1
|
Lyng FM, Azzam EI. Abscopal Effects, Clastogenic Effects and Bystander Effects: 70 Years of Non-Targeted Effects of Radiation. Radiat Res 2024; 202:355-367. [PMID: 38986531 DOI: 10.1667/rade-24-00040.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/29/2024] [Indexed: 07/12/2024]
Abstract
In vitro and in vivo observations accumulated over several decades have firmly shown that the biological effects of ionizing radiation can spread from irradiated cells/tissues to non-targeted cells/tissues. Redox-modulated intercellular communication mechanisms that include a role for secreted factors and gap junctions, can mediate these non-targeted effects. Clearly, the expression of such effects and their transmission to progeny cells has implications for issues related to radiation protection. Their elucidation is also relevant towards enhancing the efficacy of cancer radiotherapy and reducing its impact on the development of normal tissue toxicities. In addition, the study of non-targeted effects is pertinent to our basic understanding of intercellular communications under conditions of oxidative stress. This review will trace the history of non-targeted effects of radiation starting with early reports of abscopal effects which described radiation induced effects in tissues distant from the site of radiation exposure. A related effect involved the production of clastogenic factors in plasma following irradiation which can induce chromosome damage in unirradiated cells. Despite these early reports suggesting non-targeted effects of radiation, the classical paradigm that a direct deposition of energy in the nucleus was required still dominated. This paradigm was challenged by papers describing radiation induced bystander effects. This review will cover mechanisms of radiation-induced bystander effects and the potential impacts on radiation protection and radiation therapy.
Collapse
Affiliation(s)
- Fiona M Lyng
- Radiation and Environmental Science Centre, FOCAS Research Institute
- School of Physics, Clinical and Optometric Sciences, Technological University Dublin, Dublin, Ireland
| | - Edouard I Azzam
- Department of Radiology, Rutgers New Jersey Medical School Cancer Center, Newark, New Jersey
| |
Collapse
|
2
|
Radiotherapy Side Effects: Comprehensive Proteomic Study Unraveled Neural Stem Cell Degenerative Differentiation upon Ionizing Radiation. Biomolecules 2022; 12:biom12121759. [PMID: 36551187 PMCID: PMC9775306 DOI: 10.3390/biom12121759] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/11/2022] [Accepted: 11/15/2022] [Indexed: 11/29/2022] Open
Abstract
Cranial radiation therapy is one of the most effective treatments for childhood brain cancers. Despite the ameliorated survival rate of juvenile patients, radiation exposure-induced brain neurogenic region injury could markedly impair patients' cognitive functions and even their quality of life. Determining the mechanism underlying neural stem cells (NSCs) response to irradiation stress is a crucial therapeutic strategy for cognitive impairment. The present study demonstrated that X-ray irradiation arrested NSCs' cell cycle and impacted cell differentiation. To further characterize irradiation-induced molecular alterations in NSCs, two-dimensional high-resolution mass spectrometry-based quantitative proteomics analyses were conducted to explore the mechanism underlying ionizing radiation's influence on stem cell differentiation. We observed that ionizing radiation suppressed intracellular protein transport, neuron projection development, etc., particularly in differentiated cells. Redox proteomics was performed for the quantification of cysteine thiol modifications in order to profile the oxidation-reduction status of proteins in stem cells that underwent ionizing radiation treatment. Via conjoint screening of protein expression abundance and redox status datasets, several significantly expressed and oxidized proteins were identified in differentiating NSCs subjected to X-ray irradiation. Among these proteins, succinate dehydrogenase [ubiquinone] flavoprotein subunit, mitochondrial (sdha) and the acyl carrier protein, mitochondrial (Ndufab1) were highly related to neurodegenerative diseases such as Parkinson's disease, Alzheimer's disease, and Huntington's disease, illustrating the dual-character of NSCs in cell differentiation: following exposure to ionizing radiation, the normal differentiation of NSCs was compromised, and the upregulated oxidized proteins implied a degenerative differentiation trajectory. These findings could be integrated into research on neurodegenerative diseases and future preventive strategies.
Collapse
|
3
|
Nakaya G, Sakagami H, Koga-Ogawa Y, Shiroto A, Nobesawa T, Ueda D, Nakatani S, Kobata K, Iijima Y, Tone S, David-Gonzalez A, Garcia-Contreras R, Tomomura M, Kito S, Tamura N, Takeshima H. Augmentation of Neurotoxicity of Anticancer Drugs by X-Ray Irradiation. In Vivo 2021; 34:1009-1016. [PMID: 32354886 DOI: 10.21873/invivo.11869] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 01/28/2020] [Accepted: 01/30/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND In order to investigate the combination effect of anticancer drugs and X-ray irradiation on neurotoxic side-effects (neurotoxicity), a method that provides homogeneously X-ray-irradiated cells was newly established. MATERIALS AND METHODS PC12 cell suspension was irradiated by X-ray (0.5 Gy) in serum-supplemented medium, immediately inoculated into 96-microwell plates and incubated overnight. The medium was replaced with fresh serum-depleted medium containing 50 ng/ml nerve growth factor to induce differentiation toward nerve-like cells with characteristic neurites according to the overlay method without changing the medium. The differentiated cells were treated by anticancer drugs as well as antioxidants, oxaliplatin or bortezomib, and the viable cell number was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide method. RESULTS Antioxidants and anticancer drugs were cytotoxic to differentiating PC12 cells. Combination of anticancer drugs and X-ray irradiation slightly reduced cell viability. CONCLUSION The present 'population irradiation method' may be useful for the investigation of the combination effect of X-ray irradiation and any pharmaceutical drug.
Collapse
Affiliation(s)
- Giichirou Nakaya
- Faculty of Health Sciences, Nihon Institute of Medical Science, Saitama, Japan
| | | | - Yukari Koga-Ogawa
- Faculty of Health Sciences, Nihon Institute of Medical Science, Saitama, Japan
| | - Akiyoshi Shiroto
- Faculty of Health Sciences, Nihon Institute of Medical Science, Saitama, Japan
| | - Tadamasa Nobesawa
- Faculty of Health Sciences, Nihon Institute of Medical Science, Saitama, Japan
| | - Daisuke Ueda
- Faculty of Health Sciences, Nihon Institute of Medical Science, Saitama, Japan
| | - Sachie Nakatani
- Graduate School of Pharmaceutical Sciences, Josai University, Saitama, Japan
| | - Kenji Kobata
- Graduate School of Pharmaceutical Sciences, Josai University, Saitama, Japan
| | - Yosuke Iijima
- Saitama Medical Center, Saitama Medical University, Saitama, Japan
| | - Shigenobu Tone
- Graduate School of Science and Engineering, Tokyo Denki University, Saitama, Japan
| | - Angel David-Gonzalez
- National School of Higher Education, Leon Unit, National Autonomous University of Mexico, Mexico City, Mexico
| | - Rene Garcia-Contreras
- National School of Higher Education, Leon Unit, National Autonomous University of Mexico, Mexico City, Mexico
| | | | - Shinji Kito
- Meikai University School of Dentistry, Saitama, Japan
| | | | | |
Collapse
|
4
|
Mukherjee S, Chakraborty A. Radiation-induced bystander phenomenon: insight and implications in radiotherapy. Int J Radiat Biol 2019; 95:243-263. [PMID: 30496010 DOI: 10.1080/09553002.2019.1547440] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Sharmi Mukherjee
- Stress biology Lab, UGC-DAE Consortium for Scientific Research, Kolkata Centre, Kolkata, West Bengal, India
| | - Anindita Chakraborty
- Stress biology Lab, UGC-DAE Consortium for Scientific Research, Kolkata Centre, Kolkata, West Bengal, India
| |
Collapse
|
5
|
Gupta K, Burns TC. Radiation-Induced Alterations in the Recurrent Glioblastoma Microenvironment: Therapeutic Implications. Front Oncol 2018; 8:503. [PMID: 30467536 PMCID: PMC6236021 DOI: 10.3389/fonc.2018.00503] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/15/2018] [Indexed: 01/19/2023] Open
Abstract
Glioblastoma (GBM) is uniformly fatal with a median survival of just over 1 year, despite best available treatment including radiotherapy (RT). Impacts of prior brain RT on recurrent tumors are poorly understood, though increasing evidence suggests RT-induced changes in the brain microenvironment contribute to recurrent GBM aggressiveness. The tumor microenvironment impacts malignant cells directly and indirectly through stromal cells that support tumor growth. Changes in extracellular matrix (ECM), abnormal vasculature, hypoxia, and inflammation have been reported to promote tumor aggressiveness that could be exacerbated by prior RT. Prior radiation may have long-term impacts on microglia and brain-infiltrating monocytes, leading to lasting alterations in cytokine signaling and ECM. Tumor-promoting CNS injury responses are recapitulated in the tumor microenvironment and augmented following prior radiation, impacting cell phenotype, proliferation, and infiltration in the CNS. Since RT is vital to GBM management, but substantially alters the tumor microenvironment, we here review challenges, knowledge gaps, and therapeutic opportunities relevant to targeting pro-tumorigenic features of the GBM microenvironment. We suggest that insights from RT-induced changes in the tumor microenvironment may provide opportunities to target mechanisms, such as cellular senescence, that may promote GBM aggressiveness amplified in previously radiated microenvironment.
