1
|
Tavakol DN, Nash TR, Kim Y, Graney PL, Liberman M, Fleischer S, Lock RI, O'Donnell A, Andrews L, Ning D, Yeager K, Harken A, Deoli N, Amundson SA, Garty G, Leong KW, Brenner DJ, Vunjak‐Novakovic G. Modeling the Effects of Protracted Cosmic Radiation in a Human Organ-on-Chip Platform. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401415. [PMID: 38965824 PMCID: PMC11558103 DOI: 10.1002/advs.202401415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/18/2024] [Indexed: 07/06/2024]
Abstract
Galactic cosmic radiation (GCR) is one of the most serious risks posed to astronauts during missions to the Moon and Mars. Experimental models capable of recapitulating human physiology are critical to understanding the effects of radiation on human organs and developing radioprotective measures against space travel exposures. The effects of systemic radiation are studied using a multi-organ-on-a-chip (multi-OoC) platform containing engineered tissue models of human bone marrow (site of hematopoiesis and acute radiation damage), cardiac muscle (site of chronic radiation damage) and liver (site of metabolism), linked by vascular circulation with an endothelial barrier separating individual tissue chambers from the vascular perfusate. Following protracted neutron radiation, the most damaging radiation component in deep space, a greater deviation of tissue function is observed as compared to the same cumulative dose delivered acutely. Further, by characterizing engineered bone marrow (eBM)-derived immune cells in circulation, 58 unique genes specific to the effects of protracted neutron dosing are identified, as compared to acutely irradiated and healthy tissues. It propose that this bioengineered platform allows studies of human responses to extended radiation exposure in an "astronaut-on-a-chip" model that can inform measures for mitigating cosmic radiation injury.
Collapse
Affiliation(s)
| | - Trevor R. Nash
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10032USA
| | - Youngbin Kim
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10032USA
| | - Pamela L. Graney
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10032USA
| | - Martin Liberman
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10032USA
| | - Sharon Fleischer
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10032USA
| | - Roberta I. Lock
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10032USA
| | - Aaron O'Donnell
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10032USA
| | - Leah Andrews
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10032USA
| | - Derek Ning
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10032USA
| | - Keith Yeager
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10032USA
| | - Andrew Harken
- Center for Radiological ResearchColumbia UniversityNew YorkNY10032USA
| | - Naresh Deoli
- Center for Radiological ResearchColumbia UniversityNew YorkNY10032USA
| | - Sally A. Amundson
- Center for Radiological ResearchColumbia UniversityNew YorkNY10032USA
| | - Guy Garty
- Center for Radiological ResearchColumbia UniversityNew YorkNY10032USA
| | - Kam W. Leong
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10032USA
| | - David J. Brenner
- Center for Radiological ResearchColumbia UniversityNew YorkNY10032USA
| | - Gordana Vunjak‐Novakovic
- Department of Biomedical EngineeringDepartment of Medicine, and College of Dental MedicineColumbia UniversityNew YorkNY10032USA
| |
Collapse
|
2
|
Afshari N, Koturbash I, Boerma M, Newhauser W, Kratz M, Willey J, Williams J, Chancellor J. A Review of Numerical Models of Radiation Injury and Repair Considering Subcellular Targets and the Extracellular Microenvironment. Int J Mol Sci 2024; 25:1015. [PMID: 38256089 PMCID: PMC10816679 DOI: 10.3390/ijms25021015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Astronauts in space are subject to continuous exposure to ionizing radiation. There is concern about the acute and late-occurring adverse health effects that astronauts could incur following a protracted exposure to the space radiation environment. Therefore, it is vital to consider the current tools and models used to describe and study the organic consequences of ionizing radiation exposure. It is equally important to see where these models could be improved. Historically, radiobiological models focused on how radiation damages nuclear deoxyribonucleic acid (DNA) and the role DNA repair mechanisms play in resulting biological effects, building on the hypotheses of Crowther and Lea from the 1940s and 1960s, and they neglected other subcellular targets outside of nuclear DNA. The development of these models and the current state of knowledge about radiation effects impacting astronauts in orbit, as well as how the radiation environment and cellular microenvironment are incorporated into these radiobiological models, aid our understanding of the influence space travel may have on astronaut health. It is vital to consider the current tools and models used to describe the organic consequences of ionizing radiation exposure and identify where they can be further improved.
Collapse
Affiliation(s)
- Nousha Afshari
- Department of Physics and Astronomy, Louisiana State University, Baton Rouge, LA 70803, USA; (N.A.); (W.N.)
| | - Igor Koturbash
- Department of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Marjan Boerma
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Wayne Newhauser
- Department of Physics and Astronomy, Louisiana State University, Baton Rouge, LA 70803, USA; (N.A.); (W.N.)
| | - Maria Kratz
- Department of Biological and Agricultural Engineering, Louisiana State University, Baton Rouge, LA 70803, USA;
| | - Jeffrey Willey
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA;
| | - Jacqueline Williams
- School of Medicine and Dentistry, University of Rochester Medical Center, Rochester, NY 14642, USA;
| | - Jeffery Chancellor
- Department of Physics and Astronomy, Louisiana State University, Baton Rouge, LA 70803, USA; (N.A.); (W.N.)
- Department of Preventive Medicine and Population Health, University of Texas Medical Branch, Galveston, TX 77555, USA
- Outer Space Institute, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
3
|
Zhang P, Wu Y, Piao C, Song Y, Zhao Y, Lyu Y, Sun Q, Liu J. Alteration of genome-wide DNA methylation in non-uranium miners induced by high level radon exposure. MUTATION RESEARCH. GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2023; 891:503683. [PMID: 37770140 DOI: 10.1016/j.mrgentox.2023.503683] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 08/15/2023] [Accepted: 08/17/2023] [Indexed: 10/03/2023]
Abstract
In China, according to statistics about underground non-uranium mine radon levels, 15% exceed the national standard intervention level of 1000 Bq/m3, and some mines may exceed 10,000 Bq/m3. The relationship between radon exposure in underground miners and lung cancer has already been established, but the mechanisms and biological processes underlying it are poorly understood. In order to identify the genome-wide DNA methylation profile associated with long-term radon exposure, we performed the Infinium Human Methylation 850 K BeadChip measurement in whole blood samples obtained from 15 underground non-uranium miners and 10 matched aboveground control workers. Radon concentrations in the air of workplaces and living environments were measured by CR-39 radon detectors, and annual effective doses were calculated using the detection data. Under the high radon concentration with an average value of 12,700 Bq·m-3, a total of 165 significant differentially methylated positions (127 hypermethylated sites and 38 hypomethylated sites) annotated to 71 genes were identified in underground miners (|Δβ| ≥ 0.10, p < 0.05), and the average DNA methylation level of 165 DMPs was significantly higher than that of the control workers. Most DMPs were found on chromosome 1, and approximately one-quarter of them were located in genomic promoter regions. Through bioinformatics analysis and pyrosequencing validation, five candidate genes differentially methylated by radon, including TIMP2, EMP2, CPT1B, AMD1 and SLC43A2 were identified. GO and KEGG analysis implicated that long term radon exposure could induce the lung cancer related biological processes such as cell adhesion and cellular polarity maintenance. Our study provides evidence for the alterations of genome-wide DNA methylation profiles induced by long-term high level radon exposure, and new insights into searching for carcinogenic biomarkers of high radon exposure in future studies.
Collapse
Affiliation(s)
- Pinhua Zhang
- Key Laboratory of Radiological Protection and Nuclear Emergency, China CDC, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing 100088, China
| | - Yunyun Wu
- Key Laboratory of Radiological Protection and Nuclear Emergency, China CDC, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing 100088, China
| | - Chunnan Piao
- Key Laboratory of Radiological Protection and Nuclear Emergency, China CDC, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing 100088, China
| | - Yanchao Song
- Key Laboratory of Radiological Protection and Nuclear Emergency, China CDC, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing 100088, China
| | - Yanfang Zhao
- The Third People's Hospital of Henan Province, Henan Hospital for Occupational Diseases, Zhengzhou 450052, China
| | - Yumin Lyu
- The Third People's Hospital of Henan Province, Henan Hospital for Occupational Diseases, Zhengzhou 450052, China
| | - Quanfu Sun
- Key Laboratory of Radiological Protection and Nuclear Emergency, China CDC, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing 100088, China
| | - Jianxiang Liu
- Key Laboratory of Radiological Protection and Nuclear Emergency, China CDC, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing 100088, China.
| |
Collapse
|
4
|
Liu T, Wang H, Shen H, Du Z, Wan Z, Li J, Zhang X, Li Z, Yang N, Yang Y, Chen Y, Gao F, Cao K. TLR4 Agonist MPLA Ameliorates Heavy-Ion Radiation Damage via Regulating DNA Damage Repair and Apoptosis. Radiat Res 2023; 200:127-138. [PMID: 37302147 DOI: 10.1667/rade-22-00200.1] [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: 11/22/2022] [Accepted: 05/10/2023] [Indexed: 06/13/2023]
Abstract
Heavy-ion radiation received during radiotherapy as well as the heavy-ion radiation received during space flight are equally considered harmful. Our previous study showed that TLR4 low toxic agonist, monophosphoryl lipid A (MPLA), alleviated radiation injury resulting from exposure to low-LET radiation. However, the role and mechanism of MPLA in heavy-ion-radiation injury are unclear. This study aimed to investigate the role of MPLA on radiation damage. Our data showed that MPLA treatment alleviated the heavy-ion-induced damage to microstructure and the spleen and testis indexes. The number of karyocytes in the bone marrow from the MPLA-treated group was higher than that in the irradiated group. Meanwhile, western blotting analysis of intestine proteins showed that pro-apoptotic proteins (cleaved-caspase3 and Bax) were downregulated while anti-apoptotic proteins (Bcl-2) were upregulated in the MPLA-treated group. Our in vitro study demonstrated that MPLA significantly improved cell proliferation and inhibited cell apoptosis after irradiation. Moreover, immunofluorescence staining and quantification of nucleic γ-H2AX and 53BP1 foci also suggested that MPLA significantly attenuated cellular DNA damage repair. Collectively, the above evidence supports the potential ability of MPLA to protect against heavy-ion-radiation injury by inhibiting apoptosis and alleviating DNA damage in vivo and vitro, which could be a promising medical countermeasure for the prevention of heavy-ion-radiation injury.
Collapse
Affiliation(s)
- Tingting Liu
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Hang Wang
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Hui Shen
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Zhipeng Du
- School of Public Health and Management, Wenzhou Medical University, University Town, Wenzhou, Zhejiang, 325035, China
| | - Zhijie Wan
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Junshi Li
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Xide Zhang
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Zhuqing Li
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Nan Yang
- Pharmacy Department, Qingdao Special Servicemen Recuperation Center of CPLA Navy, Qingdao 266071, China
| | - Yanyong Yang
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Yuanyuan Chen
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Fu Gao
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Kun Cao
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| |
Collapse
|
5
|
More efficient induction of genotoxicity by high-LET Fe-particle radiation than low-LET X-ray radiation at low doses. RADIATION MEDICINE AND PROTECTION 2022. [DOI: 10.1016/j.radmp.2022.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
6
|
Weiss M, Nikisher B, Haran H, Tefft K, Adams J, Edwards JG. High throughput screen of small molecules as potential countermeasures to galactic cosmic radiation induced cellular dysfunction. LIFE SCIENCES IN SPACE RESEARCH 2022; 35:76-87. [PMID: 36336373 DOI: 10.1016/j.lssr.2022.06.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/23/2022] [Accepted: 06/16/2022] [Indexed: 06/16/2023]
Abstract
Space travel increases galactic cosmic ray exposure to flight crews and this is significantly elevated once travel moves beyond low Earth orbit. This includes combinations of high energy protons and heavy ions such as 56Fe or 16O. There are distinct differences in the biological response to low-energy transfer (x-rays) or high-energy transfer (High-LET). However, given the relatively low fluence rate of exposure during flight operations, it might be possible to manage these deleterious effects using small molecules currently available. Virtually all reports to date examining small molecule management of radiation exposure are based on low-LET challenges. To that end an FDA approved drug library (725 drugs) was used to perform a high throughput screen of cultured cells following exposure to galactic cosmic radiation. The H9c2 myoblasts, ES-D3 pluripotent cells, and Hy926 endothelial cell lines were exposed to a single exposure (75 cGy) using the 5-ion GCRsim protocol developed at the NASA Space Radiation Laboratory (NSRL). Following GCR exposure cells were maintained for up to two weeks. For each drug (@10µM), a hierarchical cumulative score was developed incorporating measures of mitochondrial and cellular function, oxidant stress and cell senescence. The top 160 scores were retested following a similar protocol using 1µM of each drug. Within the 160 drugs, 33 are considered to have an anti-inflammatory capacity, while others also indirectly suppressed pro-inflammatory pathways or had noted antioxidant capacity. Lead candidates came from different drug classes that included angiotensin converting enzyme inhibitors or AT1 antagonists, COX2 inhibitors, as well as drugs mediated by histamine receptors. Surprisingly, different classes of anti-diabetic medications were observed to be useful including sulfonylureas and metformin. Using a hierarchical decision structure, we have identified several lead candidates. That no one drug or even drug class was completely successful across all parameters tested suggests the complexity of managing the consequences of galactic cosmic radiation exposure.
