1
|
Zhou L, Zhu J, Liu Y, Zhou P, Gu Y. Mechanisms of radiation-induced tissue damage and response. MedComm (Beijing) 2024; 5:e725. [PMID: 39309694 PMCID: PMC11413508 DOI: 10.1002/mco2.725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 09/25/2024] Open
Abstract
Radiation-induced tissue injury (RITI) is the most common complication in clinical tumor radiotherapy. Due to the heterogeneity in the response of different tissues to radiation (IR), radiotherapy will cause different types and degrees of RITI, which greatly limits the clinical application of radiotherapy. Efforts are continuously ongoing to elucidate the molecular mechanism of RITI and develop corresponding prevention and treatment drugs for RITI. Single-cell sequencing (Sc-seq) has emerged as a powerful tool in uncovering the molecular mechanisms of RITI and for identifying potential prevention targets by enhancing our understanding of the complex intercellular relationships, facilitating the identification of novel cell phenotypes, and allowing for the assessment of cell heterogeneity and spatiotemporal developmental trajectories. Based on a comprehensive review of the molecular mechanisms of RITI, we analyzed the molecular mechanisms and regulatory networks of different types of RITI in combination with Sc-seq and summarized the targeted intervention pathways and therapeutic drugs for RITI. Deciphering the diverse mechanisms underlying RITI can shed light on its pathogenesis and unveil new therapeutic avenues to potentially facilitate the repair or regeneration of currently irreversible RITI. Furthermore, we discuss how personalized therapeutic strategies based on Sc-seq offer clinical promise in mitigating RITI.
Collapse
Affiliation(s)
- Lin Zhou
- Beijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingChina
| | - Jiaojiao Zhu
- Beijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingChina
| | - Yuhao Liu
- Beijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingChina
| | - Ping‐Kun Zhou
- Beijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingChina
| | - Yongqing Gu
- Beijing Key Laboratory for RadiobiologyBeijing Institute of Radiation MedicineBeijingChina
- Hengyang Medical CollegeUniversity of South ChinaHengyangHunanChina
- College of Life SciencesHebei UniversityBaodingChina
| |
Collapse
|
2
|
Gharib OA, Fahmy HA, Abdou FY. Role of Olive Leaf Extract, Mesenchymal Stem Cells or Low Radiation Dose in Alleviating Hepatic Injury in Rats. Dose Response 2024; 22:15593258241289301. [PMID: 39483141 PMCID: PMC11526167 DOI: 10.1177/15593258241289301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 08/28/2024] [Indexed: 11/03/2024] Open
Abstract
Objectives This study was conducted to determine the efficacy of mesenchymal stem cells (MSCs) or low-dose gamma radiation (LDR) on liver injury compared to the effect of olive leaf extract as a hepatoprotective agent. Methods Rats were allocated into six groups; group I served as the negative control. Group II received 5% dextran sodium sulfate (DSS) in its drinking water for 1 week. Group III was injected with a single dose of 1 × 106 bone marrow-derived mesenchymal stem cells (BM-MSCs) intravenously. Group IV was treated as in group III after 5% DSS treatment. Group V was given 5% DSS, followed by olive leaf extract (OLE) (1000 mg/ kg, oral). Group VI: 5% DSS for 1 week, then was exposed to low-dose gamma radiation (LDR) (0.05 Gy). Results Rats treated with OLE, BM-MSCs, or exposed to LDR exerted significant alleviation in all hepatic biomarkers, significant enhancements in oxidative stress parameters, and improvements in inflammatory biomarkers Interleukin-1 beta (IL-1β) and Interferon gamma (INF-γ) hepatic contents compared with those of the DSS group. Histological pictures emphasized the biochemical findings. Conclusions BM-MSCs might be a valuable therapeutic approach to overcome hepatic injury. Exposure to LDR provided protective mechanisms that allow the body to survive better.
Collapse
Affiliation(s)
- Ola A. Gharib
- Drug Radiation Research Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| | - Hanan A. Fahmy
- Drug Radiation Research Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| | - Fatma Y. Abdou
- Drug Radiation Research Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| |
Collapse
|
3
|
Yang KH, Yen CY, Wang SC, Chang FR, Chang MY, Chan CK, Jeng JH, Tang JY, Chang HW. 6- n-Butoxy-10-nitro-12,13-dioxa-11-azatricyclo[7.3.1.0 2,7]trideca-2,4,6,10-tetraene Improves the X-ray Sensitivity on Inhibiting Proliferation and Promoting Oxidative Stress and Apoptosis of Oral Cancer Cells. Biomedicines 2024; 12:458. [PMID: 38398060 PMCID: PMC10887088 DOI: 10.3390/biomedicines12020458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 02/09/2024] [Accepted: 02/16/2024] [Indexed: 02/25/2024] Open
Abstract
This in vitro study examines the anti-oral cancer effects and mechanisms of a combined X-ray/SK2 treatment, i.e., X-ray and 6-n-butoxy-10-nitro-12,13-dioxa-11-azatricyclo[7.3.1.02,7]trideca-2,4,6,10-tetraene (SK2). ATP cell viability and flow cytometry-based cell cycle, apoptosis, oxidative stress, and DNA damage assessments were conducted. The X-ray/SK2 treatment exhibited lower viability in oral cancer (Ca9-22 and CAL 27) cells than in normal (Smulow-Glickman, S-G) cells, i.e., 32.0%, 46.1% vs. 59.0%, which showed more antiproliferative changes than with X-ray or SK2 treatment. Oral cancer cells under X-ray/SK2 treatment showed slight subG1 and G2/M increments and induced high annexin V-monitored apoptosis compared to X-ray or SK2 treatment. The X-ray/SK2 treatment showed higher caspase 3 and 8 levels for oral cancer cells than other treatments. X-ray/SK2 showed a higher caspase 9 level in CAL 27 cells than other treatments, while Ca9-22 cells showed similar levels under X-ray and/or SK2. The X-ray/SK2 treatment showed higher reactive oxygen species (ROS) generation and mitochondrial membrane potential (MMP) depletion than other treatments. Meanwhile, the mitochondrial superoxide (MitoSOX) and glutathione levels in X-ray/SK2 treatment did not exhibit the highest rank compared to others. Moreover, oral cancer cells had higher γH2AX and/or 8-hydroxy-2-deoxyguanosine levels from X-ray/SK2 treatment than others. All these measurements for X-ray/SK2 in oral cancer cells were higher than in normal cells and attenuated by N-acetylcysteine. In conclusion, X-ray/SK2 treatment showed ROS-dependent enhanced antiproliferative, apoptotic, and DNA damage effects in oral cancer cells with a lower cytotoxic influence on normal cells.
Collapse
Affiliation(s)
- Kun-Han Yang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (K.-H.Y.); (F.-R.C.)
| | - Ching-Yu Yen
- School of Dentistry, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Oral and Maxillofacial Surgery, Chi-Mei Medical Center, Tainan 71004, Taiwan
| | - Sheng-Chieh Wang
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Fang-Rong Chang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (K.-H.Y.); (F.-R.C.)
| | - Meng-Yang Chang
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Chieh-Kai Chan
- Department of Chemistry, University of Illinois Urbana, Champaign, IL 61820, USA;
| | - Jiiang-Huei Jeng
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Department of Dentistry, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Department of Dentistry, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Jen-Yang Tang
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Hsueh-Wei Chang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (K.-H.Y.); (F.-R.C.)
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| |
Collapse
|
4
|
Averbeck D. Low-Dose Non-Targeted Effects and Mitochondrial Control. Int J Mol Sci 2023; 24:11460. [PMID: 37511215 PMCID: PMC10380638 DOI: 10.3390/ijms241411460] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/26/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023] Open
Abstract
Non-targeted effects (NTE) have been generally regarded as a low-dose ionizing radiation (IR) phenomenon. Recently, regarding long distant abscopal effects have also been observed at high doses of IR) relevant to antitumor radiation therapy. IR is inducing NTE involving intracellular and extracellular signaling, which may lead to short-ranging bystander effects and distant long-ranging extracellular signaling abscopal effects. Internal and "spontaneous" cellular stress is mostly due to metabolic oxidative stress involving mitochondrial energy production (ATP) through oxidative phosphorylation and/or anaerobic pathways accompanied by the leakage of O2- and other radicals from mitochondria during normal or increased cellular energy requirements or to mitochondrial dysfunction. Among external stressors, ionizing radiation (IR) has been shown to very rapidly perturb mitochondrial functions, leading to increased energy supply demands and to ROS/NOS production. Depending on the dose, this affects all types of cell constituents, including DNA, RNA, amino acids, proteins, and membranes, perturbing normal inner cell organization and function, and forcing cells to reorganize the intracellular metabolism and the network of organelles. The reorganization implies intracellular cytoplasmic-nuclear shuttling of important proteins, activation of autophagy, and mitophagy, as well as induction of cell cycle arrest, DNA repair, apoptosis, and senescence. It also includes reprogramming of mitochondrial metabolism as well as genetic and epigenetic control of the expression of genes and proteins in order to ensure cell and tissue survival. At low doses of IR, directly irradiated cells may already exert non-targeted effects (NTE) involving the release of molecular mediators, such as radicals, cytokines, DNA fragments, small RNAs, and proteins (sometimes in the form of extracellular vehicles or exosomes), which can induce damage of unirradiated neighboring bystander or distant (abscopal) cells as well as immune responses. Such non-targeted effects (NTE) are contributing to low-dose phenomena, such as hormesis, adaptive responses, low-dose hypersensitivity, and genomic instability, and they are also promoting suppression and/or activation of immune cells. All of these are parts of the main defense systems of cells and tissues, including IR-induced innate and adaptive immune responses. The present review is focused on the prominent role of mitochondria in these processes, which are determinants of cell survival and anti-tumor RT.
Collapse
Affiliation(s)
- Dietrich Averbeck
- Laboratory of Cellular and Molecular Radiobiology, PRISME, UMR CNRS 5822/IN2P3, IP2I, Lyon-Sud Medical School, University Lyon 1, 69921 Oullins, France
| |
Collapse
|
5
|
Beddok A, Porte B, Cottu P, Fourquet A, Kirova Y. [Biological, preclinical and clinical aspects of the association between radiation therapy and CDK4/6 inhibitors]. Cancer Radiother 2023; 27:240-248. [PMID: 37080859 DOI: 10.1016/j.canrad.2022.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 10/06/2022] [Accepted: 11/30/2022] [Indexed: 04/22/2023]
Abstract
Several clinical studies have shown that CDK4/6 inhibitors (CDK4/6i) improve survival in patients with metastatic or locally advanced HR-positive, HER-2-negative breast cancer (BC). The aim of this review was to synthesize the biological, preclinical and clinical aspects of the treatment of BC with CDK4/6i, with a focus on the combination of CDK4/6i and radiotherapy. The DNA damage induced after exposure of cells to ionizing radiation activates control pathways that inhibit cell progression in the G1 and G2 phases and induce a transient delay in progression in the S phase. These checkpoints are in particular mediated by cyclin-dependent kinases (CDK) 4/6 activated by cyclin D1. Several preclinical studies have shown that CDK4/6i could be used as radiosensitizers in non-small cell lung cancer, medulloblastoma, brainstem glioma and breast cancer. CDK4/6 inhibition also protected against radiation-induced intestinal toxicities by inducing redistribution of quiescent intestinal progenitor cells, making them less radiosensitive. Clinical data on the combination of CDK inhibitors and radiotherapy for both locoregional and metastatic irradiation are based on retrospective data. Nevertheless, the most optimal therapeutic sequence would be radiotherapy followed by palbociclib. Pending prospective clinical trials, the concomitant combination of the two treatments should be done under close supervision.
Collapse
Affiliation(s)
- A Beddok
- Institut Curie, PSL Research University, University Paris Saclay, Inserm LITO, 91898 Orsay, France; Institut Curie, PSL Research University, Radiation Oncology Department, Proton Therapy Centre, Centre Universitaire, 91898 Orsay, France.
| | - B Porte
- Service d'oncologie médicale, GHU hôpital européen Georges-Pompidou, Paris, France
| | - P Cottu
- Département d'oncologie médicale, Institut Curie, Paris, France
| | - A Fourquet
- Institut Curie, PSL Research University, Radiation Oncology Department, Paris, France
| | - Y Kirova
- Institut Curie, PSL Research University, Radiation Oncology Department, Paris, France
| |
Collapse
|
6
|
Song Y, Cheng Y, Lan T, Bai Z, Liu Y, Bi Z, Alu A, Cheng D, Wei Y, Wei X. ERK inhibitor: A candidate enhancing therapeutic effects of conventional chemo-radiotherapy in esophageal squamous cell carcinoma. Cancer Lett 2023; 554:216012. [PMID: 36470544 DOI: 10.1016/j.canlet.2022.216012] [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: 06/15/2022] [Revised: 10/25/2022] [Accepted: 11/16/2022] [Indexed: 12/12/2022]
Abstract
For patients with esophageal squamous cell carcinoma (ESCC), standard therapeutic methods (cisplatin and radiotherapy) have been found to be ineffective and severely toxic. Targeted therapy emerges as a promising solution for this dilemma. It has been reported that targeted therapies are applied alone or in combination with standard conventional therapies for the treatment of a variety of cancers. To the best of our knowledge, in patients with ESCC, the combinational methods containing standard therapy and ERK-targeted therapy have yet to be explored. To analyze the prognostic role of p-ERK in ESCC patients, the Kaplan-Meier analysis and Cox regression model were used. To assess the effects of ERK-targeted therapy (GDC0994) on ESCC cells, in vitro studies including CCK-8 assay, colony formation assay, and scratch wound healing assay were conducted. In addition, the changes in cell cycle distribution and apoptosis were analyzed by flow cytometry. Besides, to assess the efficacy of different therapies in vivo, the xenograft tumor models were established by subcutaneously inoculating tumor cells into the flank/leg of mice. In patients with ESCC, a strong correlation between the high expression level of p-ERK and the poor prognosis (p < 0.01, Log-Rank test) has been identified. By analyzing the results from CCK-8 and scratch wound healing assays, we demonstrated that the ERK inhibitor repressed the viability and migration of ESCC cells. In addition, following the treatment of GDC0994, the volumes of xenograft tumors significantly decreased (p < 0.001, one-way ANOVA). Furthermore, blocking the mitogen-activated protein kinase (MAPK/ERK) pathway enhanced the therapeutic efficacy of both cisplatin and radiotherapy (p < 0.05). These findings imply the role of p-ERK in the prognosis of ESCC patients and the therapeutic value of ERK inhibitors in ESCC.