Collapse
Affiliation(s)
- Kshama Gupta
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
| | - Terry C Burns
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
6
|
Significance and nature of bystander responses induced by various agents. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2017; 773:104-121. [DOI: 10.1016/j.mrrev.2017.05.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 05/05/2017] [Indexed: 02/07/2023]
|
7
|
Tomita M, Matsumoto H, Funayama T, Yokota Y, Otsuka K, Maeda M, Kobayashi Y. Nitric oxide-mediated bystander signal transduction induced by heavy-ion microbeam irradiation. LIFE SCIENCES IN SPACE RESEARCH 2015; 6:36-43. [PMID: 26256626 DOI: 10.1016/j.lssr.2015.06.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 06/17/2015] [Accepted: 06/22/2015] [Indexed: 06/04/2023]
Abstract
In general, a radiation-induced bystander response is known to be a cellular response induced in non-irradiated cells after receiving bystander signaling factors released from directly irradiated cells within a cell population. Bystander responses induced by high-linear energy transfer (LET) heavy ions at low fluence are an important health problem for astronauts in space. Bystander responses are mediated via physical cell-cell contact, such as gap-junction intercellular communication (GJIC) and/or diffusive factors released into the medium in cell culture conditions. Nitric oxide (NO) is a well-known major initiator/mediator of intercellular signaling within culture medium during bystander responses. In this study, we investigated the NO-mediated bystander signal transduction induced by high-LET argon (Ar)-ion microbeam irradiation of normal human fibroblasts. Foci formation by DNA double-strand break repair proteins was induced in non-irradiated cells, which were co-cultured with those irradiated by high-LET Ar-ion microbeams in the same culture plate. Foci formation was suppressed significantly by pretreatment with an NO scavenger. Furthermore, NO-mediated reproductive cell death was also induced in bystander cells. Phosphorylation of NF-κB and Akt were induced during NO-mediated bystander signaling in the irradiated and bystander cells. However, the activation of these proteins depended on the incubation time after irradiation. The accumulation of cyclooxygenase-2 (COX-2), a downstream target of NO and NF-κB, was observed in the bystander cells 6 h after irradiation but not in the directly irradiated cells. Our findings suggest that Akt- and NF-κB-dependent signaling pathways involving COX-2 play important roles in NO-mediated high-LET heavy-ion-induced bystander responses. In addition, COX-2 may be used as a molecular marker of high-LET heavy-ion-induced bystander cells to distinguish them from directly irradiated cells, although this may depend on the time after irradiation.
Collapse
Affiliation(s)
- Masanori Tomita
- Radiation Safety Research Center, Central Research Institute of Electric Power Industry, 2-11-1 Iwado Kita, Komae, Tokyo 201-8511, Japan.
| | - Hideki Matsumoto
- Division of Oncology, Biomedical Imaging Research Center, University of Fukui, 23-3 Matsuoka-Shimoaitsuki, Eiheiji-cho, Fukui 910-1193, Japan
| | - Tomoo Funayama
- Microbeam Radiation Biology Group, Radiation Biology Research Division, Quantum Beam Science Center, Japan Atomic Energy Agency, 1233 Watanuki, Takasaki, Gunma 370-1292, Japan
| | - Yuichiro Yokota
- Microbeam Radiation Biology Group, Radiation Biology Research Division, Quantum Beam Science Center, Japan Atomic Energy Agency, 1233 Watanuki, Takasaki, Gunma 370-1292, Japan
| | - Kensuke Otsuka
- Radiation Safety Research Center, Central Research Institute of Electric Power Industry, 2-11-1 Iwado Kita, Komae, Tokyo 201-8511, Japan
| | - Munetoshi Maeda
- Radiation Safety Research Center, Central Research Institute of Electric Power Industry, 2-11-1 Iwado Kita, Komae, Tokyo 201-8511, Japan; Proton Medical Research Group, Research and Development Department, The Wakasa Wan Energy Research Center, 64-52-1 Nagatani, Tsuruga-shi, Fukui 914-0192, Japan
| | - Yasuhiko Kobayashi
- Microbeam Radiation Biology Group, Radiation Biology Research Division, Quantum Beam Science Center, Japan Atomic Energy Agency, 1233 Watanuki, Takasaki, Gunma 370-1292, Japan
| |
Collapse
|
8
|
Yokota Y, Funayama T, Mutou-Yoshihara Y, Ikeda H, Kobayashi Y. The bystander cell-killing effect mediated by nitric oxide in normal human fibroblasts varies with irradiation dose but not with radiation quality. Int J Radiat Biol 2015; 91:383-8. [DOI: 10.3109/09553002.2015.1021960] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
9
|
Dong C, He M, Tu W, Konishi T, Liu W, Xie Y, Dang B, Li W, Uchihori Y, Hei TK, Shao C. The differential role of human macrophage in triggering secondary bystander effects after either gamma-ray or carbon beam irradiation. Cancer Lett 2015; 363:92-100. [PMID: 25896631 DOI: 10.1016/j.canlet.2015.04.013] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 03/21/2015] [Accepted: 04/14/2015] [Indexed: 12/30/2022]
Abstract
The abscopal effect could be an underlying factor in evaluating prognosis of radiotherapy. This study established an in vitro system to examine whether tumor-generated bystander signals could be transmitted by macrophages to further trigger secondary cellular responses after different irradiations, where human lung cancer NCI-H446 cells were irradiated with either γ-rays or carbon ions and co-cultured with human macrophage U937 cells, then these U937 cells were used as a bystander signal transmitter and co-cultured with human bronchial epithelial cells BEAS-2B. Results showed that U937 cells were only activated by γ-irradiated NCI-H446 cells so that the secondary injuries in BEAS-2B cells under carbon ion irradiation were weaker than γ-rays. Both TNF-α and IL-1α were involved in the γ-irradiation induced secondary bystander effect but only TNF-α contributed to the carbon ion induced response. Further assay disclosed that IL-1α but not TNF-α was largely responsible for the activation of macrophages and the formation of micronucleus in BEAS-2B cells. These data suggest that macrophages could transfer secondary bystander signals and play a key role in the secondary bystander effect of photon irradiation, while carbon ion irradiation has conspicuous advantage due to its reduced secondary injury.
