8:8287. [PMID: 29844494 PMCID: PMC5974424 DOI: 10.1038/s41598-018-26556-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 05/04/2018] [Indexed: 01/04/2023] Open
Abstract
During exposure to ionizing radiation, sub-lethal damage repair (SLDR) competes with DNA damage induction in cultured cells. By virtue of SLDR, cell survival increases with decrease of dose-rate, so-called dose-rate effects (DREs). Here, we focused on a wide dose-rate range and investigated the change of cell-cycle distribution during X-ray protracted exposure and dose-response curves via hybrid analysis with a combination of in vitro experiments and mathematical modelling. In the course of flow-cytometric cell-cycle analysis and clonogenic assays, we found the following responses in CHO-K1 cells: (1) The fraction of cells in S phase gradually increases during 6 h exposure at 3.0 Gy/h, which leads to radio-resistance. (2) Slight cell accumulation in S and G2/M phases is observed after exposure at 6.0 Gy/h for more than 10 hours. This suggests that an increase of SLDR rate for cells in S phase during irradiation may be a reproducible factor to describe changes in the dose-response curve at dose-rates of 3.0 and 6.0 Gy/h. By re-evaluating cell survival for various dose-rates of 0.186-60.0 Gy/h considering experimental-based DNA content and SLDR, it is suggested that the change of S phase fraction during irradiation modulates the dose-response curve and is possibly responsible for some inverse DREs.
Collapse
Affiliation(s)
- Yusuke Matsuya
- Graduate School of Health Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Stephen J McMahon
- Centre for Cancer Research & Cell Biology, Queen's University Belfast, Belfast, BT9 7AE, UK
| | - Kaori Tsutsumi
- Faculty of Health Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Kohei Sasaki
- Faculty of Health Sciences, Hokkaido University of Science, Sapporo, 006-8585, Japan
| | - Go Okuyama
- Faculty of Health Sciences, Hokkaido University of Science, Sapporo, 006-8585, Japan
| | - Yuji Yoshii
- Biological Research, Education and Instrumentation Center, Sapporo Medical University, Sapporo, 060-8556, Japan
| | - Ryosuke Mori
- Graduate School of Health Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Joma Oikawa
- Graduate School of Health Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Kevin M Prise
- Centre for Cancer Research & Cell Biology, Queen's University Belfast, Belfast, BT9 7AE, UK
| | - Hiroyuki Date
- Faculty of Health Sciences, Hokkaido University, Sapporo, 060-0812, Japan.
| |
Collapse
|
24
|
Matsuya Y, McMahon SJ, Tsutsumi K, Sasaki K, Okuyama G, Yoshii Y, Mori R, Oikawa J, Prise KM, Date H. Investigation of dose-rate effects and cell-cycle distribution under protracted exposure to ionizing radiation for various dose-rates. Sci Rep 2018. [PMID: 29844494 DOI: 10.1038/s41598a018-26556a5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023] Open
Abstract
During exposure to ionizing radiation, sub-lethal damage repair (SLDR) competes with DNA damage induction in cultured cells. By virtue of SLDR, cell survival increases with decrease of dose-rate, so-called dose-rate effects (DREs). Here, we focused on a wide dose-rate range and investigated the change of cell-cycle distribution during X-ray protracted exposure and dose-response curves via hybrid analysis with a combination of in vitro experiments and mathematical modelling. In the course of flow-cytometric cell-cycle analysis and clonogenic assays, we found the following responses in CHO-K1 cells: (1) The fraction of cells in S phase gradually increases during 6 h exposure at 3.0 Gy/h, which leads to radio-resistance. (2) Slight cell accumulation in S and G2/M phases is observed after exposure at 6.0 Gy/h for more than 10 hours. This suggests that an increase of SLDR rate for cells in S phase during irradiation may be a reproducible factor to describe changes in the dose-response curve at dose-rates of 3.0 and 6.0 Gy/h. By re-evaluating cell survival for various dose-rates of 0.186-60.0 Gy/h considering experimental-based DNA content and SLDR, it is suggested that the change of S phase fraction during irradiation modulates the dose-response curve and is possibly responsible for some inverse DREs.
Collapse
Affiliation(s)
- Yusuke Matsuya
- Graduate School of Health Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Stephen J McMahon
- Centre for Cancer Research & Cell Biology, Queen's University Belfast, Belfast, BT9 7AE, UK
| | - Kaori Tsutsumi
- Faculty of Health Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Kohei Sasaki
- Faculty of Health Sciences, Hokkaido University of Science, Sapporo, 006-8585, Japan
| | - Go Okuyama
- Faculty of Health Sciences, Hokkaido University of Science, Sapporo, 006-8585, Japan
| | - Yuji Yoshii
- Biological Research, Education and Instrumentation Center, Sapporo Medical University, Sapporo, 060-8556, Japan
| | - Ryosuke Mori
- Graduate School of Health Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Joma Oikawa
- Graduate School of Health Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Kevin M Prise
- Centre for Cancer Research & Cell Biology, Queen's University Belfast, Belfast, BT9 7AE, UK
| | - Hiroyuki Date
- Faculty of Health Sciences, Hokkaido University, Sapporo, 060-0812, Japan.
| |
Collapse
|
25
|
Cleaver JE. Transcription coupled repair deficiency protects against human mutagenesis and carcinogenesis. DNA Repair (Amst) 2017; 58:21-28. [DOI: 10.1016/j.dnarep.2017.08.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 08/07/2017] [Indexed: 11/17/2022]
|
26
|
Hong SY. DNA damage response is hijacked by human papillomaviruses to complete their life cycle. J Zhejiang Univ Sci B 2017; 18:215-232. [PMID: 28271657 DOI: 10.1631/jzus.b1600306] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The DNA damage response (DDR) is activated when DNA is altered by intrinsic or extrinsic agents. This pathway is a complex signaling network and plays important roles in genome stability, tumor transformation, and cell cycle regulation. Human papillomaviruses (HPVs) are the main etiological agents of cervical cancer. Cervical cancer ranks as the fourth most common cancer among women and the second most frequent cause of cancer-related death worldwide. Over 200 types of HPVs have been identified and about one third of these infect the genital tract. The HPV life cycle is associated with epithelial differentiation. Recent studies have shown that HPVs deregulate the DDR to achieve a productive life cycle. In this review, I summarize current findings about how HPVs mediate the ataxia-telangiectasia mutated kinase (ATM) and the ATM-and RAD3-related kinase (ATR) DDRs, and focus on the roles that ATM and ATR signalings play in HPV viral replication. In addition, I demonstrate that the signal transducer and activator of transcription-5 (STAT)-5, an important immune regulator, can promote ATM and ATR activations through different mechanisms. These findings may provide novel opportunities for development of new therapeutic targets for HPV-related cancers.