Collapse
Affiliation(s)
- M Weiss
- Department of Physiology, New York Medical College, Valhalla, New York
| | - B Nikisher
- Department of Physiology, New York Medical College, Valhalla, New York
| | - H Haran
- Department of Physiology, New York Medical College, Valhalla, New York
| | - K Tefft
- Department of Physiology, New York Medical College, Valhalla, New York
| | - J Adams
- Department of Physiology, New York Medical College, Valhalla, New York
| | - J G Edwards
- Department of Physiology, New York Medical College, Valhalla, New York.
| |
Collapse
|
7
|
Ewing LE, Pathak R, Landes RD, Skinner CM, Binz R, Young SG, Riklon S, Stahr S, Su J, Boerma M, McElfish PA, Hauer-Jensen M, Koturbash I. Cytogenetic and epigenetic aberrations in peripheral lymphocytes of northwest Arkansas Marshallese. Int J Radiat Biol 2022; 99:644-655. [PMID: 35939319 PMCID: PMC9929030 DOI: 10.1080/09553002.2022.2110319] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/28/2022] [Accepted: 07/22/2022] [Indexed: 10/15/2022]
Abstract
PURPOSE Nuclear weapons testing in the northern Marshall Islands between 1946 and 1958 resulted in ionizing radiation (IR) exposure of the thousands of Marshallese. Furthermore, numerous islands were contaminated by radioactive fallout. Significant increases in cancer and metabolic syndrome incidences have been reported among Marshallese, and potential for further increases looms due to the latency of radiation-induced health effects. The purpose of this study was to investigate the genetic and epigenetic effects of exposure to IR that could be associated with radiation-induced disease among the Northwest Arkansas (NWA) Marshallese. MATERIALS AND METHODS We performed analysis of chromosomal aberrations and DNA methylation based on residential and exposure history of NWA Marshallese. RESULTS Analysis of chromosomal aberrations demonstrated higher incidence of genetic rearrangements in women with self-reported history of radiation exposure (95% CI: 0.10, 1.22; p=.022). Further clustering of study participants based on their residential history demonstrated that participants who spent substantial amounts of time (≥6 months) in the northern atolls (thus, in the proximity of nuclear tests) before 1980 had more chromosomal aberrations than their peers who lived only in the southern atolls (95% CI: 0.08, -0.95; p=.021), and that this difference was driven by women. A relationship between the time spent in the northern atolls and increase in chromosomal aberrations was observed: 0.31 increase in chromosomal aberrations for every 10 years spent at northern atolls (95% CI: 0.06, 0.57; p=.020). Finally, significant inverse correlations between the chromosomal aberrations and the extent of DNA methylation of four LINE-1 elements L1PA2, L1PA16, L1PREC1, and L1P4B were identified. CONCLUSIONS The results of this study provide first evidence of the presence of stable genetic and epigenetic rearrangements in peripheral lymphocytes of NWA Marshallese and warrant further studies to analyze the role of radiation exposure in health disparities experienced by this Pacific Island nation.
Collapse
Affiliation(s)
- Laura E. Ewing
- Department of Environmental and Occupational Health, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, 4301 W Markham Str, Little Rock, Arkansas, USA
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, 4301 W Markham Str, Little Rock, Arkansas, USA
| | - Rupak Pathak
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, 4301 W Markham Str, Little Rock, AR, USA
| | - Reid D. Landes
- Department of Biostatistics, College of Medicine, University of Arkansas for Medical Sciences, 4301 W Markham Str, Little Rock, Arkansas, USA
| | - Charles M. Skinner
- Department of Environmental and Occupational Health, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, 4301 W Markham Str, Little Rock, Arkansas, USA
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Arkansas for Medical Sciences, 4301 W Markham Str, Little Rock, Arkansas, USA
| | - Regina Binz
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, 4301 W Markham Str, Little Rock, AR, USA
| | - Sean G. Young
- Department of Environmental and Occupational Health, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, 4301 W Markham Str, Little Rock, Arkansas, USA
| | - Sheldon Riklon
- College of Medicine, University of Arkansas for Medical Sciences Northwest, Fayetteville, AR, USA
| | - Shelbie Stahr
- Department of Epidemiology, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, 4301 W Markham Str, Little Rock, Arkansas, USA
| | - Joseph Su
- Department of Epidemiology, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, 4301 W Markham Str, Little Rock, Arkansas, USA
| | - Marjan Boerma
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, 4301 W Markham Str, Little Rock, AR, USA
| | - Pearl A. McElfish
- College of Medicine, University of Arkansas for Medical Sciences Northwest, Fayetteville, AR, USA
| | - Martin Hauer-Jensen
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, 4301 W Markham Str, Little Rock, AR, USA
| | - Igor Koturbash
- Department of Environmental and Occupational Health, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, 4301 W Markham Str, Little Rock, Arkansas, USA
| |
Collapse
|
8
|
Yushkova E. Radiobiological features in offspring of natural populations of Drosophila melanogaster after Chernobyl accident. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2022; 63:84-97. [PMID: 35275441 DOI: 10.1002/em.22476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/24/2022] [Accepted: 03/08/2022] [Indexed: 06/14/2023]
Abstract
In their natural habitats, populations of organisms are faced with different levels of chronic low-intensity radiation, causing a wide range of radiobiological effects (from radiosensitivity to radioadaptive response and hormesis). In this study, specimens of Drosophila melanogaster were selected from territories of the Chernobyl nuclear power plant with different levels of radioactive contamination. The isogenic stocks derived from these specimens represent the genetic systems of current populations and make it possible to study radioresistance and its mechanisms in future generations under controlled laboratory conditions. Previous studies have shown that transgenerational radiation effects at the level of lethal mutations and survival rate are unstable and depend not only on the level of chronic low-intensity irradiation, but also on other factors. A single acute irradiation exposure of offspring whose parents inhabited a site with a higher level of chronic irradiation made it possible to reveal pronounced radioresistant features in the offspring. And the offspring whose parents were exposed to radiation levels close to the natural radiation background, on the contrary, acquired radiosensitive features. Their response to acute exposure includes a high-frequency of lethal mutations and a short lifespan. The differential response to different levels of chronic parental exposure is caused by differences in the activities of certain transposons that destabilize the genome. Our data contribute to the understanding of genetic and epigenetic mechanisms (via transposon activity) of the effect of parental radiation exposure on the health and adaptive potential of populations affected by the technogenically increased radiation background.
Collapse
Affiliation(s)
- Elena Yushkova
- Institute of Biology of Komi Scientific Centre of the Ural Branch of the Russian Academy of Science, Syktyvkar, Russia
| |
Collapse
|
9
|
Kuzmina NS. Radiation-Induced DNA Methylation Disorders: In Vitro and In Vivo Studies. BIOL BULL+ 2022. [DOI: 10.1134/s1062359021110066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
10
|
Human mesenchymal stromal cells maintain their stem cell traits after high-LET particle irradiation - Potential implications for particle radiotherapy and manned space missions. Cancer Lett 2022; 524:172-181. [PMID: 34688844 DOI: 10.1016/j.canlet.2021.10.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/19/2021] [Accepted: 10/11/2021] [Indexed: 12/14/2022]
Abstract
The influence of high-linear energy transfer (LET) particle radiation on the functionalities of mesenchymal stromal cells (MSCs) is largely unknown. Here, we analyzed the effects of proton (1H), helium (4He), carbon (12C) and oxygen (16O) ions on human bone marrow-MSCs. Cell cycle distribution and apoptosis induction were examined by flow cytometry, and DNA damage was quantified using γH2AX immunofluorescence and Western blots. Relative biological effectiveness values of MSCs amounted to 1.0-1.1 for 1H, 1.7-2.3 for 4He, 2.9-3.4 for 12C and 2.6-3.3 for 16O. Particle radiation did not alter the MSCs' characteristic surface marker pattern, and MSCs maintained their multi-lineage differentiation capabilities. Apoptosis rates ranged low for all radiation modalities. At 24 h after irradiation, particle radiation-induced ATM and CHK2 phosphorylation as well as γH2AX foci numbers returned to baseline levels. The resistance of human MSCs to high-LET irradiation suggests that MSCs remain functional after exposure to moderate doses of particle radiation as seen in normal tissues after particle radiotherapy or during manned space flights. In the future, in vivo models focusing on long-term consequences of particle irradiation on the bone marrow niche and MSCs are needed.
Collapse
|
11
|
Sallam M, Benotmane MA, Baatout S, Guns PJ, Aerts A. Radiation-induced cardiovascular disease: an overlooked role for DNA methylation? Epigenetics 2022; 17:59-80. [PMID: 33522387 PMCID: PMC8812767 DOI: 10.1080/15592294.2021.1873628] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 11/27/2020] [Accepted: 01/04/2021] [Indexed: 11/25/2022] Open
Abstract
Radiotherapy in cancer treatment involves the use of ionizing radiation for cancer cell killing. Although radiotherapy has shown significant improvements on cancer recurrence and mortality, several radiation-induced adverse effects have been documented. Of these adverse effects, radiation-induced cardiovascular disease (CVD) is particularly prominent among patients receiving mediastinal radiotherapy, such as breast cancer and Hodgkin's lymphoma patients. A number of mechanisms of radiation-induced CVD pathogenesis have been proposed such as endothelial inflammatory activation, premature endothelial senescence, increased ROS and mitochondrial dysfunction. However, current research seems to point to a so-far unexamined and potentially novel involvement of epigenetics in radiation-induced CVD pathogenesis. Firstly, epigenetic mechanisms have been implicated in CVD pathophysiology. In addition, several studies have shown that ionizing radiation can cause epigenetic modifications, especially DNA methylation alterations. As a result, this review aims to provide a summary of the current literature linking DNA methylation to radiation-induced CVD and thereby explore DNA methylation as a possible contributor to radiation-induced CVD pathogenesis.
Collapse
Affiliation(s)
- Magy Sallam
- Radiobiology Unit, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
- Laboratory of Physiopharmacology, University of Antwerp, Wilrijk, Belgium
| | - Mohammed Abderrafi Benotmane
- Radiobiology Unit, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Sarah Baatout
- Radiobiology Unit, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
- Department of Molecular Biotechnology, Ghent University, Ghent, Belgium
| | - Pieter-Jan Guns
- Laboratory of Physiopharmacology, University of Antwerp, Wilrijk, Belgium
| | - An Aerts
- Radiobiology Unit, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| |
Collapse
|
12
|
Hermann RM, Trillmann A, Becker JN, Kaltenborn A, Nitsche M, Ruettermann M. Prospective Evaluation of Low-Dose External Beam Radiotherapy (LD-EBRT) for Painful Trapeziometacarpal Osteoarthritis (Rhizarthrosis) on Pain, Function, and Quality of Life to Calculate the Required Number of Patients for a Prospective Randomized Study. Med Sci (Basel) 2021; 9:medsci9040066. [PMID: 34842775 PMCID: PMC8628986 DOI: 10.3390/medsci9040066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/20/2021] [Accepted: 10/25/2021] [Indexed: 11/30/2022] Open
Abstract
Background: Retrospective studies have described the effectiveness of low-dose radiotherapy (LD-EBRT) in painful arthrosis of small finger joints, but two recent prospective studies have yielded ambiguous results. To generate accurate data for the planning of a trial, we conducted a prospective, monocentric, observational study to describe the effects of LD-EBRT as precisely as possible. Methods: Twenty-five consecutive patients with symptomatic trapeziometacarpal (TMC) arthrosis were irradiated with 6 × 0.5 Gy. Before, 3, and 12 months after LD-EBRT, we assessed subjective endpoints (modified “von-Pannewitz score”, 10-point visual analogue scale (VAS), “patient-rated wrist evaluation” (PRWE)), and objective measurements (“active range of motion” (AROM), Kapandji index, grip strength, pinch grip). Results: At 3/12 months, 80%/57% reported partial and 4%/18% complete remission according to the “von-Pannewitz” score. VAS “overall pain” significantly decreased from a median of seven (IQR 4) at baseline to three (IQR 6; p = 0.046) and to two (IQR 2; p = 0.013). Similar results were obtained for VAS “pain during exercise”, VAS “pain during daytime”, and VAS “function”. “PRWE overall score” was reduced from 0.5 at baseline (SD 0.19) to 0.36 (SD 0.24, p = 0.05) and to 0.27 (SD 0.18, p = 0.0009). We found no improvements of the objective endpoints (AROM, Kapandji, grip strength) except for flexion, which increased from 64° (SD 12°) at baseline to 73° (SD 9.7°, p = 0.046) at 12 months. Conclusions: We recommend the PRWE score as a useful endpoint for further studies for this indication. To prove a 15% superiority over sham irradiation, we calculated that 750 patients need to be prospectively randomized.