Collapse
Affiliation(s)
- Chen Dong
- Institute of Radiation Medicine, Fudan University, No. 2094 Xie-Tu Road, Shanghai 200032, China
| | - Mingyuan He
- Institute of Radiation Medicine, Fudan University, No. 2094 Xie-Tu Road, Shanghai 200032, China; Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Wenzhi Tu
- Institute of Radiation Medicine, Fudan University, No. 2094 Xie-Tu Road, Shanghai 200032, China
| | - Teruaki Konishi
- Research Development and Support Center, National Institute of Radiological Sciences, Inage, Chiba 263-8555, Japan
| | - Weili Liu
- Institute of Radiation Medicine, Fudan University, No. 2094 Xie-Tu Road, Shanghai 200032, China
| | - Yuexia Xie
- Institute of Radiation Medicine, Fudan University, No. 2094 Xie-Tu Road, Shanghai 200032, China; Central Laboratory, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Bingrong Dang
- Institute of Modern Physics, Chinese Academy of Sciences, No. 509 Nanchang Road, Lanzhou 730000, China
| | - Wenjian Li
- Institute of Modern Physics, Chinese Academy of Sciences, No. 509 Nanchang Road, Lanzhou 730000, China
| | - Yukio Uchihori
- Research Development and Support Center, National Institute of Radiological Sciences, Inage, Chiba 263-8555, Japan
| | - Tom K Hei
- Department of Radiation Oncology, Columbia University Medical Center, New York, NY 10032, USA
| | - Chunlin Shao
- Institute of Radiation Medicine, Fudan University, No. 2094 Xie-Tu Road, Shanghai 200032, China.
| |
Collapse
|
10
|
Dong C, He M, Ren R, Xie Y, Yuan D, Dang B, Li W, Shao C. Role of the MAPK pathway in the observed bystander effect in lymphocytes co-cultured with macrophages irradiated with γ-rays or carbon ions. Life Sci 2015; 127:19-25. [PMID: 25748424 DOI: 10.1016/j.lfs.2015.02.017] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 02/12/2015] [Accepted: 02/13/2015] [Indexed: 01/20/2023]
Abstract
AIMS The radiation-induced bystander effect (RIBE) has potential implications in cancer risks from space particle radiation; however, the mechanisms underlying RIBE are unclear. The role of the MAPK pathway in the RIBEs of different linear energy transfer (LET) was investigated. MAIN METHODS Human macrophage U937 cells were irradiated with γ-rays or carbon ions and then co-cultured with nonirradiated HMy2.CIR (HMy) lymphocytes for different periods. The activation of MAPK proteins and the generation of intracellular nitric oxide (NO) and reactive oxygen species (ROS) in the irradiated U937 cells were measured. Micronuclei (MN) formation in the HMy cells was applied to evaluate the bystander damage. Some U937 cells were pretreated with different MAPK inhibitors before irradiation. KEY FINDINGS Additional MN formation was induced in the HMy cells after co-culturing with irradiated U937 cells, and the yield of this bystander MN formation was dependent on the co-culture period with γ-ray irradiation but remained high after 1h of co-culture with carbon irradiation. Further investigations disclosed that the time response of the RIBEs had a relationship with LET, where ERK played a different role from JNK and p38 in regulating RIBEs by regulating the generation of the bystander signaling factors NO and ROS. SIGNIFICANCE The finding that the RIBE of high-LET radiation could persist for a much longer period than that of γ-rays implies that particle radiation during space flight could have a high risk of long-term harmful effects. An appropriate intervention targeting the MAPK pathway may have significant implications in reducing this risk.
Collapse
Affiliation(s)
- Chen Dong
- Institute of Radiation Medicine, Fudan University, No. 2094 Xie-Tu Road, Shanghai 200032, China
| | - Mingyuan He
- Institute of Radiation Medicine, Fudan University, No. 2094 Xie-Tu Road, Shanghai 200032, China
| | - Ruiping Ren
- Institute of Radiation Medicine, Fudan University, No. 2094 Xie-Tu Road, Shanghai 200032, China
| | - Yuexia Xie
- Institute of Radiation Medicine, Fudan University, No. 2094 Xie-Tu Road, Shanghai 200032, China
| | - Dexiao Yuan
- Institute of Radiation Medicine, Fudan University, No. 2094 Xie-Tu Road, Shanghai 200032, China
| | - Bingrong Dang
- Institute of Modern Physics, Chinese Academy of Sciences, No. 509 Nanchang Road, Lanzhou 730000, China
| | - Wenjian Li
- Institute of Modern Physics, Chinese Academy of Sciences, No. 509 Nanchang Road, Lanzhou 730000, China
| | - Chunlin Shao
- Institute of Radiation Medicine, Fudan University, No. 2094 Xie-Tu Road, Shanghai 200032, China.
| |
Collapse
|
11
|
Xiao L, Liu W, Li J, Xie Y, He M, Fu J, Jin W, Shao C. Irradiated U937 cells trigger inflammatory bystander responses in human umbilical vein endothelial cells through the p38 pathway. Radiat Res 2014; 182:111-21. [PMID: 24960416 DOI: 10.1667/rr13736.1] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Radiation-induced bystander effects are a well-known phenomenon that are observed when treating cancer and other diseases after radiotherapy, and even after occupational exposure to radiation. However, little is known about the crosstalk between irradiated macrophages and endothelial cells that line the circulatory system, which may play a role in the development of atherosclerosis. In the current study, we found that the expression of inducible nitric oxide synthase (iNOS) and the intracellular level of nitric oxide (NO) in gamma-irradiated U937 macrophage cells were significantly increased. When human umbilical vein endothelial cells (HUVECs) were co-cultured with gamma-irradiated U937 cells, additional micronuclei (MN) and apoptosis were induced so that the plating efficiency of the bystander HUVECs decreased and P38 was overexpressed in the bystander HUVECs cells. In addition, the contents of vascular cell adhesion molecule 1 (VCAM-1) and the activities of matrix metalloproteinase-9 (MMP-9) in the culture medium of bystander HUVECs were increased. Furthermore, during cell co-culture the adhesive ability of irradiated U937 cells to the bystander HUVECs increased. When U937 cells were treated with 500 μM S-methylisothiourea sulfate (SMT) (iNOS inhibitor) before irradiation, and HUVECs were treated with 10 μM SB203580 (p38 inhibitor) before cell co-culture or treated with 20 μM c-PTIO (NO scavenger) in the co-culture medium, the bystander micronuclei and the amounts of VCAM-1 and MMP-9 in the medium of bystander HUVECs were diminished, and the ability of irradiated U937 cells adhering to HUVECs was also reduced, while the plating efficiency of bystander HUVECs partially recovered. These results demonstrated that irradiated U937 cells appear to release nitric oxide and thereby further trigger apoptosis and inflammatory responses in the bystander HUVECs through a p38-dependent pathway.
Collapse
Affiliation(s)
- Linlin Xiao
- a Institute of Radiation Medicine, Fudan University, Shanghai 200032, China; and
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Seth I, Schwartz JL, Stewart RD, Emery R, Joiner MC, Tucker JD. Neutron exposures in human cells: bystander effect and relative biological effectiveness. PLoS One 2014; 9:e98947. [PMID: 24896095 PMCID: PMC4045982 DOI: 10.1371/journal.pone.0098947] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 05/09/2014] [Indexed: 11/19/2022] Open
Abstract
Bystander effects have been observed repeatedly in mammalian cells following photon and alpha particle irradiation. However, few studies have been performed to investigate bystander effects arising from neutron irradiation. Here we asked whether neutrons also induce a bystander effect in two normal human lymphoblastoid cell lines. These cells were exposed to fast neutrons produced by targeting a near-monoenergetic 50.5 MeV proton beam at a Be target (17 MeV average neutron energy), and irradiated-cell conditioned media (ICCM) was transferred to unirradiated cells. The cytokinesis-block micronucleus assay was used to quantify genetic damage in radiation-naïve cells exposed to ICCM from cultures that received 0 (control), 0.5, 1, 1.5, 2, 3 or 4 Gy neutrons. Cells grown in ICCM from irradiated cells showed no significant increase in the frequencies of micronuclei or nucleoplasmic bridges compared to cells grown in ICCM from sham irradiated cells for either cell line. However, the neutron beam has a photon dose-contamination of 5%, which may modulate a neutron-induced bystander effect. To determine whether these low doses of contaminating photons can induce a bystander effect, cells were irradiated with cobalt-60 at doses equivalent to the percent contamination for each neutron dose. No significant increase in the frequencies of micronuclei or bridges was observed at these doses of photons for either cell line when cultured in ICCM. As expected, high doses of photons induced a clear bystander effect in both cell lines for micronuclei and bridges (p<0.0001). These data indicate that neutrons do not induce a bystander effect in these cells. Finally, neutrons had a relative biological effectiveness of 2.0 ± 0.13 for micronuclei and 5.8 ± 2.9 for bridges compared to cobalt-60. These results may be relevant to radiation therapy with fast neutrons and for regulatory agencies setting standards for neutron radiation protection and safety.