Collapse
Affiliation(s)
- Yanlin Song
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yuan Cheng
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Tianxia Lan
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ziyi Bai
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yu Liu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zhenfei Bi
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Aqu Alu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Diou Cheng
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
7
|
Shen W, Yu Q, Pu Y, Xing C. Upregulation of Long Noncoding RNA MALAT1 in Colorectal Cancer Promotes Radioresistance and Aggressive Malignance. Int J Gen Med 2022; 15:8365-8380. [PMID: 36465270 PMCID: PMC9717691 DOI: 10.2147/ijgm.s393270] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 11/18/2022] [Indexed: 09/07/2023] Open
Abstract
Background Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), a conserved transcript with 8000 nt, is highly associated with malignancy of numerous cancer types. However, the function of MALAT1 plays in regulating the response to radiotherapy in colorectal cancer (CRC) remains unclear. Thus, the object of this study is to investigate the functions of MALAT1 in CRC radioresistance. Methods First, the expression of MALAT1 in colon adenocarcinoma (COAD) was analyzed through the Cancer Genome Atlas (TCGA) database. Then, we detected the expression level of MALAT1 in tumor tissues and CRC cell lines and analyzed the relevance of MALAT1 and clinicopathological parameters. In the end, the effect of silencing MALAT1 on the radiosensitivity of CRC cells was investigated, and its potential mechanism was preliminarily illustrated. Results The analysis of TCGA data showed that MALAT1 was closely related to the type of tumor, and high expression of MALAT1 was remarkably relevant to poor outcome. MALAT1 was highly expressed in CRC tissues and cell lines and related to tumor stages. Knockdown of MALAT1 could significantly suppress colony survival, proliferation, and migration and increase apoptosis, G2/M phase arrest, and formation of gamma-H2AX foci in HCT116, whether in combination with X-rays or not. Moreover, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis demonstrated that the regulated proteins were principally enriched in the glycosaminoglycan degradation pathway after silencing MALAT1. Conclusion Our results implied that MALAT1 was highly expressed in CRC and associated with tumor stage and prognosis. Silencing MALAT1 can increase HCT116 cell radiosensitivity, which may be potentially influenced by glycosaminoglycan degradation pathway.
Collapse
Affiliation(s)
- Wenqi Shen
- Department of General Surgery, Second Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| | - Qifeng Yu
- Department of General Surgery, Second Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| | - Yuwei Pu
- Department of General Surgery, Second Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| | - Chungen Xing
- Department of General Surgery, Second Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| |
Collapse
|
8
|
Polgár S, Schofield PN, Madas BG. Datasets of in vitro clonogenic assays showing low dose hyper-radiosensitivity and induced radioresistance. Sci Data 2022; 9:555. [PMID: 36075916 PMCID: PMC9458642 DOI: 10.1038/s41597-022-01653-3] [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: 05/21/2022] [Accepted: 08/19/2022] [Indexed: 11/19/2022] Open
Abstract
Low dose hyper-radiosensitivity and induced radioresistance are primarily observed in surviving fractions of cell populations exposed to ionizing radiation, plotted as the function of absorbed dose. Several biophysical models have been developed to quantitatively describe these phenomena. However, there is a lack of raw, openly available experimental data to support the development and validation of quantitative models. The aim of this study was to set up a database of experimental data from the public literature. Using Google Scholar search, 46 publications with 101 datasets on the dose-dependence of surviving fractions, with clear evidence of low dose hyper-radiosensitivity, were identified. Surviving fractions, their uncertainties, and the corresponding absorbed doses were digitized from graphs of the publications. The characteristics of the cell line and the irradiation were also recorded, along with the parameters of the linear-quadratic model and/or the induced repair model if they were provided. The database is available in STOREDB, and can be used for meta-analysis, for comparison with new experiments, and for development and validation of biophysical models.
Collapse
Affiliation(s)
- Szabolcs Polgár
- Doctoral School of Physics, ELTE Eötvös Loránd University, Budapest, Hungary
- Environmental Physics Department, Centre for Energy Research, Budapest, Hungary
| | - Paul N Schofield
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Balázs G Madas
- Environmental Physics Department, Centre for Energy Research, Budapest, Hungary.
- Department of Physical Chemistry and Materials Science, Budapest University of Technology and Economics, Budapest, Hungary.
| |
Collapse
|
9
|
Teng C, Zhu Y, Li Y, Dai L, Pan Z, Wanggou S, Li X. Recurrence- and Malignant Progression-Associated Biomarkers in Low-Grade Gliomas and Their Roles in Immunotherapy. Front Immunol 2022; 13:899710. [PMID: 35677036 PMCID: PMC9168984 DOI: 10.3389/fimmu.2022.899710] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 04/12/2022] [Indexed: 12/15/2022] Open
Abstract
Despite a generally better prognosis than high-grade glioma (HGG), recurrence and malignant progression are the main causes for the poor prognosis and difficulties in the treatment of low-grade glioma (LGG). It is of great importance to learn about the risk factors and underlying mechanisms of LGG recurrence and progression. In this study, the transcriptome characteristics of four groups, namely, normal brain tissue and recurrent LGG (rLGG), normal brain tissue and secondary glioblastoma (sGBM), primary LGG (pLGG) and rLGG, and pLGG and sGBM, were compared using Chinese Glioma Genome Atlas (CGGA) and Genotype-Tissue Expression Project (GTEx) databases. In this study, 296 downregulated and 396 upregulated differentially expressed genes (DEGs) with high consensus were screened out. Univariate Cox regression analysis of data from The Cancer Genome Atlas (TCGA) yielded 86 prognostically relevant DEGs; a prognostic prediction model based on five key genes (HOXA1, KIF18A, FAM133A, HGF, and MN1) was established using the least absolute shrinkage and selection operator (LASSO) regression dimensionality reduction and multivariate Cox regression analysis. LGG was divided into high- and low-risk groups using this prediction model. Gene Set Enrichment Analysis (GSEA) revealed that signaling pathway differences in the high- and low-risk groups were mainly seen in tumor immune regulation and DNA damage-related cell cycle checkpoints. Furthermore, the infiltration of immune cells in the high- and low-risk groups was analyzed, which indicated a stronger infiltration of immune cells in the high-risk group than that in the low-risk group, suggesting that an immune microenvironment more conducive to tumor growth emerged due to the interaction between tumor and immune cells. The tumor mutational burden and tumor methylation burden in the high- and low-risk groups were also analyzed, which indicated higher gene mutation burden and lower DNA methylation level in the high-risk group, suggesting that with the accumulation of genomic mutations and epigenetic changes, tumor cells continued to evolve and led to the progression of LGG to HGG. Finally, the value of potential therapeutic targets for the five key genes was analyzed, and findings demonstrated that KIF18A was the gene most likely to be a potential therapeutic target. In conclusion, the prediction model based on these five key genes can better identify the high- and low-risk groups of LGG and lay a solid foundation for evaluating the risk of LGG recurrence and malignant progression.
Collapse
Affiliation(s)
- Chubei Teng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China.,Department of Neurosurgery, The First Affiliated Hospital, University of South China, Hengyang, China
| | - Yongwei Zhu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Yueshuo Li
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Luohuan Dai
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Zhouyang Pan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Siyi Wanggou
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
10
|
Jiménez-Cortegana C, Klapp V, Bloy N, Galassi C, Sato A, Yamazaki T, Buqué A, Galluzzi L, Petroni G. Cytofluorometric assessment of cell cycle progression in irradiated cells. Methods Cell Biol 2022; 172:1-16. [DOI: 10.1016/bs.mcb.2021.12.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
11
|
Averbeck D, Rodriguez-Lafrasse C. Role of Mitochondria in Radiation Responses: Epigenetic, Metabolic, and Signaling Impacts. Int J Mol Sci 2021; 22:ijms222011047. [PMID: 34681703 PMCID: PMC8541263 DOI: 10.3390/ijms222011047] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/24/2021] [Accepted: 10/08/2021] [Indexed: 12/15/2022] Open
Abstract
Until recently, radiation effects have been considered to be mainly due to nuclear DNA damage and their management by repair mechanisms. However, molecular biology studies reveal that the outcomes of exposures to ionizing radiation (IR) highly depend on activation and regulation through other molecular components of organelles that determine cell survival and proliferation capacities. As typical epigenetic-regulated organelles and central power stations of cells, mitochondria play an important pivotal role in those responses. They direct cellular metabolism, energy supply and homeostasis as well as radiation-induced signaling, cell death, and immunological responses. This review is focused on how energy, dose and quality of IR affect mitochondria-dependent epigenetic and functional control at the cellular and tissue level. Low-dose radiation effects on mitochondria appear to be associated with epigenetic and non-targeted effects involved in genomic instability and adaptive responses, whereas high-dose radiation effects (>1 Gy) concern therapeutic effects of radiation and long-term outcomes involving mitochondria-mediated innate and adaptive immune responses. Both effects depend on radiation quality. For example, the increased efficacy of high linear energy transfer particle radiotherapy, e.g., C-ion radiotherapy, relies on the reduction of anastasis, enhanced mitochondria-mediated apoptosis and immunogenic (antitumor) responses.
Collapse
Affiliation(s)
- Dietrich Averbeck
- Laboratory of Cellular and Molecular Radiobiology, PRISME, UMR CNRS 5822/IN2P3, IP2I, Lyon-Sud Medical School, University Lyon 1, 69921 Oullins, France;
- Correspondence:
| | - Claire Rodriguez-Lafrasse
- Laboratory of Cellular and Molecular Radiobiology, PRISME, UMR CNRS 5822/IN2P3, IP2I, Lyon-Sud Medical School, University Lyon 1, 69921 Oullins, France;
- Department of Biochemistry and Molecular Biology, Lyon-Sud Hospital, Hospices Civils de Lyon, 69310 Pierre-Bénite, France
| |
Collapse
|
12
|
Tremi I, Spyratou E, Souli M, Efstathopoulos EP, Makropoulou M, Georgakilas AG, Sihver L. Requirements for Designing an Effective Metallic Nanoparticle (NP)-Boosted Radiation Therapy (RT). Cancers (Basel) 2021; 13:cancers13133185. [PMID: 34202342 PMCID: PMC8269428 DOI: 10.3390/cancers13133185] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Recent advances in nanotechnology gave rise to trials with various types of metallic nanoparticles (NPs) to enhance the radiosensitization of cancer cells while reducing or maintaining the normal tissue complication probability during radiation therapy. This work reviews the physical and chemical mechanisms leading to the enhancement of ionizing radiation’s detrimental effects on cells and tissues, as well as the plethora of experimental procedures to study these effects of the so-called “NPs’ radiosensitization”. The paper presents the need to a better understanding of all the phases of actions before applying metallic-based NPs in clinical practice to improve the effect of IR therapy. More physical and biological experiments especially in vivo must be performed and simulation Monte Carlo or mathematical codes based on more accurate models for all phases must be developed. Abstract Many different tumor-targeted strategies are under development worldwide to limit the side effects and improve the effectiveness of cancer therapies. One promising method is to enhance the radiosensitization of the cancer cells while reducing or maintaining the normal tissue complication probability during radiation therapy using metallic nanoparticles (NPs). Radiotherapy with MV photons is more commonly available and applied in cancer clinics than high LET particle radiotherapy, so the addition of high-Z NPs has the potential to further increase the efficacy of photon radiotherapy in terms of NP radiosensitization. Generally, when using X-rays, mainly the inner electron shells are ionized, which creates cascades of both low and high energy Auger electrons. When using high LET particles, mainly the outer shells are ionized, which give electrons with lower energies than when using X-rays. The amount of the produced low energy electrons is higher when exposing NPs to heavy charged particles than when exposing them to X-rays. Since ions traverse the material along tracks, and therefore give rise to a much more inhomogeneous dose distributions than X-rays, there might be a need to introduce a higher number of NPs when using ions compared to when using X-rays to create enough primary and secondary electrons to get the desired dose escalations. This raises the questions of toxicity. This paper provides a review of the fundamental processes controlling the outcome of metallic NP-boosted photon beam and ion beam radiation therapy and presents some experimental procedures to study the biological effects of NPs’ radiosensitization. The overview shows the need for more systematic studies of the behavior of NPs when exposed to different kinds of ionizing radiation before applying metallic-based NPs in clinical practice to improve the effect of IR therapy.
Collapse
Affiliation(s)
- Ioanna Tremi
- DNA Damage Laboratory, Department of Physics, School of Applied Mathematical and Physical Sciences, Zografou Campus, National Technical University of Athens (NTUA), 15780 Athens, Greece; (I.T.); (M.S.); (M.M.)
| | - Ellas Spyratou
- 2nd Department of Radiology, Medical School, National and Kapodistrian University of Athens, 11517 Athens, Greece; (E.S.); (E.P.E.)
| | - Maria Souli
- DNA Damage Laboratory, Department of Physics, School of Applied Mathematical and Physical Sciences, Zografou Campus, National Technical University of Athens (NTUA), 15780 Athens, Greece; (I.T.); (M.S.); (M.M.)
- Atominstitut, Technische Universität Wien, Stadionallee 2, 1020 Vienna, Austria
| | - Efstathios P. Efstathopoulos
- 2nd Department of Radiology, Medical School, National and Kapodistrian University of Athens, 11517 Athens, Greece; (E.S.); (E.P.E.)
| | - Mersini Makropoulou
- DNA Damage Laboratory, Department of Physics, School of Applied Mathematical and Physical Sciences, Zografou Campus, National Technical University of Athens (NTUA), 15780 Athens, Greece; (I.T.); (M.S.); (M.M.)
| | - Alexandros G. Georgakilas
- DNA Damage Laboratory, Department of Physics, School of Applied Mathematical and Physical Sciences, Zografou Campus, National Technical University of Athens (NTUA), 15780 Athens, Greece; (I.T.); (M.S.); (M.M.)