Collapse
Affiliation(s)
- Robert Michael Hermann
- Center for Radiotherapy and Radiooncology Bremen and Westerstede, 26655 Westerstede, Germany;
- Department of Radiotherapy and Special Oncology, Hannover Medical School, 49511 Hannover, Germany;
- Correspondence:
| | - Annika Trillmann
- Department of Anaesthesia, Federal Armed Forces Hospital Westerstede, 26655 Westerstede, Germany;
| | - Jan-Niklas Becker
- Department of Radiotherapy and Special Oncology, Hannover Medical School, 49511 Hannover, Germany;
| | - Alexander Kaltenborn
- Department of Trauma and Orthopaedic Surgery, Section for Plastic, Reconstructive and Hand Surgery, Federal Armed Forces Hospital Westerstede, 26655 Westerstede, Germany; (A.K.); (M.R.)
| | - Mirko Nitsche
- Center for Radiotherapy and Radiooncology Bremen and Westerstede, 26655 Westerstede, Germany;
- Radiotherapy, Karl-Lennert-Krebscentrum, Universität Kiel, 24105 Kiel, Germany
| | - Mike Ruettermann
- Department of Trauma and Orthopaedic Surgery, Section for Plastic, Reconstructive and Hand Surgery, Federal Armed Forces Hospital Westerstede, 26655 Westerstede, Germany; (A.K.); (M.R.)
- HPC-Institute for Hand and Plastic Surgery, 26122 Oldenburg, Germany
- University Medical Center Groningen, Department of Plastic Surgery, University of Groningen, 9713 Groningen, The Netherlands
| |
Collapse
|
13
|
Belli M, Indovina L. The Response of Living Organisms to Low Radiation Environment and Its Implications in Radiation Protection. Front Public Health 2020; 8:601711. [PMID: 33384980 PMCID: PMC7770185 DOI: 10.3389/fpubh.2020.601711] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 11/25/2020] [Indexed: 12/12/2022] Open
Abstract
Life has evolved on Earth for about 4 billion years in the presence of the natural background of ionizing radiation. It is extremely likely that it contributed, and still contributes, to shaping present form of life. Today the natural background radiation is extremely small (few mSv/y), however it may be significant enough for living organisms to respond to it, perhaps keeping memory of this exposure. A better understanding of this response is relevant not only for improving our knowledge on life evolution, but also for assessing the robustness of the present radiation protection system at low doses, such as those typically encountered in everyday life. Given the large uncertainties in epidemiological data below 100 mSv, quantitative evaluation of these health risk is currently obtained with the aid of radiobiological models. These predict a health detriment, caused by radiation-induced genetic mutations, linearly related to the dose. However a number of studies challenged this paradigm by demonstrating the occurrence of non-linear responses at low doses, and of radioinduced epigenetic effects, i.e., heritable changes in genes expression not related to changes in DNA sequence. This review is focused on the role that epigenetic mechanisms, besides the genetic ones, can have in the responses to low dose and protracted exposures, particularly to natural background radiation. Many lines of evidence show that epigenetic modifications are involved in non-linear responses relevant to low doses, such as non-targeted effects and adaptive response, and that genetic and epigenetic effects share, in part, a common origin: the reactive oxygen species generated by ionizing radiation. Cell response to low doses of ionizing radiation appears more complex than that assumed for radiation protection purposes and that it is not always detrimental. Experiments conducted in underground laboratories with very low background radiation have even suggested positive effects of this background. Studying the changes occurring in various living organisms at reduced radiation background, besides giving information on the life evolution, have opened a new avenue to answer whether low doses are detrimental or beneficial, and to understand the relevance of radiobiological results to radiation protection.
Collapse
Affiliation(s)
| | - Luca Indovina
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
14
|
Henry E, Arcangeli ML. How Hematopoietic Stem Cells Respond to Irradiation: Similarities and Differences between Low and High Doses of Ionizing Radiations. Exp Hematol 2020; 94:11-19. [PMID: 33290858 DOI: 10.1016/j.exphem.2020.12.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/12/2020] [Accepted: 12/01/2020] [Indexed: 12/12/2022]
Abstract
In this review, we will specifically address the newest insights on the effect of low doses of ionizing radiations on the hematopoietic stem cells, which are prone to long-term deleterious effects. Impact of high doses of irradiation on hematopoietic cells has been widely studied over the years, in line with the risk of accidental or terrorist exposure to irradiation and with a particular attention to the sensitivity of the hematopoietic system. Recently, more studies have focused on lower doses of irradiation on different tissues, due to the increasing exposure caused by medical imaging, radiotherapy or plane travelling for instance. Hence, we will delineate similarities and discrepancies in HSC response to high and low doses of irradiation from these studies.
Collapse
Affiliation(s)
- Elia Henry
- Team Niche and Cancer in Hematopoiesis, U1274, INSERM, 92260 Fontenay-aux-Roses, France; Laboratory of Hematopoietic Stem Cells and Leukemia/Service Stem Cells and Radiation/iRCM/JACOB/DRF, CEA, Fontenay-aux-Roses, France; UMR Stabilité Génétique Cellules Souches et Radiations, Université de Paris, CEA, Fontenay-aux-Roses, France; UMR Stabilité Génétique Cellules Souches et Radiations, Université Paris-Saclay, CEA, Fontenay-aux-Roses, France
| | - Marie-Laure Arcangeli
- Team Niche and Cancer in Hematopoiesis, U1274, INSERM, 92260 Fontenay-aux-Roses, France; Laboratory of Hematopoietic Stem Cells and Leukemia/Service Stem Cells and Radiation/iRCM/JACOB/DRF, CEA, Fontenay-aux-Roses, France; UMR Stabilité Génétique Cellules Souches et Radiations, Université de Paris, CEA, Fontenay-aux-Roses, France; UMR Stabilité Génétique Cellules Souches et Radiations, Université Paris-Saclay, CEA, Fontenay-aux-Roses, France.
| |
Collapse
|
15
|
Ionizing Radiation-Induced Epigenetic Modifications and Their Relevance to Radiation Protection. Int J Mol Sci 2020; 21:ijms21175993. [PMID: 32825382 PMCID: PMC7503247 DOI: 10.3390/ijms21175993] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/15/2020] [Accepted: 08/17/2020] [Indexed: 12/12/2022] Open
Abstract
The present system of radiation protection assumes that exposure at low doses and/or low dose-rates leads to health risks linearly related to the dose. They are evaluated by a combination of epidemiological data and radiobiological models. The latter imply that radiation induces deleterious effects via genetic mutation caused by DNA damage with a linear dose-dependence. This picture is challenged by the observation of radiation-induced epigenetic effects (changes in gene expression without altering the DNA sequence) and of non-linear responses, such as non-targeted and adaptive responses, that in turn can be controlled by gene expression networks. Here, we review important aspects of the biological response to ionizing radiation in which epigenetic mechanisms are, or could be, involved, focusing on the possible implications to the low dose issue in radiation protection. We examine in particular radiation-induced cancer, non-cancer diseases and transgenerational (hereditary) effects. We conclude that more realistic models of radiation-induced cancer should include epigenetic contribution, particularly in the initiation and progression phases, while the impact on hereditary risk evaluation is expected to be low. Epigenetic effects are also relevant in the dispute about possible "beneficial" effects at low dose and/or low dose-rate exposures, including those given by the natural background radiation.
Collapse
|
16
|
Patel R, Zhang L, Desai A, Hoenerhoff MJ, Kennedy LH, Radivoyevitch T, La Tessa C, Gerson SL, Welford SM. Protons and High-Linear Energy Transfer Radiation Induce Genetically Similar Lymphomas With High Penetrance in a Mouse Model of the Aging Human Hematopoietic System. Int J Radiat Oncol Biol Phys 2020; 108:1091-1102. [PMID: 32629081 DOI: 10.1016/j.ijrobp.2020.06.070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/22/2020] [Accepted: 06/26/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE Humans are exposed to charged particles in different scenarios. The use of protons and high-linear energy transfer (LET) in cancer treatment is steadily growing. In outer space, astronauts will be exposed to a mixed radiation field composed of both protons and heavy ions, in particularly the long-term space missions outside of earth's magnetosphere. Thus, understanding the radiobiology and transforming potential of these types of ionizing radiation are of paramount importance. METHODS AND MATERIALS We examined the effect of 10 or 100 cGy of whole-body doses of protons or 28Si ions on the hematopoietic system of a genetic model of aging based on recent studies that showed selective loss of the MLH1 protein in human hematopoietic stems with age. RESULTS We found that Mlh1 deficient animals are highly prone to develop lymphomas when exposed to either low doses of protons or 28Si ions. The lymphomas that develop are genetically indistinguishable, in spite of different types of damage elicited by low- and high-LET radiation. RNA sequencing analyses reveal similar gene expression patterns, similar numbers of altered genes, similar numbers of single nucleotide variants and insertions and deletions, and similar activation of known leukemogenic loci. CONCLUSIONS Although the incidence of malignancy is related to radiation quality, and increased due to loss of Mlh1, the phenotype of the tumors is independent of LET.
Collapse
Affiliation(s)
- Rutulkumar Patel
- Department of Radiation Oncology, Duke University, Durham, North Carolina
| | - Luchang Zhang
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Amar Desai
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio; Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Mark J Hoenerhoff
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Lucy H Kennedy
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Tomas Radivoyevitch
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio
| | - Chiara La Tessa
- University of Trento, Trento, Italy; Trento Institute for Fundamental Physics and Applications TIFPA-INFN, Trento, Italy
| | - Stanton L Gerson
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Scott M Welford
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida; Department of Radiation Oncology, University of Miami, Miami, Florida.
| |
Collapse
|
17
|
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
|
18
|
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
|
19
|
Perera BP, Faulk C, Svoboda LK, Goodrich JM, Dolinoy DC. The role of environmental exposures and the epigenome in health and disease. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2020; 61:176-192. [PMID: 31177562 PMCID: PMC7252203 DOI: 10.1002/em.22311] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/29/2019] [Accepted: 06/03/2019] [Indexed: 05/02/2023]
Abstract
The genetic material of every organism exists within the context of regulatory networks that govern gene expression, collectively called the epigenome. Epigenetics has taken center stage in the study of diseases such as cancer and diabetes, but its integration into the field of environmental health is still emerging. As the Environmental Mutagenesis and Genomics Society (EMGS) celebrates its 50th Anniversary this year, we have come together to review and summarize the seminal advances in the field of environmental epigenomics. Specifically, we focus on the role epigenetics may play in multigenerational and transgenerational transmission of environmentally induced health effects. We also summarize state of the art techniques for evaluating the epigenome, environmental epigenetic analysis, and the emerging field of epigenome editing. Finally, we evaluate transposon epigenetics as they relate to environmental exposures and explore the role of noncoding RNA as biomarkers of environmental exposures. Although the field has advanced over the past several decades, including being recognized by EMGS with its own Special Interest Group, recently renamed Epigenomics, we are excited about the opportunities for environmental epigenetic science in the next 50 years. Environ. Mol. Mutagen. 61:176-192, 2020. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Bambarendage P.U. Perera
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Christopher Faulk
- Department of Animal Sciences, University of Minnesota, St. Paul, Minnesota
| | - Laurie K. Svoboda
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Jaclyn M. Goodrich
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Dana C. Dolinoy
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan
- Correspondence to: Dana C. Dolinoy, Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan.