Collapse
Affiliation(s)
- Shi-Yuan Hong
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
27
|
Sui X, Zhang C, Zhou J, Cao S, Xu C, Tang F, Zhi X, Chen B, Wang S, Yin L. Resveratrol inhibits Extranodal NK/T cell lymphoma through activation of DNA damage response pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:133. [PMID: 28950914 PMCID: PMC5615630 DOI: 10.1186/s13046-017-0601-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 09/13/2017] [Indexed: 02/07/2023]
Abstract
Background Extranodal NK/T cell lymphoma (NKTCL) is a highly aggressive non-Hodgkin lymphoma with poor prognosis. Resveratrol (RSV, 3,5,4′-trihydroxystilbene), a natural nontoxic phenolic compound found in the skin of grapes and some other spermatophytes, performs multiple bioactivities, such as antioxidant activity, anti-aging activity, reduction of cardiovascular disease risk and anticarcinogenic effect. Here we report the anti-tumor effect of RSV in NKTCL cell lines SNT-8, SNK-10 and SNT-16. Results RSV inhibited NKTCL cell proliferation in a dose- and time-dependent manner and arrested cell cycle at S phase. It induced NKTCL cells apoptosis through mitochondrial pathway, shown as down-regulation of MCl-1 and survivin, up-regulation of Bax and Bad, and activation of caspase-9 and caspase-3. In addition, we found that RSV suppressed the phosphorylation level of AKT and Stat3, and activated DNA damage response (DDR) pathway directly or through up-regulation of Zta of Epstein-Barr virus (EBV). Furthermore, using KU55933 as the inhibitor of pATM, we verified that DDR played an important role in RSV inducing NKTCL apoptosis. RSV also showed synergistic effect on activating DDR pathway in combination with etoposide or ionizing radiation, which resulted in cell proliferation inhibition and apoptosis. Conclusions Our results provide in vitro evidence that RSV produces anti-tumor effect by activating DDR pathway in an ATM/Chk2/p53 dependent manner. So we suggest that RSV may be worthy for further study as an anti-tumor drug for NKTCL treatment. Electronic supplementary material The online version of this article (10.1186/s13046-017-0601-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xianxian Sui
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Canjing Zhang
- The Institution of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianan Zhou
- Department of Hematology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shengxuan Cao
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Chen Xu
- Department of Pathology, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Feng Tang
- Department of Pathology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiuling Zhi
- Laboratory of Medical Molecular Biology, Experimental Teaching Center, School of Basic Medical Sciences, Fudan University, 131 Dongan Rd, Shanghai, 200032, China
| | - Bobin Chen
- Department of Hematology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Songmei Wang
- Laboratory of Medical Molecular Biology, Experimental Teaching Center, School of Basic Medical Sciences, Fudan University, 131 Dongan Rd, Shanghai, 200032, China.
| | - Lianhua Yin
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China. .,Laboratory of Medical Molecular Biology, Experimental Teaching Center, School of Basic Medical Sciences, Fudan University, 131 Dongan Rd, Shanghai, 200032, China.
| |
Collapse
|
28
|
Iyer DR, Rhind N. Replication fork slowing and stalling are distinct, checkpoint-independent consequences of replicating damaged DNA. PLoS Genet 2017; 13:e1006958. [PMID: 28806726 PMCID: PMC5570505 DOI: 10.1371/journal.pgen.1006958] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 08/24/2017] [Accepted: 08/04/2017] [Indexed: 11/30/2022] Open
Abstract
In response to DNA damage during S phase, cells slow DNA replication. This slowing is orchestrated by the intra-S checkpoint and involves inhibition of origin firing and reduction of replication fork speed. Slowing of replication allows for tolerance of DNA damage and suppresses genomic instability. Although the mechanisms of origin inhibition by the intra-S checkpoint are understood, major questions remain about how the checkpoint regulates replication forks: Does the checkpoint regulate the rate of fork progression? Does the checkpoint affect all forks, or only those encountering damage? Does the checkpoint facilitate the replication of polymerase-blocking lesions? To address these questions, we have analyzed the checkpoint in the fission yeast Schizosaccharomyces pombe using a single-molecule DNA combing assay, which allows us to unambiguously separate the contribution of origin and fork regulation towards replication slowing, and allows us to investigate the behavior of individual forks. Moreover, we have interrogated the role of forks interacting with individual sites of damage by using three damaging agents-MMS, 4NQO and bleomycin-that cause similar levels of replication slowing with very different frequency of DNA lesions. We find that the checkpoint slows replication by inhibiting origin firing, but not by decreasing fork rates. However, the checkpoint appears to facilitate replication of damaged templates, allowing forks to more quickly pass lesions. Finally, using a novel analytic approach, we rigorously identify fork stalling events in our combing data and show that they play a previously unappreciated role in shaping replication kinetics in response to DNA damage.
Collapse
Affiliation(s)
- Divya Ramalingam Iyer
- Department of Biochemistry & Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Nicholas Rhind
- Department of Biochemistry & Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| |
Collapse
|
29
|
Nowak J, Świątek-Kościelna B, Kałużna EM, Rembowska J, Dzikiewicz-Krawczyk A, Zawada M, Januszkiewicz-Lewandowska D. Effect of irradiation on DNA synthesis, NBN gene expression and chromosomal stability in cells with NBN mutations. Arch Med Sci 2017; 13:283-292. [PMID: 28261280 PMCID: PMC5332466 DOI: 10.5114/aoms.2017.65452] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 04/22/2015] [Indexed: 02/04/2023] Open
Abstract
INTRODUCTION The NBN gene product is part of the MRE11/RAD50/NBN complex, which plays an essential role in genomic stability. In the study we try to answer the question what is the effect of irradiation on DNA synthesis, NBN gene expression and chromosomal stability in cells with homozygous c.657-661del, and heterozygous c.657-661del, p.I171V and p.R215W NBN gene mutations. MATERIAL AND METHODS Immortalized B-lymphocytes with NBN gene mutations were X-ray irradiated at doses of 1, 2, 5 and 8 Gy/min. Radioresistant DNA synthesis rate and the percentage of cells in phase S was analyzed by 3H thymidine and BrdU incorporation assays. NBN gene expression was quantified by real-time PCR with TaqMan fluorescent probe. RESULTS Increasing the irradiation dose resulted in gradual decrease of 3H thymidine incorporation in all cells, but significantly only in homo- and heterozygous c.657-661del cells (p-values < 0.0001). After irradiation the relative expression of NBN was significantly higher in homozygous c.657-661del and heterozygous p.R215W cells as compared to heterozygous c.657-661del, p.I171V and control cells (p < 0.01). All cells with NBN gene mutations showed significantly higher total number of chromosomal aberrations per metaphase as compared to control cells, with the highest number of aberrations in homozygous c.657-661del cells (p < 0.001). CONCLUSIONS The results obtained indicate that homozygous c.657-661del mutation affects cell sensitivity to irradiation. Moreover, homozygous variant is associated with disturbance in the activation of cell cycle checkpoints and with defects in DNA repair. In turn, heterozygous c.657-661del, p.R215W and p.I171V mutations do not substantially alter the radiosensitivity.
