1
|
Chaurasia RK, Sapra BK, Aswal DK. Interplay of immune modulation, adaptive response and hormesis: Suggestive of threshold for clinical manifestation of effects of ionizing radiation at low doses? THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 917:170178. [PMID: 38280586 DOI: 10.1016/j.scitotenv.2024.170178] [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: 08/10/2023] [Revised: 12/26/2023] [Accepted: 01/13/2024] [Indexed: 01/29/2024]
Abstract
The health impacts of low-dose ionizing radiation exposures have been a subject of debate over the last three to four decades. While there has been enough evidence of "no adverse observable" health effects at low doses and low dose rates, the hypothesis of "Linear No Threshold" continues to rule and govern the principles of radiation protection and the formulation of regulations and public policies. In adopting this conservative approach, the role of the biological processes underway in the human body is kept at abeyance. This review consolidates the available studies that discuss all related biological pathways and repair mechanisms that inhibit the progression of deleterious effects at low doses and low dose rates of ionizing radiation. It is pertinent that, taking cognizance of these processes, there is a need to have a relook at policies of radiation protection, which as of now are too stringent, leading to undue economic losses and negative public perception about radiation.
Collapse
Affiliation(s)
- R K Chaurasia
- Radiological Physics and Advisory Division, India; Health, Safety and Environment Group,Bhabha Atomic Research Centre, Mumbai 400085, India; Homi Bhabha National Institute, Mumbai 400094, India.
| | - B K Sapra
- Radiological Physics and Advisory Division, India; Health, Safety and Environment Group,Bhabha Atomic Research Centre, Mumbai 400085, India; Homi Bhabha National Institute, Mumbai 400094, India.
| | - D K Aswal
- Health, Safety and Environment Group,Bhabha Atomic Research Centre, Mumbai 400085, India; Homi Bhabha National Institute, Mumbai 400094, India.
| |
Collapse
|
2
|
Aye KT, Wattanapongpitak S, Supawat B, Kothan S, Udomtanakunchai C, Tima S, Tungjai M. Effect of pre-low-dose irradiation on anticancer activities of gallic acid in leukemic K562 and K562/Dox cells: cell viability and cellular energetic state studies. Med Oncol 2022; 39:229. [PMID: 36175689 DOI: 10.1007/s12032-022-01835-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 08/28/2022] [Indexed: 12/24/2022]
Abstract
The aim of this study was to determine the effects of pre-low-dose irradiation followed by gallic acid (GA) on cell viability and cellular energetic state of leukemic K562 and K562/Dox cells. The cells were irradiated with 0.02, 0.05, and 0.1 Gy of X-rays. For determining cell viability, pre-low-dose irradiation was followed by 10 or 100 µM GA at 24 h post-irradiation, and the cell viability was then determined at 48 h post-irradiation. For cellular energetic state, pre-low-dose irradiation was followed by 10 or 100 µM GA at 1.5 h post-irradiation and the mitochondrial activity, mitochondrial membrane potential (ΔΨm), and ATP level were determined at 3 h post-irradiation. The % cell viability was significantly decreased in both cells that were irradiated with X-rays followed by treatment with 10 or 100 µM GA at 24 h post-irradiation, when compared with control group. However, this did not happen when compared with GA alone without any pre-low-dose irradiation. The mitochondrial activity had significantly decreased in 10 µM GA-treated K562 cells and the mitochondrial activity, ΔΨm, and ATP levels had significantly decreased in 10 µM GA-treated K562/Dox cells after irradiation to X-rays when compared with GA alone group. In addition, the ΔΨm and ATP levels was significantly decreased in only 100 µM GA-treated K562/Dox cells, but was not decreased in 100 µM GA-treated K562 cells after exposure to X-rays. These findings suggest that pre-low-dose irradiation followed by GA could not kill K562 and K562/Dox cells, but could improve cellular energetic damage of GA effects possibly through mitochondrial impairment.