| |
Collapse
|
20
|
Cho YH, Jang Y, Woo HD, Kim YJ, Kim SY, Christensen S, Cole E, Choi SY, Chung HW. LINE-1 hypomethylation is associated with radiation-induced genomic instability in industrial radiographers. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2019; 60:174-184. [PMID: 30488609 PMCID: PMC6363886 DOI: 10.1002/em.22237] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 06/15/2018] [Accepted: 08/10/2018] [Indexed: 06/09/2023]
Abstract
Global DNA hypomethylation is proposed as a potential biomarker for cancer risk associated with genomic instability, which is an important factor in radiation-induced cancer. However, the associations among radiation exposure, changes in DNA methylation, and carcinogenesis are unclear. The aims of this study were (1) to examine whether low-level occupational radiation exposure induces genomic DNA hypomethylation; and (2) to determine the relationships between radiation exposure, genomic DNA hypomethylation and radiation-induced genomic instability (RIGI) in industrial radiographers. Genomic DNA methylation levels were measured in blood DNA from 40 radiographers and 28 controls using the LINE-1 pyrosequencing assay and the luminometric methylation assay. Further, the micronucleus-centromere assay was performed to measure aneuploidy of chromosomes 1 and 4 as a marker of delayed RIGI. Genomic DNA methylation levels were significantly lower in radiographers than those in controls. LINE-1 hypomethylation was not significantly correlated with recent 1-year, recent 3-year, or total cumulative radiation doses in radiographers; however, LINE-1 hypomethylation significantly correlated with the cumulative radiation dose without recent 3-year exposure data (D3dose, r = -0.39, P < 0.05). In addition, LINE-1 hypomethylation was a significant contributor to aneuploidy frequency by D3dose (F (2, 34) = 13.85, P < 0.001), in which a total of 45% of the variance in aneuploidy frequency was explained. Our results provide suggestive evidence regarding the delayed effects of low-dose occupational radiation exposure in radiographers and its association with LINE-1 hypomethylation; however, additional studies using more subjects are needed to fully understand the relationship between genomic DNA hypomethylation and RIGI. Environ. Mol. Mutagen. 60: 174-184, 2019. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yoon Hee Cho
- Departments of Biomedical and Pharmaceutical Sciences, The University of Montana, Missoula, MT, USA
| | - Yoonhee Jang
- Departments of Psychology, The University of Montana, Missoula, MT, USA
| | - Hae Dong Woo
- Molecular Epidemiology Branch, Division of Cancer Epidemiology and Prevention, Research Institute, National Cancer Center, Goyang-si, Gyeonggi-do, Korea
| | - Yang Jee Kim
- Da Vinci College of General Education, Chung-Ang University, Seoul, Korea
| | - Su Young Kim
- Departments of Preventive Medicine, School of Medicine, Jeju National University, Jeju-si, Jeju-do, Korea
| | - Sonja Christensen
- Departments of Biomedical and Pharmaceutical Sciences, The University of Montana, Missoula, MT, USA
| | - Elizabeth Cole
- Departments of Biomedical and Pharmaceutical Sciences, The University of Montana, Missoula, MT, USA
| | - Soo Yong Choi
- Laboratory of Radiation Effect, Korea Institute of Radiological and Medical Science, Seoul Korea
| | - Hai Won Chung
- School of Public Health, Seoul National University, Seoul, Korea
| |
Collapse
|
21
|
Liu F, Wang Z, Li W, Wei Y. Transcriptional response of murine bone marrow cells to total-body carbon-ion irradiation. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2019; 839:49-58. [PMID: 30744812 DOI: 10.1016/j.mrgentox.2019.01.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 01/24/2019] [Accepted: 01/24/2019] [Indexed: 12/26/2022]
Abstract
The need to understand the health effects of heavy ion irradiation is motivated by the use of this modality in radiotherapy and by the potential for exposure during space missions. We have studied the effects of carbon-ion total-body irradiation on the hematopoietic system of the mouse and, in particular, the transcriptional response of bone marrow (BM) cells. Carbon-ion irradiation caused BM cell DNA damage, apoptosis, elevated ROS, and myelosuppression. Transcriptomic analysis showed that overall gene expression in irradiated BM cells differed significantly from the controls. Of 253 genes that were modulated, 192 were up-regulated and 61 down-regulated. Gene ontology analysis showed that the modulated genes are involved in DNA damage response signaling, DNA repair, apoptosis, and the immune response. Kyoto Encyclopedia of Genes and Genomes pathway analysis indicated that these functions are regulated by the p38 MAPK, TNF, and apoptosis pathways. These findings indicate pathways that may be involved in protection against carbon ion radiation injury.
Collapse
Affiliation(s)
- Fang Liu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhuanzi Wang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China.
| | - Wenjian Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, 730000, China
| | - Yanting Wei
- School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, 730000, China
| |
Collapse
|
22
|
Fabbrizi MR, Warshowsky KE, Zobel CL, Hallahan DE, Sharma GG. Molecular and epigenetic regulatory mechanisms of normal stem cell radiosensitivity. Cell Death Discov 2018; 4:117. [PMID: 30588339 PMCID: PMC6299079 DOI: 10.1038/s41420-018-0132-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/01/2018] [Accepted: 11/20/2018] [Indexed: 12/14/2022] Open
Abstract
Ionizing radiation (IR) therapy is a major cancer treatment modality and an indispensable auxiliary treatment for primary and metastatic cancers, but invariably results in debilitating organ dysfunctions. IR-induced depletion of neural stem/progenitor cells in the subgranular zone of the dentate gyrus in the hippocampus where neurogenesis occurs is considered largely responsible for deficiencies such as learning, memory, and spatial information processing in patients subjected to cranial irradiation. Similarly, IR therapy-induced intestinal injuries such as diarrhea and malabsorption are common side effects in patients with gastrointestinal tumors and are believed to be caused by intestinal stem cell drop out. Hematopoietic stem cell transplantation is currently used to reinstate blood production in leukemia patients and pre-clinical treatments show promising results in other organs such as the skin and kidney, but ethical issues and logistic problems make this route difficult to follow. An alternative way to restore the injured tissue is to preserve the stem cell pool located in that specific tissue/organ niche, but stem cell response to ionizing radiation is inadequately understood at the molecular mechanistic level. Although embryonic and fetal hypersensity to IR has been very well known for many decades, research on embryonic stem cell models in culture concerning molecular mechanisms have been largely inconclusive and often in contradiction of the in vivo observations. This review will summarize the latest discoveries on stem cell radiosensitivity, highlighting the possible molecular and epigenetic mechanism(s) involved in DNA damage response and programmed cell death after ionizing radiation therapy specific to normal stem cells. Finally, we will analyze the possible contribution of stem cell-specific chromatin's epigenetic constitution in promoting normal stem cell radiosensitivity.
Collapse
Affiliation(s)
- Maria Rita Fabbrizi
- Cancer Biology Division, Department of Radiation Oncology, Washington University School of Medicine, 4511 Forest Park, Saint Louis, MO 63108 USA
| | - Kacie E. Warshowsky
- Cancer Biology Division, Department of Radiation Oncology, Washington University School of Medicine, 4511 Forest Park, Saint Louis, MO 63108 USA
| | - Cheri L. Zobel
- Cancer Biology Division, Department of Radiation Oncology, Washington University School of Medicine, 4511 Forest Park, Saint Louis, MO 63108 USA
| | - Dennis E. Hallahan
- Cancer Biology Division, Department of Radiation Oncology, Washington University School of Medicine, 4511 Forest Park, Saint Louis, MO 63108 USA
- Siteman Cancer Center, Washington University School of Medicine, Saint Louis, MO 63108 USA
| | - Girdhar G. Sharma
- Cancer Biology Division, Department of Radiation Oncology, Washington University School of Medicine, 4511 Forest Park, Saint Louis, MO 63108 USA
- Siteman Cancer Center, Washington University School of Medicine, Saint Louis, MO 63108 USA
| |
Collapse
|
23
|
Hoehn D, Pujol-Canadell M, Young EF, Serban G, Shuryak I, Maerki J, Xu Z, Chowdhury M, Luna AM, Vlada G, Smilenov LB. Effects of High- and Low-LET Radiation on Human Hematopoietic System Reconstituted in Immunodeficient Mice. Radiat Res 2018; 191:162-175. [PMID: 30520704 DOI: 10.1667/rr15148.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Over the last 50 years, a number of important physiological changes in humans who have traveled on spaceflights have been catalogued. Of major concern are the short- and long-term radiation-induced injuries to the hematopoietic system that may be induced by high-energy galactic cosmic rays encountered on interplanetary space missions. To collect data on the effects of space radiation on the human hematopoietic system in vivo, we used a humanized mouse model. In this study, we irradiated humanized mice with 0.4 Gy of 350 MeV/n 28Si ions, a dose that has been shown to induce tumors in tumor-prone mice and a reference dose that has a relative biological effectiveness of 1 (1 Gy of 250-kVp X rays). Cell counts, cell subset frequency and cytogenetic data were collected from bone marrow spleen and blood of irradiated and control mice at short-term (7, 30 and 60 days) and long-term ( 6 - 7 months) time points postirradiation. The data show a significant short-term effect on the human hematopoietic stem cell counts imparted by both high- and low-LET radiation exposure. The radiation effects on bone marrow, spleen and blood human cell counts and human cell subset frequency were complex but did not alter the functions of the hematopoietic system. The long-term data acquired from high-LET irradiated mice showed complete recovery of the human hematopoietic system in all hematopoietic compartments. The combined results demonstrate that, in spite of early perturbation, the longer term effects of high-LET radiation are not detrimental to human hematopoiesis in our system of study.
Collapse
Affiliation(s)
- Daniela Hoehn
- a Columbia University Medical Center, New York, New York
| | | | - Erik F Young
- a Columbia University Medical Center, New York, New York
| | - Geo Serban
- a Columbia University Medical Center, New York, New York
| | - Igor Shuryak
- a Columbia University Medical Center, New York, New York
| | | | - Zheng Xu
- a Columbia University Medical Center, New York, New York
| | | | - Aesis M Luna
- a Columbia University Medical Center, New York, New York
| | - George Vlada
- a Columbia University Medical Center, New York, New York
| | | |
Collapse
|
24
|
Ionizing Radiation and Human Health: Reviewing Models of Exposure and Mechanisms of Cellular Damage. An Epigenetic Perspective. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2018; 15:ijerph15091971. [PMID: 30201914 PMCID: PMC6163535 DOI: 10.3390/ijerph15091971] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 09/03/2018] [Accepted: 09/04/2018] [Indexed: 11/16/2022]
Abstract
We reviewed available evidence in medical literature concerning experimental models of exposure to ionizing radiations (IR) and their mechanisms of producing damages on living organisms. The traditional model is based on the theory of “stochastic breakage” of one or both strands of the DNA double helix. According to this model, high doses may cause the breaks, potentially lethal to the cell by damaging both DNA strands, while low doses of IR would cause essentially single strands breaks, easily repairable, resulting in no permanent damages. The available evidence makes this classical model increasingly less acceptable, because the exposure to low doses of IR seems to have carcinogenic effects, even after years or decades, both in the exposed individuals and in subsequent generations. In addition, the cells that survived the exposure to low doses, despite being apparently normal, accumulate damages that become evident in their progeny, such as nonclonal chromosomal aberrations, which can be found even in cells not directly irradiated due to the exchange of molecular signals and complex tissue reactions involving neighboring or distant cells. For all these reasons, a paradigm shift is needed, based on evidence and epigenetics.
Collapse
|
25
|
Wang B, Tanaka K, Ninomiya Y, Maruyama K, Varès G, Katsube T, Murakami M, Liu C, Fujimori A, Fujita K, Liu Q, Eguchi-Kasai K, Nenoi M. Increased Hematopoietic Stem Cells/Hematopoietic Progenitor Cells Measured as Endogenous Spleen Colonies in Radiation-Induced Adaptive Response in Mice (Yonezawa Effect). Dose Response 2018; 16:1559325818790152. [PMID: 30150909 PMCID: PMC6104214 DOI: 10.1177/1559325818790152] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 06/01/2018] [Accepted: 06/12/2018] [Indexed: 12/03/2022] Open
Abstract
The existence of radiation-induced adaptive response (AR) was reported in varied
biosystems. In mice, the first in vivo AR model was established using X-rays as
both the priming and the challenge doses and rescue of bone marrow death as the
end point. The underlying mechanism was due to the priming radiation-induced
resistance in the blood-forming tissues. In a series of investigations, we
further demonstrated the existence of AR using different types of ionizing
radiation (IR) including low linear energy transfer (LET) X-rays and high LET
heavy ion. In this article, we validated hematopoietic stem cells/hematopoietic
progenitor cells (HSCs/HPCs) measured as endogenous colony-forming units-spleen
(CFU-S) under AR inducible and uninducible conditions using combination of
different types of IR. We confirmed the consistency of increased CFU-S number
change with the AR inducible condition. These findings suggest that AR in mice
induced by different types of IR would share at least in part a common
underlying mechanism, the priming IR-induced resistance in the blood-forming
tissues, which would lead to a protective effect on the HSCs/HPCs and play an
important role in rescuing the animals from bone marrow death. These findings
provide a new insight into the mechanistic study on AR in vivo.
Collapse
Affiliation(s)
- Bing Wang
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Kaoru Tanaka
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Yasuharu Ninomiya
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Kouichi Maruyama
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | | | - Takanori Katsube
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Masahiro Murakami
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Cuihua Liu
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Akira Fujimori
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | | | - Qiang Liu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, People's Republic of China
| | - Kiyomi Eguchi-Kasai
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Mitsuru Nenoi
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| |
Collapse
|
26
|
Koturbash I. 2017 Michael Fry Award Lecture When DNA is Actually Not a Target: Radiation Epigenetics as a Tool to Understand and Control Cellular Response to Ionizing Radiation. Radiat Res 2018; 190:5-11. [PMID: 29697303 PMCID: PMC6036898 DOI: 10.1667/rr15027.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Aside from the generally accepted potential to cause DNA damage, it is becoming increasingly recognized that ionizing radiation has the capability to target the cellular epigenome. Epigenetics unifies the chemical marks and molecules that collectively facilitate the proper reading of genetic material. Among the epigenetic mechanisms of regulation, methylation of DNA is known to be the key player in the postirradiation response by controlling the expression of genetic information and activity of transposable elements. Radiation-induced alterations to DNA methylation may lead to cellular epigenetic reprogramming that, in turn, can substantially compromise the genomic integrity and has been proposed as one of the mechanisms of radiation-induced carcinogenesis. DNA methylation is strongly dependent on the one-carbon metabolism. This metabolic pathway is central to the support of DNA methylation by means of providing the donor of methyl groups, as well as for the synthesis of amino acids. To better understand the mechanisms of radiation-induced health effects, we study how exposure to radiation affects DNA methylation and one-carbon metabolism. Also, a tight interaction that exists between DNA methylation and one-carbon metabolism allows us to simultaneously manipulate both cellular epigenetic and metabolic profiles to modulate the normal and cancerous tissue response to radiotherapy.