Collapse
Affiliation(s)
- Jerzy Nowak
- Department of Molecular Pathology, Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | - Bogna Świątek-Kościelna
- Department of Molecular Pathology, Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | - Ewelina M. Kałużna
- Department of Molecular Pathology, Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | - Jolanta Rembowska
- Department of Molecular Pathology, Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | | | - Mariola Zawada
- Department of Molecular Pathology, Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | - Danuta Januszkiewicz-Lewandowska
- Department of Molecular Pathology, Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
- Department of Paediatric Oncology, Haematology and Bone Marrow Transplantation, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
30
|
The Intra-S Checkpoint Responses to DNA Damage. Genes (Basel) 2017; 8:genes8020074. [PMID: 28218681 PMCID: PMC5333063 DOI: 10.3390/genes8020074] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 02/08/2017] [Accepted: 02/08/2017] [Indexed: 02/03/2023] Open
Abstract
Faithful duplication of the genome is a challenge because DNA is susceptible to damage by a number of intrinsic and extrinsic genotoxins, such as free radicals and UV light. Cells activate the intra-S checkpoint in response to damage during S phase to protect genomic integrity and ensure replication fidelity. The checkpoint prevents genomic instability mainly by regulating origin firing, fork progression, and transcription of G1/S genes in response to DNA damage. Several studies hint that regulation of forks is perhaps the most critical function of the intra-S checkpoint. However, the exact role of the checkpoint at replication forks has remained elusive and controversial. Is the checkpoint required for fork stability, or fork restart, or to prevent fork reversal or fork collapse, or activate repair at replication forks? What are the factors that the checkpoint targets at stalled replication forks? In this review, we will discuss the various pathways activated by the intra-S checkpoint in response to damage to prevent genomic instability.
Collapse
|
31
|
Zhang T, Si-Hoe SL, Hudson DF, Surana U. Condensin recruitment to chromatin is inhibited by Chk2 kinase in response to DNA damage. Cell Cycle 2016; 15:3454-3470. [PMID: 27792460 DOI: 10.1080/15384101.2016.1249075] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
The DNA damage checkpoint, when activated in response to genotoxic damage during S phase, arrests cells in G2 phase of the cell cycle. ATM, ATR, Chk1 and Chk2 kinases are the main effectors of this checkpoint pathway. The checkpoint kinases prevent the onset of mitosis by eliciting well characterized inhibitory phosphorylation of Cdk1. Since Cdk1 is required for the recruitment of condensin, it is thought that upon DNA damage the checkpoint also indirectly blocks chromosome condensation via Cdk1 inhibition. Here we report that the G2 damage checkpoint prevents stable recruitment of the chromosome-packaging-machinery components condensin complex I and II onto the chromatin even in the presence of an active Cdk1. DNA damage-induced inhibition of condensin subunit recruitment is mediated specifically by the Chk2 kinase, implying that the condensin complexes are targeted by the checkpoint in response to DNA damage, independently of Cdk1 inactivation. Thus, the G2 checkpoint directly prevents stable recruitment of condensin complexes to actively prevent chromosome compaction during G2 arrest, presumably to ensure efficient repair of the genomic damage.
Collapse
Affiliation(s)
- Tao Zhang
- a Institute of Molecular and Cell Biology, Agency for Science Technology and Research , Singapore.,b Murdoch Childrens Research Institute, Royal Children's Hospital , Melbourne , Australia.,c Department of Pediatrics , University of Melbourne, Royal Children's Hospital , Melbourne , Australia
| | - San Ling Si-Hoe
- a Institute of Molecular and Cell Biology, Agency for Science Technology and Research , Singapore
| | - Damien F Hudson
- b Murdoch Childrens Research Institute, Royal Children's Hospital , Melbourne , Australia.,c Department of Pediatrics , University of Melbourne, Royal Children's Hospital , Melbourne , Australia
| | - Uttam Surana
- a Institute of Molecular and Cell Biology, Agency for Science Technology and Research , Singapore.,d Department of Pharmacology , National University of Singapore , Singapore.,e Bioprocessing Technology Institute, Agency for Science Technology and Research , Singapore
| |
Collapse
|
32
|
Morgan MA, Lawrence TS. Molecular Pathways: Overcoming Radiation Resistance by Targeting DNA Damage Response Pathways. Clin Cancer Res 2016; 21:2898-904. [PMID: 26133775 DOI: 10.1158/1078-0432.ccr-13-3229] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
DNA double-strand breaks are the critical lesions responsible for the majority of ionizing radiation-induced cell killing. Thus, the ability of tumor cells to elicit a DNA damage response following radiation, via activation of DNA repair and cell-cycle checkpoints, promotes radiation resistance and tumor cell survival. Consequently, agents that target these DNA damage response pathways are being developed to overcome radiation resistance. Overall, these agents are effective radiosensitizers; however, their mechanisms of tumor cell selectivity are not fully elucidated. In this review, we focus on the crucial radiation-induced DNA damage responses as well as clinical and translational advances with agents designed to inhibit these responses. Importantly, we describe how synthetic lethality can provide tumor cell-selective radiosensitization by these agents and expand the therapeutic window for DNA damage response-targeted agents used in combination with radiotherapy.