Collapse
Affiliation(s)
- Khin TheNu Aye
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Radiation Research and Medical Imaging, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
- Ph.D. Degree Program in Biomedical Sciences, Faculty of Associated Medical Sciences, Chiang Mai University, Under the CMU Presidential Scholarship, Chiang Mai, Thailand
| | - Sakornniya Wattanapongpitak
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Radiation Research and Medical Imaging, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Benjamaporn Supawat
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Radiation Research and Medical Imaging, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Suchart Kothan
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Radiation Research and Medical Imaging, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Chatchanok Udomtanakunchai
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Radiation Research and Medical Imaging, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Singkome Tima
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Montree Tungjai
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Center of Radiation Research and Medical Imaging, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
3
|
Welsh JS, Bevelacqua JJ, Mortazavi SMJ. Revisiting radiation hormesis: should lung adenocarcinoma patients be advised to reduce radon levels in their environment? Int J Radiat Biol 2021; 97:875-876. [PMID: 33970768 DOI: 10.1080/09553002.2021.1928788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/05/2021] [Accepted: 04/16/2021] [Indexed: 10/21/2022]
Affiliation(s)
- James S Welsh
- Department of Radiation Oncology Edward Hines Jr VA Hospital Hines, Illinois, USA
- Department of Radiation Oncology, Stritch School of Medicine, Loyola University, Chicago, IL, USA
| | | | - S M J Mortazavi
- Medical Physics and Engineering Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
4
|
Janiak MK, Pocięgiel M, Welsh JS. Time to rejuvenate ultra-low dose whole-body radiotherapy of cancer. Crit Rev Oncol Hematol 2021; 160:103286. [PMID: 33667656 DOI: 10.1016/j.critrevonc.2021.103286] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 12/11/2020] [Accepted: 02/27/2021] [Indexed: 12/24/2022] Open
Abstract
The results of clinical trials performed from the 1930s until the end of the 20th century in which total-body ultra-low level ionizing radiation (TB-LLR) was used demonstrate that this form of treatment can be equal or superior to other systemic anti-neoplastic modalities in terms of the rates of remissions, toxicity, and side effects. In this review, we provide the rationale for TB-LLR and analyze the results of reliable clinical trials in patients with predominantly lymphoproliferative disorders but also advanced solid cancers. The doses used in these trials did not exceed 0.1-0.2 Gy per fraction and cumulative totals ranged from 1 to 4 Gy. Based on the reviewed results we conclude that it is appropriate to revive interest in and resume clinical investigations of TB-LLR in order to refine and improve the effectiveness of such treatment, whether employed alone or in combination with other anticancer strategies.
Collapse
Affiliation(s)
- Marek K Janiak
- Military Institute of Hygiene and Epidemiology, Warsaw, Poland.
| | | | - James S Welsh
- Loyola University Chicago, Edward Hines Jr., VA Hospital, Stritch School of Medicine, Department of Radiation Oncology, Maywood, IL 601fv53, USA
| |
Collapse
|
5
|
Puspitasari A, Yamazaki H, Kawamura H, Nakano T, Takahashi A, Shirao T, Held KD. X-irradiation of developing hippocampal neurons causes changes in neuron population phenotypes, dendritic morphology and synaptic protein expression in surviving neurons at maturity. Neurosci Res 2019; 160:11-24. [PMID: 31711782 DOI: 10.1016/j.neures.2019.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 10/16/2019] [Accepted: 11/06/2019] [Indexed: 01/02/2023]
Abstract
The effects of X-irradiation on developing neurons and their functions are unclear. We used primary cultures of mouse hippocampal neurons to investigate the effects of X-irradiation on cell death in developing neurons by analyzing caspase-3, MAP2 and DAPI-labeled cells, and the phenotypes and function of surviving neurons, by examining GAD67-positive cells as a GABAergic marker, and the synaptic markers synapsin 1, drebrin and PSD-95 through its maturation. One-day in vitro (DIV 1) cells were exposed to 0.5 Gy or 1 Gy of X-rays. A significant increase in the percentage of activated caspase-3, a decrease in the number of MAP2/DAPI-positive cells and change in the percentage of GAD67 positive neurons, compared with sham controls, were found 6 days after 1 Gy and 13 days after 0.5 Gy of X-rays. The expression of PSD-95 and drebrin, as well as drebrin clusters, in the remaining neurons was decreased at DIV 21, in both 0.5 Gy and on 1 Gy-irradiation there was a reduced number of dendritic intersection as well. Together, our findings show that 0.5 Gy and 1 Gy of X-irradiation at DIV 1 not only causes neuronal cell death but elicits an increase in the percentage of inhibitory neurons, changes in the dendrites and decrease in expression of important synaptic proteins in the surviving neurons at maturity 3 weeks after exposure.
Collapse
Affiliation(s)
| | - Hiroyuki Yamazaki
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Hidemasa Kawamura
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Takashi Nakano
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Akihisa Takahashi
- Gunma University Heavy Ion Medical Center, Maebashi, Graduate School of Medicine, Gunma, Japan
| | - Tomoaki Shirao
- Department of Neurobiology and Behavior, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Kathryn D Held
- Gunma University Initiative for Advanced Research, Maebashi, Japan; Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
6
|
Ji K, Wang Y, Du L, Xu C, Liu Y, He N, Wang J, Liu Q. Research Progress on the Biological Effects of Low-Dose Radiation in China. Dose Response 2019; 17:1559325819833488. [PMID: 30833876 PMCID: PMC6393828 DOI: 10.1177/1559325819833488] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 12/19/2018] [Accepted: 12/29/2018] [Indexed: 01/07/2023] Open
Abstract
Human are exposed to ionizing radiation from natural and artificial sources, which consequently poses a possible risk to human health. However, accumulating evidence indicates that the biological effects of low-dose radiation (LDR) are different from those of high-dose radiation (HDR). Low-dose radiation–induced hormesis has been extensively observed in different biological systems, including immunological and hematopoietic systems. Adaptive responses in response to LDR that can induce cellular resistance to genotoxic effects from subsequent exposure to HDR have also been described and researched. Bystander effects, another type of biological effect induced by LDR, have been shown to widely occur in many cell types. Furthermore, the influence of LDR-induced biological effects on certain diseases, such as cancer and diabetes, has also attracted the interest of researchers. Many studies have suggested that LDR has the potential antitumor and antidiabetic complications effects. In addition, the researches on whether LDR could induce stochastic effects were also debated. Studies on the biological effects of LDR in China started in 1970s and considerable progress has been made since. In the present article, we provide an overview of the research progress on the biological effects of LDR in China.