Collapse
Affiliation(s)
- Igor Koturbash
- Department of Environmental and Occupational Health, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| |
Collapse
|
27
|
Galactic Cosmic Radiation Induces Persistent Epigenome Alterations Relevant to Human Lung Cancer. Sci Rep 2018; 8:6709. [PMID: 29712937 PMCID: PMC5928241 DOI: 10.1038/s41598-018-24755-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 03/27/2018] [Indexed: 12/14/2022] Open
Abstract
Human deep space and planetary travel is limited by uncertainties regarding the health risks associated with exposure to galactic cosmic radiation (GCR), and in particular the high linear energy transfer (LET), heavy ion component. Here we assessed the impact of two high-LET ions 56Fe and 28Si, and low-LET X rays on genome-wide methylation patterns in human bronchial epithelial cells. We found that all three radiation types induced rapid and stable changes in DNA methylation but at distinct subsets of CpG sites affecting different chromatin compartments. The 56Fe ions induced mostly hypermethylation, and primarily affected sites in open chromatin regions including enhancers, promoters and the edges ("shores") of CpG islands. The 28Si ion-exposure had mixed effects, inducing both hyper and hypomethylation and affecting sites in more repressed heterochromatic environments, whereas X rays induced mostly hypomethylation, primarily at sites in gene bodies and intergenic regions. Significantly, the methylation status of 56Fe ion sensitive sites, but not those affected by X ray or 28Si ions, discriminated tumor from normal tissue for human lung adenocarcinomas and squamous cell carcinomas. Thus, high-LET radiation exposure leaves a lasting imprint on the epigenome, and affects sites relevant to human lung cancer. These methylation signatures may prove useful in monitoring the cumulative biological impact and associated cancer risks encountered by astronauts in deep space.
Collapse
|
28
|
Patel R, Qing Y, Kennedy L, Yan Y, Pink J, Aguila B, Desai A, Gerson SL, Welford SM. MMR Deficiency Does Not Sensitize or Compromise the Function of Hematopoietic Stem Cells to Low and High LET Radiation. Stem Cells Transl Med 2018; 7:513-520. [PMID: 29656536 PMCID: PMC6052615 DOI: 10.1002/sctm.17-0295] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 03/20/2018] [Indexed: 12/12/2022] Open
Abstract
One of the major health concerns on long-duration space missions will be radiation exposure to the astronauts. Outside the earth's magnetosphere, astronauts will be exposed to galactic cosmic rays (GCR) and solar particle events that are principally composed of protons and He, Ca, O, Ne, Si, Ca, and Fe nuclei. Protons are by far the most common species, but the higher atomic number particles are thought to be more damaging to biological systems. Evaluation and amelioration of risks from GCR exposure will be important for deep space travel. The hematopoietic system is one of the most radiation-sensitive organ systems, and is highly dependent on functional DNA repair pathways for survival. Recent results from our group have demonstrated an acquired deficiency in mismatch repair (MMR) in human hematopoietic stem cells (HSCs) with age due to functional loss of the MLH1 protein, suggesting an additional risk to astronauts who may have significant numbers of MMR deficient HSCs at the time of space travel. In the present study, we investigated the effects gamma radiation, proton radiation, and 56 Fe radiation on HSC function in Mlh1+/+ and Mlh1-/- marrow from mice in a variety of assays and have determined that while cosmic radiation is a major risk to the hematopoietic system, there is no dependence on MMR capacity. Stem Cells Translational Medicine 2018;7:513-520.
Collapse
Affiliation(s)
| | - Yulan Qing
- Case Comprehensive Cancer Center, National Center for Regenerative Medicine, Seidman Cancer Center, University Hospitals Cleveland Medical center and Case Western Reserve University, Cleveland, Ohio, USA
| | - Lucy Kennedy
- Unit for Laboratory and Animal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Yan Yan
- Case Comprehensive Cancer Center, National Center for Regenerative Medicine, Seidman Cancer Center, University Hospitals Cleveland Medical center and Case Western Reserve University, Cleveland, Ohio, USA
| | - John Pink
- Case Comprehensive Cancer Center, National Center for Regenerative Medicine, Seidman Cancer Center, University Hospitals Cleveland Medical center and Case Western Reserve University, Cleveland, Ohio, USA
| | - Brittany Aguila
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio, USA
| | - Amar Desai
- Case Comprehensive Cancer Center, National Center for Regenerative Medicine, Seidman Cancer Center, University Hospitals Cleveland Medical center and Case Western Reserve University, Cleveland, Ohio, USA
| | - Stanton L Gerson
- Case Comprehensive Cancer Center, National Center for Regenerative Medicine, Seidman Cancer Center, University Hospitals Cleveland Medical center and Case Western Reserve University, Cleveland, Ohio, USA
| | - Scott M Welford
- Department of Radiation Oncology, Sylvester Cancer Center, University of Miami, Miami, Florida, USA
| |
Collapse
|
29
|
Cardelli M. The epigenetic alterations of endogenous retroelements in aging. Mech Ageing Dev 2018; 174:30-46. [PMID: 29458070 DOI: 10.1016/j.mad.2018.02.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/05/2018] [Accepted: 02/08/2018] [Indexed: 02/06/2023]
Abstract
Endogenous retroelements, transposons that mobilize through RNA intermediates, include some of the most abundant repetitive sequences of the human genome, such as Alu and LINE-1 sequences, and human endogenous retroviruses. Recent discoveries demonstrate that these mobile genetic elements not only act as intragenomic parasites, but also exert regulatory roles in living cells. The risk of genomic instability represented by endogenous retroelements is normally counteracted by a series of epigenetic control mechanisms which include, among the most important, CpG DNA methylation. Indeed, most of the genomic CpG sites subjected to DNA methylation in the nuclear DNA are carried by these repetitive elements. As other parts of the genome, endogenous retroelements and other transposable elements are subjected to deep epigenetic alterations during aging, repeatedly observed in the context of organismal and cellular senescence, in human and other species. This review summarizes the current status of knowledge about the epigenetic alterations occurring in this large, non-genic portion of the genome in aging and age-related conditions, with a focus on the causes and the possible functional consequences of these alterations.
Collapse
Affiliation(s)
- Maurizio Cardelli
- Advanced Technology Center for Aging Research, Scientific Technological Area, Italian National Research Center on Aging (INRCA), via Birarelli 8, 60121 Ancona, Italy.
| |
Collapse
|
30
|
Miousse IR, Ewing LE, Kutanzi KR, Griffin RJ, Koturbash I. DNA Methylation in Radiation-Induced Carcinogenesis: Experimental Evidence and Clinical Perspectives. Crit Rev Oncog 2018; 23:1-11. [PMID: 29953365 PMCID: PMC6369919 DOI: 10.1615/critrevoncog.2018025687] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Ionizing radiation is a valuable tool in many spheres of human life. At the same time, it is a genotoxic agent with a well-established carcinogenic potential. Progress achieved in the last two decades has demonstrated convincingly that ionizing radiation can also target the cellular epigenome. Epigenetics is defined as heritable changes in the expression of genes that are not due to alterations of DNA sequence but consist of specific covalent modifications of chromatin components, such as methylation of DNA, histone modifications, and control performed by non-coding RNAs. Accumulating evidence suggests that DNA methylation, a key epigenetic mechanism involved in the control of expression of genetic information, may serve as one of the driving mechanisms of radiation-induced carcinogenesis. Here, we review the literature on the effects of ionizing radiation on DNA methylation in various biological systems, discuss the role of DNA methylation in radiation carcinogenesis, and provide our opinion on the potential utilization of this knowledge in radiation oncology.
Collapse
Affiliation(s)
- Isabelle R. Miousse
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Laura E. Ewing
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Kristy R. Kutanzi
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Robert J. Griffin
- Department of Radiation Oncology, Radiation Biology Division, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Igor Koturbash
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| |
Collapse
|
31
|
Schofield PN, Kondratowicz M. Evolving paradigms for the biological response to low dose ionizing radiation; the role of epigenetics. Int J Radiat Biol 2017; 94:769-781. [PMID: 29157078 DOI: 10.1080/09553002.2017.1388548] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE In the late 1990s, it had become clear that the long-standing paradigm for the action of radiation on living cells and organisms did not have sufficient power to explain the observed effects of low dose ionizing radiation. The purpose of this commentary is to examine the experiments that lead up to the modification of the classic paradigm consequent on these observations, their historical precedents, and the development of our understanding of the role of epigenetics in low dose radiation effects. RESULTS AND CONCLUSIONS We discuss how parallel advances in epigenetics from developmental biology and cancer studies, and the discovery of epigenetic modifications of chromatin, such as DNA methylation, impacted on the development of an epigenetic paradigm for low dose effects. We also assess the impact of technology development in supporting the paradigm shift. We then examine recent accumulated data on epigenetic modification in response to irradiation since that shift took place, and identify areas where bringing together data from developmental biology and cancer might answer some of the paradoxes and contradictions in this data. We predict that further paradigm shifts are imminent.
Collapse
Affiliation(s)
- Paul N Schofield
- a Department of Physiology, Development, and Neuroscience , University of Cambridge , Cambridge , UK
| | - Monika Kondratowicz
- a Department of Physiology, Development, and Neuroscience , University of Cambridge , Cambridge , UK
| |
Collapse
|
32
|
Koturbash I. LINE-1 in response to exposure to ionizing radiation. Mob Genet Elements 2017; 7:e1393491. [PMID: 29209557 DOI: 10.1080/2159256x.2017.1393491] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 10/13/2017] [Indexed: 12/15/2022] Open
Abstract
It is becoming increasingly recognized that Long Interspersed Nuclear Element, 1 (LINE-1), the most ubiquitous repetitive element in the mammalian genomes, plays an important role in the pathogenesis of disease and in the response to exposure to environmental stressors. Ionizing radiation is a known genotoxic stressor, but it is capable of targeting the cellular epigenome as well. Radiation-induced alterations in LINE-1 DNA methylation are the most frequently observed epigenetic effects of exposure. The extent of this aberrant DNA methylation, however, strongly depends on a number of factors, including the type and dose of radiation. Two other factors are being discussed in this commentary - the evolutionary age and type of the LINE-1 promoter, as well as the type of irradiated cell. This knowledge will further aid in elucidating the mechanisms of response to ionizing radiation exposure, as well in understanding the pathogenesis of the negative health effects associated with exposure.
Collapse
Affiliation(s)
- Igor Koturbash
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
33
|
How Will the Hematopoietic System Deal with Space Radiation on the Way to Mars? CURRENT STEM CELL REPORTS 2017. [DOI: 10.1007/s40778-017-0104-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
34
|
Asselin-Labat ML, Rampersad R, Xu X, Ritchie ME, Michalski J, Huang L, Onaitis MW. High-LET Radiation Increases Tumor Progression in a K-Ras-Driven Model of Lung Adenocarcinoma. Radiat Res 2017; 188:562-570. [PMID: 28952911 DOI: 10.1667/rr14794.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
High-linear energy transfer (LET) radiation encountered by astronauts in space generates clustered DNA damage that is potentially oncogenic. Analysis of the impact of exposure to space radiation on cancer formation is necessary to determine the best ways to prepare astronauts for space travel so they are protected for the duration of the space mission. A mouse model of lung adenocarcinoma driven by oncogenic K-Ras was used to ascertain the effect of low- and high-LET radiation on tumor formation. We observed increased tumor progression and tumor cell proliferation after single dose or fractionated high-LET doses, which was not observed in mice exposed to low-LET radiation. Location of the tumor nodules was not affected by radiation, indicating that the cell of origin of K-Ras-driven tumors was the same in irradiated or nonirradiated mice. Gene expression analysis revealed an upregulation of genes involved in cell proliferation and DNA damage repair. This study provides evidence that exposure to a single dose or fractionated doses of high-LET radiation induces molecular and cellular changes that accelerate lung tumor growth.
