1
|
Matarèse BFE, Desai R, Oughton DH, Mothersill C. EGO to ECO: Tracing the History of Radioecology from the 1950's to the Present Day. Radiat Res 2024; 202:273-288. [PMID: 39021078 DOI: 10.1667/rade-24-00035.1] [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: 01/30/2024] [Accepted: 05/09/2024] [Indexed: 07/20/2024]
Abstract
This paper starts with a brief history of the birth of the field of radioecology during the Cold War with a focus on US activity. We review the establishment of the international system for radiation protection and the science underlying the guidelines. We then discuss the famous ICRP 60 statement that if "Man" is protected, so is everything else and show how this led to a focus in radioecology on pathways to "Man" rather than concern about impacts on environments or ecosystems. We then review the contributions of Radiation Research Society members and papers published in Radiation Research which contributed to the knowledge base about effects on non-human species. These fed into international databases and computer-based tools such as ERICA and ResRad Biota to guide regulators. We then examine the origins of the concern that ICRP 60 is not sufficient to protect ecosystems and discuss the establishment of ICRP Committee 5 and its recommendations to establish reference animals and plants. The review finishes with current concerns that reference animals and plants (RAPs) are not sufficient to protect ecosystems, given the complexity of interacting factors such as the climate emergency and discusses the efforts of ICRP, the International Union of Radioecologists and other bodies to capture the concepts of ecosystem services and ecosystem complexity modelling in radioecology.
Collapse
Affiliation(s)
- Bruno F E Matarèse
- Department of Haematology, University of Cambridge, Cambridge CB2 1TN, United Kingdom
- Department of Physics, University of Cambridge, Cambridge CB2 1TN, United Kingdom
| | - Rhea Desai
- Department of Biology, McMaster University, Hamilton, ON L8S 4L8, Canada
| | | | - Carmel Mothersill
- Department of Biology, McMaster University, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
2
|
Role of p53 in Regulating Radiation Responses. LIFE (BASEL, SWITZERLAND) 2022; 12:life12071099. [PMID: 35888186 PMCID: PMC9319710 DOI: 10.3390/life12071099] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/15/2022] [Accepted: 07/16/2022] [Indexed: 12/12/2022]
Abstract
p53 is known as the guardian of the genome and plays various roles in DNA damage and cancer suppression. The p53 gene was found to express multiple p53 splice variants (isoforms) in a physiological, tissue-dependent manner. The various genes that up- and down-regulated p53 are involved in cell viability, senescence, inflammation, and carcinogenesis. Moreover, p53 affects the radioadaptive response. Given that several studies have already been published on p53, this review presents its role in the response to gamma irradiation by interacting with MDM2, NF-κB, and miRNA, as well as in the inflammation processes, senescence, carcinogenesis, and radiation adaptive responses. Finally, the potential of p53 as a biomarker is discussed.
Collapse
|
3
|
Nasrollahzadeh E, Razi S, Keshavarz-Fathi M, Mazzone M, Rezaei N. Pro-tumorigenic functions of macrophages at the primary, invasive and metastatic tumor site. Cancer Immunol Immunother 2020; 69:1673-1697. [PMID: 32500231 DOI: 10.1007/s00262-020-02616-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 05/16/2020] [Indexed: 12/19/2022]
Abstract
The tumor microenvironment (TME) not only facilitates cancer progression from the early formation to distant metastasis, but also it differs itself from time to time alongside the tumor evolution. Tumor-associated macrophages (TAMs), whether as pre-existing tissue-resident macrophages or recruited monocytes, are an inseparable part of this microenvironment. As their parents are broadly classified into a dichotomic, simplistic M1 and M2 subtypes, TAMs also exert paradoxical and diverse phenotypes as they are settled in different regions of TME and receive different microenvironmental signals. Briefly, M1 macrophages induce an inflammatory precancerous niche and flame the early oncogenic mutations, whereas their M2 counterparts are reprogrammed to release various growth factors and providing an immunosuppressive state in TME as long as abetting hypoxic cancer cells to set up a new vasculature. Further, they mediate stromal micro-invasion and co-migrate with invasive cancer cells to invade the vascular wall and neural sheath, while another subtype of TAMs prepares suitable niches much earlier than metastatic cells arrive at the target tissues. Accordingly, at the neoplastic transformation, during the benign-to-malignant transition and through the metastatic cascade, macrophages are involved in shaping the primary, micro-invasive and pre-metastatic TMEs. Whether their behavioral plasticity is derived from distinct genotypes or is fueled by microenvironmental cues, it could define these cells as remarkably interesting therapeutic targets.
Collapse
Affiliation(s)
- Elaheh Nasrollahzadeh
- School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.,Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Sepideh Razi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahsa Keshavarz-Fathi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, Center for Cancer Biology, VIB, KU Leuven, Louvain, B3000, Belgium
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr Qarib St, Keshavarz Blvd, 14194, Tehran, Iran. .,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran. .,Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Stockholm, Sweden.
| |
Collapse
|
4
|
Helm JS, Rudel RA. Adverse outcome pathways for ionizing radiation and breast cancer involve direct and indirect DNA damage, oxidative stress, inflammation, genomic instability, and interaction with hormonal regulation of the breast. Arch Toxicol 2020. [PMID: 32399610 DOI: 10.1007/s00204-020-02752-z)] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Abstract
Knowledge about established breast carcinogens can support improved and modernized toxicological testing methods by identifying key mechanistic events. Ionizing radiation (IR) increases the risk of breast cancer, especially for women and for exposure at younger ages, and evidence overall supports a linear dose-response relationship. We used the Adverse Outcome Pathway (AOP) framework to outline and evaluate the evidence linking ionizing radiation with breast cancer from molecular initiating events to the adverse outcome through intermediate key events, creating a qualitative AOP. We identified key events based on review articles, searched PubMed for recent literature on key events and IR, and identified additional papers using references. We manually curated publications and evaluated data quality. Ionizing radiation directly and indirectly causes DNA damage and increases production of reactive oxygen and nitrogen species (RONS). RONS lead to DNA damage and epigenetic changes leading to mutations and genomic instability (GI). Proliferation amplifies the effects of DNA damage and mutations leading to the AO of breast cancer. Separately, RONS and DNA damage also increase inflammation. Inflammation contributes to direct and indirect effects (effects in cells not directly reached by IR) via positive feedback to RONS and DNA damage, and separately increases proliferation and breast cancer through pro-carcinogenic effects on cells and tissue. For example, gene expression changes alter inflammatory mediators, resulting in improved survival and growth of cancer cells and a more hospitable tissue environment. All of these events overlap at multiple points with events characteristic of "background" induction of breast carcinogenesis, including hormone-responsive proliferation, oxidative activity, and DNA damage. These overlaps make the breast particularly susceptible to ionizing radiation and reinforce that these biological activities are important characteristics of carcinogens. Agents that increase these biological processes should be considered potential breast carcinogens, and predictive methods are needed to identify chemicals that increase these processes. Techniques are available to measure RONS, DNA damage and mutation, cell proliferation, and some inflammatory proteins or processes. Improved assays are needed to measure GI and chronic inflammation, as well as the interaction with hormonally driven development and proliferation. Several methods measure diverse epigenetic changes, but it is not clear which changes are relevant to breast cancer. In addition, most toxicological assays are not conducted in mammary tissue, and so it is a priority to evaluate if results from other tissues are generalizable to breast, or to conduct assays in breast tissue. Developing and applying these assays to identify exposures of concern will facilitate efforts to reduce subsequent breast cancer risk.