Collapse
Affiliation(s)
- Meredith A Morgan
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, Michigan.
| | - Theodore S Lawrence
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
33
|
Activation of the DNA Damage Response by RNA Viruses. Biomolecules 2016; 6:2. [PMID: 26751489 PMCID: PMC4808796 DOI: 10.3390/biom6010002] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 11/17/2015] [Accepted: 11/24/2015] [Indexed: 12/11/2022] Open
Abstract
RNA viruses are a genetically diverse group of pathogens that are responsible for some of the most prevalent and lethal human diseases. Numerous viruses introduce DNA damage and genetic instability in host cells during their lifecycles and some species also manipulate components of the DNA damage response (DDR), a complex and sophisticated series of cellular pathways that have evolved to detect and repair DNA lesions. Activation and manipulation of the DDR by DNA viruses has been extensively studied. It is apparent, however, that many RNA viruses can also induce significant DNA damage, even in cases where viral replication takes place exclusively in the cytoplasm. DNA damage can contribute to the pathogenesis of RNA viruses through the triggering of apoptosis, stimulation of inflammatory immune responses and the introduction of deleterious mutations that can increase the risk of tumorigenesis. In addition, activation of DDR pathways can contribute positively to replication of viral RNA genomes. Elucidation of the interactions between RNA viruses and the DDR has provided important insights into modulation of host cell functions by these pathogens. This review summarises the current literature regarding activation and manipulation of the DDR by several medically important RNA viruses.
Collapse
|
34
|
Cordeiro-Stone M, McNulty JJ, Sproul CD, Chastain PD, Gibbs-Flournoy E, Zhou Y, Carson C, Rao S, Mitchell DL, Simpson DA, Thomas NE, Ibrahim JG, Kaufmann WK. Effective intra-S checkpoint responses to UVC in primary human melanocytes and melanoma cell lines. Pigment Cell Melanoma Res 2015; 29:68-80. [PMID: 26437005 DOI: 10.1111/pcmr.12426] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 09/25/2015] [Indexed: 11/29/2022]
Abstract
The objective of this study was to assess potential functional attenuation or inactivation of the intra-S checkpoint during melanoma development. Proliferating cultures of skin melanocytes, fibroblasts, and melanoma cell lines were exposed to increasing fluences of UVC and intra-S checkpoint responses were quantified. Melanocytes displayed stereotypic intra-S checkpoint responses to UVC qualitatively and quantitatively equivalent to those previously demonstrated in skin fibroblasts. In comparison with fibroblasts, primary melanocytes displayed reduced UVC-induced inhibition of DNA strand growth and enhanced degradation of p21Waf1 after UVC, suggestive of enhanced bypass of UVC-induced DNA photoproducts. All nine melanoma cell lines examined, including those with activating mutations in BRAF or NRAS oncogenes, also displayed proficiency in activation of the intra-S checkpoint in response to UVC irradiation. The results indicate that bypass of oncogene-induced senescence during melanoma development was not associated with inactivation of the intra-S checkpoint response to UVC-induced DNA replication stress.
Collapse
Affiliation(s)
- Marila Cordeiro-Stone
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA.,Curriculum in Toxicology, University of North Carolina, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA.,Center for Environmental Health and Susceptibility, University of North Carolina, Chapel Hill, NC, USA
| | - John J McNulty
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | | | - Paul D Chastain
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Eugene Gibbs-Flournoy
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Yingchun Zhou
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Craig Carson
- Department of Dermatology, University of North Carolina, Chapel Hill, NC, USA
| | - Shangbang Rao
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA
| | - David L Mitchell
- Science Park - Research Division, The University of Texas MD Anderson Cancer Center, Smithville, TX, USA
| | - Dennis A Simpson
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Nancy E Thomas
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA.,Center for Environmental Health and Susceptibility, University of North Carolina, Chapel Hill, NC, USA.,Department of Dermatology, University of North Carolina, Chapel Hill, NC, USA
| | - Joseph G Ibrahim
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA
| | - William K Kaufmann
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA.,Curriculum in Toxicology, University of North Carolina, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA.,Center for Environmental Health and Susceptibility, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
35
|
Beccafico S, Morozzi G, Marchetti MC, Riccardi C, Sidoni A, Donato R, Sorci G. Artesunate induces ROS- and p38 MAPK-mediated apoptosis and counteracts tumor growth in vivo in embryonal rhabdomyosarcoma cells. Carcinogenesis 2015; 36:1071-83. [PMID: 26153023 DOI: 10.1093/carcin/bgv098] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 07/01/2015] [Indexed: 12/12/2022] Open
Abstract
Rhabdomyosarcoma represents about 50% of soft-tissue sarcomas and 10% of malignant solid tumors in childhood. Embryonal rhabdomyosarcoma (ERMS) is the most frequent subtype, suggested to have an origin in muscle precursor cells that fail to exit the cell cycle and terminally differentiate mainly because of overexpression of the transcription factor, PAX7, which sustains proliferation, migration and invasiveness in ERMS cells. Artesunate (ARS) is a semi-synthetic derivative of artemisinin (ART), a natural compound well known as an antimalarial drug. However, ART and its derivatives have been found efficacious even as anticancer drugs that induce cell cycle arrest and/or apoptosis in several kinds of cancer. Here, we show that ARS dose-dependently induces DNA damage and apoptosis in ERMS cell lines. Production of reactive oxygen species (ROS) and activation of p38 MAPK have a central role in triggering ARS-mediated apoptosis in ERMS cells; indeed either the antioxidant, N-acetylcysteine or the p38 MAPK inhibitor, SB203580, protects ERMS cells from ARS-induced apoptosis. Moreover, ARS treatment in ERMS cells ROS-dependently induces the expression of the myo-miRs, miR-133a and miR-206, which are down-regulated in RMS, and reduces PAX7 protein levels. Finally, ARS upregulates the expression of the adhesion molecules, NCAM and integrin β1, and reduces migration and invasiveness of ERMS cells in vitro, and ARS treatment reduces of about 50% the growth of ERMS xenografts in vivo. Our results are the first evidence of efficacy of ART derivatives in restraining ERMS growth in vivo, and suggest ARS as a potential candidate for therapeutic treatment of ERMS.