Collapse
Affiliation(s)
- Marie-Liesse Asselin-Labat
- a University California San Diego, Moores Cancer Center, La Jolla, California.,b ACRF Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,d Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Rishi Rampersad
- f Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - Xia Xu
- f Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - Matthew E Ritchie
- c Molecular Medicine Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,e School of Mathematics and Statistics, The University of Melbourne, Parkville, Victoria, Australia
| | - Jacob Michalski
- f Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - Lingling Huang
- f Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - Mark W Onaitis
- a University California San Diego, Moores Cancer Center, La Jolla, California.,f Department of Surgery, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
35
|
Miousse IR, Chang J, Shao L, Pathak R, Nzabarushimana É, Kutanzi KR, Landes RD, Tackett AJ, Hauer-Jensen M, Zhou D, Koturbash I. Inter-Strain Differences in LINE-1 DNA Methylation in the Mouse Hematopoietic System in Response to Exposure to Ionizing Radiation. Int J Mol Sci 2017; 18:ijms18071430. [PMID: 28677663 PMCID: PMC5535921 DOI: 10.3390/ijms18071430] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 06/28/2017] [Accepted: 06/30/2017] [Indexed: 12/15/2022] Open
Abstract
Long Interspersed Nuclear Element 1 (LINE-1) retrotransposons are the major repetitive elements in mammalian genomes. LINE-1s are well-accepted as driving forces of evolution and critical regulators of the expression of genetic information. Alterations in LINE-1 DNA methylation may lead to its aberrant activity and are reported in virtually all human cancers and in experimental carcinogenesis. In this study, we investigated the endogenous DNA methylation status of the 5′ untranslated region (UTR) of LINE-1 elements in the bone marrow hematopoietic stem cells (HSCs), hematopoietic progenitor cells (HPCs), and mononuclear cells (MNCs) in radioresistant C57BL/6J and radiosensitive CBA/J mice and in response to ionizing radiation (IR). We demonstrated that basal levels of DNA methylation within the 5′-UTRs of LINE-1 elements did not differ significantly between the two mouse strains and were negatively correlated with the evolutionary age of LINE-1 elements. Meanwhile, the expression of LINE-1 elements was higher in CBA/J mice. At two months after irradiation to 0.1 or 1 Gy of 137Cs (dose rate 1.21 Gy/min), significant decreases in LINE-1 DNA methylation in HSCs were observed in prone to radiation-induced carcinogenesis CBA/J, but not C57BL/6J mice. At the same time, no residual DNA damage, increased ROS, or changes in the cell cycle were detected in HSCs of CBA/J mice. These results suggest that epigenetic alterations may potentially serve as driving forces of radiation-induced carcinogenesis; however, future studies are needed to demonstrate the direct link between the LINE-1 DNA hypomethylation and radiation carcinogenesis.
Collapse
Affiliation(s)
- Isabelle R Miousse
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Jianhui Chang
- Department of Pharmaceutical Sciences, Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Lijian Shao
- Department of Pharmaceutical Sciences, Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Rupak Pathak
- Department of Pharmaceutical Sciences, Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Étienne Nzabarushimana
- Department of Bioinformatics, School of Informatics and Computing, Indiana University, Bloomington, IN 47408, USA.
| | - Kristy R Kutanzi
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Reid D Landes
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Alan J Tackett
- Department of Biochemistry, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Martin Hauer-Jensen
- Department of Pharmaceutical Sciences, Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Daohong Zhou
- Department of Pharmaceutical Sciences, Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Igor Koturbash
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| |
Collapse
|
36
|
McLaughlin MF, Donoviel DB, Jones JA. Novel Indications for Commonly Used Medications as Radiation Protectants in Spaceflight. Aerosp Med Hum Perform 2017. [PMID: 28641684 DOI: 10.3357/amhp.4735.2017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND In the space environment, the traditional radioprotective principles of time, distance, and shielding become difficult to implement. Additionally, the complex radiation environment inherent in space, the chronic exposure timeframe, and the presence of numerous confounding variables complicate the process of creating appropriate risk models for astronaut exposure. Pharmaceutical options hold tremendous promise to attenuate acute and late effects of radiation exposure in the astronaut population. Pharmaceuticals currently approved for other indications may also offer radiation protection, modulation, or mitigation properties along with a well-established safety profile. Currently there are only three agents which have been clinically approved to be employed for radiation exposure, and these only for very narrow indications. This review identifies a number of agents currently approved by the U.S. Food and Drug Administration (FDA) which could warrant further investigation for use in astronauts. Specifically, we examine preclinical and clinical evidence for statins, nonsteroidal anti-inflammatory drugs (NSAIDs), angiotensin converting enzyme inhibitors (ACEIs), angiotensin II receptor blockers (ARBs), metformin, calcium channel blockers, β adrenergic receptor blockers, fingolimod, N-acetylcysteine, and pentoxifylline as potential radiation countermeasures.McLaughlin MF, Donoviel DB, Jones JA. Novel indications for commonly used medications as radiation protectants in spaceflight. Aerosp Med Hum Perform. 2017; 88(7):665-676.
Collapse
|
37
|
Miousse IR, Tobacyk J, Melnyk S, James SJ, Cheema AK, Boerma M, Hauer-Jensen M, Koturbash I. One-carbon metabolism and ionizing radiation: a multifaceted interaction. Biomol Concepts 2017; 8:83-92. [PMID: 28574375 PMCID: PMC6693336 DOI: 10.1515/bmc-2017-0003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 05/03/2017] [Indexed: 01/20/2023] Open
Abstract
Ionizing radiation (IR) is a ubiquitous component of our environment and an important tool in research and medical treatment. At the same time, IR is a potent genotoxic and epigenotoxic stressor, exposure to which may lead to negative health outcomes. While the genotoxocity is well described and characterized, the epigenetic effects of exposure to IR and their mechanisms remain under-investigated. In this conceptual review, we propose the IR-induced changes to one-carbon metabolism as prerequisites to alterations in the cellular epigenome. We also provide evidence from both experimental and clinical studies describing the interactions between IR and one-carbon metabolism. We further discuss the potential for the manipulation of the one-carbon metabolism in clinical applications for the purpose of normal tissue protection and for increasing the radiosensitivity of cancerous cells.
Collapse
Affiliation(s)
- Isabelle R. Miousse
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Julia Tobacyk
- Departments of Environmental and Occupational Health, and Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Stepan Melnyk
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - S. Jill James
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Amrita K. Cheema
- Departments of Oncology and Biochemistry, Molecular and Cellular Biology, Georgetown University Medical Center, Washington DC 20057, USA
| | - Marjan Boerma
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Martin Hauer-Jensen
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Igor Koturbash
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
38
|
Miousse IR, Kutanzi KR, Koturbash I. Effects of ionizing radiation on DNA methylation: from experimental biology to clinical applications. Int J Radiat Biol 2017; 93:457-469. [PMID: 28134023 PMCID: PMC5411327 DOI: 10.1080/09553002.2017.1287454] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE Ionizing radiation (IR) is a ubiquitous environmental stressor with genotoxic and epigenotoxic capabilities. Terrestrial IR, predominantly a low-linear energy transfer (LET) radiation, is being widely utilized in medicine, as well as in multiple industrial applications. Additionally, an interest in understanding the effects of high-LET irradiation is emerging due to the potential of exposure during space missions and the growing utilization of high-LET radiation in medicine. CONCLUSIONS In this review, we summarize the current knowledge of the effects of IR on DNA methylation, a key epigenetic mechanism regulating the expression of genetic information. We discuss global, repetitive elements and gene-specific DNA methylation in light of exposure to high and low doses of high- or low-LET IR, fractionated IR exposure, and bystander effects. Finally, we describe the mechanisms of IR-induced alterations to DNA methylation and discuss ways in which that understanding can be applied clinically, including utilization of DNA methylation as a predictor of response to radiotherapy and in the manipulation of DNA methylation patterns for tumor radiosensitization.
Collapse
Affiliation(s)
- Isabelle R Miousse
- a Department of Environmental and Occupational Health, Fay W. Boozman College of Public Health , University of Arkansas for Medical Sciences , Little Rock , AR , USA
| | - Kristy R Kutanzi
- a Department of Environmental and Occupational Health, Fay W. Boozman College of Public Health , University of Arkansas for Medical Sciences , Little Rock , AR , USA
| | - Igor Koturbash
- a Department of Environmental and Occupational Health, Fay W. Boozman College of Public Health , University of Arkansas for Medical Sciences , Little Rock , AR , USA
| |
Collapse
|
39
|
Wang Y, Liu Q, Zhao W, Zhou X, Miao G, Sun C, Zhang H. NADPH Oxidase Activation Contributes to Heavy Ion Irradiation-Induced Cell Death. Dose Response 2017; 15:1559325817699697. [PMID: 28473742 PMCID: PMC5407528 DOI: 10.1177/1559325817699697] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Increased oxidative stress plays an important role in heavy ion radiation-induced cell death. The mechanism involved in the generation of elevated reactive oxygen species (ROS) is not fully illustrated. Here we show that NADPH oxidase activation is closely related to heavy ion radiation-induced cell death via excessive ROS generation. Cell death and cellular ROS can be greatly reduced in irradiated cancer cells with the preincubation of diphenyleneiodium, an inhibitor of NADPH oxidase. Most of the NADPH oxidase (NOX) family proteins (NOX1, NOX2, NOX3, NOX4, and NOX5) showed increased expression after heavy ion irradiation. Meanwhile, the cytoplasmic subunit p47phox was translocated to the cell membrane and localized with NOX2 to form reactive NADPH oxidase. Our data suggest for the first time that ROS generation, as mediated by NADPH oxidase activation, could be an important contributor to heavy ion irradiation-induced cell death.
Collapse
Affiliation(s)
- Yupei Wang
- Department of Heavy Ion Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, Gansu, China.,Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou, Gansu, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Qing Liu
- Department of Heavy Ion Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, Gansu, China.,Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou, Gansu, China.,School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Weiping Zhao
- Department of Heavy Ion Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, Gansu, China.,Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou, Gansu, China
| | - Xin Zhou
- Department of Heavy Ion Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, Gansu, China.,Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou, Gansu, China
| | - Guoying Miao
- Department of Heavy Ion Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, Gansu, China.,Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou, Gansu, China.,Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Chao Sun
- Department of Heavy Ion Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, Gansu, China.,Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou, Gansu, China
| | - Hong Zhang
- Department of Heavy Ion Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, China.,Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, Gansu, China.,Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou, Gansu, China.,Gansu Wuwei Institute of Medical Sciences, Wuwei, China
| |
Collapse
|
40
|
Zhou Q, Song W, Xiao W. Dioscin induces demethylation of DAPK-1 and RASSF-1alpha genes via the antioxidant capacity, resulting in apoptosis of bladder cancer T24 cells. EXCLI JOURNAL 2017; 16:101-112. [PMID: 28435431 PMCID: PMC5379114 DOI: 10.17179/excli2016-571] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 01/12/2017] [Indexed: 12/28/2022]
Abstract
DNA methylation at CpG rich regions often occurs at tumor suppressor gene promoters, resulting in reduced gene expression and final carcinogenesis. Hypermethylation of tumor suppressor genes, including DAPK-1 and RASSF-1α genes, have been found in patients with bladder carcinoma (BC) in some western countries. Reactive oxygen species (ROS) was reported to play a causative role in gene hypermethylation. In this study, we detected the methylation status and expression of DAPK1 and RASSF-1α genes in tissue samples from Chinese BC patients, using methylation-specific PCR, reverse transcription PCR and western blotting. Further, we examined the ability of dioscin, a natural antioxidant, to regulate methylation status and expression of DAPK-1 and RASSF-1α genes in BC cell lines. In our results, DAPK-1 and RASSF-1α genes showed higher methylation level and lower express level in BC tissues than matched normal tissues. Treatment with dioscin decreased viability of BC 5637 and T24 cells, but not non-cancer bladder epithelial cell, SV-HUC-1. Dioscin triggered demethylation of DAPK1 and RASSF-1α genes in T24 cells and increased the gene and protein expression in 5637 and T24 cells. Both dioscin and substituted antioxidants (N-acetyl cysteine and Vitamin E) decreased intracellular ROS, but the effect of dioscin was abolished by adding H2O2. Similar to dioscin, the substituted antioxidants also induced the gene demethylation and T24 cell apoptosis. Co-treatment with dioscin and H2O2 had no such effects. Collectively, dioscin induces demethylation of DAPK-1 and RASSF-1α genes via the antioxidant capacity, resulting in apoptosis of bladder cancer T24 cells or inhibitory cell viability.