- Correspondence: (A.G.G.); (L.S.)
| | - Lembit Sihver
- Atominstitut, Technische Universität Wien, Stadionallee 2, 1020 Vienna, Austria
- Department of Physics, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden
- Correspondence: (A.G.G.); (L.S.)
| |
Collapse
|
13
|
Abd-ElRaouf A, Nada AS, Mohammed NEDA, Amer HA, Abd-ElRahman SS, Abdelsalam RM, Salem HA. Low dose gamma irradiation attenuates cyclophosphamide-induced cardiotoxicity in rats: role of NF-κB signaling pathway. Int J Radiat Biol 2021; 97:632-641. [PMID: 33635746 DOI: 10.1080/09553002.2021.1893856] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/24/2021] [Accepted: 02/14/2021] [Indexed: 01/03/2023]
Abstract
PURPOSE Cyclophosphamide (Cyp) is one of the most commonly used, wide spectrum chemotherapeutic agents. Cyp has multi-organ toxicities that are dose limiting, thus it's mostly used in chemotherapeutic combinations. Radiation is well known as a hazardous sort of energy, recent studies are interested in studying the beneficial therapeutic effects of low-dose gamma radiation. This study examined the protective effect of two different doses/dose-rates of irradiation either alone or combined with telmisartan against Cyp-induced cardiotoxicity. MATERIALS AND METHODS Rats were divided into seven groups; (1): Control, (2): Cyp, (3-4): 0.05 Gy low dose rate (LDR) irradiation, 0.25 Gy high dose rate (HDR) irradiation, respectively, prior to Cyp dose, (5-7): telmisartan either alone or with 0.05 Gy LDR-irradiation or 0.25 Gy HDR-irradiation, respectively, prior to Cyp dose. The current investigation studied the effect of Cyp alone or combined with different treatment regimens on serum cTn-I and LDH, nuclear factor-κB (NF-κB) pathway (p65/IκB/IKK-α/IKK-ß) in the myocardium. Pro-inflammatory cytokines IL-1ß, IL-6 and TNF-α were assessed in addition to histopathological examination of the heart. RESULTS Low-dose irradiation attenuated cardiac enzymes, pro-inflammatory cytokines, NF-κB content, and histology, in both low and HDRs. Furthermore, the combination of low-dose irradiation with telmisartan (an angiotensin-II receptor type-1 blocker and a known cardio-protective drug) offered the best histological results. CONCLUSIONS Low-dose irradiation-induced amelioration is partially but not completely through canonical activation of NF-κB, and may have another atypical pathway. While telmisartan probably ameliorates NF-κB totally through canonical pathway.
Collapse
Affiliation(s)
- Amira Abd-ElRaouf
- National Centre of Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Ahmed S Nada
- National Centre of Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Nour El-Din A Mohammed
- National Centre of Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Hany A Amer
- National Centre for Nuclear Safety and Radiation Control, Atomic Energy Authority, Cairo, Egypt
| | - Sahar S Abd-ElRahman
- Department of Pathology, Faculty of Veterinary, Cairo University, Giza Square, Egypt
| | - Rania M Abdelsalam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Hesham A Salem
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
14
|
Fernando C, Byun SH, Shi X, Seymour CB, Mothersill CE. Isolation of the effects of alpha-related components from total effects of radium at low doses. Int J Radiat Biol 2021; 98:1168-1175. [PMID: 33332185 DOI: 10.1080/09553002.2020.1866226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
PURPOSE Radium is the most common source of alpha radiation exposure to humans and non-human species in the environment but the dosimetry is complicated by the decay chain which involves gamma exposure due to radon daughters. This paper seeks to determine the separate contributions of alpha and gamma doses to the total dose and total direct and non-targeted effect in a fish and a human cell line. MATERIALS AND METHODS This study aimed to isolate the effect of alpha particles following exposure to low doses of radium in cells, and their progeny which received no further exposure. This was initially done by comparing the survival values of a human keratinocyte cell line (HaCaT) and an embryonic Chinook salmon cell line (CHSE-214) exposed to gamma radiation, from survival of the same cell lines exposed to mixed alpha and gamma radiation through exposure to Ra-226 and its decay products. A Monte Carlo simulation was later performed to determine the contributions of radium decay products including radon daughters. RESULTS The human cell line showed increased radioresistance when exposed to low doses of alpha particles. In contrast the fish cell line, which demonstrated radioresistance to low dose gamma radiation, showed increased lethality when exposed to low doses of alpha particles. Significant and complex levels of non-targeted effects were induced in progeny of irradiated cells. The simulation showed that gamma and beta decay products did not contribute significant dose and the highest beta dose was below the threshold for inducing non-targeted effects. CONCLUSIONS The results confirm the need to consider the dose-response relationship when developing radiation weighting factors for low dose exposures, as well as the need to be aware of possible cell line and species differences.
Collapse
Affiliation(s)
- Chandula Fernando
- Radiation Sciences Graduate Program, McMaster University, Hamilton, Canada
| | - Soo Hyun Byun
- Department of Physics and Astronomy, McMaster University, Hamilton, Canada
| | - Xiaopei Shi
- Department of Biology, McMaster University, Hamilton, Canada
| | - Colin B Seymour
- Department of Biology, McMaster University, Hamilton, Canada
| | | |
Collapse
|
15
|
Asadian S, Mirzaei H, Kalantari BA, Davarpanah MR, Mohamadi M, Shpichka A, Nasehi L, Es HA, Timashev P, Najimi M, Gheibi N, Hassan M, Vosough M. β-radiating radionuclides in cancer treatment, novel insight into promising approach. Pharmacol Res 2020; 160:105070. [PMID: 32659429 DOI: 10.1016/j.phrs.2020.105070] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 12/17/2022]
Abstract
Targeted radionuclide therapy, known as molecular radiotherapy is a novel therapeutic module in cancer medicine. β-radiating radionuclides have definite impact on target cells via interference in cell cycle and particular signalings that can lead to tumor regression with minimal off-target effects on the surrounding tissues. Radionuclides play a remarkable role not only in apoptosis induction and cell cycle arrest, but also in the amelioration of other characteristics of cancer cells. Recently, application of novel β-radiating radionuclides in cancer therapy has been emerged as a promising therapeutic modality. Several investigations are ongoing to understand the underlying molecular mechanisms of β-radiating elements in cancer medicine. Based on the radiation dose, exposure time and type of the β-radiating element, different results could be achieved in cancer cells. It has been shown that β-radiating radioisotopes block cancer cell proliferation by inducing apoptosis and cell cycle arrest. However, physical characteristics of the β-radiating element (half-life, tissue penetration range, and maximum energy) and treatment protocol determine whether tumor cells undergo cell cycle arrest, apoptosis or both and to which extent. In this review, we highlighted novel therapeutic effects of β-radiating radionuclides on cancer cells, particularly apoptosis induction and cell cycle arrest.
Collapse
Affiliation(s)
- Samieh Asadian
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran; Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | | | | | - Morteza Mohamadi
- Department of Physical Chemistry, Faculty of Science, University of Tehran, Tehran, Iran
| | - Anastasia Shpichka
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia; Chemistry Department, Lomonosov Moscow State University, Moscow, Russia
| | - Leila Nasehi
- Department of Medical Laboratory Sciences, Zanjan University of Medical Sciences, Zanjan, Iran
| | | | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia; Chemistry Department, Lomonosov Moscow State University, Moscow, Russia; Department of Polymers and Composites, NN Semenov Institute of Chemical Physics, Moscow, Russia.
| | - Mustapha Najimi
- Laboratory of Pediatric Hepatology and Cell Therapy, Institute of Experimental and Clinical Research, Université Catholique de Louvain, B-1200 Brussels, Belgium
| | - Nematollah Gheibi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran.
| | - Moustapha Hassan
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
16
|
The low dose effects of human mammary epithelial cells induced by internal exposure to low radioactive tritiated water. Toxicol In Vitro 2019; 61:104608. [PMID: 31348984 DOI: 10.1016/j.tiv.2019.104608] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 06/16/2019] [Accepted: 07/22/2019] [Indexed: 02/06/2023]
Abstract
Tritium is an important radioactive waste which needs to be monitored for radiation protection. Due to long biological half-life of organically bound tritium (OBT), the adverse consequence caused by chronic exposure of tritiated water (HTO) attracts concern. In this study, fibroblast cells were exposed to 2 × 106 Bq/ml HTO to investigate the cellular behaviors. The dose relationship of survival fraction and γH2AX foci was a "U-shaped" curve. And the results of γH2AX intensity produced by ICCM, which was obtained from different doses, demonstrated bystander signal accounted for the protective effects induced by intermediate dose of 100 mGy. The comparison of temporal kinetics and spatial dynamics of DNA repair between tritium β-rays and γ-rays showed longer time was need for the dephosphorylation of H2AX protein after HTO exposure. It indicated complex cluster DSBs induced by tritium β-rays at the low dose impaired efficient recovery of DNA damage, which bear responsibility for the persistence of residual foci after low dose expsoure. It suggests after exposed to low dose radiation cells prefer to eliminate damage population to avoid DNA damage increasing the mutation potential.
Collapse
|
17
|
Epsilon-Globin HBE1 Enhances Radiotherapy Resistance by Down-Regulating BCL11A in Colorectal Cancer Cells. Cancers (Basel) 2019; 11:cancers11040498. [PMID: 30965648 PMCID: PMC6521047 DOI: 10.3390/cancers11040498] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/27/2019] [Accepted: 04/02/2019] [Indexed: 12/28/2022] Open
Abstract
Resistance to radiotherapy is considered an important obstacle in the treatment of colorectal cancer. However, the mechanisms that enable tumor cells to tolerate the effects of radiation remain unclear. Moreover, radiotherapy causes accumulated mutations in transcription factors, which can lead to changes in gene expression and radiosensitivity. This phenomenon reduces the effectiveness of radiation therapy towards cancer cells. In the present study, radiation-resistant (RR) cancer cells were established by sequential radiation exposure, and hemoglobin subunit epsilon 1 (HBE1) was identified as a candidate radiation resistance-associated protein based on RNA-sequencing analysis. Then, compared to radiosensitive (RS) cell lines, the overexpression of HBE1 in RR cell lines was used to measure various forms of radiation-induced cellular damage. Consequently, HBE1-overexpressing cell lines were found to exhibit decreased radiation-induced intracellular reactive oxygen species (ROS) production and cell mortality. Conversely, HBE1 deficiency in RR cell lines increased intracellular ROS production, G2/M arrest, and apoptosis, and decreased clonogenic survival rate. These effects were reversed by the ROS scavenger N-acetyl cysteine. Moreover, HBE1 overexpression was found to attenuate radiation-induced endoplasmic reticulum stress and apoptosis via an inositol-requiring enzyme 1(IRE1)-Jun amino-terminal kinase (JNK) signaling pathway. In addition, increased HBE1 expression induced by γ-irradiation in RS cells attenuated expression of the transcriptional regulator BCL11A, whereas its depletion in RR cells increased BCL11A expression. Collectively, these observations indicate that the expression of HBE1 during radiotherapy might potentiate the survival of radiation-exposed colorectal cancer cells.
Collapse
|
18
|
Tharmalingam S, Sreetharan S, Brooks AL, Boreham DR. Re-evaluation of the linear no-threshold (LNT) model using new paradigms and modern molecular studies. Chem Biol Interact 2019; 301:54-67. [PMID: 30763548 DOI: 10.1016/j.cbi.2018.11.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 11/13/2018] [Accepted: 11/22/2018] [Indexed: 02/06/2023]
Abstract
The linear no-threshold (LNT) model is currently used to estimate low dose radiation (LDR) induced health risks. This model lacks safety thresholds and postulates that health risks caused by ionizing radiation is directly proportional to dose. Therefore even the smallest radiation dose has the potential to cause an increase in cancer risk. Advances in LDR biology and cell molecular techniques demonstrate that the LNT model does not appropriately reflect the biology or the health effects at the low dose range. The main pitfall of the LNT model is due to the extrapolation of mutation and DNA damage studies that were conducted at high radiation doses delivered at a high dose-rate. These studies formed the basis of several outdated paradigms that are either incorrect or do not hold for LDR doses. Thus, the goal of this review is to summarize the modern cellular and molecular literature in LDR biology and provide new paradigms that better represent the biological effects in the low dose range. We demonstrate that LDR activates a variety of cellular defense mechanisms including DNA repair systems, programmed cell death (apoptosis), cell cycle arrest, senescence, adaptive memory, bystander effects, epigenetics, immune stimulation, and tumor suppression. The evidence presented in this review reveals that there are minimal health risks (cancer) with LDR exposure, and that a dose higher than some threshold value is necessary to achieve the harmful effects classically observed with high doses of radiation. Knowledge gained from this review can help the radiation protection community in making informed decisions regarding radiation policy and limits.
Collapse
Affiliation(s)
- Sujeenthar Tharmalingam
- Northern Ontario School of Medicine, Laurentian University, 935 Ramsey Lake Rd, Sudbury, ON, P3E 2C6, Canada.
| | - Shayenthiran Sreetharan
- Department of Medical Physics and Applied Radiation Sciences, McMaster University, 1280 Main Street W, Hamilton ON, L8S 4K1, Canada
| | - Antone L Brooks
- Environmental Science, Washington State University, Richland, WA, USA
| | - Douglas R Boreham
- Northern Ontario School of Medicine, Laurentian University, 935 Ramsey Lake Rd, Sudbury, ON, P3E 2C6, Canada; Bruce Power, Tiverton, ON(3), UK.
| |
Collapse
|
19
|
Branter J, Basu S, Smith S. Tumour treating fields in a combinational therapeutic approach. Oncotarget 2018; 9:36631-36644. [PMID: 30564303 PMCID: PMC6290966 DOI: 10.18632/oncotarget.26344] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 10/24/2018] [Indexed: 12/15/2022] Open
Abstract
The standard of care for patients with newly diagnosed Glioblastoma multiforme (GBM) has remained unchanged since 2005, with patients undergoing maximal surgical resection, followed by radiotherapy plus concomitant and maintenance Temozolomide. More recently, Tumour treating fields (TTFields) therapy has become FDA approved for adult recurrent and adult newly-diagnosed GBM following the EF-11 and EF-14 trials, respectively. TTFields is a non-invasive anticancer treatment which utilizes medium frequency alternating electric fields to target actively dividing cancerous cells. TTFields selectively targets cells within mitosis through interacting with key mitotic proteins to cause mitotic arrest and cell death. TTFields therapy presents itself as a candidate for the combinational therapy route due to the lack of overlapping toxicities associated with electric fields. Here we review current literature pertaining to TTFields in combination with alkylating agents, radiation, anti-angiogenics, mitotic inhibitors, immunotherapies, and also with novel agents. This review highlights the observed synergistic and additive effects of combining TTFields with various other therapies, as well highlighting the strategies relating to combinations with electric fields.