Collapse
Affiliation(s)
- Sara Beccafico
- Department of Experimental Medicine, University of Perugia 06132 Perugia, Italy, Interuniversity Institute of Myology (IIM), Padova, Italy and
| | - Giulio Morozzi
- Department of Experimental Medicine, University of Perugia 06132 Perugia, Italy
| | | | - Carlo Riccardi
- Department of Medicine, University of Perugia 06132 Perugia, Italy
| | - Angelo Sidoni
- Department of Experimental Medicine, University of Perugia 06132 Perugia, Italy
| | - Rosario Donato
- Department of Experimental Medicine, University of Perugia 06132 Perugia, Italy, Interuniversity Institute of Myology (IIM), Padova, Italy and
| | - Guglielmo Sorci
- Department of Experimental Medicine, University of Perugia 06132 Perugia, Italy, Interuniversity Institute of Myology (IIM), Padova, Italy and
| |
Collapse
|
36
|
Zhong X, Luo G, Zhou X, Luo W, Wu X, Zhong R, Wang Y, Xu F, Wang J. Rad51 in regulating the radiosensitivity of non-small cell lung cancer with different epidermal growth factor receptor mutation status. Thorac Cancer 2015; 7:50-60. [PMID: 26816539 PMCID: PMC4718133 DOI: 10.1111/1759-7714.12274] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Accepted: 04/12/2015] [Indexed: 02/05/2023] Open
Abstract
Background Non‐small cell lung cancer (NSCLC) harboring kinase‐domain mutations in epidermal growth factor receptors (EGFR) has been observed to be sensitive to ionizing radiation (IR). We explore Rad51‐dependent homologous recombination (HR) DNA repair in regulating radiosensitivity in two NSCLC cell lines with different EGFR mutation status. Methods NSCLC cell lines, wild‐type EGFR A549 and mutant EGFR H820 with an in‐frame deletion in exon 19 of EGFR (ΔE746–E750), were cultured. Radiosensitivity was estimated by colony forming assay. Rad51 expression was evaluated by quantitative real time‐polymerase chain reaction and Western‐blot. Lentiviral small hairpin ribonucleic acid‐Rad51 and ΔE746–E750 deletion mutant EGFR were constructed and transfected into cells. Flowcytometry assay was used to analyze DNA double strand breaks, cell cycle alterations, and apoptosis. Results A549 had a higher survival factor (SF)2 (0.66 vs. 0.44) and lower α/β value (4.07 vs. 9.01). Compared with the A549 cell, the H820 cell exhibited defective arrest in the S‐phase, a higher rate of G2/M accumulation, early apoptosis, and residual γ‐H2AX. Downregulated Rad51 expression decreased SF2 (0.42 vs. 0.31) and increased the α/β ratio (7.51 vs. 10.5), G2/M accumulation, early apoptosis, and γ‐H2AX in two cell lines. H820 had a low IR‐induced Rad51 expression and nuclear translocation. Exogenous expression of the ΔE746–E750 deletion mutant EGFR caused the A549 cell to become more radiosensitive. Conclusions An EGFR mutated NSCLC cell line is sensitive to IR, which is correlated with reduced IR‐induced Rad51 expression and nuclear translocation. The signaling pathway of EGFR maintaining Rad51 protein levels maybe a novel lung cancer therapeutic target to overcome radioresistance.
Collapse
Affiliation(s)
- Xing Zhong
- Thoracic Oncology West China Hospital Sichuan University Chengdu China; State Key Laboratory of Biotherapy West China Hospital Sichuan University Chengdu China
| | - Guomin Luo
- Thoracic Oncology West China Hospital Sichuan University Chengdu China; State Key Laboratory of Biotherapy West China Hospital Sichuan University Chengdu China
| | - Xiaojuan Zhou
- Thoracic Oncology West China Hospital Sichuan University Chengdu China; State Key Laboratory of Biotherapy West China Hospital Sichuan University Chengdu China
| | - Wen Luo
- Thoracic Oncology West China Hospital Sichuan University Chengdu China; State Key Laboratory of Biotherapy West China Hospital Sichuan University Chengdu China
| | - Xia Wu
- Thoracic Oncology West China Hospital Sichuan University Chengdu China; State Key Laboratory of Biotherapy West China Hospital Sichuan University Chengdu China
| | - Renming Zhong
- State Key Laboratory of Biotherapy West China Hospital Sichuan University Chengdu China
| | - Yanping Wang
- Laboratory of Molecular Diagnosis of Cancer West China Hospital Sichuan University Chengdu China
| | - Feng Xu
- State Key Laboratory of Biotherapy West China Hospital Sichuan University Chengdu China; Abdominal oncology West China Hospital Sichuan University Chengdu China
| | - Jin Wang
- Thoracic Oncology West China Hospital Sichuan University Chengdu China; State Key Laboratory of Biotherapy West China Hospital Sichuan University Chengdu China
| |
Collapse
|
37
|
Gou Q, Xie Y, Liu L, Xie K, Wu Y, Wang Q, Wang Z, Li P. Downregulation of MDC1 and 53BP1 by short hairpin RNA enhances radiosensitivity in laryngeal carcinoma cells. Oncol Rep 2015; 34:251-7. [PMID: 25976740 DOI: 10.3892/or.2015.3980] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Accepted: 02/10/2015] [Indexed: 02/05/2023] Open
Abstract
DNA double-strand breaks (DSBs) induced by ionizing radiation (IR) are among the most cytotoxic types of DNA damage. The DNA damage response (DDR) may be a reason for the cancer cell resistance to radiotherapy using IR. Identified as critical upstream mediators of the phosphorylation of ataxia telangiectasia-mutated (ATM) pathway, mediator of DNA damage checkpoint 1 (MDC1) and p53-binding proteins 1 (53BP1) may affect the radiosensitivity of tumor cells. In the present study, we generated two HEP-2 cell lines with a stable knockdown of MDC1 or 53BP1 with short hairpin RNA (shRNA), respectively, and investigated the effect of MDC1 and 53BP1 on cell radiosensitivity, cell cycle distribution and the formation of cell foci. Downregulation of the two proteins reduced the number of clonogenic cells that treated with IR. Accumulation of G2/M phase cells was detected after the MDC1 and 53BP1 downregulation. These results indicated that the expression of MDC1 or 53BP1 limited tumor cell sensitivity to radiotherapy and may play an important role in the DNA repair progression. Furthermore, the MDC1 foci was identified and presented in the 53BP1-inhibited cells. By contrast, the 53BP1 foci was absent from the MDC1-inhibited cells. The results confirmed that the recruitment of 53BP1 into the foci occurred in an MDC1-dependent manner.
Collapse
Affiliation(s)
- Qiheng Gou
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yuxin Xie
- Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Lei Liu
- Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Keqi Xie
- Department of Anesthesiology, Mianyang Central Hospital, Mianyang, Sichuan 621000, P.R. China
| | - Yanxia Wu
- Laboratory of Molecular Diagnosis of Cancer, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Qianqian Wang
- Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Zhu Wang
- Laboratory of Molecular Diagnosis of Cancer, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Ping Li
- Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
38
|
Dobbelstein M, Sørensen CS. Exploiting replicative stress to treat cancer. Nat Rev Drug Discov 2015; 14:405-23. [PMID: 25953507 DOI: 10.1038/nrd4553] [Citation(s) in RCA: 218] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
DNA replication in cancer cells is accompanied by stalling and collapse of the replication fork and signalling in response to DNA damage and/or premature mitosis; these processes are collectively known as 'replicative stress'. Progress is being made to increase our understanding of the mechanisms that govern replicative stress, thus providing ample opportunities to enhance replicative stress for therapeutic purposes. Rather than trying to halt cell cycle progression, cancer therapeutics could aim to increase replicative stress by further loosening the checkpoints that remain available to cancer cells and ultimately inducing the catastrophic failure of proliferative machineries. In this Review, we outline current and future approaches to achieve this, emphasizing the combination of conventional chemotherapy with targeted approaches.