Collapse
Affiliation(s)
- Qiang Zhou
- Department of Urology, Hunan Provincial People's Hospital, 61#, West Jiefang Road, Changsha, Hunan, P.R. China
| | - Wie Song
- Department of Urology, Hunan Provincial People's Hospital, 61#, West Jiefang Road, Changsha, Hunan, P.R. China
| | - Wei Xiao
- Department of Urology, Hunan Provincial People's Hospital, 61#, West Jiefang Road, Changsha, Hunan, P.R. China
| |
Collapse
|
41
|
Prior S, Miousse IR, Nzabarushimana E, Pathak R, Skinner C, Kutanzi KR, Allen AR, Raber J, Tackett AJ, Hauer-Jensen M, Nelson GA, Koturbash I. Densely ionizing radiation affects DNA methylation of selective LINE-1 elements. ENVIRONMENTAL RESEARCH 2016; 150:470-481. [PMID: 27419368 PMCID: PMC5003736 DOI: 10.1016/j.envres.2016.06.043] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 06/15/2016] [Accepted: 06/27/2016] [Indexed: 05/26/2023]
Abstract
Long Interspersed Nucleotide Element 1 (LINE-1) retrotransposons are heavily methylated and are the most abundant transposable elements in mammalian genomes. Here, we investigated the differential DNA methylation within the LINE-1 under normal conditions and in response to environmentally relevant doses of sparsely and densely ionizing radiation. We demonstrate that DNA methylation of LINE-1 elements in the lungs of C57BL6 mice is dependent on their evolutionary age, where the elder age of the element is associated with the lower extent of DNA methylation. Exposure to 5-aza-2'-deoxycytidine and methionine-deficient diet affected DNA methylation of selective LINE-1 elements in an age- and promoter type-dependent manner. Exposure to densely IR, but not sparsely IR, resulted in DNA hypermethylation of older LINE-1 elements, while the DNA methylation of evolutionary younger elements remained mostly unchanged. We also demonstrate that exposure to densely IR increased mRNA and protein levels of LINE-1 via the loss of the histone H3K9 dimethylation and an increase in the H3K4 trimethylation at the LINE-1 5'-untranslated region, independently of DNA methylation. Our findings suggest that DNA methylation is important for regulation of LINE-1 expression under normal conditions, but histone modifications may dictate the transcriptional activity of LINE-1 in response to exposure to densely IR.
Collapse
Affiliation(s)
- Sara Prior
- Department of Environmental and Occupational Health, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Isabelle R Miousse
- Department of Environmental and Occupational Health, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Etienne Nzabarushimana
- Department of Environmental and Occupational Health, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; Department of Bioinformatics, School of Informatics and Computing, Indiana University, Bloomington, IN 47405, USA
| | - Rupak Pathak
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Charles Skinner
- Department of Environmental and Occupational Health, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Kristy R Kutanzi
- Department of Environmental and Occupational Health, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Antiño R Allen
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Jacob Raber
- Departments of Behavioral Neuroscience, Neurology, and Radiation Medicine, Division of Neuroscience, ONPRC, Oregon Health & Science University, Portland, OR 97239, USA
| | - Alan J Tackett
- Department of Biochemistry, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Martin Hauer-Jensen
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Gregory A Nelson
- Department of Basic Sciences, Division of Radiation Research, Loma Linda University, Loma Linda, CA 92350, USA
| | - Igor Koturbash
- Department of Environmental and Occupational Health, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| |
Collapse
|
42
|
56Fe ion irradiation induced apoptosis through Nrf2 pathway in mouse testis. Life Sci 2016; 157:32-37. [DOI: 10.1016/j.lfs.2016.05.043] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 05/19/2016] [Accepted: 05/30/2016] [Indexed: 12/18/2022]
|
43
|
Low Doses of Oxygen Ion Irradiation Cause Acute Damage to Hematopoietic Cells in Mice. PLoS One 2016; 11:e0158097. [PMID: 27367604 PMCID: PMC4930193 DOI: 10.1371/journal.pone.0158097] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 06/11/2016] [Indexed: 12/29/2022] Open
Abstract
One of the major health risks to astronauts is radiation on long-duration space missions. Space radiation from sun and galactic cosmic rays consists primarily of 85% protons, 14% helium nuclei and 1% high-energy high-charge (HZE) particles, such as oxygen (16O), carbon, silicon, and iron ions. HZE particles exhibit dense linear tracks of ionization associated with clustered DNA damage and often high relative biological effectiveness (RBE). Therefore, new knowledge of risks from HZE particle exposures must be obtained. In the present study, we investigated the acute effects of low doses of 16O irradiation on the hematopoietic system. Specifically, we exposed C57BL/6J mice to 0.1, 0.25 and 1.0 Gy whole body 16O (600 MeV/n) irradiation and examined the effects on peripheral blood (PB) cells, and bone marrow (BM) hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) at two weeks after the exposure. The results showed that the numbers of white blood cells, lymphocytes, monocytes, neutrophils and platelets were significantly decreased in PB after exposure to 1.0 Gy, but not to 0.1 or 0.25 Gy. However, both the frequency and number of HPCs and HSCs were reduced in a radiation dose-dependent manner in comparison to un-irradiated controls. Furthermore, HPCs and HSCs from irradiated mice exhibited a significant reduction in clonogenic function determined by the colony-forming and cobblestone area-forming cell assays. These acute adverse effects of 16O irradiation on HSCs coincided with an increased production of reactive oxygen species (ROS), enhanced cell cycle entry of quiescent HSCs, and increased DNA damage. However, none of the 16O exposures induced apoptosis in HSCs. These data suggest that exposure to low doses of 16O irradiation induces acute BM injury in a dose-dependent manner primarily via increasing ROS production, cell cycling, and DNA damage in HSCs. This finding may aid in developing novel strategies in the protection of the hematopoietic system from space radiation.
Collapse
|
44
|
Rithidech KN, Jangiam W, Tungjai M, Gordon C, Honikel L, Whorton EB. Induction of Chronic Inflammation and Altered Levels of DNA Hydroxymethylation in Somatic and Germinal Tissues of CBA/CaJ Mice Exposed to (48)Ti Ions. Front Oncol 2016; 6:155. [PMID: 27446801 PMCID: PMC4921787 DOI: 10.3389/fonc.2016.00155] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 06/10/2016] [Indexed: 11/17/2022] Open
Abstract
Although the lung is one of the target organs at risk for cancer induction from exposure to heavy ions found in space, information is insufficient on cellular/molecular responses linked to increased cancer risk. Knowledge of such events may aid in the development of new preventive measures. Furthermore, although it is known that germinal cells are sensitive to X- or γ-rays, there is little information on the effects of heavy ions on germinal cells. Our goal was to investigate in vivo effects of 1 GeV/n (48)Ti ions (one of the important heavy ions found in the space environment) on somatic (lung) and germinal (testis) tissues collected at various times after a whole body irradiation of CBA/CaJ mice (0, 0.1, 0.25, or 0.5 Gy, delivered at 1 cGy/min). We hypothesized that (48)Ti-ion-exposure induced damage in both tissues. Lung tissue was collected from each mouse from each treatment group at 1 week, 1 month, and 6 months postirradiation. For the testis, we collected samples at 6 months postirradiation. Hence, only late-occurring effects of (48)Ti ions in the testis were studied. There were five mice per treatment group at each harvest time. We investigated inflammatory responses after exposure to (48)Ti ions by measuring the levels of activated nuclear factor kappa B and selected pro-inflammatory cytokines in both tissues of the same mouse. These measurements were coupled with the quantitation of the levels of global 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC). Our data clearly showed the induction of chronic inflammation in both tissues of exposed mice. A dose-dependent reduction in global 5hmC was found in the lung at all time-points and in testes collected at 6 months postirradiation. In contrast, significant increases in global 5mC were found only in lung and testes collected at 6 months postirradiation from mice exposed to 0.5 Gy of 1 GeV/n (48)Ti ions. Overall, our data showed that (48)Ti ions may create health risks in both lung and testicular tissues.
Collapse
Affiliation(s)
| | - Witawat Jangiam
- Department of Pathology, Stony Brook University, Stony Brook, NY, USA
- Department of Chemical Engineering, Faculty of Engineering, Burapha University, Chonburi, Thailand
| | - Montree Tungjai
- Department of Pathology, Stony Brook University, Stony Brook, NY, USA
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Center of Excellence for Molecular Imaging, Chiang Mai University, Chiang Mai, Thailand
| | - Chris Gordon
- Department of Pathology, Stony Brook University, Stony Brook, NY, USA
| | - Louise Honikel
- Department of Pathology, Stony Brook University, Stony Brook, NY, USA
| | | |
Collapse
|
45
|
Radiation-induced changes in DNA methylation of repetitive elements in the mouse heart. Mutat Res 2016; 787:43-53. [PMID: 26963372 DOI: 10.1016/j.mrfmmm.2016.02.009] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 02/02/2016] [Accepted: 02/28/2016] [Indexed: 01/04/2023]
Abstract
DNA methylation is a key epigenetic mechanism, needed for proper control over the expression of genetic information and silencing of repetitive elements. Exposure to ionizing radiation, aside from its strong genotoxic potential, may also affect the methylation of DNA, within the repetitive elements, in particular. In this study, we exposed C57BL/6J male mice to low absorbed mean doses of two types of space radiation-proton (0.1 Gy, 150 MeV, dose rate 0.53 ± 0.08 Gy/min), and heavy iron ions ((56)Fe) (0.5 Gy, 600 MeV/n, dose rate 0.38 ± 0.06 Gy/min). Radiation-induced changes in cardiac DNA methylation associated with repetitive elements were detected. Specifically, modest hypomethylation of retrotransposon LINE-1 was observed at day 7 after irradiation with either protons or (56)Fe. This was followed by LINE-1, and other retrotransposons, ERV2 and SINE B1, as well as major satellite DNA hypermethylation at day 90 after irradiation with (56)Fe. These changes in DNA methylation were accompanied by alterations in the expression of DNA methylation machinery and affected the one-carbon metabolism pathway. Furthermore, loss of transposable elements expression was detected in the cardiac tissue at the 90-day time-point, paralleled by substantial accumulation of mRNA transcripts, associated with major satellites. Given that the one-carbon metabolism pathway can be modulated by dietary modifications, these findings suggest a potential strategy for the mitigation and, possibly, prevention of the negative effects exerted by ionizing radiation on the cardiovascular system. Additionally, we show that the methylation status and expression of repetitive elements may serve as early biomarkers of exposure to space radiation.
Collapse
|
46
|
Pirela SV, Miousse IR, Lu X, Castranova V, Thomas T, Qian Y, Bello D, Kobzik L, Koturbash I, Demokritou P. Effects of Laser Printer-Emitted Engineered Nanoparticles on Cytotoxicity, Chemokine Expression, Reactive Oxygen Species, DNA Methylation, and DNA Damage: A Comprehensive in Vitro Analysis in Human Small Airway Epithelial Cells, Macrophages, and Lymphoblasts. ENVIRONMENTAL HEALTH PERSPECTIVES 2016; 124:210-9. [PMID: 26080392 PMCID: PMC4749083 DOI: 10.1289/ehp.1409582] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 06/12/2015] [Indexed: 05/20/2023]
Abstract
BACKGROUND Engineered nanomaterials (ENMs) incorporated into toner formulations of printing equipment become airborne during consumer use. Although information on the complex physicochemical and toxicological properties of both toner powders and printer-emitted particles (PEPs) continues to grow, most toxicological studies have not used the actual PEPs but rather have primarily used raw toner powders, which are not representative of current exposures experienced at the consumer level during printing. OBJECTIVES We assessed the biological responses of a panel of human cell lines to PEPs. METHODS Three physiologically relevant cell lines--small airway epithelial cells (SAECs), macrophages (THP-1 cells), and lymphoblasts (TK6 cells)--were exposed to PEPs at a wide range of doses (0.5-100 μg/mL) corresponding to human inhalation exposure durations at the consumer level of 8 hr or more. Following treatment, toxicological parameters reflecting distinct mechanisms were evaluated. RESULTS PEPs caused significant membrane integrity damage, an increase in reactive oxygen species (ROS) production, and an increase in pro-inflammatory cytokine release in different cell lines at doses equivalent to exposure durations from 7.8 to 1,500 hr. Furthermore, there were differences in methylation patterns that, although not statistically significant, demonstrate the potential effects of PEPs on the overall epigenome following exposure. CONCLUSIONS The in vitro findings obtained in this study suggest that laser printer-emitted engineered nanoparticles may be deleterious to lung cells and provide preliminary evidence of epigenetic modifications that might translate to pulmonary disorders.