Collapse
Affiliation(s)
- Kaihua Ji
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Department of Radiobiology, Institute of Radiation Medicine of Chinese Academy of Medical Science & Pecking Union Medical College, Tianjin, PR China
| | - Yan Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Department of Radiobiology, Institute of Radiation Medicine of Chinese Academy of Medical Science & Pecking Union Medical College, Tianjin, PR China
| | - Liqing Du
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Department of Radiobiology, Institute of Radiation Medicine of Chinese Academy of Medical Science & Pecking Union Medical College, Tianjin, PR China
| | - Chang Xu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Department of Radiobiology, Institute of Radiation Medicine of Chinese Academy of Medical Science & Pecking Union Medical College, Tianjin, PR China
| | - Yang Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Department of Radiobiology, Institute of Radiation Medicine of Chinese Academy of Medical Science & Pecking Union Medical College, Tianjin, PR China
| | - Ningning He
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Department of Radiobiology, Institute of Radiation Medicine of Chinese Academy of Medical Science & Pecking Union Medical College, Tianjin, PR China
| | - Jinhan Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Department of Radiobiology, Institute of Radiation Medicine of Chinese Academy of Medical Science & Pecking Union Medical College, Tianjin, PR China
| | - Qiang Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Department of Radiobiology, Institute of Radiation Medicine of Chinese Academy of Medical Science & Pecking Union Medical College, Tianjin, PR China
| |
Collapse
|
7
|
Abdelrazzak AB, El-Missiry MA, Ahmed MT, Elnady BF. Effect of low-dose X-rays on the liver of whole-body irradiated rats. Int J Radiat Biol 2019; 95:264-273. [DOI: 10.1080/09553002.2019.1554925] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
| | | | - Moustafa T. Ahmed
- Physics Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Basma F. Elnady
- Physics Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| |
Collapse
|
8
|
Devic C, Ferlazzo ML, Foray N. Influence of Individual Radiosensitivity on the Adaptive Response Phenomenon: Toward a Mechanistic Explanation Based on the Nucleo-Shuttling of ATM Protein. Dose Response 2018; 16:1559325818789836. [PMID: 30093841 PMCID: PMC6081762 DOI: 10.1177/1559325818789836] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 05/29/2018] [Accepted: 06/12/2018] [Indexed: 02/03/2023] Open
Abstract
The adaptive response (AR) phenomenon generally describes a protective effect caused by a "priming" low dose (dAR) delivered after a period of time (ΔtAR) before a higher "challenging" dose (DAR). The AR is currently observed in human cells if dAR, ΔtAR, and DAR belong to (0.001-0.5 Gy), (2-24 hours), (0.1-5 Gy), respectively. In order to investigate the molecular mechanisms specific to AR in human cells, we have systematically reviewed the experimental AR protocols, the cellular models, and the biological endpoints used from the 1980s. The AR appears to be preferentially observed in radiosensitive cells and is strongly dependent on individual radiosensitivity. To date, the model of the nucleo-shuttling of the ATM protein provides a relevant mechanistic explanation of the AR molecular and cellular events. Indeed, the priming dose dAR may result in the diffusion of a significant amount of active ATM monomers in the nucleus. These ATM monomers, added to those induced directly by the challenging dose DAR, may increase the efficiency of the response to DAR by a better ATM-dependent DNA damage recognition. Such mechanistic model would also explain why AR is not observed in radioresistant or hyperradiosensitive cells. Further investigations at low dose are needed to consolidate our hypotheses.
Collapse
Affiliation(s)
- Clément Devic
- Institut National de la Santé et de la Recherche Médicale (INSERM), Lyon, France.,Fibermetrix Company, Strasbourg, France
| | - Mélanie L Ferlazzo
- Institut National de la Santé et de la Recherche Médicale (INSERM), Lyon, France
| | - Nicolas Foray
- Institut National de la Santé et de la Recherche Médicale (INSERM), Lyon, France
| |
Collapse
|
9
|
Paraswani N, Thoh M, Bhilwade HN, Ghosh A. Early antioxidant responses via the concerted activation of NF-κB and Nrf2 characterize the gamma-radiation-induced adaptive response in quiescent human peripheral blood mononuclear cells. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2018; 831:50-61. [PMID: 29875077 DOI: 10.1016/j.mrgentox.2018.04.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 04/25/2018] [Accepted: 04/26/2018] [Indexed: 11/16/2022]
Abstract
The radiation-induced adaptive response (RI-AR) is a non-targeted effect which is outside the scope of the classical Linear-No-Threshold (LNT) dose-response paradigm. However, the mechanisms of the RI-AR are not well understood. We have studied the RI-AR in quiescent human peripheral blood mononuclear cells (PBMCs). PBMCs in G0 phase were 'primed' with a low dose (100 mGy gamma radiation) and then, after an 'adaptive window' of 4 h, 'challenged' with a high dose (2 Gy). A small (5.7%) increase in viability and a decrease in DNA strand breaks were seen in primed cells, compared to non-primed cells. This was consistent with lower levels of reactive oxygen species, higher mitochondrial membrane potential, and increased activity of antioxidant enzymes such as catalase, superoxide dismutase, thioredoxin reductase, and glutathione peroxidase, in the primed cells. Reduced oxidative stress in primed PBMCs correlated with greater nuclear translocation of the redox-sensitive transcription factors Nuclear factor kappa B (NF-κB) and Nuclear factor E2-related factor 2 (Nrf2). Distinct differences in responses were seen in PBMCs irradiated with low dose (100 mGy) and high dose (2 Gy). These findings provide insight into the mechanisms of radioadaptation in human cells.