Collapse
Affiliation(s)
- Matthias Dobbelstein
- Institute of Molecular Oncology, Göttingen Center of Molecular Biosciences, Ernst Caspari Haus, University of Göttingen, 37077 Göttingen, Germany
| | | |
Collapse
|
39
|
Pan X, Mou J, Liu S, Sun Z, Meng R, Zhou Z, Wu G, Peng G. SHP-1 overexpression increases the radioresistance of NPC cells by enhancing DSB repair, increasing S phase arrest and decreasing cell apoptosis. Oncol Rep 2015; 33:2999-3005. [PMID: 25962492 DOI: 10.3892/or.2015.3939] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 03/24/2015] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to investigate the influence of SHP-1 on the radioresistance of the nasopharyngeal carcinoma (NPC) cell line CNE-2 and the relevant underlying mechanisms. The human NPC cell line CNE-2 was transfected with a lentivirus that contained the SHP-1 gene or a nonsense sequence (referred to as LP-H1802Lv201 and LP-NegLv201 cells, respectively). Cells were irradiated with different ionizing radiation (IR) doses. Cell survival, DNA double-strand breaks (DSBs), apoptosis, cell cycle distribution, and the expression of related proteins were assessed using colony formation assay, immunofluorescent assays (IFAs), flow cytometry (FCM) and western blot analyses, respectively. Compared with the control (CNE-2 cells) and LP-NegLv201 cells, LP-H1802Lv201 cells were more resistant to IR. IFAs showed that IR caused less histone H2AX phosphorylation (γH2AX) and RAD51 foci in the LP-H1802Lv201 cells. Compared with the control and LP-NegLv201 cells, LP-H1802Lv201 cells showed increased S phase arrest. After IR, the apoptotic rate of the LP-H1802Lv201 cells was lower in contrast to the control and LP-NegLv201 cells. Western blot analyses showed that IR increased the phosphorylation of ataxia telangiectasia mutated (ATM) kinase, checkpoint kinase 2 (CHK2), ataxia telangiectasia and Rad3-related (ATR) protein, checkpoint kinase 1 (CHK1) and p53. In LP-H1802Lv201 cells, the phosphorylation levels of ATM and CHK2 were significantly increased while the p53 phosphorylation level was decreased compared to these levels in the control and LP-NegLv201 cells. Phosphorylation of ATR and CHK1 did not show significant differences in the three cell groups. Overexpression of SHP-1 in the CNE-2 cells led to radioresistance and the radioresistance was related to enhanced DNA DSB repair, increased S phase arrest and decreased cell apoptosis.
Collapse
Affiliation(s)
- Xiaofen Pan
- Cancer Center, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jingjing Mou
- Cancer Center, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Sha Liu
- Cancer Center, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Ziyi Sun
- Cancer Center, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Rui Meng
- Cancer Center, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Zhenwei Zhou
- Cancer Center, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Gang Wu
- Cancer Center, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Gang Peng
- Cancer Center, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
40
|
Dicks N, Gutierrez K, Michalak M, Bordignon V, Agellon LB. Endoplasmic reticulum stress, genome damage, and cancer. Front Oncol 2015; 5:11. [PMID: 25692096 PMCID: PMC4315039 DOI: 10.3389/fonc.2015.00011] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Accepted: 01/12/2015] [Indexed: 01/30/2023] Open
Abstract
Endoplasmic reticulum (ER) stress has been linked to many diseases, including cancer. A large body of work has focused on the activation of the ER stress response in cancer cells to facilitate their survival and tumor growth; however, there are some studies suggesting that the ER stress response can also mitigate cancer progression. Despite these contradictions, it is clear that the ER stress response is closely associated with cancer biology. The ER stress response classically encompasses activation of three separate pathways, which are collectively categorized the unfolded protein response (UPR). The UPR has been extensively studied in various cancers and appears to confer a selective advantage to tumor cells to facilitate their enhanced growth and resistance to anti-cancer agents. It has also been shown that ER stress induces chromatin changes, which can also facilitate cell survival. Chromatin remodeling has been linked with many cancers through repression of tumor suppressor and apoptosis genes. Interplay between the classic UPR and genome damage repair mechanisms may have important implications in the transformation process of normal cells into cancer cells.
Collapse
Affiliation(s)
- Naomi Dicks
- Department of Animal Science, McGill University , Montréal, QC , Canada
| | - Karina Gutierrez
- Department of Animal Science, McGill University , Montréal, QC , Canada
| | - Marek Michalak
- Department of Biochemistry, University of Alberta , Edmonton, AB , Canada
| | - Vilceu Bordignon
- Department of Animal Science, McGill University , Montréal, QC , Canada
| | - Luis B Agellon
- School of Dietetics and Human Nutrition, McGill University , Montréal, QC , Canada
| |
Collapse
|
41
|
Differential S-phase progression after irradiation of p53 functional versus non-functional tumour cells. Radiol Oncol 2014; 48:354-60. [PMID: 25435848 PMCID: PMC4230555 DOI: 10.2478/raon-2014-0032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 07/24/2014] [Indexed: 11/20/2022] Open
Abstract
Background Many pathways seem to be involved in the regulation of the intra-S-phase checkpoint after exposure to ionizing radiation, but the role of p53 has proven to be rather elusive. Here we have a closer look at the progression of irradiated cells through S-phase in dependence of their p53 status. Materials and methods. Three pairs of tumour cell lines were used, each consisting of one p53 functional and one p53 non-functional line. Cells were labelled with bromodeoxyuridine(BrdU) immediately after irradiation, they were then incubated in label-free medium, and at different times afterwards their position within the S-phase was determined by means of flow cytometry. Results While in the p53 deficient cells progression through S-phase was slowed significantly over at least a few hours, it was halted for just about an hour in the p53 proficient cells and then proceeded without further delay or even at a slightly accelerated pace. Conclusions It is clear from the experiments presented here that p53 does play a role for the progress of cells through the S-phase after X-ray exposure, but the exact mechanisms by which replicon initiation and elongation is controlled in irradiated cells remain to be elucidated.