Collapse
Affiliation(s)
- Jessica S Helm
- Silent Spring Institute, 320 Nevada Street, Suite 302, Newton, MA, 02460, USA
| | - Ruthann A Rudel
- Silent Spring Institute, 320 Nevada Street, Suite 302, Newton, MA, 02460, USA.
| |
Collapse
|
5
|
Helm JS, Rudel RA. Adverse outcome pathways for ionizing radiation and breast cancer involve direct and indirect DNA damage, oxidative stress, inflammation, genomic instability, and interaction with hormonal regulation of the breast. Arch Toxicol 2020; 94:1511-1549. [PMID: 32399610 PMCID: PMC7261741 DOI: 10.1007/s00204-020-02752-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 04/16/2020] [Indexed: 12/15/2022]
Abstract
Knowledge about established breast carcinogens can support improved and modernized toxicological testing methods by identifying key mechanistic events. Ionizing radiation (IR) increases the risk of breast cancer, especially for women and for exposure at younger ages, and evidence overall supports a linear dose-response relationship. We used the Adverse Outcome Pathway (AOP) framework to outline and evaluate the evidence linking ionizing radiation with breast cancer from molecular initiating events to the adverse outcome through intermediate key events, creating a qualitative AOP. We identified key events based on review articles, searched PubMed for recent literature on key events and IR, and identified additional papers using references. We manually curated publications and evaluated data quality. Ionizing radiation directly and indirectly causes DNA damage and increases production of reactive oxygen and nitrogen species (RONS). RONS lead to DNA damage and epigenetic changes leading to mutations and genomic instability (GI). Proliferation amplifies the effects of DNA damage and mutations leading to the AO of breast cancer. Separately, RONS and DNA damage also increase inflammation. Inflammation contributes to direct and indirect effects (effects in cells not directly reached by IR) via positive feedback to RONS and DNA damage, and separately increases proliferation and breast cancer through pro-carcinogenic effects on cells and tissue. For example, gene expression changes alter inflammatory mediators, resulting in improved survival and growth of cancer cells and a more hospitable tissue environment. All of these events overlap at multiple points with events characteristic of "background" induction of breast carcinogenesis, including hormone-responsive proliferation, oxidative activity, and DNA damage. These overlaps make the breast particularly susceptible to ionizing radiation and reinforce that these biological activities are important characteristics of carcinogens. Agents that increase these biological processes should be considered potential breast carcinogens, and predictive methods are needed to identify chemicals that increase these processes. Techniques are available to measure RONS, DNA damage and mutation, cell proliferation, and some inflammatory proteins or processes. Improved assays are needed to measure GI and chronic inflammation, as well as the interaction with hormonally driven development and proliferation. Several methods measure diverse epigenetic changes, but it is not clear which changes are relevant to breast cancer. In addition, most toxicological assays are not conducted in mammary tissue, and so it is a priority to evaluate if results from other tissues are generalizable to breast, or to conduct assays in breast tissue. Developing and applying these assays to identify exposures of concern will facilitate efforts to reduce subsequent breast cancer risk.
Collapse
Affiliation(s)
- Jessica S Helm
- Silent Spring Institute, 320 Nevada Street, Suite 302, Newton, MA, 02460, USA
| | - Ruthann A Rudel
- Silent Spring Institute, 320 Nevada Street, Suite 302, Newton, MA, 02460, USA.
| |
Collapse
|
6
|
Torabizadeh SA, Rezaeifar M, Jomehzadeh A, Nabizadeh Haghighi F, Ansari M. Radioprotective Potential of Sulindac Sulfide to Prevent DNA Damage Due to Ionizing Radiation. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:4127-4134. [PMID: 31827319 PMCID: PMC6902880 DOI: 10.2147/dddt.s218022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 11/15/2019] [Indexed: 11/23/2022]
Abstract
Introduction: The ionizing radiation exposure of the normal cell causes damage to DNA, which leads to cell dysfunction or even cell death. However, it is necessary to identify new radio protectives in order to protect normal cells. Sulindac sulfide (SS) is a metabolite of sulindac (a non-steroidal anti-inflammatory drug) known as a cyclooxygenase inhibitor. Free radicals and reactive oxygen species are generated in the IR-exposed cells. Also, the induced inflammation process causes damage in DNA. Purpose In this research, the radioprotective effect of SS was investigated against genotoxicity and lipid peroxidation induced by ionizing radiation in the human blood lymphocytes. Methods In this study, the human blood samples were pretreated with SS at different concentrations (10, 25, 50, 100 and 250 μM) and then were exposed to IR at a dose of 1.5 Gy. The micronucleus (MN) assay was used to indicate the radioprotective effects of SS on exposed cells. Total antioxidant activity of the SS was measured by using FRAP and DPPH assay. Also, the malondialdehyde (MDA) levels and the activity of superoxide dismutase (SOD) on the exposed cells were evaluated. Results It was found that SS decreased the percentage of MN induced by IR in exposed cells. Maximum reduction in the frequency of MN was observed at 250 μM of SS (87%) that provides the highest degree of protection against IR. On the other hand, pretreatment at 250 μM of SS inhibited IR-induced oxidative stress, which led to a decrease in the MN frequencies and MDA levels, while SOD activity showed an increase in the exposed cells. Conclusion It could be concluded that SS as a good radioprotective agent protects the human normal cells against the oxidative stress and genetic damage induced by IR.