Collapse
Affiliation(s)
- Isheeta Seth
- Department of Biological Sciences, Wayne State University, Detroit, Michigan, United States of America
| | - Jeffrey L. Schwartz
- Department of Radiation Oncology, School of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Robert D. Stewart
- Department of Radiation Oncology, School of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Robert Emery
- Department of Radiation Oncology, School of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Michael C. Joiner
- Department of Radiation Oncology, Wayne State University, Detroit, Michigan, United States of America
| | - James D. Tucker
- Department of Biological Sciences, Wayne State University, Detroit, Michigan, United States of America
- * E-mail:
| |
Collapse
|
13
|
|
14
|
Suzuki K, Yamashita S. Radiation-Induced Bystander Response: Mechanism and Clinical Implications. Adv Wound Care (New Rochelle) 2014; 3:16-24. [PMID: 24761341 DOI: 10.1089/wound.2013.0468] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 06/21/2013] [Indexed: 01/05/2023] Open
Abstract
Significance: Absorption of energy from ionizing radiation (IR) to the genetic material in the cell gives rise to damage to DNA in a dose-dependent manner. There are two types of DNA damage; by a high dose (causing acute or deterministic effects) and by a low dose (related to chronic or stochastic effects), both of which induce different health effects. Among radiation effects, acute cutaneous radiation syndrome results from cell killing as a consequence of high-dose exposure. Recent advances: Recent advances in radiation biology and oncology have demonstrated that bystander effects, which are emerged in cells that have never been exposed, but neighboring irradiated cells, are also involved in radiation effects. Bystander effects are now recognized as an indispensable component of tissue response related to deleterious effects of IR. Critical issues: Evidence has indicated that nonapoptotic premature senescence is commonly observed in various tissues and organs. Senesced cells were found to secrete various proteins, including cytokines, chemokines, and growth factors, most of which are equivalent to those identified as bystander factors. Secreted factors could trigger cell proliferation, angiogenesis, cell migration, inflammatory response, etc., which provide a tissue microenvironment assisting tissue repair and remodeling. Future directions: Understandings of the mechanisms and physiological relevance of radiation-induced bystander effects are quite essential for the beneficial control of wound healing and care. Further studies should extend our knowledge of the mechanisms of bystander effects and mode of cell death in response to IR.
Collapse
Affiliation(s)
- Keiji Suzuki
- Department of Radiation Medical Sciences, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Shunichi Yamashita
- Department of Radiation Medical Sciences, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
15
|
Zhang J, Xie Y, Xu Y, Shao C. Suppression of endogenous hydrogen sulfide contributes to the radiation-induced bystander effects on hypoxic HepG2 cells. Radiat Res 2012; 178:395-402. [PMID: 23020085 DOI: 10.1667/rr2967.1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Radiation-induced bystander effects may have important implications in radiotherapy, but it is still not well known if radiation-induced bystander effects can be triggered in hypoxic tumor cells and what are the related bystander signals. Using human hepatoma cells of HepG2, the present study found that micronuclei (MN) could be induced in the nonirradiated cells after treatment with conditioned medium (CM) harvested from irradiated cells under hypoxic conditions. Bystander effects were diminished when the irradiated cells were pretreated with sodium hydrosulfide (NaHS, an exogenous H(2)S donor) (≤100 μM). However, the bystander effects were increased when the irradiated cells were pretreated with an inhibitor of cystathionine β-synthase (CBS) and cystathionine γ-lyase (CSE), the synthases of endogenous hydrogen sulfide (H(2)S). Western blotting showed that the expressions of CSE and CBS were increased in the irradiated hypoxic cells, but were reduced in the CM treated bystander cells. The ratio of Bcl-2/Bax, a molecular marker of apoptosis, decreased with CM treatment time. However, the activity of caspase-3 increased in the hypoxic bystander cells, and this could be regulated by both NaHS and the inhibitor of endogenous H(2)S. These results demonstrate that under hypoxic conditions irradiated hepatoma cells induce bystander responses by depressing the generation of endogenous H(2)S and altering Bcl-2/Bax ratios as well as caspase-3 dependent damage in the bystander cells.
Collapse
Affiliation(s)
- Jianghong Zhang
- Institute of Radiation Medicine, Fudan University, Shanghai 200032, China
| | | | | | | |
Collapse
|
16
|
Cytochrome-c mediated a bystander response dependent on inducible nitric oxide synthase in irradiated hepatoma cells. Br J Cancer 2012; 106:889-95. [PMID: 22274409 PMCID: PMC3305951 DOI: 10.1038/bjc.2012.9] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Radiation-induced bystander effect (RIBE) has important implication in tumour radiotherapy, but the bystander signals are still not well known. METHODS The role of cytochrome-c (cyt-c) and free radicals in RIBE on human hepatoma cells HepG2 was investigated by detecting the formation of bystander micronuclei (MN) and the generation of endogenous cyt-c, inducible nitric oxide (NO) synthase (iNOS), NO, and reactive oxygen species (ROS) molecules. RESULTS When HepG2 cells were cocultured with an equal number of irradiated HepG2 cells, the yield of MN in the nonirradiated bystander cells was increased in a manner depended on radiation dose and cell coculture time, but it was diminished when the cells were treated with cyclosporin A (CsA), an inhibitor of cyt-c release. Meanwhile the CsA treatment inhibited radiation-induced NO but not ROS. Both of the depressed bystander effect and NO generation in the CsA-treated cells were reversed when 5 μM cyt-c was added in the cell coculture medium. But these exogenous cyt-c-mediated overproductions of NO and bystander MN were abolished when the cells were pretreated with s-methylisothiourea sulphate, an iNOS inhibitor. CONCLUSION Radiation-induced cyt-c has a profound role in regulating bystander response through an iNOS-triggered NO signal but not ROS in HepG2 cells.
Collapse
|
17
|
Blyth BJ, Sykes PJ. Radiation-induced bystander effects: what are they, and how relevant are they to human radiation exposures? Radiat Res 2011; 176:139-57. [PMID: 21631286 DOI: 10.1667/rr2548.1] [Citation(s) in RCA: 156] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The term radiation-induced bystander effect is used to describe radiation-induced biological changes that manifest in unirradiated cells remaining within an irradiated cell population. Despite their failure to fit into the framework of classical radiobiology, radiation-induced bystander effects have entered the mainstream and have become established in the radiobiology vocabulary as a bona fide radiation response. However, there is still no consensus on a precise definition of radiation-induced bystander effects, which currently encompasses a number of distinct signal-mediated effects. These effects are classified here into three classes: bystander effects, abscopal effects and cohort effects. In this review, the data have been evaluated to define, where possible, various features specific to radiation-induced bystander effects, including their timing, range, potency and dependence on dose, dose rate, radiation quality and cell type. The weight of evidence supporting these defining features is discussed in the context of bystander experimental systems that closely replicate realistic human exposure scenarios. Whether the manifestation of bystander effects in vivo is intrinsically limited to particular radiation exposure scenarios is considered. The conditions under which radiation-induced bystander effects are induced in vivo will ultimately determine their impact on radiation-induced carcinogenic risk.