Collapse
|
42
|
González Besteiro MA, Gottifredi V. The fork and the kinase: a DNA replication tale from a CHK1 perspective. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2014; 763:168-80. [PMID: 25795119 DOI: 10.1016/j.mrrev.2014.10.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Revised: 10/07/2014] [Accepted: 10/10/2014] [Indexed: 11/30/2022]
Abstract
Replication fork progression is being continuously hampered by exogenously introduced and naturally occurring DNA lesions and other physical obstacles. Checkpoint kinase 1 (Chk1) is activated at replication forks that encounter damaged DNA. Subsequently, Chk1 inhibits the initiation of new replication factories and stimulates the firing of dormant origins (those in the vicinity of stalled forks). Chk1 also avoids fork collapse into DSBs (double strand breaks) and promotes fork elongation. At the molecular level, the current model considers stalled forks as the site of Chk1 activation and the nucleoplasm as the location where Chk1 phosphorylates target proteins. This model certainly serves to explain how Chk1 modulates origin firing, but how Chk1 controls the fate of stalled forks is less clear. Interestingly, recent reports demonstrating that Chk1 phosphorylates chromatin-bound proteins and even holds kinase-independent functions might shed light on how Chk1 contributes to the elongation of damaged DNA. Indeed, such findings have unveiled a puzzling connection between Chk1 and DNA lesion bypass, which might be central to promoting fork elongation and checkpoint attenuation. In summary, Chk1 is a multifaceted and versatile signaling factor that acts at ongoing forks and replication origins to determine the extent and quality of the cellular response to replication stress.
Collapse
Affiliation(s)
- Marina A González Besteiro
- Cell Cycle and Genomic Stability Laboratory, Fundación Instituto Leloir, CONICET, Buenos Aires, Argentina
| | - Vanesa Gottifredi
- Cell Cycle and Genomic Stability Laboratory, Fundación Instituto Leloir, CONICET, Buenos Aires, Argentina.
| |
Collapse
|
43
|
Liu T, Huang J. Quality control of homologous recombination. Cell Mol Life Sci 2014; 71:3779-97. [PMID: 24858417 PMCID: PMC11114062 DOI: 10.1007/s00018-014-1649-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 05/09/2014] [Indexed: 12/21/2022]
Abstract
Exogenous and endogenous genotoxic agents, such as ionizing radiation and numerous chemical agents, cause DNA double-strand breaks (DSBs), which are highly toxic and lead to genomic instability or tumorigenesis if not repaired accurately and efficiently. Cells have over evolutionary time developed certain repair mechanisms in response to DSBs to maintain genomic integrity. Major DSB repair mechanisms include non-homologous end joining and homologous recombination (HR). Using sister homologues as templates, HR is a high-fidelity repair pathway that can rejoin DSBs without introducing mutations. However, HR execution without appropriate guarding may lead to more severe gross genome rearrangements. Here we review current knowledge regarding the factors and mechanisms required for accomplishment of accurate HR.
Collapse
Affiliation(s)
- Ting Liu
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058 Zhejiang China
| | - Jun Huang
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058 Zhejiang China
| |
Collapse
|
44
|
Abstract
Cell-cycle checkpoints are generally global in nature: one unattached kinetochore prevents the segregation of all chromosomes; stalled replication forks inhibit late origin firing throughout the genome. A potential exception to this rule is the regulation of replication fork progression by the S-phase DNA damage checkpoint. In this case, it is possible that the checkpoint is global, and it slows all replication forks in the genome. However, it is also possible that the checkpoint acts locally at sites of DNA damage, and only slows those forks that encounter DNA damage. Whether the checkpoint regulates forks globally or locally has important mechanistic implications for how replication forks deal with damaged DNA during S-phase.
Collapse
|
45
|
Multifunctional role of ATM/Tel1 kinase in genome stability: from the DNA damage response to telomere maintenance. BIOMED RESEARCH INTERNATIONAL 2014; 2014:787404. [PMID: 25247188 PMCID: PMC4163350 DOI: 10.1155/2014/787404] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 07/28/2014] [Accepted: 08/07/2014] [Indexed: 12/19/2022]
Abstract
The mammalian protein kinase ataxia telangiectasia mutated (ATM) is a key regulator of the DNA double-strand-break response and belongs to the evolutionary conserved phosphatidylinositol-3-kinase-related protein kinases. ATM deficiency causes ataxia telangiectasia (AT), a genetic disorder that is characterized by premature aging, cerebellar neuropathy, immunodeficiency, and predisposition to cancer. AT cells show defects in the DNA damage-response pathway, cell-cycle control, and telomere maintenance and length regulation. Likewise, in Saccharomyces cerevisiae, haploid strains defective in the TEL1 gene, the ATM ortholog, show chromosomal aberrations and short telomeres. In this review, we outline the complex role of ATM/Tel1 in maintaining genomic stability through its control of numerous aspects of cellular survival. In particular, we describe how ATM/Tel1 participates in the signal transduction pathways elicited by DNA damage and in telomere homeostasis and its importance as a barrier to cancer development.
Collapse
|
46
|
The role of nibrin in doxorubicin-induced apoptosis and cell senescence in Nijmegen Breakage Syndrome patients lymphocytes. PLoS One 2014; 9:e104964. [PMID: 25119968 PMCID: PMC4132076 DOI: 10.1371/journal.pone.0104964] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 07/16/2014] [Indexed: 11/19/2022] Open
Abstract
Nibrin plays an important role in the DNA damage response (DDR) and DNA repair. DDR is a crucial signaling pathway in apoptosis and senescence. To verify whether truncated nibrin (p70), causing Nijmegen Breakage Syndrome (NBS), is involved in DDR and cell fate upon DNA damage, we used two (S4 and S3R) spontaneously immortalized T cell lines from NBS patients, with the founding mutation and a control cell line (L5). S4 and S3R cells have the same level of p70 nibrin, however p70 from S4 cells was able to form more complexes with ATM and BRCA1. Doxorubicin-induced DDR followed by cell senescence could only be observed in L5 and S4 cells, but not in the S3R ones. Furthermore the S3R cells only underwent cell death, but not senescence after doxorubicin treatment. In contrary to doxorubicin treatment, cells from all three cell lines were able to activate the DDR pathway after being exposed to γ-radiation. Downregulation of nibrin in normal human vascular smooth muscle cells (VSMCs) did not prevent the activation of DDR and induction of senescence. Our results indicate that a substantially reduced level of nibrin or its truncated p70 form is sufficient to induce DNA-damage dependent senescence in VSMCs and S4 cells, respectively. In doxorubicin-treated S3R cells DDR activation was severely impaired, thus preventing the induction of senescence.