Collapse
Affiliation(s)
- Seyedeh Atekeh Torabizadeh
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mehdi Rezaeifar
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Ali Jomehzadeh
- Department of Medical Physics, Faculty of Medicine, Medical Physics Department, Radiotherapy & Oncology Unit, Shafa Kerman Hospital, Kerman University of Medical Sciences, Kerman, Iran
| | - Farzaneh Nabizadeh Haghighi
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mehdi Ansari
- Drug and Food Control Department, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
7
|
Zhang A, Steen TY. Gut Microbiomics-A Solution to Unloose the Gordian Knot of Biological Effects of Ionizing Radiation. J Hered 2019; 109:212-221. [PMID: 29452420 DOI: 10.1093/jhered/esx059] [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: 01/23/2017] [Accepted: 07/05/2017] [Indexed: 12/24/2022] Open
Abstract
The Chernobyl and Fukushima nuclear accidents have called forth a growing body of research on their biological aftermaths. A variety of wild organisms, including primates, birds, fish, insects, and worms are being studied in the affected areas, with emerging morphological, physiological, and genetic aberrations ascribed to ionizing radiation. Despite the effort in surveying Chernobyl and Fukushima wildlife, little is known about the microorganisms associated with these radiation-contaminated animals. The microbiota, especially the gut commensal, plays an important role in shaping the metabolic reservoir and immune system of the host, and is sensitive to a wide array of environmental factors, including ionizing radiation. Humans and limited numbers of laboratory species have been the main subjects of microbiome studies, however, a more practical insight on host-gut microbiota dynamics under environmental impact should be explored in natural habitats. In this analysis, we introduced a working model explaining possible mechanisms of ionizing radiation on the gut microbiota, with an evaluation of the gut microbiota as a potential biomarker for exposure to ionizing radiation.
Collapse
Affiliation(s)
- Amy Zhang
- Department of Biological Sciences, Binghamton University, State University of New York, Binghamton, NY
| | - Tomoko Y Steen
- Department of Microbiology and Immunology, School of Medicine, Georgetown University, Washington, DC
| |
Collapse
|
8
|
Electromagnetic Fields, Genomic Instability and Cancer: A Systems Biological View. Genes (Basel) 2019; 10:genes10060479. [PMID: 31242701 PMCID: PMC6627294 DOI: 10.3390/genes10060479] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/19/2019] [Accepted: 06/22/2019] [Indexed: 12/12/2022] Open
Abstract
This review discusses the use of systems biology in understanding the biological effects of electromagnetic fields, with particular focus on induction of genomic instability and cancer. We introduce basic concepts of the dynamical systems theory such as the state space and attractors and the use of these concepts in understanding the behavior of complex biological systems. We then discuss genomic instability in the framework of the dynamical systems theory, and describe the hypothesis that environmentally induced genomic instability corresponds to abnormal attractor states; large enough environmental perturbations can force the biological system to leave normal evolutionarily optimized attractors (corresponding to normal cell phenotypes) and migrate to less stable variant attractors. We discuss experimental approaches that can be coupled with theoretical systems biology such as testable predictions, derived from the theory and experimental methods, that can be used for measuring the state of the complex biological system. We also review potentially informative studies and make recommendations for further studies.
Collapse
|
9
|
Radiation-induced inflammatory cascade and its reverberating crosstalks as potential cause of post-radiotherapy second malignancies. Cancer Metastasis Rev 2017; 36:375-393. [DOI: 10.1007/s10555-017-9669-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
10
|
Bannister LA, Mantha RR, Devantier Y, Petoukhov ES, Brideau CLA, Serran ML, Klokov DY. Dose and Radioadaptive Response Analysis of Micronucleus Induction in Mouse Bone Marrow. Int J Mol Sci 2016; 17:ijms17091548. [PMID: 27649149 PMCID: PMC5037821 DOI: 10.3390/ijms17091548] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 09/01/2016] [Accepted: 09/05/2016] [Indexed: 12/12/2022] Open
Abstract
Enhanced cellular DNA repair efficiency and suppression of genomic instability have been proposed as mechanisms underlying radio-adaptive responses following low-dose radiation exposures. We previously showed that low-dose γ irradiation does not generate radio-adaptation by lowering radiation-induced cytogenetic damage in mouse spleen. Since radiation may exert tissue-specific effects, we extended these results here by examining the effects of γ radiation on cytogenetic damage and proliferative index in bone marrow erythrocytes of C57BL/6 and BALB/c mice. In C57BL/6 mice, the induction of micronuclei in polychromatic erythrocytes (MN-PCE) was observed at radiation doses of 100 mGy and greater, and suppression of erythroblast maturation occurred at doses of >500 mGy. A linear dose-response relationship for MN-PCE frequencies in C57BL/6 mice was established for radiation doses between 100 mGy and 1 Gy, with departure from linearity at doses of >1 Gy. BALB/c mice exhibited increased MN-PCE frequencies above baseline following a 20 mGy radiation exposure but did not exhibit radio-sensitivity relative to C57BL/6 mice following 2 Gy exposure. Radio-adaptation of bone marrow erythrocytes was not observed in either strain of mice exposed to low-dose priming γ irradiation (single doses of 20 mGy or 100 mGy or multiple 20 mGy doses) administered at various times prior to acute 2 Gy irradiation, confirming the lack of radio-adaptive response for induction of cytogenetic damage or suppression or erythrocyte proliferation/maturation in bone marrow of these mouse strains.