Collapse
Affiliation(s)
- Joshua Branter
- Children's Brain Tumour Research Centre, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | - Surajit Basu
- Queen's Medical Centre, Department of Neurosurgery, Nottingham, UK
| | - Stuart Smith
- Children's Brain Tumour Research Centre, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| |
Collapse
|
20
|
Fei Z, Gu W, Xie R, Su H, Jiang Y. Artesunate enhances radiosensitivity of esophageal cancer cells by inhibiting the repair of DNA damage. J Pharmacol Sci 2018; 138:131-137. [PMID: 30337244 DOI: 10.1016/j.jphs.2018.09.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 08/16/2018] [Accepted: 09/20/2018] [Indexed: 01/24/2023] Open
Abstract
Radiotherapy plays an important therapeutic role in esophageal cancer (EC). However, acquired radioresistance impairs the efficacy of radiotherapy, often leading to treatment failure. Therefore, it is important to develop novel radiosensitizers to enhance the clinical treatment of EC. The purpose of this study was to investigate the role of artesunate (ART) on radiosensitivity of human EC cell line TE-1. We found that ART inhibited the proliferation of EC cells and enhanced the radiosensitivity of TE-1 cells (SER = 1.24). In vivo tumor growth of xenografts was inhibited markedly by irradiation (IR) combined with ART, with a tumor inhibition rate of 53.76% in IR + ART group vs. 41.13% in IR-alone group. Pretreatment with ART significantly prompted cell apoptosis and reversed the IR-induced G2/M arrest. ART treatment could aggravate DNA damage of EC cells and prolong the formation of γ-H2AX foci induced by IR. ART up-regulated P21 and down-regulated the expression of cyclin D1, RAD51, RAD54, Ku70 and Ku86 protein of irradiated TE-1 cells. These findings support that ART induce radiosensitivity of TE-1 cells in vitro and in vivo, and may prove to be a promising radiosensitizer for EC treatment.
Collapse
Affiliation(s)
- Zhenhua Fei
- Department of Oncology, The 1st Affiliated Hospital of Wenzhou Medical University, No.2 Fuxue Lane, Wenzhou, Zhejiang, 325000, PR China
| | - Wenyue Gu
- Department of Pathology, Yancheng Hospital Affiliated Southeast University, No.2 Xingdu Road, Yancheng, Jiangsu, 224000, PR China
| | - Raoying Xie
- Department of Oncology, The 1st Affiliated Hospital of Wenzhou Medical University, No.2 Fuxue Lane, Wenzhou, Zhejiang, 325000, PR China
| | - Huafang Su
- Department of Oncology, The 1st Affiliated Hospital of Wenzhou Medical University, No.2 Fuxue Lane, Wenzhou, Zhejiang, 325000, PR China
| | - Yiyan Jiang
- Department of Tumor Rehabilitation, The 1st Affiliated Hospital of Wenzhou Medical University, No.2 Fuxue Lane, Wenzhou, Zhejiang Province, 325000, China.
| |
Collapse
|
21
|
Differential miRNA expression profiling reveals miR-205-3p to be a potential radiosensitizer for low- dose ionizing radiation in DLD-1 cells. Oncotarget 2018; 9:26387-26405. [PMID: 29899866 PMCID: PMC5995186 DOI: 10.18632/oncotarget.25405] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 04/28/2018] [Indexed: 12/12/2022] Open
Abstract
Enhanced radiosensitivity at low doses of ionizing radiation (IR) (0.2 to 0.6 Gy) has been reported in several cell lines. This phenomenon, known as low doses hyper-radiosensitivity (LDHRS), appears as an opportunity to decrease toxicity of radiotherapy and to enhance the effects of chemotherapy. However, the effect of low single doses IR on cell death is subtle and the mechanism underlying LDHRS has not been clearly explained, limiting the utility of LDHRS for clinical applications. To understand the mechanisms responsible for cell death induced by low-dose IR, LDHRS was evaluated in DLD-1 human colorectal cancer cells and the expression of 80 microRNAs (miRNAs) was assessed by qPCR array. Our results show that DLD-1 cells display an early DNA damage response and apoptotic cell death when exposed to 0.6 Gy. miRNA expression profiling identified 3 over-expressed (miR-205-3p, miR-1 and miR-133b) and 2 down-regulated miRNAs (miR-122-5p, and miR-134-5p) upon exposure to 0.6 Gy. This miRNA profile differed from the one in cells exposed to high-dose IR (12 Gy), supporting a distinct low-dose radiation-induced cell death mechanism. Expression of a mimetic miR-205-3p, the most overexpressed miRNA in cells exposed to 0.6 Gy, induced apoptotic cell death and, more importantly, increased LDHRS in DLD-1 cells. Thus, we propose miR-205-3p as a potential radiosensitizer to low-dose IR.
Collapse
|
22
|
Olobatuyi O, de Vries G, Hillen T. Effects of G2-checkpoint dynamics on low-dose hyper-radiosensitivity. J Math Biol 2018; 77:1969-1997. [PMID: 29679122 DOI: 10.1007/s00285-018-1236-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 02/17/2018] [Indexed: 02/03/2023]
Abstract
In experimental studies, it has been found that certain cell lines are more sensitive to low-dose radiation than would be expected from the classical Linear-Quadratic model (LQ model). In fact, it is frequently observed that cells incur more damage at low dose (say 0.3 Gy) than at higher dose (say 1 Gy). This effect has been termed hyper-radiosensitivity (HRS). The effect depends on the type of cells and on their phase in the cell cycle when radiation is applied. Experiments have shown that the G2-checkpoint plays an important role in the HRS effects. Here we design and analyze a differential equation model for the cell cycle that includes G2-checkpoint dynamics and radiation treatment. We fit the model to surviving fraction data for different cell lines including glioma cells, prostate cancer cells, as well as to cell populations that are enriched in certain phases of the cell cycle. The HRS effect is measured in the literature through [Formula: see text], the ratio of slope [Formula: see text] of the surviving fraction curve at zero dose to slope [Formula: see text] of the corresponding LQ model. We derive an explicit formula for this ratio and we show that it corresponds very closely to experimental observations. Finally, we identify the dependence of this ratio on the surviving fraction at 2 Gy. It was speculated in the literature that such dependence exists. Our theoretical analysis will help to more systematically identify the HRS in cell lines, and opens doors to analyze its use in cancer treatment.
Collapse
Affiliation(s)
- Oluwole Olobatuyi
- Collaborative Mathematical Biology Group (formerly Center for Mathematical Biology), Department of Mathematical and Statistical Sciences, University of Alberta, Edmonton, AB, T6G 2G1, Canada.
| | - Gerda de Vries
- Collaborative Mathematical Biology Group (formerly Center for Mathematical Biology), Department of Mathematical and Statistical Sciences, University of Alberta, Edmonton, AB, T6G 2G1, Canada
| | - Thomas Hillen
- Collaborative Mathematical Biology Group (formerly Center for Mathematical Biology), Department of Mathematical and Statistical Sciences, University of Alberta, Edmonton, AB, T6G 2G1, Canada
| |
Collapse
|
23
|
Zhang X, Peng L, Liu A, Ji J, Zhao L, Zhai G. The enhanced effect of tetrahydrocurcumin on radiosensitivity of glioma cells. J Pharm Pharmacol 2018; 70:749-759. [PMID: 29492979 DOI: 10.1111/jphp.12891] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 01/13/2018] [Indexed: 12/22/2022]
Abstract
Abstract
Objectives
To evaluate the effects of tetrahydrocurcumin (THC) on the radiosensitivity of glioma cells and the possible molecular mechanism.
Methods
MTT assay, colony forming and wound healing assays were performed to detect the proliferation, radiosensitivity and migration of cells with various treatments. Cell apoptosis, cell cycle and GHS level were determined for exploring potent sensitization mechanism of THC. Meanwhile, protein expressions of cyclin D1 and PCNA were also measured. Furthermore, both orthotopic C6 mouse models and C6 subcutaneously grafted mouse models were established to test the tumour inhibitory effects of combined treatment in vivo.
Key findings
Cells treated with combined THC and radiation demonstrated lower cell viability and higher apoptosis rate as compared to radiation group. Moreover, the intracellular GSH was also decreased in the THC co-treated C6 cells. More importantly, combinatorial treatment group significantly induced G0/G1 cell cycle arrest and a decrease in the S phase cell through the down-regulation of cyclin D1 and PCNA. The in-vivo therapeutic efficacy assay indicated that the growth of tumour was greatly inhibited in combinatorial group.
Conclusions
Tetrahydrocurcumin can synergistically enhance the radiosensitivity of glioma cells by inhibiting the expressions of cyclin D1 and PCNA.
Collapse
Affiliation(s)
- Xingzhen Zhang
- Department of Pharmaceutics, College of Pharmacy, Shandong University, Shandong University, Jinan, China
| | - Lei Peng
- Department of Hepatology and Gastroenterology, Qilu Hospital of Shandong University, Jinan, China
| | - Anchang Liu
- Department of Pharmacy, Qilu Hospital of Shandong University, Jinan, China
| | - Jianbo Ji
- Department of Pharmaceutics, College of Pharmacy, Shandong University, Shandong University, Jinan, China
| | - Lixia Zhao
- Department of Pharmacy, Qilu Hospital of Shandong University, Jinan, China
| | - Guangxi Zhai
- Department of Pharmaceutics, College of Pharmacy, Shandong University, Shandong University, Jinan, China
| |
Collapse
|
24
|
Słonina D, Kowalczyk A, Janecka-Widła A, Kabat D, Szatkowski W, Biesaga B. Low-Dose Hypersensitive Response for Residual pATM and γH2AX Foci in Normal Fibroblasts of Cancer Patients. Int J Radiat Oncol Biol Phys 2017; 100:756-766. [PMID: 29248168 DOI: 10.1016/j.ijrobp.2017.10.054] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 10/10/2017] [Accepted: 10/30/2017] [Indexed: 01/04/2023]
Abstract
PURPOSE To define the dose-response relationship for initial and residual pATM and γH2AX foci and temporal response of pATM foci in fibroblasts of 4 hyper-radiosensitivity (HRS)-positive cancer patients and 8 HRS-negative cancer patients and answer the question regarding the role of DNA double-strand break (DSB) recognition and repair in the mechanism of HRS. METHODS AND MATERIALS The cells were irradiated with single doses (0.1-4 Gy) of 6-MV X rays. The number of initial and residual pATM and γH2AX foci was assessed 1 hour and 24 hours after irradiation, respectively. Kinetics of DSB recognition and repair was estimated by pATM foci assay after irradiation with 0.2 and 2 Gy. RESULTS Hyper-radiosensitivity response (confirmed by the induced-repair model) was clearly evident for residual pATM and γH2AX foci in fibroblasts of HRS-positive patients but not in fibroblasts of HRS-negative patients. Significantly less DSB was recognized by pATM early (10-30 minutes) after irradiation with 0.2 Gy in HRS-positive compared with HRS-negative fibroblasts. CONCLUSIONS The present results provide evidence for the role of DSB recognition by pATM and repair in the mechanism of HRS and seem to support the idea of nucleo-shuttling of the pATM protein to be involved in HRS response.
Collapse
Affiliation(s)
- Dorota Słonina
- Department of Applied Radiobiology, Maria Sklodowska-Curie Institute - Oncology Center, Cracow Branch, Cracow, Poland.
| | - Aleksandra Kowalczyk
- Department of Applied Radiobiology, Maria Sklodowska-Curie Institute - Oncology Center, Cracow Branch, Cracow, Poland
| | - Anna Janecka-Widła
- Department of Applied Radiobiology, Maria Sklodowska-Curie Institute - Oncology Center, Cracow Branch, Cracow, Poland
| | - Damian Kabat
- Department of Medical Physics, Maria Sklodowska-Curie Institute - Oncology Center, Cracow Branch, Cracow, Poland
| | - Wiktor Szatkowski
- Department of Gynecologic Oncology, Maria Sklodowska-Curie Institute - Oncology Center, Cracow Branch, Cracow, Poland
| | - Beata Biesaga
- Department of Applied Radiobiology, Maria Sklodowska-Curie Institute - Oncology Center, Cracow Branch, Cracow, Poland
| |
Collapse
|
25
|
Piotrowski I, Kulcenty K, Suchorska WM, Skrobała A, Skórska M, Kruszyna-Mochalska M, Kowalik A, Jackowiak W, Malicki J. Carcinogenesis Induced by Low-dose Radiation. Radiol Oncol 2017; 51:369-377. [PMID: 29333114 PMCID: PMC5765312 DOI: 10.1515/raon-2017-0044] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 09/25/2017] [Indexed: 01/10/2023] Open
Abstract
Background Although the effects of high dose radiation on human cells and tissues are relatively well defined, there is no consensus regarding the effects of low and very low radiation doses on the organism. Ionizing radiation has been shown to induce gene mutations and chromosome aberrations which are known to be involved in the process of carcinogenesis. The induction of secondary cancers is a challenging long-term side effect in oncologic patients treated with radiation. Medical sources of radiation like intensity modulated radiotherapy used in cancer treatment and computed tomography used in diagnostics, deliver very low doses of radiation to large volumes of healthy tissue, which might contribute to increased cancer rates in long surviving patients and in the general population. Research shows that because of the phenomena characteristic for low dose radiation the risk of cancer induction from exposure of healthy tissues to low dose radiation can be greater than the risk calculated from linear no-threshold model. Epidemiological data collected from radiation workers and atomic bomb survivors confirms that exposure to low dose radiation can contribute to increased cancer risk and also that the risk might correlate with the age at exposure. Conclusions Understanding the molecular mechanisms of response to low dose radiation is crucial for the proper evaluation of risks and benefits that stem from these exposures and should be considered in the radiotherapy treatment planning and in determining the allowed occupational exposures.