Collapse
|
47
|
Olcina MM, Grand RJ, Hammond EM. ATM activation in hypoxia - causes and consequences. Mol Cell Oncol 2014; 1:e29903. [PMID: 27308313 PMCID: PMC4905164 DOI: 10.4161/mco.29903] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 06/14/2014] [Accepted: 06/30/2014] [Indexed: 01/07/2023]
Abstract
The DNA damage response is a complex signaling cascade that is triggered by cellular stress. This response is essential for the maintenance of genomic integrity and is considered to act as a barrier to the early stages of tumorigenesis. The integral role of ataxia telangiectasia mutated (ATM) kinase in the response to DNA damaging agents is well characterized; however, ATM can also be activated by non-DNA damaging agents. In fact, much has been learnt recently about the mechanism of ATM activation in response to physiologic stresses such as hypoxia that do not induce DNA damage. Regions of low oxygen concentrations that occur in solid tumors are associated with a poor prognostic outcome irrespective of treatment modality. Severe levels of hypoxia induce replication stress and trigger the activation of DNA damage response pathways including ataxia telangiectasia and Rad3-related (ATR)- and ATM-mediated signaling. In this review, we discuss hypoxia-driven ATM signaling and the possible contribution of ATM activation in this context to tumorigenesis.
Collapse
Affiliation(s)
- Monica M Olcina
- Cancer Research UK/MRC Oxford Institute for Radiation Oncology; Department of Oncology; University of Oxford; Oxford, UK
| | - Roger Ja Grand
- School of Cancer Sciences; College of Medical and Dental Sciences; University of Birmingham; Birmingham, UK
| | - Ester M Hammond
- Cancer Research UK/MRC Oxford Institute for Radiation Oncology; Department of Oncology; University of Oxford; Oxford, UK
| |
Collapse
|
48
|
Hong S, Laimins LA. Regulation of the life cycle of HPVs by differentiation and the DNA damage response. Future Microbiol 2014; 8:1547-57. [PMID: 24266355 DOI: 10.2217/fmb.13.127] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
HPVs are the causative agents of cervical and other anogenital cancers. HPVs infect stratified epithelia and link their productive life cycles to cellular differentiation. Low levels of viral genomes are stably maintained in undifferentiated cells and productive replication or amplification is restricted to differentiated suprabasal cells. Amplification is dependent on the activation of the ATM DNA damage factors that are recruited to viral replication centers and inhibition of this pathway blocks productive replication. The STAT-5 protein appears to play a critical role in mediating activation of the ATM pathway in HPV-positive cells. While HPVs need to activate the DNA damage pathway for replication, cervical cancers contain many genomic alterations suggesting that this pathway is circumvented during progression to malignancy.
Collapse
Affiliation(s)
- Shiyuan Hong
- Department of Microbiology-Immunology, Northwestern University, Feinberg, School of Medicine, Chicago Avenue, Morton 6-681, Chicago, IL 60611, USA
| | | |
Collapse
|
49
|
Co-abrogation of Chk1 and Chk2 by potent oncolytic adenovirus potentiates the antitumor efficacy of cisplatin or irradiation. Cancer Gene Ther 2014; 21:209-17. [PMID: 24853623 DOI: 10.1038/cgt.2014.20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Revised: 04/14/2014] [Accepted: 04/14/2014] [Indexed: 11/08/2022]
Abstract
Mammalian checkpoint kinases 1 and 2 (Chk1 and Chk2) are essential kinases that are involved in cell cycle checkpoint control, and the abrogation of either has been proposed as one way to sensitize cancer cells to DNA-damaging agents. However, it remains unclear which kinase is the most therapeutically relevant target, and whether multiple kinases might need to be targeted to achieve the best efficacy because of their overlapping substrate spectra and redundant functions. To clarify this issue, we established asynchronous cell cycle arrest models to investigate the therapeutic outcomes of silencing Chk1 and Chk2 in the presence of irradiation or cisplatin. Our results showed that Chk1- and Chk2-targeting small interference RNAs (siRNAs) demonstrated synergistic effects when both siRNAs were used simultaneously. Interestingly, Chk1 and Chk2 co-expression occurred in ∼90% of neoplastic tissues examined and showed no difference in neoplastic versus non-neoplastic tissues. Therefore, the selective targeting of Chk1 and Chk2 by oncolytic adenovirus mutants was chosen to treat resistant tumor xenograft mice, and the maximum antitumoral efficacy was achieved with the combined co-abrogation of Chk1 and Chk2 in the presence of low-dose cisplatin. This work deepens our understanding of novel strategies that target checkpoint pathways and contributes to the development of novel, potent and safe checkpoint abrogators.
Collapse
|
50
|
Zhao T, Sun Q, del Rincon SV, Lovato A, Marques M, Witcher M. Gallotannin imposes S phase arrest in breast cancer cells and suppresses the growth of triple-negative tumors in vivo. PLoS One 2014; 9:e92853. [PMID: 24658335 PMCID: PMC3962455 DOI: 10.1371/journal.pone.0092853] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 02/27/2014] [Indexed: 12/18/2022] Open
Abstract
Triple-negative breast cancers are associated with poor clinical outcomes and new therapeutic strategies are clearly needed. Gallotannin (Gltn) has been previously demonstrated to have potent anti-tumor properties against cholangiocarcinoma in mice, but little is known regarding its capacity to suppress tumor outgrowth in breast cancer models. We tested Gltn for potential growth inhibitory properties against a variety of breast cancer cell lines in vitro. In particular, triple-negative breast cancer cells display higher levels of sensitivity to Gltn. The loss of proliferative capacity in Gltn exposed cells is associated with slowed cell cycle progression and S phase arrest, dependent on Chk2 phosphorylation and further characterized by changes to proliferation related genes, such as cyclin D1 (CcnD1) as determined by Nanostring technology. Importantly, Gltn administered orally or via intraperitoneal (IP) injections greatly reduced tumor outgrowth of triple-negative breast cells from mammary fat pads without signs of toxicity. In conclusion, these data strongly suggest that Gltn represents a novel approach to treat triple-negative breast carcinomas.
Collapse
Affiliation(s)
- Tiejun Zhao
- The Lady Davis Institute and Segal Cancer Center of the Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Qiang Sun
- The Lady Davis Institute and Segal Cancer Center of the Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Sonia V. del Rincon
- The Lady Davis Institute and Segal Cancer Center of the Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Amanda Lovato
- The Lady Davis Institute and Segal Cancer Center of the Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Maud Marques
- The Lady Davis Institute and Segal Cancer Center of the Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Michael Witcher
- The Lady Davis Institute and Segal Cancer Center of the Jewish General Hospital, McGill University, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|