Collapse
Affiliation(s)
- Laura A Bannister
- Canadian Nuclear Laboratories, Radiobiology and Health, Chalk River, ON K0J1J0, Canada.
| | - Rebecca R Mantha
- Canadian Nuclear Laboratories, Radiobiology and Health, Chalk River, ON K0J1J0, Canada.
| | - Yvonne Devantier
- Canadian Nuclear Laboratories, Radiobiology and Health, Chalk River, ON K0J1J0, Canada.
| | - Eugenia S Petoukhov
- Canadian Nuclear Laboratories, Radiobiology and Health, Chalk River, ON K0J1J0, Canada.
| | - Chantal L A Brideau
- Canadian Nuclear Laboratories, Radiobiology and Health, Chalk River, ON K0J1J0, Canada.
| | - Mandy L Serran
- Canadian Nuclear Laboratories, Radiobiology and Health, Chalk River, ON K0J1J0, Canada.
| | - Dmitry Y Klokov
- Canadian Nuclear Laboratories, Radiobiology and Health, Chalk River, ON K0J1J0, Canada.
| |
Collapse
|
11
|
Werner E, Wang H, Doetsch PW. Role of Pro-inflammatory Cytokines in Radiation-Induced Genomic Instability in Human Bronchial Epithelial Cells. Radiat Res 2015; 184:621-9. [PMID: 26579942 DOI: 10.1667/rr14045.1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Inflammatory cytokines have been implicated in the regulation of radiation-induced genomic instability in the hematopoietic system and have also been shown to induce chronic DNA damage responses in radiation-induced senescence. We have previously shown that human bronchial epithelial cells (HBEC3-KT) have increased genomic instability and IL-8 production persisting at day 7 after exposure to high-LET (600 MeV/nucleon (56)Fe ions) compared to low-LET (320 keV X rays) radiation. Thus, we investigated whether IL-8 induction is part of a broader pro-inflammatory response produced by the epithelial cells in response to damage, which influences genomic instability measured by increased micronuclei and DNA repair foci frequencies. We found that exposure to radiation induced the release of multiple inflammatory cytokines into the media, including GM-CSF, GROα, IL-1α, IL-8 and the inflammation modulator, IL-1 receptor antagonist (IL-1RA). Our results suggest that this is an IL-1α-driven response, because an identical signature was induced by the addition of recombinant IL-1α to nonirradiated cells and functional interference with recombinant IL-1RA (Anakinra) or anti-IL-1α function-blocking antibody, decreased IL-8 production induced by radiation exposure. However, genomic instability was not influenced by this pathway as addition of recombinant IL-1α to naive or irradiated cells or the presence of IL-1 RA under the same conditions as those that interfered with the function of IL-8, did not affect micronuclei or DNA repair foci frequencies measured at day 7 after exposure. While dose-response studies revealed that genomic instability and IL-8 production are the consequences of targeted effects, experiments employing a co-culture transwell system revealed the propagation of pro-inflammatory responses but not genomic instability from irradiated to nonirradiated cells. Collectively, these results point to a cell-autonomous mechanism sustaining radiation-induced genomic instability in this model system and suggest that while molecules associated with these mechanisms could be markers for persisting damage, they reflect two different outcomes.
Collapse
Affiliation(s)
- Erica Werner
- a Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia
| | - Huichen Wang
- b Department of Physics, Radiation Institute for Science and Engineering (RaISE), Prairie View A&M University, Prairie View, Texas; and
| | - Paul W Doetsch
- a Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia;,c Departments of Radiation Oncology and Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
12
|
Rithidech KN, Honikel LM, Reungpathanaphong P, Tungjai M, Jangiam W, Whorton EB. Late-occurring chromosome aberrations and global DNA methylation in hematopoietic stem/progenitor cells of CBA/CaJ mice exposed to silicon ((28)Si) ions. Mutat Res 2015; 781:22-31. [PMID: 26398320 DOI: 10.1016/j.mrfmmm.2015.09.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 07/18/2015] [Accepted: 09/07/2015] [Indexed: 06/05/2023]
Abstract
Although myeloid leukemia (ML) is one of the major health concerns from exposure to space radiation, the risk prediction for developing ML is unsatisfactory. To increase the reliability of predicting ML risk, a much improved understanding of space radiation-induced changes in the target cells, i.e. hematopoietic stem/progenitor cells (HSPCs), is important. We focused on the in vivo induction of late-occurring damage in HSPCs of mice exposed to (28)Si ions since such damage is associated with radiation-induced genomic instability (a key event of carcinogenesis). We gave adult male CBA/CaJ mice, known to be sensitive to radiation-induced ML, a whole-body exposure (2 fractionated exposures, 15 days apart, that totaled each selected dose, delivered at the dose-rate of 1 cGy/min) to various doses of 300 MeV/n (28)Si ions, i.e. 0 (sham controls), 0.1, 0.25, or 0.5 Gy. At 6 months post-irradiation, we collected bone marrow cells from each mouse (five mice per treatment-group) for obtaining the myeloid-lineage of HSPC-derived clones for analyses. We measured the frequencies of late-occurring chromosome aberrations (CAs), using the genome-wide multicolor fluorescence in situ hybridization method. The measurement of CAs was coupled with the characterization of the global DNA methylation patterns, i.e. 5-methylcytosine (5 mC) and 5-hydroxymethylcytosine (5 hmC). A dose-dependent increase in the frequencies of CAs was detected (Analysis of Variance or ANOVA, p<0.01), indicating the induction of genomic instability after exposure of mice to 300 MeV/n (28)Si ions. Slight increases in the levels of 5 mC were observed in all treatment groups, as compared to the sham-control level. In contrast, there was a significant reduction in levels of 5 hmC (ANOVA, p<0.01). Since these endpoints were evaluated in the same mouse, our data suggested for the first time a link between a reduction in 5 hmC and genomic instability in HSPC-derived myeloid colonies of CBA/CaJ mice exposed to 300 MeV/n (28)Si ions.
Collapse
Affiliation(s)
| | - Louise M Honikel
- Pathology Department, Stony Brook University, Stony Brook, NY 11794-8691, USA
| | - Paiboon Reungpathanaphong
- Pathology Department, Stony Brook University, Stony Brook, NY 11794-8691, USA; Department of Applied Radiation and Isotopes, Faculty of Sciences, Kasetsart University, Chatuchuck, Bangkok 10900, Thailand
| | - Montree Tungjai
- Pathology Department, Stony Brook University, Stony Brook, NY 11794-8691, USA; Department of Radiologic Technology, Faculty of Associated Medical Sciences, Center of Excellence for Molecular Imaging, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Witawat Jangiam
- Pathology Department, Stony Brook University, Stony Brook, NY 11794-8691, USA; Department of Chemical Engineering, Faculty of Engineering, Burapha University, Chonburi 20131, Thailand
| | | |
Collapse
|
13
|
Hosseinimehr SJ, Nobakht R, Ghasemi A, Pourfallah TA. Radioprotective effect of mefenamic acid against radiation-induced genotoxicity in human lymphocytes. Radiat Oncol J 2015; 33:256-60. [PMID: 26484310 PMCID: PMC4607580 DOI: 10.3857/roj.2015.33.3.256] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 06/20/2015] [Accepted: 06/29/2015] [Indexed: 12/23/2022] Open
Abstract
Purpose Mefenamic acid (MEF) as a non-steroidal anti-inflammatory drug is used as a medication for relieving of pain and inflammation. Radiation-induced inflammation process is involved in DNA damage and cell death. In this study, the radioprotective effect of MEF was investigated against genotoxicity induced by ionizing radiation in human blood lymphocytes. Materials and Methods Peripheral blood samples were collected from human volunteers and incubated with MEF at different concentrations (5, 10, 50, or 100 µM) for two hours. The whole blood was exposed to ionizing radiation at a dose 1.5 Gy. Lymphocytes were cultured with mitogenic stimulation to determine the micronuclei in cytokinesis blocked binucleated lymphocyte. Results A significant decreasing in the frequency of micronuclei was observed in human lymphocytes irradiated with MEF as compared to irradiated lymphocytes without MEF. The maximum decreasing in frequency of micronuclei was observed at 100 µM of MEF (38% decrease), providing maximal protection against ionizing radiation. Conclusion The radioprotective effect of MEF is probably related to anti-inflammatory property of MEF on human lymphocytes.