Collapse
Affiliation(s)
- Sandra V. Pirela
- Department of Environmental Health, Center for Nanotechnology and Nanotoxicology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
| | - Isabelle R. Miousse
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Xiaoyan Lu
- Department of Environmental Health, Center for Nanotechnology and Nanotoxicology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
| | - Vincent Castranova
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, West Virginia, USA
| | - Treye Thomas
- Office of Hazard Identification and Reduction, U.S. Consumer Product Safety Commission, Rockville, Maryland, USA
| | - Yong Qian
- Pathology and Physiology Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, West Virginia, USA
| | - Dhimiter Bello
- Department of Environmental Health, Center for Nanotechnology and Nanotoxicology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
- Department of Work Environment, University of Massachusetts-Lowell, Lowell, Massachusetts, USA
| | - Lester Kobzik
- Department of Environmental Health, Center for Nanotechnology and Nanotoxicology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
| | - Igor Koturbash
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Philip Demokritou
- Department of Environmental Health, Center for Nanotechnology and Nanotoxicology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
- Address correspondence to P. Demokritou, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Harvard University, 665 Huntington Ave., Room 1310B, Boston, MA 02115 USA. (617) 432-3481. E-mail:
| |
Collapse
|
47
|
Pirela SV, Lu X, Miousse I, Sisler JD, Qian Y, Guo N, Koturbash I, Castranova V, Thomas T, Godleski J, Demokritou P. Effects of intratracheally instilled laser printer-emitted engineered nanoparticles in a mouse model: A case study of toxicological implications from nanomaterials released during consumer use. NANOIMPACT 2016; 1:1-8. [PMID: 26989787 PMCID: PMC4791579 DOI: 10.1016/j.impact.2015.12.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Incorporation of engineered nanomaterials (ENMs) into toners used in laser printers has led to countless quality and performance improvements. However, the release of ENMs during printing (consumer use) has raised concerns about their potential adverse health effects. The aim of this study was to use "real world" printer-emitted particles (PEPs), rather than raw toner powder, and assess the pulmonary responses following exposure by intratracheal instillation. Nine-week old male Balb/c mice were exposed to various doses of PEPs (0.5, 2.5 and 5 mg/kg body weight) by intratracheal instillation. These exposure doses are comparable to real world human inhalation exposures ranging from 13.7 to 141.9 h of printing. Toxicological parameters reflecting distinct mechanisms of action were evaluated, including lung membrane integrity, inflammation and regulation of DNA methylation patterns. Results from this in vivo toxicological analysis showed that while intratracheal instillation of PEPs caused no changes in the lung membrane integrity, there was a pulmonary immune response, indicated by an elevation in neutrophil and macrophage percentage over the vehicle control and low dose PEPs groups. Additionally, exposure to PEPs upregulated expression of the Ccl5 (Rantes), Nos1 and Ucp2 genes in the murine lung tissue and modified components of the DNA methylation machinery (Dnmt3a) and expression of transposable element (TE) LINE-1 compared to the control group. These genes are involved in both the repair process from oxidative damage and the initiation of immune responses to foreign pathogens. The results are in agreement with findings from previous in vitro cellular studies and suggest that PEPs may cause immune responses in addition to modifications in gene expression in the murine lung at doses that can be comparable to real world exposure scenarios, thereby raising concerns of deleterious health effects.
Collapse
Affiliation(s)
- Sandra V. Pirela
- Department of Environmental Health, Center for Nanotechnology and Nanotoxicology, T. H. Chan School of Public Health, Harvard University, Boston, MA, United States
| | - Xiaoyan Lu
- Department of Environmental Health, Center for Nanotechnology and Nanotoxicology, T. H. Chan School of Public Health, Harvard University, Boston, MA, United States
| | - Isabelle Miousse
- Department of Environmental and Occupational Health, College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Jennifer D. Sisler
- Pathology and Physiology Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, United States
| | - Yong Qian
- Pathology and Physiology Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, United States
| | - Nancy Guo
- Department of Pharmaceutical Sciences/Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV, United States
| | - Igor Koturbash
- Department of Environmental and Occupational Health, College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Vincent Castranova
- Department of Pharmaceutical Sciences/Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV, United States
| | - Treye Thomas
- U.S. Consumer Product Safety Commission, Office of Hazard Identification and Reduction, Rockville, MD, United States
| | - John Godleski
- Department of Environmental Health, Center for Nanotechnology and Nanotoxicology, T. H. Chan School of Public Health, Harvard University, Boston, MA, United States
| | - Philip Demokritou
- Department of Environmental Health, Center for Nanotechnology and Nanotoxicology, T. H. Chan School of Public Health, Harvard University, Boston, MA, United States
- Corresponding author at: Department of Environmental Health, Center for Nanotechnology and Nanotoxicology, T. H. Chan School of Public Health, Harvard University, 665 Huntington Avenue, Room 1310, Boston, MA 02115, United States. Tel.: +1 917 432 3481. (P. Demokritou)
| |
Collapse
|
48
|
Miousse IR, Chalbot MCG, Pathak R, Lu X, Nzabarushimana E, Krager K, Aykin-Burns N, Hauer-Jensen M, Demokritou P, Kavouras IG, Koturbash I. In Vitro Toxicity and Epigenotoxicity of Different Types of Ambient Particulate Matter. Toxicol Sci 2015; 148:473-87. [PMID: 26342214 PMCID: PMC5009441 DOI: 10.1093/toxsci/kfv200] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Exposure to ambient particulate matter (PM) has been associated with adverse health effects, including pulmonary and cardiovascular disease. Studies indicate that ambient PM originated from different sources may cause distinct biological effects. In this study, we sought to investigate the potential of various types of PM to cause epigenetic alterations in the in vitro system. RAW264.7 murine macrophages were exposed for 24 and 72 h to 5- and 50-μg/ml doses of the water soluble extract of 6 types of PM: soil dust, road dust, agricultural dust, traffic exhausts, biomass burning, and pollen, collected in January-April of 2014 in the area of Little Rock, Arkansas. Cytotoxicity, oxidative potential, epigenetic endpoints, and chromosomal aberrations were addressed. Exposure to 6 types of PM resulted in induction of cytotoxicity and oxidative stress in a type-, time-, and dose-dependent manner. Epigenetic alterations were characterized by type-, time-, and dose-dependent decreases of DNA methylation/demethylation machinery, increased DNA methyltransferases enzymatic activity and protein levels, and transcriptional activation and subsequent silencing of transposable elements LINE-1, SINE B1/B2. The most pronounced changes were observed after exposure to soil dust that were also characterized by hypomethylation and reactivation of satellite DNA and structural chromosomal aberrations in the exposed cells. The results of our study indicate that the water-soluble fractions of the various types of PM have differential potential to target the cellular epigenome.
Collapse
Affiliation(s)
- Isabelle R Miousse
- *Department of Environmental and Occupational Health, Fay W. Boozman College of Public Health and
| | - Marie-Cecile G Chalbot
- *Department of Environmental and Occupational Health, Fay W. Boozman College of Public Health and
| | - Rupak Pathak
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Xiaoyan Lu
- Center for Nanotechnology and Nanotoxicology, Department of Environmental Health, Harvard School of Public Health, Boston, Massachusetts; and
| | - Etienne Nzabarushimana
- *Department of Environmental and Occupational Health, Fay W. Boozman College of Public Health and Department of Biology, Indiana University, Bloomington, Indiana 47405
| | - Kimberly Krager
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Nukhet Aykin-Burns
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Martin Hauer-Jensen
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205
| | - Philip Demokritou
- Center for Nanotechnology and Nanotoxicology, Department of Environmental Health, Harvard School of Public Health, Boston, Massachusetts; and
| | - Ilias G Kavouras
- *Department of Environmental and Occupational Health, Fay W. Boozman College of Public Health and
| | - Igor Koturbash
- *Department of Environmental and Occupational Health, Fay W. Boozman College of Public Health and
| |
Collapse
|
49
|
Nzabarushimana E, Prior S, Miousse IR, Pathak R, Allen AR, Latendresse J, Olsen RHJ, Raber J, Hauer-Jensen M, Nelson GA, Koturbash I. Combined exposure to protons and (56)Fe leads to overexpression of Il13 and reactivation of repetitive elements in the mouse lung. LIFE SCIENCES IN SPACE RESEARCH 2015; 7:1-8. [PMID: 26553631 PMCID: PMC4641818 DOI: 10.1016/j.lssr.2015.08.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 08/08/2015] [Accepted: 08/15/2015] [Indexed: 05/15/2023]
Abstract
Interest in deep space exploration underlines the needs to investigate the effects of exposure to combined sources of space radiation. The lung is a target organ for radiation, and exposure to protons and heavy ions as radiation sources may lead to the development of degenerative disease and cancer. In this study, we evaluated the pro-fibrotic and epigenetic effects of exposure to protons (150 MeV/nucleon, 0.1 Gy) and heavy iron ions ((56)Fe, 600 MeV/nucleon, 0.5 Gy) alone or in combination (protons on Day 1 and (56)Fe on Day 2) in C57BL/6 male mice 4 weeks after irradiation. Exposure to (56)Fe, proton or in combination, did not result in histopathological changes in the murine lung. At the same time, combined exposure to protons and (56)Fe resulted in pronounced molecular alterations in comparison with either source of radiation alone. Specifically, we observed a substantial increase in the expression of cytokine Il13, loss of expression of DNA methyltransferase Dnmt1, and reactivation of LINE-1, SINE B1 retrotransposons, and major and minor satellites. Given the deleterious potential of the observed effects that may lead to development of chronic lung injury, pulmonary fibrosis, and cancer, future studies devoted to the investigation of the long-term effects of combined exposures to proton and heavy ions are clearly needed.
Collapse
Affiliation(s)
- Etienne Nzabarushimana
- Department of Environmental Health, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA; Department of Biology, Indiana University, Bloomington, IN, 47405, USA
| | - Sara Prior
- Department of Environmental Health, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Isabelle R Miousse
- Department of Environmental Health, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Rupak Pathak
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Antiño R Allen
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | | | - Reid H J Olsen
- Department of Behavioral Neuroscience, Oregon Health & Science University Portland, OR, 97239, USA
| | - Jacob Raber
- Departments of Neurology and Radiation Medicine, Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University Portland, OR, 97239, USA
| | - Martin Hauer-Jensen
- Division of Radiation Health, Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Gregory A Nelson
- Department of Basic Sciences, Division of Radiation Research, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Igor Koturbash
- Department of Environmental Health, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| |
Collapse
|
50
|
Muralidharan S, Sasi SP, Zuriaga MA, Hirschi KK, Porada CD, Coleman MA, Walsh KX, Yan X, Goukassian DA. Ionizing Particle Radiation as a Modulator of Endogenous Bone Marrow Cell Reprogramming: Implications for Hematological Cancers. Front Oncol 2015; 5:231. [PMID: 26528440 PMCID: PMC4604322 DOI: 10.3389/fonc.2015.00231] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 10/01/2015] [Indexed: 12/15/2022] Open
Abstract
Exposure of individuals to ionizing radiation (IR), as in the case of astronauts exploring space or radiotherapy cancer patients, increases their risk of developing secondary cancers and other health-related problems. Bone marrow (BM), the site in the body where hematopoietic stem cell (HSC) self-renewal and differentiation to mature blood cells occurs, is extremely sensitive to low-dose IR, including irradiation by high-charge and high-energy particles. Low-dose IR induces DNA damage and persistent oxidative stress in the BM hematopoietic cells. Inefficient DNA repair processes in HSC and early hematopoietic progenitors can lead to an accumulation of mutations whereas long-lasting oxidative stress can impair hematopoiesis itself, thereby causing long-term damage to hematopoietic cells in the BM niche. We report here that low-dose 1H- and 56Fe-IR significantly decreased the hematopoietic early and late multipotent progenitor (E- and L-MPP, respectively) cell numbers in mouse BM over a period of up to 10 months after exposure. Both 1H- and 56Fe-IR increased the expression of pluripotent stem cell markers Sox2, Nanog, and Oct4 in L-MPPs and 10 months post-IR exposure. We postulate that low doses of 1H- and 56Fe-IR may induce endogenous cellular reprogramming of BM hematopoietic progenitor cells to assume a more primitive pluripotent phenotype and that IR-induced oxidative DNA damage may lead to mutations in these BM progenitors. This could then be propagated to successive cell lineages. Persistent impairment of BM progenitor cell populations can disrupt hematopoietic homeostasis and lead to hematologic disorders, and these findings warrant further mechanistic studies into the effects of low-dose IR on the functional capacity of BM-derived hematopoietic cells including their self-renewal and pluripotency.
Collapse
Affiliation(s)
- Sujatha Muralidharan
- Whitaker Cardiovascular Institute, Boston University School of Medicine , Boston, MA , USA
| | - Sharath P Sasi
- Cardiovascular Research Center, GeneSys Research Institute , Boston, MA , USA
| | - Maria A Zuriaga
- Whitaker Cardiovascular Institute, Boston University School of Medicine , Boston, MA , USA
| | - Karen K Hirschi
- Yale Cardiovascular Research Center, Yale School of Medicine , New Haven, CT , USA
| | - Christopher D Porada
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine , Winston-Salem, NC , USA
| | - Matthew A Coleman
- Radiation Oncology, School of Medicine, University of California Davis , Sacramento, CA , USA ; Lawrence Livermore National Laboratory , Livermore, CA , USA
| | - Kenneth X Walsh
- Whitaker Cardiovascular Institute, Boston University School of Medicine , Boston, MA , USA
| | - Xinhua Yan
- Cardiovascular Research Center, GeneSys Research Institute , Boston, MA , USA ; Tufts University School of Medicine , Boston, MA , USA
| | - David A Goukassian
- Whitaker Cardiovascular Institute, Boston University School of Medicine , Boston, MA , USA ; Cardiovascular Research Center, GeneSys Research Institute , Boston, MA , USA ; Tufts University School of Medicine , Boston, MA , USA
| |
Collapse
|