Collapse
Affiliation(s)
- Neha Paraswani
- Radiation Signaling Group, Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Mumbai 400 085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai, 400 094, India
| | - Maikho Thoh
- Free Radical Biology Section, Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Mumbai 400 085, India
| | - Hari N Bhilwade
- Free Radical Biology Section, Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Mumbai 400 085, India
| | - Anu Ghosh
- Radiation Signaling Group, Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Mumbai 400 085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai, 400 094, India.
| |
Collapse
|
10
|
Feng L, Qin L, Guo D, Deng D, Lu F, Li H, Bao N, Yang X, Ding H, Li J. Immunological mechanism of low-dose priming radiation resistance in walker-256 tumor model mice. Exp Ther Med 2017; 14:3868-3873. [PMID: 29042994 DOI: 10.3892/etm.2017.4975] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 06/08/2017] [Indexed: 12/27/2022] Open
Abstract
The aim of the present study was to investigate whether low-dose priming radiation induces antitumor immunity that can be augmented by the modulation of natural killer (NK) cell and cytokine activity using a mouse tumor model. Walker-256 cells were injected into the right flank of male BALB/c mice. At 7 days after inoculation, mice were divided into three groups, including group 1,2,3. In group 1 the mice were without radiation, in group 2 the mice were received 2 Gy radiation only, and in group 3 the mice were radiated with a priming dose of 75 mGy followed by 2 Gy radiation after 24 h. On day 21 following the radiation, the tumors were removed and the tumor index (tumor weight as a percentage of body weight) was calculated. At 1, 7, 14 and 21 days following the 2 Gy radiation, mouse splenocytes were isolated to analyze the NK activity and measure the production of the cytokines interleukin-1β, interferon-γ and tumor necrosis factor-α by ELISA. Apoptosis was also measured by flow cytometry. The results demonstrated that priming radiation significantly delayed the tumor growth and prolonged the median survival time to 38 days compared with the 31-day survival in the 2 Gy radiation group. The percentage of apoptotic cells was significantly higher in the mice that received 75 mGy + 2 Gy radiation compared with that in the mice that received 2 Gy alone; by contrast, mice that were not irradiated exhibited a relatively low level of apoptosis. The primed mice had a higher level of NK activity as compared with the mice exposed to 2 Gy radiation only or mice that were not irradiated. Furthermore, cytokine expression remained at a higher level in mice receiving priming dose of radiation compared that in the mice receiving only 2 Gy radiation. In conclusion, the results indicated that low-dose priming X-ray radiation may enhance the NK activity and the levels of cytokines, and that the immune response serves an important role in anticancer therapy, including radiotherapy.
Collapse
Affiliation(s)
- Li Feng
- Ultrasound Department, Qianfoshan Hospital of Shandong, Jinan, Shandong 250014, P.R. China
| | - Ling Qin
- Ultrasound Department, Qianfoshan Hospital of Shandong, Jinan, Shandong 250014, P.R. China
| | - Dan Guo
- Graduate Department, Taishan Medical University, Taian, Shandong 271016, P.R. China
| | - Daping Deng
- Laboratory of Radiation Biology, The Radiation Medical Institute, Shandong Academy of Medical Sciences, Jinan, Shandong 250062, P.R. China
| | - Feng Lu
- Laboratory of Radiation Biology, The Radiation Medical Institute, Shandong Academy of Medical Sciences, Jinan, Shandong 250062, P.R. China
| | - Hailiang Li
- Laboratory of Radiation Biology, The Radiation Medical Institute, Shandong Academy of Medical Sciences, Jinan, Shandong 250062, P.R. China
| | - Narisu Bao
- Department of Human Anatomy, The School of Medicine of Inner Mongolia University for The Nationalities, Tongliao, Inner Mongolia 028041, P.R. China
| | - Xiting Yang
- Department of Human Anatomy, The School of Medicine of Inner Mongolia University for The Nationalities, Tongliao, Inner Mongolia 028041, P.R. China
| | - Hongyu Ding
- Ultrasound Department, Qianfoshan Hospital of Shandong, Jinan, Shandong 250014, P.R. China
| | - Jianguo Li
- Department of Human Anatomy, The School of Medicine of Inner Mongolia University for The Nationalities, Tongliao, Inner Mongolia 028041, P.R. China.,The Key Laboratory of Bioactive Materials, Ministry of Education, School of Medicine, Nankai University, Tianjin 300071, P.R. China
| |
Collapse
|
11
|
Tungjai M, Phathakanon N, Rithidech KN. Effects of Medical Diagnostic Low-dose X Rays on Human Lymphocytes: Mitochondrial Membrane Potential, Apoptosis and Cell Cycle. HEALTH PHYSICS 2017; 112:458-464. [PMID: 28350700 DOI: 10.1097/hp.0000000000000647] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Low-dose radiation is widely used across the world for the diagnosis of many diseases by means of a variety of imaging technologies. However, the harmful effects of exposure to low-dose radiation during medical examination remain controversial. The authors studied the effects of medical diagnostic low-dose x rays (i.e., 0.03, 0.05, or 0.1 mGy) after an in vitro exposure of human lymphocytes. Cells with no irradiation served as the non-irradiated control group. Three biological indicators were used to determine the effects of medical diagnostic low-dose x rays at 4, 8, 24, 48, and 72 h post-irradiation. These biological endpoints were mitochondrial membrane potential (ΔΨm), cell cycle, and apoptosis. Results indicated no changes in the ΔΨm, number of apoptotic cells, and cell cycle in lymphocytes exposed to these low doses of radiation, as compared to the corresponding non-irradiated lymphocytes at all harvest time-points. These results suggested that there were no harmful effects of the diagnostic low-dose x rays when human lymphocytes were exposed in an in vitro condition.