Collapse
Affiliation(s)
- Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Pharmaceutical Sciences Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Reyhaneh Nobakht
- Department of Radiopharmacy, Faculty of Pharmacy, Pharmaceutical Sciences Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Arash Ghasemi
- Department of Radiology and Radiation Oncology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Tayyeb Allahverdi Pourfallah
- Department of Biochemistry and Biophysics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
14
|
Abstract
The immune system has the power to modulate the expression of radiation-induced normal and tumor tissue damage. On the one hand, it can contribute to cancer cure, and on the other hand, it can influence acute and late radiation side effects, which in many ways resemble acute and chronic inflammatory disease states. The way radiation-induced inflammation feeds into adaptive antigen-specific immune responses adds another dimension to the tumor-host cross talk during radiation therapy and to possible radiation-driven autoimmune responses. Understanding how radiation affects inflammation and immunity is therefore critical if we are to effectively manipulate these forces for benefit in radiation oncology treatments.
Collapse
Affiliation(s)
- Dörthe Schaue
- Department of Radiation Oncology, University of California at Los Angeles, Los Angeles, CA
| | - Ewa D Micewicz
- Department of Radiation Oncology, University of California at Los Angeles, Los Angeles, CA
| | - Josephine A Ratikan
- Department of Radiation Oncology, University of California at Los Angeles, Los Angeles, CA
| | - Michael W Xie
- Department of Radiation Oncology, University of California at Los Angeles, Los Angeles, CA
| | - Genhong Cheng
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA
| | - William H McBride
- Department of Radiation Oncology, University of California at Los Angeles, Los Angeles, CA.
| |
Collapse
|
15
|
Brandão PDTJ, Gomes-Filho IS, Cruz SS, Passos-Soares JDS, Trindade SC, Souza LDCM, Meireles JRC, Cerqueira EDMM. Can periodontal infection induce genotoxic effects? Acta Odontol Scand 2015; 73:219-25. [PMID: 25428625 DOI: 10.3109/00016357.2014.982705] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE This study aimed to evaluate the occurrence of chromosomal abnormalities, through micronuclei, and apoptosis by the sum of karyorrhexis, pyknosis and condensed chromatin in individuals with chronic periodontitis, gingivitis associated with biofilm and no periodontal disease. MATERIALS AND METHODS This study included 72 individuals divided into three groups: gingivitis (n = 21), periodontitis (n = 24) and control (n = 27). Information on sociodemographic characteristics, health and lifestyle was obtained. Full mouth clinical examination was performed to define the periodontal condition. Exfoliated cells from gingival mucosa were collected for computation of micronuclei and nuclear changes indicative of apoptosis. The differences in the occurrence of endpoints (micronucleus, karyorrhexis, pyknosis and condensed chromatin) were evaluated using the conditional test to compare proportions in a rare events situation. RESULTS There was no statistically significant difference in the occurrence of micronucleus (p > 0.1) between gingivitis, periodontitis and control groups. The occurrence of apoptosis was significantly higher among individuals with periodontitis compared to individuals with gingivitis (p < 0.05) and controls (p < 0.025). CONCLUSIONS The findings showed that the inflammatory process generated by gingivitis and periodontitis is not related to a higher occurrence of chromosomal damage. However, the higher occurrence of apoptosis in individuals with periodontitis points to genotoxic effects induced by periodontal infection.
Collapse
|
16
|
Sridharan DM, Asaithamby A, Bailey SM, Costes SV, Doetsch PW, Dynan WS, Kronenberg A, Rithidech KN, Saha J, Snijders AM, Werner E, Wiese C, Cucinotta FA, Pluth JM. Understanding cancer development processes after HZE-particle exposure: roles of ROS, DNA damage repair and inflammation. Radiat Res 2015; 183:1-26. [PMID: 25564719 DOI: 10.1667/rr13804.1] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
During space travel astronauts are exposed to a variety of radiations, including galactic cosmic rays composed of high-energy protons and high-energy charged (HZE) nuclei, and solar particle events containing low- to medium-energy protons. Risks from these exposures include carcinogenesis, central nervous system damage and degenerative tissue effects. Currently, career radiation limits are based on estimates of fatal cancer risks calculated using a model that incorporates human epidemiological data from exposed populations, estimates of relative biological effectiveness and dose-response data from relevant mammalian experimental models. A major goal of space radiation risk assessment is to link mechanistic data from biological studies at NASA Space Radiation Laboratory and other particle accelerators with risk models. Early phenotypes of HZE exposure, such as the induction of reactive oxygen species, DNA damage signaling and inflammation, are sensitive to HZE damage complexity. This review summarizes our current understanding of critical areas within the DNA damage and oxidative stress arena and provides insight into their mechanistic interdependence and their usefulness in accurately modeling cancer and other risks in astronauts exposed to space radiation. Our ultimate goals are to examine potential links and crosstalk between early response modules activated by charged particle exposure, to identify critical areas that require further research and to use these data to reduced uncertainties in modeling cancer risk for astronauts. A clearer understanding of the links between early mechanistic aspects of high-LET response and later surrogate cancer end points could reveal key nodes that can be therapeutically targeted to mitigate the health effects from charged particle exposures.