Collapse
Affiliation(s)
- Benjamin J Blyth
- Haematology and Genetic Pathology, Flinders University, Bedford Park, South Australia 5042, Australia
| | | |
Collapse
|
18
|
Coulter JA, Page NL, Worthington J, Robson T, Hirst DG, McCarthy HO. Transcriptional regulation of inducible nitric oxide synthase gene therapy: targeting early stage and advanced prostate cancer. J Gene Med 2010; 12:755-65. [PMID: 20821746 DOI: 10.1002/jgm.1495] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Using the tumour type specific human osteocalcin (hOC) promoter, we have previously reported strong promoter activation in hormone independent prostate cancer cells in vitro. In the present study, we present a comparative study of the tissue specific promoter prostate specific membrane antigen (PSMA), and the tumour-type specific hOC promoter driving the inducible nitric oxide synthase (iNOS) transgene using both in vitro and in vivo models. METHODS In vitro cytotoxicity was assessed by clonogenic assay. Quantification of nitric oxide expression was determined by the Griess test. In vivo anti-tumour efficacy was determined by tumour growth delay following direct intra-tumoural injection of the constructs into PC3 xenografts. In addition, tumours were dissected post mortem and examined for morphological differences as well as changes in apoptotic protein expression. RESULTS PSMA/iNOS produced cytotoxicity in both androgen dependant and independent cell lines. Nitric oxide quantification confirmed that increased cytotoxicity was directly associated with nitric oxide production. Tumour growth delays were observed in all groups treated with the iNOS-expressing constructs ranging from 10.7 days for the hOC/iNOS single dose treatment group to a maximum of 52.2 days for the hOC/iNOS multiple dose group. Intra-tumoural assessment of iNOS and cleaved poly (ADP-ribose) polymerase protein expression demonstrated a significant up-regulation of both proteins, indicating cytotoxicity mediated through the intrinsic apoptotic pathway. CONCLUSIONS Highly significant tumour growth delay coupled with no detrimental side-effects were observed following treatment with the PSMA/iNOS and hOC/iNOS constructs. We consider that these findings provide a basis for the development of systemically delivered PSMA/iNOS or hOC/iNOS targeting early stage and advanced prostate cancer.
Collapse
Affiliation(s)
- Jonathan A Coulter
- School of Pharmacy, McClay Research Centre, Queen's University, Belfast, Ireland, UK
| | | | | | | | | | | |
Collapse
|
19
|
Maeda M, Tomita M, Usami N, Kobayashi K. Bystander cell death is modified by sites of energy deposition within cells irradiated with a synchrotron X-ray microbeam. Radiat Res 2010; 174:37-45. [PMID: 20681797 DOI: 10.1667/rr2086.1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Radiation-induced bystander effects are the biological responses exhibited by cells adjacent to cells that have been traversed by charged particles. Using a synchrotron X-ray microbeam irradiation system, we irradiated five cells in two different ways: by targeting the nuclei with 10 microm x 10-microm 5.35 keV X-ray beams and by irradiating the whole cells with 50 microm x 50-microm 5.35 keV X-ray beams. Then we measured the clonogenic survival of the bystander cells. When only the nuclei were irradiated, a parabolic enhancement of bystander cell death was observed in a dose-dependent manner in the low-dose region around 1 Gy. In contrast, the surviving fraction of bystander cells decreased monotonically when whole cells were irradiated. Addition of carboxy-PTIO, a specific scavenger of nitric oxide (NO), suppressed bystander cell death in both cases. These results indicate that NO is a mediator in the induction of the parabolic and monotonic types of bystander cell death. Moreover, from the spatial analysis, we found that the parabolic type of bystander cell death was induced primarily within 1 mm of irradiated cells. Our findings demonstrate that the induction of bystander cell death depends on the sites of energy deposition in irradiated cells.
Collapse
Affiliation(s)
- Munetoshi Maeda
- Radiation Safety Research Center, Nuclear Technology Research Laboratory, Central Research Institute of Electric Power Industry, CRIEPI, Komae-shi, Tokyo 201-8511, Japan.
| | | | | | | |
Collapse
|
20
|
Tomita M, Maeda M, Maezawa H, Usami N, Kobayashi K. Bystander cell killing in normal human fibroblasts is induced by synchrotron X-ray microbeams. Radiat Res 2010; 173:380-5. [PMID: 20199223 DOI: 10.1667/rr1995.1] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Abstract The radiation-induced bystander response is defined as a response in cells that have not been directly targeted by radiation but that are in the neighborhood of cells that have been directly exposed. In the work described here, it is shown that bystander cell killing of normal human fibroblast WI-38 cells was induced by synchrotron microbeam X radiation. Cell nuclei in confluent WI-38 cells were irradiated with the microbeam. All of the cells on the dish were harvested and plated 24 h after irradiation. It was found that the bystander cell killing effect showed a parabolic relationship to the radiation dose when five cells were irradiated. At doses above 1.9 Gy, the surviving fraction increased to approximately 1.0. This suggests that induction of bystander cell killing may require some type of activity in the targeted cells, because the dose resulting in 37% cell survival was about 2.0 Gy. Bystander cell killing was suppressed by a pretreatment with aminoguanidine [an inhibitor of inducible nitric oxide (NO) synthase] or carboxy-PTIO (a scavenger of NO). These results suggest that NO is the chief initiator/mediator of bystander cell killing induced by X-ray microbeams.
Collapse
Affiliation(s)
- Masanori Tomita
- Radiation Safety Research Center, Central Research Institute of Electric Power Industry, Tokyo, Japan.
| | | | | | | | | |
Collapse
|
21
|
Lyng FM, Maguire P, Kilmurray N, Mothersill C, Shao C, Folkard M, Prise KM. Apoptosis is initiated in human keratinocytes exposed to signalling factors from microbeam irradiated cells. Int J Radiat Biol 2009; 82:393-9. [PMID: 16846974 DOI: 10.1080/09553000600803904] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
PURPOSE There is now no doubt that bystander signalling from irradiated cells occurs and causes a variety of responses in cells not targeted by the ionizing track. However, the mechanisms underlying these processes are unknown and the relevance to radiotherapy and risk assessment remains controversial. Previous research by our laboratory has shown bystander effects in a human keratinocyte cell line, HPV-G cells, exposed to medium from gamma irradiated HPV-G cells. The aim of this work was to investigate if similar mechanisms to those identified in medium transfer experiments occurred in these HPV-G cells when they are in the vicinity of microbeam irradiated cells. Demonstration of a commonality of mechanisms would support the idea that the process is not artifactual. MATERIALS AND METHODS HPV-G cells were plated as two separate populations on mylar dishes. One population was directly irradiated using a charged particle microbeam (1 - 10 protons). The other population was not irradiated. Bystander factor-induced apoptosis was investigated in both populations following treatment by monitoring the levels of reactive oxygen species and mitochondrial membrane potential using fluorescent probes. Expression of the anti-apoptotic protein, bcl-2, and cytochrome c were determined, as well as apoptosis levels. RESULTS Microbeam irradiation induced increases in reactive oxygen species and decreases in mitochondrial membrane potential at 6 h post-exposure, increased expression of bcl-2 and cytochrome c release at 6.5 h and increased apoptosis at 24 h. CONCLUSION This study shows that similar bystander signalling pathways leading to apoptosis are induced following microbeam irradiation and following medium transfer. This demonstrates that the mechanisms involved are common across different radiation qualities and conditions and indicates that they may be relevant in vivo.
Collapse
Affiliation(s)
- Fiona M Lyng
- Radiation and Environmental Science Centre, Focas Institute, Dublin Institute of Technology, Dublin, Ireland.
| | | | | | | | | | | | | |
Collapse
|
22
|
Kanasugi Y, Hamada N, Wada S, Funayama T, Sakashita T, Kakizaki T, Kobayashi Y, Takakura K. Role of DNA-PKcs in the bystander effect after low- or high-LET irradiation. Int J Radiat Biol 2009; 83:73-80. [PMID: 17357428 DOI: 10.1080/09553000601121116] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE To investigate the role of the catalytic subunit of DNA-dependent protein kinase (DNA-PKcs) in the medium-mediated bystander effect for chromosomal aberrations induced by low-linear energy transfer (LET) X-rays and high-LET heavy ions in normal human fibroblast cells. MATERIALS AND METHODS The recipient cells were treated for 12 h with conditioned medium, which was harvested from donor cells at 24 h after exposure to 10 Gy of soft X-rays (5 keV/microm) and 20Ne ions (437 keV/microm), followed by analyses of chromosome aberrations in recipient cells with premature chromosome condensation methods. To examine the role of DNA-PKcs and nitric oxide (NO), cells were treated with its inhibitor LY294002 (LY) and its scavenger 2-(4-carboxyphenyl)-4,4,5,5-tetramethyl-imidazoline-1-oxyl-3-oxide (c-PTIO), respectively. RESULTS Increased frequency of chromosome aberrations in recipient cells treated with conditioned medium from irradiated but not from un-irradiated donor cells was observed which was independent of radiation type. Bystander induction of chromosome aberrations in recipient cells was mitigated when donor cells were treated with LY before irradiation and with c-PTIO after irradiation, and was enhanced when recipient cells were treated with LY before treatment of recipient cells with conditioned medium from irradiated donor cells. CONCLUSION Irradiated normal human cells secrete NO and other molecules which in turn transmit radiation signals to unirradiated bystander cells, leading to the induction of bystander chromosome aberrations partially repairable by DNA-PKcs-mediated DNA damage repair machinery, such as non-homologous end-joining repair pathways.