Collapse
Affiliation(s)
- Montree Tungjai
- *Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, 110 Intawaroroj Rd., Sripoom, Chiang Mai, 50200, Thailand; †Department of Pathology, Stony Brook University, Stony Brook, NY 11794-8691
| | | | | |
Collapse
|
12
|
Morel KL, Ormsby RJ, Bezak E, Sweeney CJ, Sykes PJ. Parthenolide Selectively Sensitizes Prostate Tumor Tissue to Radiotherapy while Protecting Healthy Tissues In Vivo. Radiat Res 2017; 187:501-512. [PMID: 28398879 DOI: 10.1667/rr14710.1] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Radiotherapy is widely used in cancer treatment, however the benefits can be limited by radiation-induced damage to neighboring normal tissues. Parthenolide (PTL) exhibits anti-inflammatory and anti-tumor properties and selectively induces radiosensitivity in prostate cancer cell lines, while protecting primary prostate epithelial cell lines from radiation-induced damage. Low doses of radiation have also been shown to protect from subsequent high-dose-radiation-induced apoptosis as well as DNA damage. These properties of PTL and low-dose radiation could be used to improve radiotherapy by killing more tumor cells and less normal cells. Sixteen-week-old male Transgenic Adenocarcinoma of the Mouse Prostate (TRAMP) and C57BL/6J mice were treated with PTL (40 mg/kg), dimethylaminoparthenolide (DMAPT, a PTL analogue with increased bioavailability) (100 mg/kg), or vehicle control three times over one week prior to combinations of low (10 mGy) and high (6 Gy) doses of whole-body X-irradiation. Tissues were analyzed for apoptosis at a range of time points up to 72 h postirradiation. Both PTL and DMAPT protected normal tissues, but not prostate tumor tissues, from a significant proportion of high-dose-radiation-induced apoptosis. DMAPT provided superior protection compared to PTL in normal dorsolateral prostate (71.7% reduction, P = 0.026), spleen (48.2% reduction, P = 0.0001) and colorectal tissue (38.0% reduction, P = 0.0002), and doubled radiation-induced apoptosis in TRAMP prostate tumor tissue (101.3% increase, P = 0.039). Both drugs induced the greatest radiosensitivity in TRAMP prostate tissue in areas with higher grade prostatic intraepithelial neoplasia (PIN) lesions. A 10 mGy dose delivered 3 h prior to a 6 Gy dose induced a radioadaptive apoptosis response in normal C57Bl/6J prostate (28.4% reduction, P = 0.045) and normal TRAMP spleen (13.6% reduction, P = 0.047), however the low-dose-adaptive radioprotection did not significantly add to the PTL/DMAPT-induced protection in normal tissues, nor did it affect tumor kill. These results support the use of the more bioavailable DMAPT and low-dose radiation, alone or in combination as useful radioprotectors of normal tissues to alleviate radiotherapy-induced side-effects in patients. The enhanced radiosensitisation in prostate tissues displaying high-grade PIN suggests that DMAPT also holds promise for targeted therapy of advanced prostate cancer, which may go on to become metastatic. The redox mechanisms involved in the differential radioprotection observed here suggest that increased radiotherapy efficacy by DMAPT is more broadly applicable to a range of cancer types.