Collapse
Affiliation(s)
- D M Sridharan
- a Lawrence Berkeley National Laboratory, Berkeley, California
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Grygoryev D, Dan C, Gauny S, Eckelmann B, Ohlrich AP, Connolly M, Lasarev M, Grossi G, Kronenberg A, Turker MS. Autosomal mutants of proton-exposed kidney cells display frequent loss of heterozygosity on nonselected chromosomes. Radiat Res 2014; 181:452-63. [PMID: 24758577 DOI: 10.1667/rr13654.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
High-energy protons found in the space environment can induce mutations and cancer, which are inextricably linked. We hypothesized that some mutants isolated from proton-exposed kidneys arose through a genome-wide incident that causes loss of heterozygosity (LOH)-generating mutations on multiple chromosomes (termed here genomic LOH). To test this hypothesis, we examined 11 pairs of nonselected chromosomes for LOH events in mutant cells isolated from the kidneys of mice exposed to 4 or 5 Gy of 1 GeV protons. The mutant kidney cells were selected for loss of expression of the chromosome 8-encoded Aprt gene. Genomic LOH events were also assessed in Aprt mutants isolated from isogenic cultured kidney epithelial cells exposed to 5 Gy of protons in vitro. Control groups were spontaneous Aprt mutants and clones isolated without selection from the proton-exposed kidneys or cultures. The in vivo results showed significant increases in genomic LOH events in the Aprt mutants from proton-exposed kidneys when compared with spontaneous Aprt mutants and when compared with nonmutant (i.e., nonselected) clones from the proton-exposed kidneys. A bias for LOH events affecting chromosome 14 was observed in the proton-induced Aprt mutants, though LOH for this chromosome did not confer increased radiation resistance. Genomic LOH events were observed in Aprt mutants isolated from proton-exposed cultured kidney cells; however the incidence was fivefold lower than in Aprt mutants isolated from exposed intact kidneys, suggesting a more permissive environment in the intact organ and/or the evolution of kidney clones prior to their isolation from the tissue. We conclude that proton exposure creates a subset of viable cells with LOH events on multiple chromosomes, that these cells form and persist in vivo, and that they can be isolated from an intact tissue by selection for a mutation on a single chromosome.
Collapse
Affiliation(s)
- Dmytro Grygoryev
- a Center for Research on Occupational and Environmental Toxicology (CROET), Oregon Health & Science University, Portland, Oregon 97239
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Kma L. Plant Extracts and Plant-Derived Compounds: Promising Players in Countermeasure Strategy Against Radiological Exposure: A Review. Asian Pac J Cancer Prev 2014; 15:2405-25. [DOI: 10.7314/apjcp.2014.15.6.2405] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
19
|
Mukherjee D, Coates PJ, Lorimore SA, Wright EG. Responses to ionizing radiation mediated by inflammatory mechanisms. J Pathol 2014; 232:289-99. [PMID: 24254983 DOI: 10.1002/path.4299] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 10/30/2013] [Accepted: 11/05/2013] [Indexed: 01/20/2023]
Abstract
Since the early years of the twentieth century, the biological consequences of exposure to ionizing radiation have been attributed solely to mutational DNA damage or cell death induced in irradiated cells at the time of exposure. However, numerous observations have been at variance with this dogma. In the 1950s, attention was drawn to abscopal effects in areas of the body not directly irradiated. In the 1960s reports began appearing that plasma factors induced by irradiation could affect unirradiated cells, and since 1990 a growing literature has documented an increased rate of DNA damage in the progeny of irradiated cells many cell generations after the initial exposure (radiation-induced genomic instability) and responses in non-irradiated cells neighbouring irradiated cells (radiation-induced bystander effects). All these studies have in common the induction of effects not in directly irradiated cells but in unirradiated cells as a consequence of intercellular signalling. Recently, it has become clear that all the various effects demonstrated in vivo may reflect an ongoing inflammatory response to the initial radiation-induced injury that, in a genotype-dependent manner, has the potential to contribute primary and/or ongoing damage displaced in time and/or space from the initial insult. Importantly, there is direct evidence that non-steroidal anti-inflammatory drug treatment reduces such damage in vivo. These new findings highlight the importance of tissue responses and indicate additional mechanisms of radiation action, including the likelihood that radiation effects are not restricted to the initiation stage of neoplastic diseases, but may also contribute to tumour promotion and progression. The various developments in understanding the responses to radiation exposures have implications not only for radiation pathology but also for therapeutic interventions.
Collapse
Affiliation(s)
- Debayan Mukherjee
- Centre for Oncology and Molecular Medicine, University of Dundee Medical School, Dundee, DD1 9SY, UK
| | | | | | | |
Collapse
|
20
|
Mothersill C, Fernandez-Palomo C, Fazzari J, Smith R, Schültke E, Bräuer-Krisch E, Laissue J, Schroll C, Seymour C. Transmission of signals from rats receiving high doses of microbeam radiation to cage mates: an inter-mammal bystander effect. Dose Response 2014; 12:72-92. [PMID: 24659934 PMCID: PMC3960955 DOI: 10.2203/dose-response.13-011.mothersill] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Inter-animal signaling from irradiated to non-irradiated organisms has been demonstrated for whole body irradiated mice and also for fish. The aim of the current study was to look at radiotherapy style limited exposure to part of the body using doses relevant in preclinical therapy. High dose homogenous field irradiation and the use of irradiation in the microbeam radiation therapy mode at the European Synchrotron Radiation Facility (ESRF) at Grenoble was tested by giving high doses to the right brain hemisphere of the rat. The right and left cerebral hemispheres and the urinary bladder were later removed to determine whether abscopal effects could be produced in the animals and also whether effects occurred in cage mates housed with them. The results show strong bystander signal production in the contra-lateral brain hemisphere and weaker effects in the distant bladder of the irradiated rats. Signal strength was similar or greater in each tissue in the cage mates housed for 48hrs with the irradiated rats. Our results support the hypothesis that proximity to an irradiated animal induces signalling changes in an unirradiated partner. If similar signaling occurs between humans, the results could have implications for caregivers and hospital staff treating radiotherapy patients.