Collapse
Affiliation(s)
- Igor Piotrowski
- Radiobiology Laboratory, Department of Medical Physics, Greater Poland Cancer Centre, Garbary 15 Street, Poznań, Poland
| | - Katarzyna Kulcenty
- Radiobiology Laboratory, Department of Medical Physics, Greater Poland Cancer Centre, Garbary 15 Street, Poznań, Poland.,Department of Electroradiology, University of Medical Sciences, Poznań, Poland
| | - Wiktoria Maria Suchorska
- Radiobiology Laboratory, Department of Medical Physics, Greater Poland Cancer Centre, Garbary 15 Street, Poznań, Poland.,Department of Electroradiology, University of Medical Sciences, Poznań, Poland
| | - Agnieszka Skrobała
- Department of Electroradiology, University of Medical Sciences, Poznań, Poland.,Department of Medical Physics, Greater Poland Cancer Centre, Poznań, Poland
| | - Małgorzata Skórska
- Department of Medical Physics, Greater Poland Cancer Centre, Poznań, Poland
| | - Marta Kruszyna-Mochalska
- Department of Electroradiology, University of Medical Sciences, Poznań, Poland.,Department of Medical Physics, Greater Poland Cancer Centre, Poznań, Poland
| | - Anna Kowalik
- Department of Medical Physics, Greater Poland Cancer Centre, Poznań, Poland
| | | | - Julian Malicki
- Department of Electroradiology, University of Medical Sciences, Poznań, Poland.,Department of Medical Physics, Greater Poland Cancer Centre, Poznań, Poland
| |
Collapse
|
26
|
Paths from DNA damage and signaling to genome rearrangements via homologous recombination. Mutat Res 2017; 806:64-74. [PMID: 28779875 DOI: 10.1016/j.mrfmmm.2017.07.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 07/21/2017] [Indexed: 12/31/2022]
Abstract
DNA damage is a constant threat to genome integrity. DNA repair and damage signaling networks play a central role maintaining genome stability, suppressing tumorigenesis, and determining tumor response to common cancer chemotherapeutic agents and radiotherapy. DNA double-strand breaks (DSBs) are critical lesions induced by ionizing radiation and when replication forks encounter damage. DSBs can result in mutations and large-scale genome rearrangements reflecting mis-repair by non-homologous end joining or homologous recombination. Ionizing radiation induces genetic change immediately, and it also triggers delayed events weeks or even years after exposure, long after the initial damage has been repaired or diluted through cell division. This review covers DNA damage signaling and repair pathways and cell fate following genotoxic insult, including immediate and delayed genome instability and cell survival/cell death pathways.
Collapse
|
27
|
Sharma R, Kumar C, Mallia MB, Kameswaran M, Sarma HD, Banerjee S, Dash A. In Vitro Evaluation of 188Re-HEDP: A Mechanistic View of Bone Pain Palliations. Cancer Biother Radiopharm 2017; 32:184-191. [PMID: 28598690 DOI: 10.1089/cbr.2017.2200] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Skeletal metastasis is common in advanced stages of various cancers, particularly of the prostate and breast carcinoma. 188Re-HEDP (1-hydroxyethane 1, 1-diphosphonic acid) is a clinically established radiopharmaceutical for bone pain palliation of osseous metastasis, and it takes advantage of high bone affinity. The present work aims at elucidating the possible mechanisms of cell killing by 188Re-HEDP in osteosarcoma cells and biodistribution studies in mice.188Re-HEDP complex was prepared by using lyophilized HEDP kits prepared in-house. In vitro cellular uptake in mineralized bone matrix was found to be 13.41% ± 0.46% (at 2 hours), which was reduced to 2.44% ± 0.12% in the presence of excess amounts of unlabeled HEDP ligand. Uptake of 188Re-HEDP in bones of normal Swiss mice in vivo and mineralized bone in vitro indicated its affinity toward the bone matrix. The study also revealed that cellular toxicity and G2/M cell cycle arrest were dose dependent. At higher doses, G2/M cell cycle arrest was observed, which might be the major cause of cell death and a possible mechanism of bone pain relief.
Collapse
Affiliation(s)
- Rohit Sharma
- 1 Radiopharmaceuticals Division, Bhabha Atomic Research Centre , Mumbai, India
| | - Chandan Kumar
- 1 Radiopharmaceuticals Division, Bhabha Atomic Research Centre , Mumbai, India
| | - Madhava B Mallia
- 1 Radiopharmaceuticals Division, Bhabha Atomic Research Centre , Mumbai, India
| | - Mythili Kameswaran
- 1 Radiopharmaceuticals Division, Bhabha Atomic Research Centre , Mumbai, India
| | - Haladhar D Sarma
- 2 Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre , Mumbai, India
| | - Sharmila Banerjee
- 3 Radiation Medicine Centre, Bhabha Atomic Research Centre , Mumbai, India
| | - Ashutosh Dash
- 1 Radiopharmaceuticals Division, Bhabha Atomic Research Centre , Mumbai, India
| |
Collapse
|
28
|
Hanu C, Wong R, Sur RK, Hayward JE, Seymour C, Mothersill C. Low-dose non-targeted radiation effects in human esophageal adenocarcinoma cell lines. Int J Radiat Biol 2016; 93:165-173. [PMID: 27653785 DOI: 10.1080/09553002.2017.1237057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
PURPOSE To investigate non-targeted radiation effects in esophageal adenocarcinoma cell lines (OE19 and OE33) using human keratinocyte and colorectal cancer cell reporters following γ-ray exposure. MATERIALS AND METHODS Both clonogenic assays and ratiometric calcium endpoints were used to check for the occurrence of bystander signals in reporter cells. RESULTS We report data suggesting that γ-irradiation increases cell killing over the expected linear quadratic (LQ) model levels in the OE19 cell line exposed to doses below 1 Gy, i.e. which may be suggestive to be a low hyper-radiosensitive (HRS) response to direct irradiation. Both EAC cell lines (OE19 and OE33) have the ability to produce bystander signals when irradiated cell conditioned medium (ICCM) is placed onto human keratinocyte reporters, but do not seem to be capable of responding to bystander signals when placed on their autologous reporters. Further work with human keratinocyte reporter models showed statistically significant intracellular calcium fluxes following exposure of the reporters to ICCM harvested from both EAC cell lines exposed to 0.5 Gy. CONCLUSION These experiments suggest that the OE19 and OE33 cell lines produce bystander signals in human keratinocyte reporter cells. However, the radiosensitivity of the EAC cell lines used in this study cannot be enhanced by the bystander response since both cell lines could not respond to bystander signals.
Collapse
Affiliation(s)
- Christine Hanu
- a Medical Physics & Applied Radiation Sciences , McMaster University , Hamilton , ON , Canada
| | - Raimond Wong
- b Department of Oncology and McMaster University , Hamilton , ON , Canada
| | - Ranjan K Sur
- b Department of Oncology and McMaster University , Hamilton , ON , Canada
| | - Joseph E Hayward
- a Medical Physics & Applied Radiation Sciences , McMaster University , Hamilton , ON , Canada.,c Department of Radiology , McMaster University , Hamilton , ON , Canada
| | - Colin Seymour
- a Medical Physics & Applied Radiation Sciences , McMaster University , Hamilton , ON , Canada
| | - Carmel Mothersill
- a Medical Physics & Applied Radiation Sciences , McMaster University , Hamilton , ON , Canada
| |
Collapse
|
29
|
Chiker S, Pennaneach V, Loew D, Dingli F, Biard D, Cordelières FP, Gemble S, Vacher S, Bieche I, Hall J, Fernet M. Cdk5 promotes DNA replication stress checkpoint activation through RPA-32 phosphorylation, and impacts on metastasis free survival in breast cancer patients. Cell Cycle 2016; 14:3066-78. [PMID: 26237679 DOI: 10.1080/15384101.2015.1078020] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cyclin dependent kinase 5 (Cdk5) is a determinant of PARP inhibitor and ionizing radiation (IR) sensitivity. Here we show that Cdk5-depleted (Cdk5-shRNA) HeLa cells show higher sensitivity to S-phase irradiation, chronic hydroxyurea exposure, and 5-fluorouracil and 6-thioguanine treatment, with hydroxyurea and IR sensitivity also seen in Cdk5-depleted U2OS cells. As Cdk5 is not directly implicated in DNA strand break repair we investigated in detail its proposed role in the intra-S checkpoint activation. While Cdk5-shRNA HeLa cells showed altered basal S-phase dynamics with slower replication velocity and fewer active origins per DNA megabase, checkpoint activation was impaired after a hydroxyurea block. Cdk5 depletion was associated with reduced priming phosphorylations of RPA32 serines 29 and 33 and SMC1-Serine 966 phosphorylation, lower levels of RPA serine 4 and 8 phosphorylation and DNA damage measured using the alkaline Comet assay, gamma-H2AX signal intensity, RPA and Rad51 foci, and sister chromatid exchanges resulting in impaired intra-S checkpoint activation and subsequently higher numbers of chromatin bridges. In vitro kinase assays coupled with mass spectrometry demonstrated that Cdk5 can carry out the RPA32 priming phosphorylations on serines 23, 29, and 33 necessary for this checkpoint activation. In addition we found an association between lower Cdk5 levels and longer metastasis free survival in breast cancer patients and survival in Cdk5-depleted breast tumor cells after treatment with IR and a PARP inhibitor. Taken together, these results show that Cdk5 is necessary for basal replication and replication stress checkpoint activation and highlight clinical opportunities to enhance tumor cell killing.
Collapse
Affiliation(s)
- Sara Chiker
- a Institut Curie; Centre de Recherche; Centre Universitaire ; Orsay Cedex , France.,b Inserm; U612; Centre Universitaire ; Orsay Cedex , France.,c Université Paris-XI; Faculté de Médecine ; Le Kremlin Bicêtre , France
| | - Vincent Pennaneach
- a Institut Curie; Centre de Recherche; Centre Universitaire ; Orsay Cedex , France.,b Inserm; U612; Centre Universitaire ; Orsay Cedex , France
| | - Damarys Loew
- d Institut Curie; Centre de Recherche; Laboratoire de Spectrométrie de Masse Protéomique ; Paris , France
| | - Florent Dingli
- d Institut Curie; Centre de Recherche; Laboratoire de Spectrométrie de Masse Protéomique ; Paris , France
| | - Denis Biard
- e Commissariat à l'Energie Atomique; DSV; iMETI; SEPIA; Team Cellular Engineering and Human Syndromes ; Fontenay aux Roses , France
| | - Fabrice P Cordelières
- a Institut Curie; Centre de Recherche; Centre Universitaire ; Orsay Cedex , France.,f CNRS; UMR3348; Centre Universitaire ; Orsay Cedex , France.,g Plateforme IBiSA d'Imagerie Cellulaire et Tissulaire; Institut Curie; Centre Universitaire ; Orsay , France
| | - Simon Gemble
- a Institut Curie; Centre de Recherche; Centre Universitaire ; Orsay Cedex , France.,f CNRS; UMR3348; Centre Universitaire ; Orsay Cedex , France
| | - Sophie Vacher
- h Pharmacogenetics Unit; Genetics Service ; Department of Tumour Biology ; Institut Curie ; Paris , France
| | - Ivan Bieche
- h Pharmacogenetics Unit; Genetics Service ; Department of Tumour Biology ; Institut Curie ; Paris , France
| | - Janet Hall
- a Institut Curie; Centre de Recherche; Centre Universitaire ; Orsay Cedex , France.,b Inserm; U612; Centre Universitaire ; Orsay Cedex , France.,i Centre de Recherche en Cancérologie de Lyon -UMR Inserm 1052 - CNRS 5286 ; Lyon , France
| | - Marie Fernet
- a Institut Curie; Centre de Recherche; Centre Universitaire ; Orsay Cedex , France.,b Inserm; U612; Centre Universitaire ; Orsay Cedex , France
| |
Collapse
|
30
|
|
31
|
Yang G, Li W, Jiang H, Liang X, Zhao Y, Yu D, Zhou L, Wang G, Tian H, Han F, Cai L, Cui J. Low-dose radiation may be a novel approach to enhance the effectiveness of cancer therapeutics. Int J Cancer 2016; 139:2157-68. [PMID: 27299986 DOI: 10.1002/ijc.30235] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 05/01/2016] [Accepted: 06/06/2016] [Indexed: 12/26/2022]
Abstract
It has been generally accepted that both natural and man-made sources of ionizing radiation contribute to human exposure and consequently pose a possible risk to human health. However, accumulating evidence has shown that the biological effects of low-dose radiation (LDR) are different from those of high-dose radiation. LDR can stimulate proliferation of normal cells and activate their defense systems, while these biological effects are not observed in some cancer cell types. Although there is still no concordance on this matter, the fact that LDR has the potential to enhance the effects of cancer therapeutics and reduce the toxic side effects of anti-cancer therapy has garnered significant interest. Here, we provide an overview of the current knowledge regarding the experimental data detailing the different responses of normal and cancer tissues to LDR, the underlying mechanisms, and its significance in clinical application.
Collapse
Affiliation(s)
- Guozi Yang
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China.,Department of Radiation-Oncology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Wei Li
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Hongyu Jiang
- Health Examination Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Xinyue Liang
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Yuguang Zhao
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Dehai Yu
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Lei Zhou
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Guanjun Wang
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Huimin Tian
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Fujun Han
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Lu Cai
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China. .,Kosair Children's Hospital Research Institute, Departments of Pediatrics, Radiation Oncology, Pharmacology and Toxicology of the University of Louisville, Louisville, KY, 40202.
| | - Jiuwei Cui
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
32
|
Pan Y, Liu G, Xiao J, Su B, Zhou F, Wei Y. A novel curcuminoid exhibits enhanced antitumor activity in nasopharyngeal carcinoma. Int J Oncol 2016; 48:2175-83. [PMID: 26983360 DOI: 10.3892/ijo.2016.3425] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 02/18/2016] [Indexed: 11/05/2022] Open
Abstract
Curcumin shows growth-inhibition against tumor cells through multi-target mechanisms. Nevertheless, the poor stability and pharmacokinetics considerably limit its clinical functions. Increased effort has been put into the chemical alteration of curcumin to find potential analogues with improved bioavailability and antitumor activities. In this study, the antitumor activity of a novel curcuminoid (B63) in nasopharyngeal carcinoma (NPC) was examined. The MTT and colony formation assays were used to detect NPC cell viability and proliferation. Flow cytometry was used to detect cell cycle distribution. The Annexin V/PI staining assay and cleavage PARP and cleavage caspase-3 expression were used to examine apoptosis. Western blotting was used to examine the protein expression of endoplasmic reticulum (ER) stress pathway markers, XBP-1, ATF-4 and CHOP. The suppressive effect of B63 on tumor growth was examined in vivo by subcutaneously inoculated NPC in a tumor model using nude mice. Treatment with B63 potentially caused growth inhibition and apoptosis in NPC cells in a dose- and time-responsive manner. Its antitumor effect was associated with the ER stress activation. Nevertheless, the same dose of curcumin did not activate ER stress. In addition, knockdown of Chop attenuated B63-induced cell viability inhibition, suggesting that the apoptotic pathway is ER stress-dependent. The tumor volume and weight were significantly reduced by pretreating the NPC cells with B63 before implantation in the in vivo mouse model. B63 exhibited a more potent antitumor action than curcumin in NPC. These observations on the novel compound B63 indicate a novel candidate for NPC therapy.