Collapse
Affiliation(s)
- Katherine L Morel
- a Molecular Medicine and Pathology, Flinders Centre for Innovation in Cancer, Flinders University and Medical Centre, Bedford Park, Adelaide, South Australia
| | - Rebecca J Ormsby
- a Molecular Medicine and Pathology, Flinders Centre for Innovation in Cancer, Flinders University and Medical Centre, Bedford Park, Adelaide, South Australia
| | - Eva Bezak
- b Medical Radiation, School of Health Sciences, University of South Australia, Adelaide, South Australia
| | | | - Pamela J Sykes
- a Molecular Medicine and Pathology, Flinders Centre for Innovation in Cancer, Flinders University and Medical Centre, Bedford Park, Adelaide, South Australia
| |
Collapse
|
13
|
Cui J, Yang G, Pan Z, Zhao Y, Liang X, Li W, Cai L. Hormetic Response to Low-Dose Radiation: Focus on the Immune System and Its Clinical Implications. Int J Mol Sci 2017; 18:280. [PMID: 28134809 PMCID: PMC5343816 DOI: 10.3390/ijms18020280] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 01/17/2017] [Indexed: 12/12/2022] Open
Abstract
The interrelationship between ionizing radiation and the immune system is complex, multifactorial, and dependent on radiation dose/quality and immune cell type. High-dose radiation usually results in immune suppression. On the contrary, low-dose radiation (LDR) modulates a variety of immune responses that have exhibited the properties of immune hormesis. Although the underlying molecular mechanism is not fully understood yet, LDR has been used clinically for the treatment of autoimmune diseases and malignant tumors. These advancements in preclinical and clinical studies suggest that LDR-mediated immune modulation is a well-orchestrated phenomenon with clinical potential. We summarize recent developments in the understanding of LDR-mediated immune modulation, with an emphasis on its potential clinical applications.
Collapse
Affiliation(s)
- Jiuwei Cui
- Cancer Center, the First Hospital of Jilin University, Changchun 130021, China.
| | - Guozi Yang
- Cancer Center, the First Hospital of Jilin University, Changchun 130021, China.
- Department of Radiation-Oncology, the First Hospital of Jilin University, Changchun 130021, China.
| | - Zhenyu Pan
- Department of Radiation-Oncology, the First Hospital of Jilin University, Changchun 130021, China.
| | - Yuguang Zhao
- Cancer Center, the First Hospital of Jilin University, Changchun 130021, China.
| | - Xinyue Liang
- Cancer Center, the First Hospital of Jilin University, Changchun 130021, China.
| | - Wei Li
- Cancer Center, the First Hospital of Jilin University, Changchun 130021, China.
| | - Lu Cai
- Cancer Center, the First Hospital of Jilin University, Changchun 130021, China.
- The Pediatric Research Institute, the Departments of Pediatrics, Radiation Oncology, Pharmacology and Toxicology of the University of Louisville, Louisville, KY 40202, USA.
| |
Collapse
|
14
|
Yang G, Li W, Jiang H, Liang X, Zhao Y, Yu D, Zhou L, Wang G, Tian H, Han F, Cai L, Cui J. Low-dose radiation may be a novel approach to enhance the effectiveness of cancer therapeutics. Int J Cancer 2016; 139:2157-2168. [PMID: 27299986 DOI: 10.1002/ijc.30235] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 05/01/2016] [Accepted: 06/06/2016] [Indexed: 12/26/2022]
Abstract
It has been generally accepted that both natural and man-made sources of ionizing radiation contribute to human exposure and consequently pose a possible risk to human health. However, accumulating evidence has shown that the biological effects of low-dose radiation (LDR) are different from those of high-dose radiation. LDR can stimulate proliferation of normal cells and activate their defense systems, while these biological effects are not observed in some cancer cell types. Although there is still no concordance on this matter, the fact that LDR has the potential to enhance the effects of cancer therapeutics and reduce the toxic side effects of anti-cancer therapy has garnered significant interest. Here, we provide an overview of the current knowledge regarding the experimental data detailing the different responses of normal and cancer tissues to LDR, the underlying mechanisms, and its significance in clinical application.
Collapse
Affiliation(s)
- Guozi Yang
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China
- Department of Radiation-Oncology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Wei Li
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Hongyu Jiang
- Health Examination Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Xinyue Liang
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Yuguang Zhao
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Dehai Yu
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Lei Zhou
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Guanjun Wang
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Huimin Tian
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Fujun Han
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Lu Cai
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China.
- Kosair Children's Hospital Research Institute, Departments of Pediatrics, Radiation Oncology, Pharmacology and Toxicology of the University of Louisville, Louisville, KY, 40202.
| | - Jiuwei Cui
- Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
15
|
XIN YONG, ZHANG HAIBIN, TANG TIANYOU, LIU GUIHONG, WANG JIANSHE, JIANG GUAN, ZHANG LONGZHEN. Low-dose radiation-induced apoptosis in human leukemia K562 cells through mitochondrial pathways. Mol Med Rep 2014; 10:1569-75. [DOI: 10.3892/mmr.2014.2381] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Accepted: 10/23/2013] [Indexed: 11/06/2022] Open
|
16
|
Different responses of tumor and normal cells to low-dose radiation. Contemp Oncol (Pozn) 2013; 17:356-62. [PMID: 24592123 PMCID: PMC3934051 DOI: 10.5114/wo.2013.35289] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Revised: 11/12/2012] [Accepted: 02/12/2013] [Indexed: 02/05/2023] Open
Abstract
AIM OF THE STUDY We demonstrated stimulation of both erythrocyte immune function and superoxide dismutase activity in tumor-bearing mice in response to whole-body 75 mGy X-rays. In addition, we enhanced the chemotherapeutic effect by exposing tumor-bearing mice to low-dose radiation (LDR). This study aims to investigate the different responses of tumor cells and normal cells to LDR. MATERIAL AND METHODS Survival fraction, micronucleus frequency, and cell cycle of Lewis cells and primary human fibroblast AG01522 cells were measured. S180 sarcoma cells were implanted in mice, and tumor sizes were measured in vivo. RESULTS In response to LDR exposure in vitro, a stimulating effect was observed in AG01522 cells but not in Lewis cells. Low-dose radiation did not cause an adaptive response in the Lewis cell cycle. Lack of an LDR-induced radioadaptive response in tumor cells was observed in tumor-bearing mouse models. Furthermore, a higher apoptotic effect and lower expression of the anti-apoptosis gene Bcl-2 were found in tumor cells of tumor-bearing mice exposed to D1 + D2 than those in tumor cells of tumor-bearing mice exposed to D2 alone. CONCLUSIONS Different responses of tumor cells and normal cells to LDR were found. Low-dose radiation was found to stimulate the growth of normal cells but not of tumor cells in vitro and in vivo, which is a very important and clinically relevant phenomenon.