Collapse
Affiliation(s)
- Carmel Mothersill
- Medical Physics and Applied Radiation Sciences Department, McMaster University, Hamilton, Ontario, Canada
| | - Cristian Fernandez-Palomo
- Medical Physics and Applied Radiation Sciences Department, McMaster University, Hamilton, Ontario, Canada
| | - Jennifer Fazzari
- Medical Physics and Applied Radiation Sciences Department, McMaster University, Hamilton, Ontario, Canada
| | - Richard Smith
- Medical Physics and Applied Radiation Sciences Department, McMaster University, Hamilton, Ontario, Canada
| | - Elisabeth Schültke
- Stereotactic Neurosurgery and Laboratory for Molecular Neurosurgery, Freiburg University Medical Centre, Freiburg, Germany
| | | | - Jean Laissue
- Institute of Pathology, University of Bern, Switzerland
| | - Christian Schroll
- Stereotactic Neurosurgery and Laboratory for Molecular Neurosurgery, Freiburg University Medical Centre, Freiburg, Germany
| | - Colin Seymour
- Medical Physics and Applied Radiation Sciences Department, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
21
|
Ha CT, Li XH, Fu D, Xiao M, Landauer MR. Genistein nanoparticles protect mouse hematopoietic system and prevent proinflammatory factors after gamma irradiation. Radiat Res 2013; 180:316-25. [PMID: 23952576 DOI: 10.1667/rr3326.1] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Previous studies demonstrated that genistein protects mice from radiation-induced bone marrow failure. To overcome genistein's extremely low water solubility, a nanoparticle suspension of genistein has been formulated for more rapid dissolution. In the current study, we evaluated the radioprotective effects of a nanoparticle formulation of genistein on survival and hematopoietic recovery in mice exposed to total-body gamma irradiation. A single intramuscular injection of a saline-based genistein nanosuspension (150 mg/kg) administered to CD2F1 mice 24 h before 9.25 Gy (60)Co radiation exposure resulted in a 30-day survival rate of 95% compared to 25% in vehicle-treated animals. In mice irradiated at 7 Gy, the genistein nanosuspension increased mouse bone marrow cellularity from approximately 2.9% (vehicle treated) to 28.3% on day 7 postirradiation. Flow cytometry analysis demonstrated decreased radiation-induced hematopoietic stem and progenitor cell (HSPC, Lineage(-)/cKit(+)) death from 77.0% (vehicle) to 43.9% (genistein nanosuspension) with a significant recovery of clonogenicity 7 days after irradiation. The genistein nanosuspension also attenuated the radiation-induced elevation of proinflammatory factors interleukin 1 beta (IL-1β), IL-6 and cyclooxygenase-2 (COX-2) in mouse bone marrow and spleen, which may contribute to protecting HSPCs.
Collapse
Affiliation(s)
- Cam T Ha
- Radiation Countermeasures Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| | | | | | | | | |
Collapse
|
22
|
Abstract
The concept of the tumour microenvironment recognizes that the interplay between cancer cells and stromal cells is a crucial determinant of cancer growth. In this Perspectives article, we propose the novel concept that the tumour microenvironment is built through rate-limiting steps during multistage carcinogenesis. Construction of a 'precancer niche' is a necessary and early step that is required for initiated cells to survive and evolve; subsequent niche expansion and maturation accompany tumour promotion and progression, respectively. As such, cancer niches represent an emergent property of a tumour that could be a robust target for cancer prevention and therapy.
Collapse
Affiliation(s)
- Mary Helen Barcellos-Hoff
- The Department of Radiation Oncology, New York University School of Medicine, New York, New York 10016, USA.
| | | | | |
Collapse
|
23
|
Lorimore SA, Rastogi S, Mukherjee D, Coates PJ, Wright EG. The influence of p53 functions on radiation-induced inflammatory bystander-type signaling in murine bone marrow. Radiat Res 2013; 179:406-15. [PMID: 23578188 DOI: 10.1667/rr3158.2] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Radiation-induced bystander and abscopal effects, in which DNA damage is produced by inter-cellular communication, indicate mechanisms of generating damage in addition to those observed in directly irradiated cells. In this article, we show that the bone marrow of irradiated p53(+/+) mice, but not p53(-/-) mice, produces the inflammatory pro-apoptotic cytokines FasL and TNF-α able to induce p53-independent apoptosis in vitro in nonirradiated p53(-/-) bone marrow cells. Using a congenic sex-mismatch bone marrow transplantation protocol to generate chimeric mice, p53(-/-) hemopoietic cells functioning in a p53(+/+) bone marrow stromal microenvironment exhibited greater cell killing after irradiation than p53(-/-) hemopoietic cells in a p53(-/-) microenvironment. Cytogenetic analysis demonstrated fewer damaged p53(-/-) cells in a p53(+/+) microenvironment than p53(-/-) cells in a p53(-/-) microenvironment. Using the two different model systems, the findings implicate inflammatory tissue processes induced as a consequence of p53-dependent cellular responses to the initial radiation damage, producing cytokines that subsequently induce ongoing p53-independent apoptosis. As inactivation of the p53 tumor suppressor pathway is a common event in malignant cells developing in a stromal microenvironment that has normal p53 function, the signaling processes identified in the current investigations have potential implications for disease pathogenesis and therapy.
Collapse
Affiliation(s)
- Sally A Lorimore
- University of Dundee, Centre for Oncology and Molecular Medicine, Division of Medical Science, Ninewells Hospital and Medical School, Dundee, Scotland, United Kingdom
| | | | | | | | | |
Collapse
|
24
|
Rastogi S, Boylan M, Wright EG, Coates PJ. Interactions of apoptotic cells with macrophages in radiation-induced bystander signaling. Radiat Res 2012; 179:135-45. [PMID: 23237586 DOI: 10.1667/rr2969.1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Nontargeted effects that result in ongoing cellular and tissue damage show genotype-dependency in murine models with CBA/Ca, but not C57BL/6, exhibiting sensitivity to induced genomic instability. In vivo, radiation exposure is associated with genotype-dependent macrophage activation, and these cells are a source of bystander signaling involving cytokines and reactive oxygen and nitrogen species. The mechanisms responsible for macrophage activation and production of damaging bystander signals after irradiation are unclear. Macrophages from CBA/Ca exhibit an M1 (proinflammatory) phenotype compared to the M2 (anti-inflammatory) phenotype of C57BL/6 macrophages. Using the murine RAW264.7 macrophage-like cell line, we show that the ability of macrophages to interact with apoptotic cells and their responses to interaction varies significantly according to macrophage phenotype. Nonstimulated and M2 macrophages induce anti-inflammatory markers arginase and TGFβ after engulfment of apoptotic cells. In contrast, M1 macrophages do not induce anti-inflammatory responses, but express the proinflammatory markers NOS2, IL-6, TNFα, superoxide and NO, able to contribute to a damaging microenvironment. Macrophages stimulated with both inflammatory and anti-inflammatory agents prior to exposure to apoptotic cells induce a mixed response. The results indicate a complex cross-talk between macrophages and apoptotic cells and demonstrate that phagocytic clearance of apoptotic cells induced by genotoxic stress can produce microenvironmental responses consistent with the induction of a chromosomal instability phenotype in sensitive CBA/Ca mice with M1 macrophage activation, but not in resistant C57BL/6 mice with M2 macrophage activation. Modulation of macrophage phenotypes may represent a novel approach for reducing the nontargeted effects of radiation.