Collapse
Affiliation(s)
- Yuichi Kanasugi
- Physics Department, International Christian University, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Hamada N. The Bystander Response to Heavy-Ion Radiation: Intercellular Signaling Between Irradiated and Non-Irradiated Cells. ACTA ACUST UNITED AC 2009. [DOI: 10.2187/bss.23.195] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
24
|
Hamada N. Recent insights into the biological action of heavy-ion radiation. JOURNAL OF RADIATION RESEARCH 2009; 50:1-9. [PMID: 18838844 DOI: 10.1269/jrr.08070] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Biological effectiveness varies with the linear energy transfer (LET) of ionizing radiation. During cancer therapy or long-term interplanetary manned explorations, humans are exposed to high-LET energetic heavy ions that inactivate cells more effectively than low-LET photons like X-rays and gamma-rays. Recent biological studies have illustrated that heavy ions overcome tumor radioresistance caused by Bcl-2 overexpression, p53 mutations and intratumor hypoxia, and possess antiangiogenic and antimetastatic potential. Compared with heavy ions alone, the combination with chemical agents (a Bcl-2 inhibitor HA14-1, an anticancer drug docetaxel, and a halogenated pyrimidine analogue 5-iodo-2'-deoxyuridine) or hyperthermia further enhances tumor cell killing. Beer, its certain constituents, or melatonin ameliorate heavy ion-induced damage to normal cells. In addition to effects in cells directly targeted with heavy ions, there is mounting evidence for nontargeted biological effects in cells that have not themselves been directly irradiated. The bystander effect of heavy ions manifests itself as the loss of clonogenic potential, a transient apoptotic response, delayed p53 phosphorylation, alterations in gene expression profiles, and the elevated frequency of gene mutations, micronuclei and chromosome aberrations, which arise in nonirradiated cells having received signals from irradiated cells. Proposed mediating mechanisms involve gap junctional intercellular communication, reactive oxygen species and nitric oxide. This paper reviews briefly the current knowledge of the biological effects of heavy-ion irradiation with a focus on recent findings regarding its potential benefits for therapeutic use as well as on the bystander effect.
Collapse
Affiliation(s)
- Nobuyuki Hamada
- Department of Quantum Biology, Division of Bioregulatory Medicine, Gunma University Graduate School of Medicine, Gunma, Japan.
| |
Collapse
|
25
|
Shao C, Prise KM, Folkard M. Signaling factors for irradiated glioma cells induced bystander responses in fibroblasts. Mutat Res 2008; 638:139-145. [PMID: 17977565 DOI: 10.1016/j.mrfmmm.2007.09.007] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2007] [Revised: 09/16/2007] [Accepted: 09/19/2007] [Indexed: 05/25/2023]
Abstract
The aim of this study was to investigate the signaling factor and its pathway involved in the targeted irradiation-induced bystander response from glioblastoma cells to primary fibroblasts. After co-culturing with a glioblastoma T98G population where a fraction of cells had been individually irradiated with a precise number of helium particles, additional micronucleus (MN) were induced in the non-irradiated human fibroblasts AG01522 cells and its yield was independent of irradiation dose. This bystander MN induction was eliminated by treating the cells with either aminoguanidine (AG), an iNOS inhibitor, or anti-transforming growth factor-beta1 (anti-TGF-beta1). In addition, TGF-beta1 could be released from irradiated T98G cells but this release was inhibited by AG. In consistent, TGF-beta1 could also be induced from T98G cells treated with diethylamine nitric oxide (DEANO), a donor of nitric oxide (NO). Moreover, the effect of TGF-beta1 on bystander AG01522 cells was investigated. It was found that reactive oxygen species (ROS) and MN were induced in AG01522 cells after TGF-beta1 treatment. Our results indicate that, downstream of NO, TGF-beta1 plays an important role in the targeted T98G cells induced bystander response to AG0 cells by further causing DNA damage in vicinal fibroblasts through a ROS related pathway. This study may have implications for properly evaluating the secondary effects of radiotherapy.
Collapse
Affiliation(s)
- Chunlin Shao
- Institute of Radiation Medicine, Fudan University, No. 2094 Xie-Tu Road, Shanghai 200032, China.
| | | | | |
Collapse
|
26
|
Shao C, Folkard M, Prise KM. Role of TGF-beta1 and nitric oxide in the bystander response of irradiated glioma cells. Oncogene 2008; 27:434-40. [PMID: 17621264 PMCID: PMC3016606 DOI: 10.1038/sj.onc.1210653] [Citation(s) in RCA: 166] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2007] [Revised: 05/14/2007] [Accepted: 06/01/2007] [Indexed: 12/29/2022]
Abstract
The radiation-induced bystander effect (RIBE) increases the probability of cellular response and therefore has important implications for cancer risk assessment following low-dose irradiation and for the likelihood of secondary cancers after radiotherapy. However, our knowledge of bystander signaling factors, especially those having long half-lives, is still limited. The present study found that, when a fraction of cells within a glioblastoma population were individually irradiated with helium ions from a particle microbeam, the yield of micronuclei (MN) in the nontargeted cells was increased, but these bystander MN were eliminated by treating the cells with either aminoguanidine (an inhibitor of inducible nitric oxide (NO) synthase) or anti-transforming growth factor beta1 (anti-TGF-beta1), indicating that NO and TGF-beta1 are involved in the RIBE. Intracellular NO was detected in the bystander cells, and additional TGF-beta1 was detected in the medium from irradiated T98G cells, but it was diminished by aminoguanidine. Consistent with this, an NO donor, diethylamine nitric oxide (DEANO), induced TGF-beta1 generation in T98G cells. Conversely, treatment of cells with recombinant TGF-beta1 could also induce NO and MN in T98G cells. Treatment of T98G cells with anti-TGF-beta1 inhibited the NO production when only 1% of cells were targeted, but not when 100% of cells were targeted. Our results indicate that, downstream of radiation-induced NO, TGF-beta1 can be released from targeted T98G cells and plays a key role as a signaling factor in the RIBE by further inducing free radicals and DNA damage in the nontargeted bystander cells.
Collapse
Affiliation(s)
- C Shao
- Gray Cancer Institute, Mount Vernon Hospital, Northwood, Middlesex, UK
- Institute of Radiation Medicine, Fudan University, Shanghai, China
| | - M Folkard
- Gray Cancer Institute, Mount Vernon Hospital, Northwood, Middlesex, UK
| | - KM Prise
- Gray Cancer Institute, Mount Vernon Hospital, Northwood, Middlesex, UK
- Centre for Cancer Research & Cell Biology, Queen's University Belfast, Belfast, UK
| |
Collapse
|
27
|
Fukamoto K, Shirai K, Sakata T, Sakashita T, Funayama T, Hamada N, Wada S, Kakizaki T, Shimura S, Kobayashi Y, Kiguchi K. Development of the irradiation method for the first instar silkworm larvae using locally targeted heavy-ion microbeam. JOURNAL OF RADIATION RESEARCH 2007; 48:247-53. [PMID: 17327687 DOI: 10.1269/jrr.06066] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
To carry out the radio-microsurgery study using silkworm, Bombyx mori, we have already developed the specific irradiation systems for eggs and third to fifth instar larvae. In this study, a modified application consisting of the first instar silkworm larvae was further developed using heavy-ion microbeams. This system includes aluminum plates with holes specially designed to fix the first instar silkworm larvae during irradiation, and Mylar films were used to adjust energy deposited for planning radiation doses at certain depth. Using this system, the suppression of abnormal proliferation of epidermal cells in the knob mutant was examined. Following target irradiation of the knob-forming region at the first instar stage with 180-mum-diameter microbeam of 220 MeV carbon (12C) ions, larvae were reared to evaluate the effects of irradiation. The results indicated that the knob formation at the irradiated segment was specially suppressed in 5.9, 56.4, 66.7 and 73.6% of larvae irradiated with 120, 250, 400 and 600 Gy, respectively, but the other knob formations at the non-irradiated segments were not suppressed in either irradiation. Although some larva did not survive undesired non-targeted exposure, our present results indicate that this method would be useful to investigate the irradiation effect on a long developmental period of time. Moreover, our system could also be applied to other species by targeting tissues, or organs during development and metamorphosis in insect and animals.