Collapse
|
17
|
Abstract
AbstractThe biological effects of low-dose radiation have attracted attention, but data are currently insufficient to fully understand the beneficial role of the phenomenon. In the present study, we have investigated the effects of low doses of gamma-irradiation alone and in combination with all-trans-retinoic acid (RA) on proliferation, apoptosis and differentiation of the human promyelocytic leukemia HL-60 cells. Changes in cell behavior and protein expression were determined with the use of light and fluorescent microscopy, immunocytochemical and Western blot analysis. Low-dose irradiation with 1–100 cGy caused a dose-dependent inhibition of HL-60 cell proliferation, and induced apoptosis and differentiation to granulocytes with an increase in the number of CD15-positive cells. Pre-irradiation with 1–100 cGy for 24 h before treatment with RA promoted apoptosis but did not impair RA-induced differentiation. Both processes were associated with a decrease in the expression of the proliferating cell nuclear antigen (PCNA), BCL-2, c-MYC, and changes in both cytosolic and nuclear levels of protein tyrosine-phosphorylation as well as protein kinase C alpha or beta isoforms. These results demonstrate the beneficial role of low-dose irradiation in modulating leukemia cell proliferation, differentiation and apoptosis.
Collapse
|
18
|
Matsumoto H, Tomita M, Otsuka K, Hatashita M. A new paradigm in radioadaptive response developing from microbeam research. JOURNAL OF RADIATION RESEARCH 2009; 50 Suppl A:A67-A79. [PMID: 19346687 DOI: 10.1269/jrr.09003s] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
A classic paradigm in radiation biology asserts that all radiation effects on cells, tissues and organisms are due to the direct action of radiation on living tissue. Using this model, possible risks from exposure to low dose ionizing radiation (below 100 mSv) are estimated by extrapolating from data obtained after exposure to higher doses of radiation, using a linear non-threshold model (LNT model). However, the validity of using this dose-response model is controversial because evidence accumulated over the past decade has indicated that living organisms, including humans, respond differently to low dose/low dose-rate radiation than they do to high dose/high dose-rate radiation. These important responses to low dose/low dose-rate radiation are the radiation-induced adaptive response, the bystander response, low-dose hypersensitivity, and genomic instability. The mechanisms underlying these responses often involve biochemical and molecular signals generated in response to targeted and non-targeted events. In order to define and understand the bystander response to provide a basis for the understanding of non-targeted events and to elucidate the mechanisms involved, recent sophisticated research has been conducted with X-ray microbeams and charged heavy particle microbeams, and these studies have produced many new observations. Based on these observations, associations have been suggested to exist between the radioadaptive and bystander responses. The present review focuses on these two phenomena, and summarizes observations supporting their existence, and discusses the linkage between them in light of recent results obtained from experiments utilizing microbeams.
Collapse
Affiliation(s)
- Hideki Matsumoto
- Division of Oncology, Biomedical Imaging Research Center, University of Fukui, 23-3 Matsuoka-Shimoaitsuki, Eiheiji-cho, Fukui 910-1193, Japan.
| | | | | | | |
Collapse
|
19
|
Jiang H, Li W, Li X, Cai L, Wang G. Low-dose radiation induces adaptive response in normal cells, but not in tumor cells: in vitro and in vivo studies. JOURNAL OF RADIATION RESEARCH 2008; 49:219-230. [PMID: 18296871 DOI: 10.1269/jrr.07072] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Biological effects of low-dose radiation (LDR) are distinguishable from those of high-dose radiation. Hormetic and adaptive responses are such two examples. However, whether adaptive response could be induced in tumor cells by LDR, especially under in vivo condition, remains elusive, and was systemically investigated in the present study. Four tumor cell lines: two human leukemia cell lines (erythroleukemia cell line K562, and acute promyelocytic leukemia cell line HL60), and two human solid tumor cell lines (lung carcinoma cell line NCI-H446 and glioma cell line U251), along with one normal cell line (human fibroblast cells, MRC-5), were irradiated with LDR at 75 mGy of X-rays as D1 and then 4 Gy of X-rays as D2 (i.e.: D1 + D2) or only 4 Gy of X-rays (D2 alone). Three tumor-bearing animal models were also used to further define whether LDR induces adaptive response in tumor cells in vivo. Adaptive response was observed only in normal cell line, but not in four tumor cell lines, in response to LDR, showing a resistance to subsequent D2-induced cell growth inhibition. Three tumor-bearing mouse models with U251, NCI-H446 or S180 tumor cells were used to confirm that pre-exposure of tumor-bearing mice to D1 did not induce the resistance of tumor cells in vivo to D2-induced tumor growth inhibition. Furthermore, a higher apoptotic effect, along with higher expression of apoptosis-related genes P53 and Bax and lower expression of anti-apoptosis gene Bcl-2, was found in tumor cells of the tumor-bearing mice exposed to D1 + D2 than those in the tumor cells of the tumor-bearing mice exposed to D2 alone. These results suggest that LDR does not induce adaptive response in the tumor cells under both in vitro and in vivo conditions, which is a very important, clinic-relevant phenomenon.