Collapse
Affiliation(s)
- Shubhra Rastogi
- Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee, DD1 9SY, United Kingdom.
| | | | | | | |
Collapse
|
25
|
Karotki AV, Baverstock K. What mechanisms/processes underlie radiation-induced genomic instability? Cell Mol Life Sci 2012; 69:3351-60. [PMID: 22955377 PMCID: PMC11115179 DOI: 10.1007/s00018-012-1148-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 08/22/2012] [Accepted: 08/23/2012] [Indexed: 01/11/2023]
Abstract
Radiation-induced genomic instability is a modification of the cell genome found in the progeny of irradiated somatic and germ cells but that is not confined on the initial radiation-induced damage and may occur de novo many generations after irradiation. Genomic instability in the germ line does not follow Mendelian segregation and may have unpredictable outcomes in every succeeding generation. This phenomenon, for which there is extensive experimental data and some evidence in human populations exposed to ionising radiation, is not taken into account in health risk assessments. It poses an unknown morbidity/mortality burden. Based on experimental data derived over the last 20 years (up to January 2012) six mechanistic explanations for the phenomenon have been proposed in the peer-reviewed literature. This article compares these hypotheses with the empirical data to test their fitness to explain the phenomenon. As a conclusion, the most convincing explanation of radiation-induced genomic instability attributes it to an irreversible regulatory change in the dynamic interaction network of the cellular gene products, as a response to non-specific molecular damage, thus entailing the rejection of the machine metaphor for the cell in favour of one appropriate to a complex dissipative dynamic system, such as a whirlpool. It is concluded that in order to evaluate the likely morbidity/mortality associated with radiation-induced genomic instability, it will be necessary to study the damage to processes by radiation rather than damage to molecules.
Collapse
Affiliation(s)
- Andrei V. Karotki
- Radiation Group, International Agency for Research on Cancer, International Agency for Research on Cancer, 150 Cours A. Thomas, 69372 Lyon, France
| | - Keith Baverstock
- Department of Environmental Science, University of Eastern Finland, Kuopio Campus, PL 1627, 70211 Kuopio, Finland
| |
Collapse
|
26
|
McAllister KA, Lorimore SA, Wright EG, Coates PJ. In vivo interactions between ionizing radiation, inflammation and chemical carcinogens identified by increased DNA damage responses. Radiat Res 2012; 177:584-93. [PMID: 22463680 DOI: 10.1667/rr2690.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Exposure to ionizing radiation or a variety of chemical agents is known to increase the risk of developing malignancy and many tumors have been linked to inflammatory processes. In most studies, the potentially harmful effects of ionizing radiation or other agents are considered in isolation, mainly due to the large number of experiments required to assess the effects of mixed exposures with different doses and different schedules, and the length of time and expense of studies using disease as the measure of outcome. Here, we have used short-term DNA damage responses to identify interactive effects of mixed exposures. The data demonstrate that exposure to ionizing radiation on two separate occasions ten days apart leads to an increase in the percentage of cells with a sub-G(0) DNA content compared to cells exposed only once, and this is a greater than additive effect. Short-term measurements of p53 stabilization, induction of p21/Cdkn1a and of apoptosis also identify these interactive effects. We also demonstrate similar interactive effects of radiation with the mutagenic chemical methyl-nitrosourea and with a nonspecific pro-inflammatory agent, lipopolysaccharide. The magnitude of the interactive effects is greater in cells taken from mice first exposed as juveniles compared to adults. These data indicate that short-term measurements of DNA damage and response to damage are useful for the identification of interactions between ionizing radiation and other agents.
Collapse
Affiliation(s)
- K A McAllister
- Centre for Oncology and Molecular Medicine, Division of Medical Sciences, University of Dundee, Ninewells Hospital and Medical School, Dundee DD1 9SY, United Kingdom
| | | | | | | |
Collapse
|
27
|
Kuffler DP. Hyperbaric oxygen therapy: can it prevent irradiation-induced necrosis? Exp Neurol 2012; 235:517-27. [PMID: 22465460 DOI: 10.1016/j.expneurol.2012.03.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 02/27/2012] [Accepted: 03/17/2012] [Indexed: 10/28/2022]
Abstract
Radiosurgery is an important non-invasive procedure for the treatment of tumors and vascular malformations. However, in addition to killing target tissues, cranial irradiation induces damage to adjacent healthy tissues leading to neurological deterioration in both pediatric and adult patients, which is poorly understood and insufficiently treatable. To minimize irradiation damage to healthy tissue, not the optimal therapeutic irradiation dose required to eliminate the target lesion is used but lower doses. Although the success rate of irradiation surgery is about 95%, 5% of patients suffer problems, most commonly neurological, that are thought to be a direct consequence of irradiation-induced inflammation. Although no direct correlation has been demonstrated, the appearance and disappearance of inflammation that develops following irradiation commonly parallel the appearance and disappearance of neurological side effects that are associated with the neurological function of the irradiated brain regions. These observations have led to the hypothesis that brain inflammation is causally related to the observed neurological side effects. Studies indicate that hyperbaric oxygen therapy (HBOT) applied after the appearance of irradiation-induced neurological side effects reduces the incidence and severity of those side effects. This may result from HBOT reducing inflammation, promoting angiogenesis, and influencing other cellular functions thereby suppressing events that cause the neurological side effects. However, it would be significantly better for the patient if rather than waiting for neurological side effects to become manifest they could be avoided. This review examines irradiation-induced neurological side effects, methods that minimize or resolve those side effects, and concludes with a discussion of whether HBOT applied following irradiation, but before manifestation of neurological side effects may prevent or reduce the appearance of irradiation-induced neurological side effects.
Collapse
Affiliation(s)
- Damien P Kuffler
- Institute of Neurobiology, University of Puerto Rico, Medical Sciences Campus, Puerto Rico.
| |
Collapse
|