Collapse
Affiliation(s)
- Kana Fukamoto
- Microbeam Radiation Biology Group, Japan Atomic Energy Agency, 1233 Watanuki-machi, Takasaki, Gunma 370-1292, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Wu H, Hada M, Meador J, Hu X, Rusek A, Cucinotta FA. Induction of micronuclei in human fibroblasts across the Bragg curve of energetic heavy ions. Radiat Res 2006; 166:583-9. [PMID: 17007550 DOI: 10.1667/rr0535.1] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2006] [Accepted: 06/23/2006] [Indexed: 11/03/2022]
Abstract
The space environment consists of a varying field of radiation particles including high-energy ions, with spacecraft shielding material providing the major protection to astronauts from harmful exposure. Unlike low-LEpsilonTau gamma or X rays, the presence of shielding does not always reduce the radiation risks for energetic charged-particle exposure. The dose delivered by the charged particle increases sharply as the particle approaches the end of its range, a position known as the Bragg peak. However, the Bragg curve does not necessarily represent the biological damage along the particle path since biological effects are influenced by the track structures of both primary and secondary particles. Therefore, the "biological Bragg curve" is dependent on the energy and the type of the primary particle and may vary for different biological end points. Here we report measurements of the biological response across the Bragg curve in human fibroblasts exposed to energetic silicon and iron ions in vitro at two different energies, 300 MeV/nucleon and 1 GeV/nucleon. A quantitative biological response curve generated for micronuclei per binucleated cell across the Bragg curve did not reveal an increased yield of micronuclei at the location of the Bragg peak. However, the ratio of mono- to binucleated cells, which indicates inhibition of cell progression, increased at the Bragg peak location. These results confirm the hypothesis that severely damaged cells at the Bragg peak are more likely to go through reproductive death and not be evaluated for micronuclei.
Collapse
Affiliation(s)
- H Wu
- NASA Johnson Space Center, Houston, Texas 77058, USA.
| | | | | | | | | | | |
Collapse
|
29
|
Shao C, Lyng FM, Folkard M, Prise KM. Calcium Fluxes Modulate the Radiation-Induced Bystander Responses in Targeted Glioma and Fibroblast Cells. Radiat Res 2006; 166:479-87. [PMID: 16953666 DOI: 10.1667/rr3600.1] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Bystander responses have been reported to be a major determinant of the response of cells to radiation exposure at low doses, including those of relevance to therapy. This study investigated the role of changes in calcium levels in bystander responses leading to chromosomal damage in nonirradiated T98G glioma cells and AG01522 fibroblasts that had been either exposed to conditioned medium from irradiated cells or co-cultured with a population where a fraction of cells were individually targeted through the nucleus or cytoplasm with a precise number of microbeam helium-3 particles. After the recipient cells were treated with conditioned medium from T98G or AG01522 cells that had been irradiated through either nucleus or cytoplasm, rapid calcium fluxes were monitored in the nonirradiated recipient cells. Their characteristics were dependent on the source of the conditioned medium but had no dependence on radiation dose. When recipient cells were co-cultured with an irradiated population of either T98G or AG01522 cells, micronuclei were induced in the nonirradiated cells, but this response was eliminated by treating the cells with calcicludine (CaC), a potent blocker of Ca(2+) channels. Moreover, both the calcium fluxes and the bystander effect were inhibited when the irradiated T98G cells were treated with aminoguanidine, an inhibitor of nitric oxide synthase (NOS), and when the irradiated AG01522 cells were treated with DMSO, a scavenger of reactive oxygen species (ROS), which indicates that NO and ROS were involved in the bystander responses generated from irradiated T98G and AG01522 cells, respectively. Our findings indicate that calcium signaling may be an early response in radiation-induced bystander effects leading to chromosome damage.
Collapse
Affiliation(s)
- Chunlin Shao
- Gray Cancer Institute, Mount Vernon Hospital, Northwood, Middlesex, UK
| | | | | | | |
Collapse
|
30
|
Müller WEG, Ushijima H, Batel R, Krasko A, Borejko A, Müller IM, Schröder HC. Novel mechanism for the radiation-induced bystander effect: nitric oxide and ethylene determine the response in sponge cells. Mutat Res 2006; 597:62-72. [PMID: 16427660 DOI: 10.1016/j.mrfmmm.2005.09.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2005] [Revised: 06/17/2005] [Accepted: 09/14/2005] [Indexed: 05/06/2023]
Abstract
Until now the bystander effect had only been described in vertebrates. In the present study the existence of this effect has been demonstrated for the phylogenetically oldest metazoan phylum, the Porifera. We used the demosponge Suberites domuncula for the experiments in the two-chamber-system. The lower dish contained irradiated "donor" cells (single cells) and the upper dish the primmorphs ("recipient" primmorphs). The "donor" cells were treated with UV-B light (40 mJ/cm2) and 100 microM hydrogen peroxide (H2O2), factors that exist also in the natural marine aquatic environment of sponges; these factors caused a high level of DNA strand breaks followed by a reduced viability of the cells. If these cells were added to the "recipient" primmorphs these 3D-cell cultures started to undergo apoptosis. This effect could be abolished by the NO-specific scavenger PTIO and ethylene. The conclusion that NO is synthesized by the UV-B/H2O2-treated cells was supported analytically. The cDNA encoding the enzyme dimethylarginine dimethylaminohydrolase (DDAH) was isolated from the "donor" cells. High levels of DDAH transcripts were measured in UV-B/H2O2-treated "donor" cells while after ethylene treatment the steady-state level of expression drops drastically. We conclude that in the absence of ethylene the concentration of the physiological inhibitor for the NO synthase ADMA is low, due to the high level of DDAH. In consequence, high amounts of NO are released from "donor" cells which cause apoptosis in "recipient" primmorphs. In contrast, ethylene reduces the DDAH expression with the consequence of higher levels of ADMA which prevent the formation of larger amounts of NO. This study describes the radiation-induced bystander effect also for the most basal metazoans and demonstrates that this effect is controlled by the two gases NO and ethylene.
Collapse
Affiliation(s)
- Werner E G Müller
- Institut für Physiologische Chemie, Abteilung Angewandte Molekularbiologie, Universität, Duesbergweg 6, D-55099 Mainz, Germany.
| | | | | | | | | | | | | |
Collapse
|
31
|
Wang R, Coderre JA. A Bystander Effect in Alpha-Particle Irradiations of Human Prostate Tumor Cells. Radiat Res 2005; 164:711-22. [PMID: 16296877 DOI: 10.1667/3475.1] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Alpha-particle exposures were used to determine whether cells of the human prostate carcinoma cell line DU-145 can produce and respond to a bystander effect signal. An apparatus for alpha-particle irradiation of cells growing as a monolayer on a 1.4-microm-thick Mylar membrane directly above an 241Am alpha-particle source was constructed and calibrated. At the cell irradiation position, the alpha-particle fluence was 998 counts/mm2 s(-1), the average alpha-particle energy was 3.14 MeV, and the average linear energy transfer was 128 keV/microm. The average dose rate to the cells growing on the Mylar surface was 1.2 Gy/min. A co-culture system was used to examine bystander effects transmitted through the medium from the directly targeted cells to tumor cells growing on an insert well beyond the range of the alpha particles. Alpha-particle doses from 0.1 to 6.0 Gy to the targeted cells on the Mylar membrane, followed by a 2-h co-incubation of the cells on the insert in the irradiated medium above the irradiated cells, all caused an approximately 50% increase in micronucleus formation in the nontargeted co-cultured cells. Addition of the radical scavenger DMSO to the medium during the irradiation and the 2-h postirradiation incubation period completely blocked the bystander effect, whereas addition of a nitric oxide scavenger had no effect. Irradiation of medium containing serum, followed by a 2-h incubation, caused no bystander effect in the co-cultured cells. When the co-cultured cells on the insert were placed into the irradiated medium above the directly targeted cells immediately (approximately 1 min) after the irradiation and co-incubated for 2 h, there was no bystander effect. These data indicate that the observed bystander effect requires that the co-cultured cells be present in the medium during the irradiation of the directly targeted cells and suggest the involvement of a short-lived radical species.