Collapse
Affiliation(s)
- Yunbao Pan
- Department of Pathology, Affiliated Hospital, Jiangnan University, Wuxi, Jiangsu 214062, P.R. China
| | - Guohong Liu
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, P.R. China
| | - Jian Xiao
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, P.R. China
| | - Bojin Su
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, P.R. China
| | - Fuling Zhou
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Yongchang Wei
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
33
|
Shi HM, Ding KK, Zhou PK, Guo DM, Chen D, Li YS, Zhao CL, Zhao CC, Zhang X. Radiation-induced expression of IER5 is dose-dependent and not associated with the clinical outcomes of radiotherapy in cervical cancer. Oncol Lett 2016; 11:1309-1314. [PMID: 26893736 PMCID: PMC4734294 DOI: 10.3892/ol.2016.4086] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 10/30/2015] [Indexed: 11/08/2022] Open
Abstract
This study aimed to investigate the expression of the immediate-early response 5 (IER5) gene in cervical cancer tissues and explore the association between the expression of IER5 and the clinical outcomes of radiotherapy. We collected specimens by surgery or biopsy and obtained 53 specimens from tissues after radiotherapy and 16 specimens from tissues before radiotherapy. Immunohistochemistry and western blotting were used to assess the protein expression levels of IER5. Quantitative polymerase chain reaction (qPCR) was performed to assess the mRNA expression levels of IER5. The protein and mRNA expression levels of IER5 in cervical cancer patients treated with radiation doses ≥20 Gy were significantly higher than in those treated with radiation doses <20 Gy (P<0.05) and before treatment with radiotherapy. Moreover, the expression of IER5 was significantly positively correlated with the radiation dose (immunohistochemistry: r=0.548, P=0.019; qPCR: r=0.671, P=0.002; western blotting: r=0.573, P<0.0001). Radiotherapy induced the upregulated expression of IER5 and this was dependent on the radiation dose. However, the radiation-induced expression of IER5 was not associated with the clinical outcomes of radiotherapy in cervical cancer.
Collapse
Affiliation(s)
- Hai-Min Shi
- Department of Gynaecology, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Ku-Ke Ding
- National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing 100088, P.R. China
| | - Ping-Kun Zhou
- Department of Radiation Toxicology and Oncology, Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| | - Dong-Mei Guo
- Department of Gynaecology, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Dan Chen
- Department of Gynaecology, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Yan-Sha Li
- Department of Gynaecology, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Chun-Li Zhao
- Department of Gynaecology, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Chen-Chen Zhao
- Department of Gynaecology, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Xin Zhang
- Department of Gynaecology, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| |
Collapse
|
34
|
Meade AD, Howe O, Unterreiner V, Sockalingum GD, Byrne HJ, Lyng FM. Vibrational spectroscopy in sensing radiobiological effects: analyses of targeted and non-targeted effects in human keratinocytes. Faraday Discuss 2016; 187:213-34. [DOI: 10.1039/c5fd00208g] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Modern models of radiobiological effects include mechanisms of damage initiation, sensing and repair, for those cells that directly absorb ionizing radiation as well as those that experience molecular signals from directly irradiated cells. In the former case, the effects are termed targeted effects while, in the latter, non-targeted effects. It has emerged that phenomena occur at low doses below 1 Gy in directly irradiated cells that are associated with cell-cycle-dependent mechanisms of DNA damage sensing and repair. Likewise in non-targeted bystander-irradiated cells the effect saturates at 0.5 Gy. Both effects at these doses challenge the limits of detection of vibrational spectroscopy. In this paper, a study of the sensing of both targeted and non-targeted effects in HaCaT human keratinocytes irradiated with gamma ray photons is conducted with vibrational spectroscopy. In the case of directly irradiated cells, it is shown that the HaCaT cell line does exhibit both hyperradiosensitivity and increased radioresistance at low doses, a transition between the two effects occurring at a dose of 200 mGy, and that cell survival and other physiological effects as a function of dose follow the induced repair model. Both Raman and FTIR signatures are shown to follow a similar model, suggesting that the spectra include signatures of DNA damage sensing and repair. In bystander-irradiated cells, pro- and anti-apoptotic signalling and mechanisms of ROS damage were inhibited in the mitogen-activated protein kinase (MAPK) transduction pathway. It is shown that Raman spectral profiles of bystander-irradiated cells are correlated with markers of bystander signalling and molecular transduction. This work demonstrates for the first time that both targeted and non-targeted effects of ionizing radiation damage are detected by vibrational spectroscopy in vitro.
Collapse
Affiliation(s)
- Aidan D. Meade
- School of Physics
- Dublin Institute of Technology
- Dublin 8
- Ireland
- DIT Centre for Radiation and Environmental Science
| | - Orla Howe
- DIT Centre for Radiation and Environmental Science
- Focas Research Institute
- Dublin Institute of Technology
- Dublin 8
- Ireland
| | - Valérie Unterreiner
- Plateforme en Imagerie Cellulaire et Tissulaire (PICT)
- Université de Reims Champagne-Ardenne
- 51095 Reims Cedex
- France
| | - Ganesh D. Sockalingum
- Université de Reims Champagne-Ardenne
- MéDIAN-Biophotonique et Technologies pour la Santé
- UFR de Pharmacie
- 51095 Reims Cedex
- France
| | - Hugh J. Byrne
- Focas Research Institute
- Dublin Institute of Technology
- Dublin 8
- Ireland
| | - Fiona M. Lyng
- School of Physics
- Dublin Institute of Technology
- Dublin 8
- Ireland
- DIT Centre for Radiation and Environmental Science
| |
Collapse
|
35
|
Słonina D, Gasińska A, Biesaga B, Janecka A, Kabat D. An association between low-dose hyper-radiosensitivity and the early G2-phase checkpoint in normal fibroblasts of cancer patients. DNA Repair (Amst) 2015; 39:41-5. [PMID: 26725161 DOI: 10.1016/j.dnarep.2015.12.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 12/11/2015] [Accepted: 12/11/2015] [Indexed: 10/22/2022]
Abstract
In our previous study, low-dose hyper-radiosensitivity (HRS) effect was demonstrated for normal fibroblasts (asynchronous and G2-phase enriched) of 4 of the 25 cancer patients investigated. For the rest of patients, HRS was not defined in either of the 2 fibroblast populations. Thus, the study indicated that G2-phase enrichment had no influence on HRS identification. The conclusion contradicts that reported for human tumor cells, and suggests different mechanism of HRS in normal human cells. In the present paper we report, for the first time, the activity of early G2-phase checkpoint after low-dose irradiation in normal fibroblasts of these 4 HRS-positive patients and 4 HRS-negative patients and answer the question regarding the role of this checkpoint in normal human cells. The response of the early G2-phase checkpoint was determined by assessment of the progression of irradiated cells into mitosis using the mitotic marker, phosphorylated histone H3. We found evident differences in the activity of the early G2-phase checkpoint between HRS-positive and HRS-negative fibroblasts. In HRS-positive fibroblasts the checkpoint was not triggered and DNA damage was not recognized after doses lower than 0.2Gy resulting in HRS response. On the contrary, in HRS-negative fibroblasts the early G2-phase checkpoint was activated regardless of the dose in the range 0.1-2Gy. In conclusion, although cell cycle phase has no effect on the presence of HRS effect in normal human fibroblasts, the data reported here indicate that HRS response in these cells is associated with the functioning of early G2-phase checkpoint in a threshold-dose dependent manner, similarly as it takes place in most of human tumor and other cells.
Collapse
Affiliation(s)
- Dorota Słonina
- Department of Applied Radiobiology, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology Cracow Branch, 11 Garncarska Street, Cracow 31-115, Poland.
| | - Anna Gasińska
- Department of Applied Radiobiology, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology Cracow Branch, 11 Garncarska Street, Cracow 31-115, Poland
| | - Beata Biesaga
- Department of Applied Radiobiology, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology Cracow Branch, 11 Garncarska Street, Cracow 31-115, Poland
| | - Anna Janecka
- Department of Applied Radiobiology, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology Cracow Branch, 11 Garncarska Street, Cracow 31-115, Poland
| | - Damian Kabat
- Department of Medical Physics, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology Cracow Branch, 11 Garncarska Street, Cracow 31-115, Poland
| |
Collapse
|
36
|
Allen CP, Tinganelli W, Sharma N, Nie J, Sicard C, Natale F, King M, Keysar SB, Jimeno A, Furusawa Y, Okayasu R, Fujimori A, Durante M, Nickoloff JA. DNA Damage Response Proteins and Oxygen Modulate Prostaglandin E2 Growth Factor Release in Response to Low and High LET Ionizing Radiation. Front Oncol 2015; 5:260. [PMID: 26697402 PMCID: PMC4670845 DOI: 10.3389/fonc.2015.00260] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 11/09/2015] [Indexed: 12/20/2022] Open
Abstract
Common cancer therapies employ chemicals or radiation that damage DNA. Cancer and normal cells respond to DNA damage by activating complex networks of DNA damage sensor, signal transducer, and effector proteins that arrest cell cycle progression, and repair damaged DNA. If damage is severe enough, the DNA damage response (DDR) triggers programed cell death by apoptosis or other pathways. Caspase 3 is a protease that is activated upon damage and triggers apoptosis, and production of prostaglandin E2 (PGE2), a potent growth factor that can enhance growth of surviving cancer cells leading to accelerated tumor repopulation. Thus, dying tumor cells can promote growth of surviving tumor cells, a pathway aptly named Phoenix Rising. In the present study, we surveyed Phoenix Rising responses in a variety of normal and established cancer cell lines, and in cancer cell lines freshly derived from patients. We demonstrate that IR induces a Phoenix Rising response in many, but not all cell lines, and that PGE2 production generally correlates with enhanced growth of cells that survive irradiation, and of unirradiated cells co-cultured with irradiated cells. We show that PGE2 production is stimulated by low and high LET ionizing radiation, and can be enhanced or suppressed by inhibitors of key DDR proteins. PGE2 is produced downstream of caspase 3 and the cyclooxygenases COX1 and COX2, and we show that the pan COX1–2 inhibitor indomethacin blocks IR-induced PGE2 production in the presence or absence of DDR inhibitors. COX1–2 require oxygen for catalytic activity, and we further show that PGE2 production is markedly suppressed in cells cultured under low (1%) oxygen concentration. Thus, Phoenix Rising is most likely to cause repopulation of tumors with relatively high oxygen, but not in hypoxic tumors. This survey lays a foundation for future studies to further define tumor responses to radiation and inhibitors of the DDR and Phoenix Rising to enhance the efficacy of radiotherapy with the ultimate goal of precision medicine informed by deep understanding of specific tumor responses to radiation and adjunct chemotherapy targeting key factors in the DDR and Phoenix Rising pathways.
Collapse
Affiliation(s)
- Christopher P Allen
- Department of Environmental and Radiological Health Sciences, Colorado State University , Fort Collins, CO , USA
| | - Walter Tinganelli
- GSI Helmholtzzentrum für Schwerionenforschung GmbH , Darmstadt , Germany ; Research Development and Support Center, National Institute of Radiological Sciences , Chiba , Japan
| | - Neelam Sharma
- Department of Environmental and Radiological Health Sciences, Colorado State University , Fort Collins, CO , USA
| | - Jingyi Nie
- Department of Environmental and Radiological Health Sciences, Colorado State University , Fort Collins, CO , USA
| | - Cory Sicard
- Department of Environmental and Radiological Health Sciences, Colorado State University , Fort Collins, CO , USA
| | - Francesco Natale
- GSI Helmholtzzentrum für Schwerionenforschung GmbH , Darmstadt , Germany
| | - Maurice King
- Department of Environmental and Radiological Health Sciences, Colorado State University , Fort Collins, CO , USA
| | - Steven B Keysar
- Division of Medical Oncology, University of Colorado School of Medicine , Aurora, CO , USA
| | - Antonio Jimeno
- Division of Medical Oncology, University of Colorado School of Medicine , Aurora, CO , USA
| | - Yoshiya Furusawa
- Research Development and Support Center, National Institute of Radiological Sciences , Chiba , Japan ; Research Center for Radiation Protection, National Institute of Radiological Sciences , Chiba , Japan
| | - Ryuichi Okayasu
- Research Center for Charged Particle Therapy, National Institute of Radiological Sciences , Chiba , Japan
| | - Akira Fujimori
- Research Center for Charged Particle Therapy, National Institute of Radiological Sciences , Chiba , Japan
| | - Marco Durante
- GSI Helmholtzzentrum für Schwerionenforschung GmbH , Darmstadt , Germany
| | - Jac A Nickoloff
- Department of Environmental and Radiological Health Sciences, Colorado State University , Fort Collins, CO , USA
| |
Collapse
|
37
|
Enns L, Rasouli-Nia A, Hendzel M, Marples B, Weinfeld M. Association of ATM activation and DNA repair with induced radioresistance after low-dose irradiation. RADIATION PROTECTION DOSIMETRY 2015; 166:131-6. [PMID: 25904696 PMCID: PMC4572139 DOI: 10.1093/rpd/ncv203] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Mammalian cells often exhibit a hyper-radiosensitivity (HRS) to radiation doses <20 cGy, followed by increased radioresistance (IRR) at slightly higher doses (∼20-30 cGy). Here, the influence of DNA double-strand break repair (DSBR) on IRR was examined. The failure of Ataxia telangiectasia (AT) cells to undergo IRR reported by others was confirmed. Flow cytometric analysis indicated that normal cells fail to show a measurable increase in serine 1981 phosphorylated AT-mutated (ATM) protein after 10 cGy up to 4 h post irradiation, but a two- to fourfold increase after 25 cGy. Similarly, more proficient reduction of phosphorylated histone H2AX was observed 24 h after 25 cGy than after 10 cGy, suggesting that DSBR is more efficient during IRR than HRS. A direct examination of the consequences of inefficient DNA repair per se (as opposed to ATM-mediated signal transduction/cell cycle responses), by determining the clonogenic survival of cells lacking the DNA repair enzyme polynucleotide kinase/phosphatase, indicated that these cells have a response similar to AT cells, i.e. HRS but no IRR, strongly linking IRR to DSBR.