Collapse
Affiliation(s)
- Rong Wang
- Department of Nuclear Science and Engineering, Massachusetts Institute of Technology, Cambridge 02139, USA
| | | |
Collapse
|
32
|
Funayama T, Wada S, Kobayashi Y, Watanabe H. Irradiation of mammalian cultured cells with a collimated heavy-ion microbeam. Radiat Res 2005; 163:241-6. [PMID: 15658901 DOI: 10.1667/rr3301] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
As the first step for the analysis of the biological effect of heavy charged-particle radiation, we established a method for the irradiation of individual cells with a heavy-ion microbeam apparatus at JAERI-Takasaki. CHO-K1 cells attached on a thin film of an ion track detector, CR-39, were automatically detected under a fluorescence microscope and irradiated individually with an 40Ar13+ ion (11.5 MeV/nucleon, LET 1260 keV/microm) microbeam. Without killing the irradiated cells, trajectories of irradiated ions were visualized as etch pits by treatment of the CR-39 with an alkaline-ethanol solution at 37 degrees C. The exact positions of ion hits were determined by overlaying images of both cells and etch pits. The cells that were irradiated with argon ions showed a reduced growth in postirradiation observations. Moreover, a single hit of an argon ion to the cell nucleus resulted in strong growth inhibition. These results tell us that our verified irradiation method enables us to start a precise study of the effects of high-LET radiation on cells.
Collapse
Affiliation(s)
- Tomoo Funayama
- Research Group for Biotechnology Development, JAERI-Takasaki, Takasaki, Gunma 370-1292, Japan.
| | | | | | | |
Collapse
|
33
|
Shao C, Folkard M, Michael BD, Prise KM. Bystander signaling between glioma cells and fibroblasts targeted with counted particles. Int J Cancer 2005; 116:45-51. [PMID: 15756683 DOI: 10.1002/ijc.21003] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Radiation-induced bystander effects may play an important role in cancer risks associated with environmental, occupational and medical exposures and they may also present a therapeutic opportunity to modulate the efficacy of radiotherapy. However, the mechanisms underpinning these responses between tumor and normal cells are poorly understood. Using a microbeam, we investigated interactions between T98G malignant glioma cells and AG01522 normal fibroblasts by targeting cells through their nuclei in one population, then detecting cellular responses in the other co-cultured non-irradiated population. It was found that when a fraction of cells was individually irradiated with exactly 1 or 5 helium particles ((3)He(2+)), the yield of micronuclei (MN) in the non-irradiated population was significantly increased. This increase was not related to the fraction of cells targeted or the number of particles delivered to those cells. Even when one cell was targeted with a single (3)He(2+), the induction of MN in the bystander non-irradiated population could be increased by 79% for AG01522 and 28% for T98G. Furthermore, studies showed that nitric oxide (NO) and reactive oxygen species (ROS) were involved in these bystander responses. Following nuclear irradiation in only 1% of cells, the NO level in the T98G population was increased by 31% and the ROS level in the AG0 population was increased by 18%. Treatment of cultures with 2-(4-carboxyphenyl)-4,4,5,5-tetramethyl-imidazoline-1-oxyl-3-oxide (c-PTIO), an NO scavenger, abolished the bystander MN induction in non-irradiated AG01522 cells but only partially in non-irradiated T98G cells, and this could be eliminated by treatment with either DMSO or antioxidants. Our findings indicate that differential mechanisms involving NO and ROS signaling factors play a role in bystander responses generated from targeted T98G glioma and AG0 fibroblasts, respectively. These bystander interactions suggest that a mechanistic control of the bystander effect could be of benefit to radiotherapy.
Collapse
Affiliation(s)
- Chunlin Shao
- Gray Cancer Institute, Mount Vernon Hospital, Northwood, Middlesex, United Kingdom
| | | | | | | |
Collapse
|
34
|
Kobayashi Y, Funayama T, Wada S, Furusawa Y, Aoki M, Shao C, Yokota Y, Sakashita T, Matsumoto Y, Kakizaki T, Hamada N. Microbeams of heavy charged particles. UCHU SEIBUTSU KAGAKU 2004; 18:235-40. [PMID: 15858390 DOI: 10.2187/bss.18.235] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
We have established a single cell irradiation system, which allows selected cells to be individually hit with defined number of heavy charged particles, using a collimated heavy-ion microbeam apparatus at JAERI-Takasaki. This system has been developed to study radiobiological processes in hit cells and bystander cells exposed to low dose and low dose-rate high-LET radiations, in ways that cannot be achieved using conventional broad-field exposures. Individual cultured cells grown in special dishes were irradiated in the atmosphere with a single or defined numbers of 18.3 MeV/amu 12C, 13.0 MeV/amu 20Ne, and 11.5 MeV/amu 40Ar ions. Targeting and irradiation of the cells were performed automatically at the on-line microscope of the microbeam apparatus according to the positional data of the target cells obtained at the off-line microscope before irradiation. The actual number of particle tracks that pass through cell nuclei was detected with prompt etching of the bottom of the cell dish made of ion track detector TNF-1 (modified CR-39), with alkaline-ethanol solution at 37 degrees C for 15-30 minutes. Using this system, separately inoculated Chinese hamster ovary cells, confluent normal human fibroblasts, and single plant cells (tobacco protoplasts) have been irradiated. These are the first studies in which single-ion direct hit effect and the bystander effect have been investigated using a high-LET heavy particle microbeam.
Collapse
Affiliation(s)
- Yasuhiko Kobayashi
- Research Group for Biotechnology Development, Japan Atomic Energy Research Institute (JAERI-Takasaki), Takasaki, Gunma, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Banerjee G, Gupta N, Kapoor A, Raman G. UV induced bystander signaling leading to apoptosis. Cancer Lett 2004; 223:275-84. [PMID: 15896462 DOI: 10.1016/j.canlet.2004.09.035] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2004] [Accepted: 09/13/2004] [Indexed: 12/17/2022]
Abstract
Human keratinocytes (HaCaT) were exposed to UV (A+B) (UVA-350-400 mJ/cm2 and UVB-30 mJ/cm2) which induces apoptosis as evidenced by MTT assay, DNA laddering, Bax and Fas up-regulation. UV induced apoptotic conditioned media (6 h or earlier) did not cause apoptosis in unexposed cells. However, treatment with conditioned medium collected post UV exposure (1 h) induced Bax in unexposed cells as observed by RT-PCR. The induction of cell death was initiated by conditioned medium collected 12 h after UV exposure and the extent of death was increased progressively when conditioned medium collected 24 or 72 h post UV exposure was used. Medium collected 24 h after UV exposure also increased mitochondrial membrane permeability as determined by rhodamine uptake. Conditioned medium induced apoptosis did not involve reactive oxygen species (ROS) unlike UV induced apoptosis indicating that the apoptosis pathway could be different. Interestingly, at high dilution apototic conditioned medium did not induce apoptosis but actually protected cells from UV insult. The role of nerve growth factor (NGF) in UV induced bystander effects are also discussed.
Collapse
Affiliation(s)
- Gautam Banerjee
- Cell and Molecular Biology, Environmental Safety Laboratory, Hindustan Lever Research Centre, Mumbai, India.
| | | | | | | |
Collapse
|