Collapse
Affiliation(s)
- L Enns
- Department of Oncology, University of Alberta, Cross Cancer Institute, 11560 University Ave., Edmonton, Alberta T6G 1Z2, Canada
| | - A Rasouli-Nia
- Department of Oncology, University of Alberta, Cross Cancer Institute, 11560 University Ave., Edmonton, Alberta T6G 1Z2, Canada
| | - M Hendzel
- Department of Oncology, University of Alberta, Cross Cancer Institute, 11560 University Ave., Edmonton, Alberta T6G 1Z2, Canada
| | - B Marples
- Department of Radiation Oncology, William Beaumont Hospital, 3811 W. Thirteen Mile Rd., 105-RI, Royal Oak, MI 48073, USA
| | - M Weinfeld
- Department of Oncology, University of Alberta, Cross Cancer Institute, 11560 University Ave., Edmonton, Alberta T6G 1Z2, Canada
| |
Collapse
|
38
|
Nguyen DM, Parekh PR, Chang ET, Sharma NK, Carrier F. Contribution of Dual Oxidase 2 (DUOX2) to Hyper-Radiosensitivity in Human Gastric Cancer Cells. Radiat Res 2015. [PMID: 26207686 DOI: 10.1667/rr13661.1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Whole-abdominal radiotherapy (WART) is a primary method for managing gastrointestinal cancers that have disseminated into intra-abdominal tissues. While effective, this approach is limited because of the increased toxicity to normal tissue associated with combined WART and full-dose chemotherapy regimens. Recent studies have demonstrated a survival advantage in a novel treatment paradigm that allows for the safe use of full-dose systemic chemotherapy in combination with low-dose fractionated radiotherapy (LDFRT). Traditionally, radiation doses greater than 120 cGy have been used in radiotherapy because lower doses were thought to be ineffective for tumor therapy. However, we now know that LDFRT can produce hyper-radiosensitivity (HRS), a phenomenon where cells undergo apoptosis at radiation doses as low as 15 cGy, in a number of proliferating cells. The objectives of our current study were to determine whether LDFRT can induce HRS in gastrointestinal cancer cells and to identify biomarkers of chemopotentiation by LDFRT. Our data indicate that three consecutive daily fractions of 15 cGy produced HRS in gastric cancer cells and potentiated a modified regimen of docetaxel, cisplatin and 5'-fluorouracil (mDCF). Colony survival assays indicated that 15 cGy was sufficient to kill 90% of the cells when LDFRT was combined with mDCF whereas a dose almost 10 times higher (135 cGy) was needed to achieve the same rate when using conventional radiotherapy alone. RT(2) PCR Profiler™ array analysis indicated that this combined regimen upregulated dual oxidase 2 (DUOX2), an enzyme functioning in the production of hydrogen peroxide, without upregulating genes involved in DNA repair. Moreover, downregulation of DUOX2 increased radioresistance at every radiation dose tested. In addition, our data indicate that reactive oxygen species (ROS) increase up to 3.5-fold in cells exposed to LDFRT and mDCF. Furthermore, inhibition of NADPH oxidase abrogated the killing efficiency of this combined regimen. Taken together these data suggest that chemopotentiation by LDFRT in gastric cancer cells may be due, at least in part, to increased ROS production (DUOX2) without upregulation of the DNA repair machinery. These data thus provide a rationale for further explorations of potential clinical applications of LDFRT, such as in WART, as a chemopotentiator for advanced and metastatic gastric cancers.
Collapse
Affiliation(s)
- Duc M Nguyen
- Marlene and Stewart Greenebaum Cancer Center, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Palak R Parekh
- Marlene and Stewart Greenebaum Cancer Center, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Elizabeth T Chang
- Marlene and Stewart Greenebaum Cancer Center, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Navesh K Sharma
- Marlene and Stewart Greenebaum Cancer Center, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - France Carrier
- Marlene and Stewart Greenebaum Cancer Center, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| |
Collapse
|
39
|
Xue L, Furusawa Y, Yu D. ATR signaling cooperates with ATM in the mechanism of low dose hypersensitivity induced by carbon ion beam. DNA Repair (Amst) 2015; 34:1-8. [PMID: 26246317 DOI: 10.1016/j.dnarep.2015.07.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 07/04/2015] [Accepted: 07/14/2015] [Indexed: 02/04/2023]
Abstract
Little work has been done on the mechanism of low dose hyper-radiosensitivity (HRS) and later appeared radioresistance (termed induced radioresistance (IRR)) after irradiation with medium and high linear energy transfer (LET) particles. The aim of this study was to find out whether ATR pathway is involved in the mechanism of HRS induced by high LET radiation. GM0639 cells and two ATM deficient/mutant cells, AT5BIVA and AT2KY were irradiated by carbon ion beam. Thymidine block technique was developed to enrich the G2-phase population. Radiation induced early G2/M checkpoint was quantitatively assess with dual-parameter flow cytometry by detecting the cells positive for phospho-histone H3. The involvement of ATR pathway in HRS/IRR response was detected with pretreatment of specific inhibitors prior to carbon ion beam. The link between the early G2/M checkpoint and HRS/IRR under carbon ion beam was first confirmed in GM0639 cells, through the enrichment of cell population in G2-phase or with Aurora kinase inhibitor that attenuates the transition from G2 to M phase. Interestingly, the early G2/M arrest could still be observed in ATM deficient/mutant cells with an effect of ATR signaling, which was discovered to function in an LET-dependent manner, even as low as 0.2Gy for carbon ion radiation. The involvement of ATR pathway in heavy particles induced HRS/IRR was determined with the specific ATR inhibitor in GM0639 cells, which affected the HRS/IRR occurrence similarly as ATM inhibitor. These data demonstrate that ATR pathway may cooperate with ATM in the mechanism of low dose hypersensitivity induced by carbon ion beam.
Collapse
Affiliation(s)
- Lian Xue
- School of Public Health, Medical College of Soochow University, Suzhou, China; Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Soochow University, Suzhou, China
| | - Yoshiya Furusawa
- Microbeam Development Office, Research/Development/Support Center, National Institute of Radiological Sciences, Chiba, Japan
| | - Dong Yu
- School of Radiological Medicine and Protection, Medical College of Soochow University, Suzhou, China.
| |
Collapse
|
40
|
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
|
41
|
Zhang W, Gao R, Yu Y, Guo K, Hou P, Yu M, Liu Y, Yang A. Iodine-131 induces apoptosis in HTori-3 human thyrocyte cell line and G2/M phase arrest in a p53-independent pathway. Mol Med Rep 2015; 11:3148-54. [PMID: 25515142 DOI: 10.3892/mmr.2014.3096] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 11/07/2014] [Indexed: 11/05/2022] Open
Abstract
Iodine‑131 is known to destroy residual thyroid tissue following surgical resection of differentiated thyroid carcinoma and is widely used to treat hyperthyroidism. However, the mechanism by which iodine‑131 induces apoptosis and cell cycle arrest in the human thyrocyte cell line, Htori‑3, remains to be elucidated. In the present study, the cytotoxic effect of iodine‑131 on the HTori‑3 cell line and the underlying mechanism of iodine‑131‑induced cell apoptosis were investigated. Cell viability was analyzed using an MTT assay, while cell apoptosis and cell cycle arrest were determined using flow cytometry. Reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) and western blot analyses were performed to determine the changes in the expression levels of p53, B‑cell lymphoma 2 (Bcl‑2), Fas and growth arrest and DNA damage‑inducible 45 (GADD45), following iodine‑131 treatment. The results demonstrated that iodine‑131 may inhibit HTori‑3 cell growth via cell apoptosis and G2/M phase arrest in a time‑ and dose‑dependent manner. The iodine‑131 dose required for 50% growth inhibition of HTori‑3 cell viability 48 h after treatment was 27.75±2.22 MBq/ml. Upregulation of Fas and downregulation of Bcl‑2 expression levels were observed following iodine‑131 treatment. The results of RT‑qPCR revealed an increase in the GADD45 mRNA expression following HTori‑3 cell exposure to iodine‑131. Notably, the mRNA and protein expression levels of p53 were not altered following iodine‑131 treatment. In conclusion, iodine‑131 may induce apoptosis in HTori‑3 cells by downregulating the expression of Bcl‑2 and upregulating the expression of Fas. In addition, iodine‑131 may upregulate GADD45 mRNA expression in HTori‑3 cells, resulting in G2/M phase arrest in a p53‑independent pathway.
Collapse
Affiliation(s)
- Weixiao Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Rui Gao
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Yan Yu
- Department of Public Health, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Kun Guo
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Peng Hou
- Endocrinology Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Mingqi Yu
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Yan Liu
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Aimin Yang
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
42
|
Boudra MT, Bolin C, Chiker S, Fouquin A, Zaremba T, Vaslin L, Biard D, Cordelières FP, Mégnin-Chanet F, Favaudon V, Fernet M, Pennaneach V, Hall J. PARP-2 depletion results in lower radiation cell survival but cell line-specific differences in poly(ADP-ribose) levels. Cell Mol Life Sci 2015; 72:1585-97. [PMID: 25336152 PMCID: PMC11113491 DOI: 10.1007/s00018-014-1765-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 10/15/2014] [Accepted: 10/17/2014] [Indexed: 02/02/2023]
Abstract
Poly(ADP-ribose) polymerase-2 (PARP-2) activity contributes to a cells' poly(ADP-ribosyl)ating potential and like PARP-1, has been implicated in several DNA repair pathways including base excision repair and DNA single strand break repair. Here the consequences of its stable depletion in HeLa, U20S, and AS3WT2 cells were examined. All three PARP-2 depleted models showed increased sensitivity to the cell killing effects on ionizing radiation as reported in PARP-2 depleted mouse embryonic fibroblasts providing further evidence for a role in DNA strand break repair. The PARP-2 depleted HeLa cells also showed both higher constitutive and DNA damage-induced levels of polymers of ADP-ribose (PAR) associated with unchanged PARP-1 protein levels, but higher PARP activity and a concomitant lower PARG protein levels and activity. These changes were accompanied by a reduced maximal recruitment of PARP-1, XRCC1, PCNA, and PARG to DNA damage sites. This PAR-associated phenotype could be reversed in HeLa cells on re-expression of PARP-2 and was not seen in U20S and AS3WT2 cells. These results highlight the complexity of the relationship between different members of the PARP family on PAR metabolism and suggest that cell model dependent phenotypes associated with the absence of PARP-2 exist within a common background of radiation sensitivity.
Collapse
Affiliation(s)
- Mohammed-Tayyib Boudra
- Institut Curie, Centre de Recherche, Bât. 110-112, Centre Universitaire, 91405 Orsay, France
- Inserm, U612, Bât. 110-112, Centre Universitaire, 91405 Orsay, France
- Faculté de Médecine, Université Paris-XI, 94270 Le Kremlin Bicêtre, France
| | - Celeste Bolin
- Institut Curie, Centre de Recherche, Bât. 110-112, Centre Universitaire, 91405 Orsay, France
- Inserm, U612, Bât. 110-112, Centre Universitaire, 91405 Orsay, France
- Present Address: Department of Biology, The College of Idaho, 2112 Cleveland Boulevard, Caldwell, ID 83605 USA
| | - Sara Chiker
- Institut Curie, Centre de Recherche, Bât. 110-112, Centre Universitaire, 91405 Orsay, France
- Inserm, U612, Bât. 110-112, Centre Universitaire, 91405 Orsay, France
- Faculté de Médecine, Université Paris-XI, 94270 Le Kremlin Bicêtre, France
| | - Alexis Fouquin
- Institut Curie, Centre de Recherche, Bât. 110-112, Centre Universitaire, 91405 Orsay, France
- Inserm, U612, Bât. 110-112, Centre Universitaire, 91405 Orsay, France
- Faculté de Médecine, Université Paris-XI, 94270 Le Kremlin Bicêtre, France
| | - Tomasz Zaremba
- Institut Curie, Centre de Recherche, Bât. 110-112, Centre Universitaire, 91405 Orsay, France
- Inserm, U612, Bât. 110-112, Centre Universitaire, 91405 Orsay, France
- Present Address: AstraZeneca Pharma Poland Sp. z o.o.ul., Postępu 18, 02-676 Warsaw, Poland
| | - Laurence Vaslin
- Institut Curie, Centre de Recherche, Bât. 110-112, Centre Universitaire, 91405 Orsay, France
- Inserm, U612, Bât. 110-112, Centre Universitaire, 91405 Orsay, France
| | - Denis Biard
- Commissariat à l’Energie Atomique, DSV-iMETI-SEPIA, 92265 Fontenay Aux Roses, France
| | - Fabrice P. Cordelières
- Institut Curie, Centre de Recherche, Bât. 110-112, Centre Universitaire, 91405 Orsay, France
- CNRS, UMR3348, Bât. 110-112, Centre Universitaire, 91405 Orsay, France
- Plateforme IBiSA d’Imagerie Cellulaire et Tissulaire, Institut Curie, Bât. 110-112, Centre Universitaire, 91405 Orsay, France
- Present Address: Pôle d’imagerie photonique, Institut François Magendie, Bordeaux Imaging Center, UMS 3420 CNRS-Université de Bordeaux-US4 INSERM, 146 Rue Léo-Saignat, 33077 Bordeaux, France
| | - Frédérique Mégnin-Chanet
- Institut Curie, Centre de Recherche, Bât. 110-112, Centre Universitaire, 91405 Orsay, France
- Inserm, U612, Bât. 110-112, Centre Universitaire, 91405 Orsay, France
- Present Address: Inserm U1030, Gustave Roussy Cancer Campus Grand Paris, 114 rue Edouard-Vaillant, 94805 Villejuif, France
| | - Vincent Favaudon
- Institut Curie, Centre de Recherche, Bât. 110-112, Centre Universitaire, 91405 Orsay, France
- Inserm, U612, Bât. 110-112, Centre Universitaire, 91405 Orsay, France
| | - Marie Fernet
- Institut Curie, Centre de Recherche, Bât. 110-112, Centre Universitaire, 91405 Orsay, France
- Inserm, U612, Bât. 110-112, Centre Universitaire, 91405 Orsay, France
| | - Vincent Pennaneach
- Institut Curie, Centre de Recherche, Bât. 110-112, Centre Universitaire, 91405 Orsay, France
- Inserm, U612, Bât. 110-112, Centre Universitaire, 91405 Orsay, France
| | - Janet Hall
- Institut Curie, Centre de Recherche, Bât. 110-112, Centre Universitaire, 91405 Orsay, France
- Inserm, U612, Bât. 110-112, Centre Universitaire, 91405 Orsay, France
- Inserm U612, Institut Curie-Recherche, Bât. 110-112, Centre Universitaire, 91405 Orsay, France
| |
Collapse
|
43
|
Bae S, Kim K, Cha HJ, Choi Y, Shin SH, An IS, Lee JH, Lee SJ, Kim JY, Nam SY, An S. Low-dose γ-irradiation induces dual radio-adaptive responses depending on the post-irradiation time by altering microRNA expression profiles in normal human dermal fibroblasts. Int J Mol Med 2014; 35:227-37. [PMID: 25384363 DOI: 10.3892/ijmm.2014.1994] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 10/27/2014] [Indexed: 11/06/2022] Open
Abstract
Exposure to high-dose ionizing radiation, including γ-radiation, induces severe skin disorders. However, the biological consequences and molecular mechanisms responsible for the response of human skin to low-dose γ-radiation (LDR) are largely unknown. In the present study, we demonstrate that LDR (0.1 Gy) induces distinct cellular responses in normal human dermal fibroblasts (NHDFs) depending on the post-irradiation time point. A MTT-based cell viability assay and propidium iodide staining-based cell cycle assay revealed that the viability and proportion of the cells in the G2/M phase were differed at 6 and 24 h post-irradiation. Reverse transcription quantitative PCR (RT-qPCR) revealed that LDR significantly upregulated the mRNA expression of collagen type I alpha 1 (COL1A1), but downregulated the mRNA expression of matrix metalloproteinase 1 (MMP1) at 24 h post-irradiation. MicroRNA (miRNA) microarray analysis further demonstrated that LDR induced changes in the expression profiles of specific miRNAs and that some of the deregulated miRNAs were specific to either the early or late radio-adaptive response. Our results suggest that LDR generates dual radio-adaptive responses depending on the post-irradiation time by altering specific miRNA expression profiles in NHDFs.
Collapse
Affiliation(s)
- Seunghee Bae
- Molecular-Targeted Drug Research Center and Korea Institute for Skin and Clinical Sciences, Konkuk University, Seoul 143-701, Republic of Korea
| | - Karam Kim
- Molecular-Targeted Drug Research Center and Korea Institute for Skin and Clinical Sciences, Konkuk University, Seoul 143-701, Republic of Korea
| | - Hwa Jun Cha
- Molecular-Targeted Drug Research Center and Korea Institute for Skin and Clinical Sciences, Konkuk University, Seoul 143-701, Republic of Korea
| | - Yeongmin Choi
- Molecular-Targeted Drug Research Center and Korea Institute for Skin and Clinical Sciences, Konkuk University, Seoul 143-701, Republic of Korea
| | - Shang Hun Shin
- Molecular-Targeted Drug Research Center and Korea Institute for Skin and Clinical Sciences, Konkuk University, Seoul 143-701, Republic of Korea
| | - In-Sook An
- Molecular-Targeted Drug Research Center and Korea Institute for Skin and Clinical Sciences, Konkuk University, Seoul 143-701, Republic of Korea
| | - Jae Ho Lee
- Laboratory of Molecular Oncology, Cheil General Hospital and Women's Healthcare Center, Kwandong University, College of Medicine, Seoul 100-380, Republic of Korea
| | - Su Jae Lee
- Department of Chemistry, Hanyang University, Seoul 133-791, Republic of Korea
| | - Ji Young Kim
- Radiation Effect Research Team, Radiation Health Research Institute, Korea Hydro and Nuclear Power Co., Ltd., Seoul 132-703, Republic of Korea
| | - Seon Young Nam
- Radiation Effect Research Team, Radiation Health Research Institute, Korea Hydro and Nuclear Power Co., Ltd., Seoul 132-703, Republic of Korea
| | - Sungkwan An
- Molecular-Targeted Drug Research Center and Korea Institute for Skin and Clinical Sciences, Konkuk University, Seoul 143-701, Republic of Korea
| |
Collapse
|
44
|
Choi SH, Yang H, Lee SH, Ki JH, Nam DH, Yoo HY. TopBP1 and Claspin contribute to the radioresistance of lung cancer brain metastases. Mol Cancer 2014; 13:211. [PMID: 25216549 PMCID: PMC4168047 DOI: 10.1186/1476-4598-13-211] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2014] [Accepted: 09/09/2014] [Indexed: 01/03/2023] Open
Abstract
Background Radiation therapy is one of the most effective therapeutic tools for brain metastasis. However, it is inevitable that some cancer cells become resistant to radiation. This study is focused on the identification of genes associated with radioresistance in metastatic brain tumor from lung cancer and the functional examination of the selected genes with regards to altered sensitivity of cancer cells to radiation. Methods After establishing radioresistant cells from the xenograft model, we explored the significant transcriptional changes by performing DNA microarray profiling. Functional analyses in vitro and in vivo performed to validate the gene responsible for radioresistance. Results Transcriptional changes induced by radiation therapy are much more extensive in H460 cells than in PC14PE6 cells. The expression levels of TopBP1 and Claspin were increased in the cancer cells that survived radiation therapy. Depletion of TopBP1 or Claspin using shRNA showed an enhancement of sensitivity to radiation in radioresistant lung cancer cells (PC14PE6). Moreover, increased levels of TopBP1 or Claspin endowed cells a higher resistance to radiation. In xenograft models, the knock-down of TopBP1 or Claspin significantly prolonged the median survival time post radiation therapy. Conclusions We analyzed the gene expression profiles of the radiosensitive cells and the radioresistant cells to define a set of genes that may be involved in endowing lung cancer cells radioresistance post brain metastasis. Functional analyses indicated that the expression TopBP1 and Claspin positively affects the survival of cancer cells and thus negatively the xenograft metastasis model animals in response to radiation. These results show that TopBP1 and Claspin can be potential targets for the enhanced efficacy of radiotherapy. Electronic supplementary material The online version of this article (doi:10.1186/1476-4598-13-211) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | - Do-Hyun Nam
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, 50 Ilwon-Dong, Gangnam-Gu, Seoul 135-710, South Korea.
| | | |
Collapse
|
45
|
Martin LM, Marples B, Lynch TH, Hollywood D, Marignol L. Exposure to low dose ionising radiation: Molecular and clinical consequences. Cancer Lett 2014; 349:98-106. [DOI: 10.1016/j.canlet.2013.12.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
46
|
TACC3 deregulates the DNA damage response and confers sensitivity to radiation and PARP inhibition. Oncogene 2014; 34:1667-78. [PMID: 24769898 DOI: 10.1038/onc.2014.105] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 02/28/2014] [Accepted: 03/12/2014] [Indexed: 12/21/2022]
Abstract
Deregulation of the transforming acidic coiled-coil protein 3 (TACC3), an important factor in the centrosome-microtubule system, has been linked to a variety of human cancer types. We have recently reported on the oncogenic potential of TACC3; however, the molecular mechanisms by which TACC3 mediates oncogenic function remain to be elucidated. In this study, we show that high levels of TACC3 lead to the accumulation of DNA double-strand breaks (DSBs) and disrupt the normal cellular response to DNA damage, at least in part, by negatively regulating the expression of ataxia telangiectasia mutated (ATM) and the subsequent DNA damage response (DDR) signaling cascade. Cells expressing high levels of TACC3 display defective checkpoints and DSB-mediated homologous recombination (HR) and non-homologous end joining (NHEJ) repair systems, leading to genomic instability. Importantly, high levels of TACC3 confer cellular sensitization to radiation and poly(ADP-ribose) polymerase (PARP) inhibition. Overall, our findings provide critical information regarding the mechanisms by which TACC3 contributes to genomic instability, potentially leading to cancer development, and suggest a novel prognostic, diagnostic and therapeutic strategy for the treatment of cancer types expressing high levels of TACC3.
Collapse
|
47
|
Luo J, Zhu W, Tang Y, Cao H, Zhou Y, Ji R, Zhou X, Lu Z, Yang H, Zhang S, Cao J. Artemisinin derivative artesunate induces radiosensitivity in cervical cancer cells in vitro and in vivo. Radiat Oncol 2014; 9:84. [PMID: 24666614 PMCID: PMC3987175 DOI: 10.1186/1748-717x-9-84] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 03/18/2014] [Indexed: 12/18/2022] Open
Abstract
Objective Cervical cancer is the third most common type of cancer in women worldwide and radiotherapy remains its predominant therapeutic treatment. Artesunate (ART), a derivative of artemisinin, has shown radiosensitization effect in previous studies. However, such effects of ART have not yet been revealed for cervical cancer cells. Methods The effect of ART on radiosensitivity of human cervical cancer cell lines HeLa and SiHa was assessed using the clonogenic assay. Cell cycle progression and apoptosis alterations were analyzed by flow cytometry. For in vivo study, HeLa or SiHa cells were inoculated into nude mice to establish tumors. Tissues from xenografts were obtained to detect the changes of microvessel density, apoptosis and cell cycle distribution. Microarray was used to analyze differentially expressed genes. Results ART increased the radiosensitivity of HeLa cells (SER = 1.43, P < 0.001) but not of SiHa cells. Apoptosis and the G2-M phase transition induced by X-ray irradiation (IR) were enhanced by ART via increased Cyclin B1 expression in HeLa cells. Tumor growth of xenografts from HeLa but not SiHa cells was significantly inhibited by irradiation combined with ART (tumor volume reduction of 72.34% in IR + ART group vs. 41.22% in IR group in HeLa cells and 48.79% in IR + ART group vs. 44.03% in IR alone group in SiHa cells). Compared with the irradiated group, cell apoptosis was increased and the G2/M cell cycle arrest was enhanced in the group receiving irradiation combined with ART. Furthermore, compared with radiation alone, X-ray irradiation plus ART affected the expression of 203 genes that function in multiple pathways including RNA transport, the spliceosome, RNA degradation and p53 signaling. Conclusion ART potently abrogates the G2 checkpoint control in HeLa cells. ART can induce radiosensitivity of HeLa cells in vitro and in vivo.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Jianping Cao
- School of Radiation Medicine and Protection and Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China.
| |
Collapse
|
48
|
Słonina D, Biesaga B, Janecka A, Kabat D, Bukowska-Strakova K, Gasińska A. Low-Dose Hyper-Radiosensitivity Is Not a Common Effect in Normal Asynchronous and G2-Phase Fibroblasts of Cancer Patients. Int J Radiat Oncol Biol Phys 2014; 88:369-76. [DOI: 10.1016/j.ijrobp.2013.10.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 10/21/2013] [Accepted: 10/22/2013] [Indexed: 11/17/2022]
|
49
|
El-Saghire H, Vandevoorde C, Ost P, Monsieurs P, Michaux A, De Meerleer G, Baatout S, Thierens H. Intensity modulated radiotherapy induces pro-inflammatory and pro-survival responses in prostate cancer patients. Int J Oncol 2014; 44:1073-83. [PMID: 24435511 PMCID: PMC3977809 DOI: 10.3892/ijo.2014.2260] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 11/11/2013] [Indexed: 12/21/2022] Open
Abstract
Intensity modulated radiotherapy (IMRT) is one of the modern conformal radiotherapies that is widely used within the context of cancer patient treatment. It uses multiple radiation beams targeted to the tumor, however, large volumes of the body receive low doses of irradiation. Using γ-H2AX and global genome expression analysis, we studied the biological responses induced by low doses of ionizing radiation in prostate cancer patients following IMRT. By means of different bioinformatics analyses, we report that IMRT induced an inflammatory response via the induction of viral, adaptive, and innate immune signaling. In response to growth factors and immune-stimulatory signaling, positive regulation in the progression of cell cycle and DNA replication were induced. This denotes pro-inflammatory and pro-survival responses. Furthermore, double strand DNA breaks were induced in every patient 30 min after the treatment and remaining DNA repair and damage signaling continued after 18-24 h. Nine genes belonging to inflammatory responses (TLR3, SH2D1A and IL18), cell cycle progression (ORC4, SMC2 and CCDC99) and DNA damage and repair (RAD17, SMC6 and MRE11A) were confirmed by quantitative RT-PCR. This study emphasizes that the risk assessment of health effects from the out-of-field low doses during IMRT should be of concern, as these may increase the risk of secondary cancers and/or systemic inflammation.
Collapse
Affiliation(s)
- Houssein El-Saghire
- Radiobiology Unit, Molecular and Cellular Biology, Belgian Nuclear Research Centre (SCK·CEN), Mol, Belgium
| | | | - Piet Ost
- Department of Radiation Oncology, Ghent University Hospital, Gent, Belgium
| | - Pieter Monsieurs
- Radiobiology Unit, Molecular and Cellular Biology, Belgian Nuclear Research Centre (SCK·CEN), Mol, Belgium
| | - Arlette Michaux
- Radiobiology Unit, Molecular and Cellular Biology, Belgian Nuclear Research Centre (SCK·CEN), Mol, Belgium
| | - Gert De Meerleer
- Department of Radiation Oncology, Ghent University Hospital, Gent, Belgium
| | - Sarah Baatout
- Radiobiology Unit, Molecular and Cellular Biology, Belgian Nuclear Research Centre (SCK·CEN), Mol, Belgium
| | - Hubert Thierens
- Department of Basic Medical Sciences, Ghent University, Gent, Belgium
| |
Collapse
|
50
|
Alexander BM, Ligon KL, Wen PY. Enhancing radiation therapy for patients with glioblastoma. Expert Rev Anticancer Ther 2013; 13:569-81. [PMID: 23617348 DOI: 10.1586/era.13.44] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Radiation therapy has been the foundation of therapy following maximal surgical resection in patients with newly diagnosed glioblastoma for decades and the primary therapy for unresected tumors. Using the standard approach with radiation and temozolomide, however, outcomes are poor, and glioblastoma remains an incurable disease with the majority of recurrences and progression within the radiation treatment field. As such, there is much interest in elucidating the mechanisms of resistance to radiation therapy and in developing novel approaches to overcoming this treatment resistance.
Collapse
Affiliation(s)
- Brian M Alexander
- Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, 75 Francis Street, ASB1-L2, Boston, MA 02115, USA.
| | | | | |
Collapse
|