Collapse
Affiliation(s)
- Hongyu Jiang
- Department of Hematology at the First Clinical College, Jilin University, Changchum, China
| | | | | | | | | |
Collapse
|
20
|
Okazaki R, Ootsuyama A, Norimura T. TP53 and TP53-Related Genes Associated with Protection from Apoptosis in the Radioadaptive Response. Radiat Res 2007; 167:51-7. [PMID: 17214514 DOI: 10.1667/rr0623.1] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2006] [Accepted: 09/21/2006] [Indexed: 11/03/2022]
Abstract
We investigated the effect of administering priming low-dose radiation prior to high-dose radiation on the level of apoptosis and on the expression of TP53 and TP53-related genes in mouse splenocytes. The percentage of apoptotic cells was significantly lower in TP53(+/+) mice receiving priming radiation 2 to 168 h before the high-dose irradiation, compared to TP53(+/+) mice exposed to 2 Gy alone. In contrast, TP53(+/-) mice exhibited a reduced level of apoptosis only when priming was performed for 2 or 4 h prior to the high-dose irradiation. In TP53(+/+) mice, primed mice had higher TP53 expression than mice exposed to 2 Gy. Phospho-TP53 (ser15/18) expression was the highest in mice exposed to 2 Gy and intermediate in primed mice. Expression of p21 (CDKN1A) was higher in primed mice compared with mice exposed to 2 Gy. MDM2 expression remained at a high level in all mice receiving 2 Gy. Elevated phospho-ATM expression was observed only in mice exposed to 2 Gy. We conclude that TP53 plays a critical role in the radioadaptive response and that TP53 and TP53-related genes might protect cells from apoptosis through activation of the intracellular repair system.
Collapse
Affiliation(s)
- Ryuji Okazaki
- Department of Radiation Biology and Health, School of Medicine, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Yahatanishi-ku, Kitakyushu 807-8555, Japan.
| | | | | |
Collapse
|
21
|
Liu G, Gong P, Bernstein LR, Bi Y, Gong S, Cai L. Apoptotic cell death induced by low-dose radiation in male germ cells: hormesis and adaptation. Crit Rev Toxicol 2007; 37:587-605. [PMID: 17674213 DOI: 10.1080/10408440701493061] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Biological effects of low-dose radiation (LDR) in somatic cells have captured the interest of radiobiologists for the last two decades. Apoptosis of germ cells is required for normal spermatogenesis and often occurs through highly conserved events, including the transfer of vital cellular materials to the growing gametes following death of neighboring cells. Apoptosis of germ cells also functions in diverse processes, including removal of abnormal or superfluous cells at specific checkpoints, establishment of caste differentiation, and individualization of gametes. Moreover, germ cells are very sensitive to radiation-induced genomic and cytological effects. Therefore, induction of germ-cell apoptosis has been observed in the testis of animals exposed to both high-dose radiation (HDR) and LDR. Exposure of male germ cells to LDR induces a stimulating effect, while exposure to HDR causes an inhibitory effect on the metabolism, antioxidant capacity, and proliferation and maturation of cells, a phenomenon termed hormesis. Preexposure to LDR also protects cells from subsequently HDR-induced genomic and cytological effects, a phenomenon termed adaptive response. This review describes the features of male germ-cell apoptosis. It reviews the evidence that LDR induces the hormesis and adaptive responses in the male germ cells in terms of apoptosis. This review also discusses the possible effects of LDR-induced apoptotic hormesis and adaptive response on the modulation of inheritable genomic damage caused by subsequent radiation exposure to male germ cells.
Collapse
Affiliation(s)
- Guangwei Liu
- Ministry of Health Radiobiology Research Unit, School of Public Health, Jilin University, Changchun, China
| | | | | | | | | | | |
Collapse
|
22
|
Mohammadi S, Taghavi-Dehaghani M, Gharaati MR, Masoomi R, Ghiassi-Nejad M. Adaptive response of blood lymphocytes of inhabitants residing in high background radiation areas of ramsar- micronuclei, apoptosis and comet assays. JOURNAL OF RADIATION RESEARCH 2006; 47:279-85. [PMID: 16988494 DOI: 10.1269/jrr.0575] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
The hot springs in certain areas of Ramsar contain (226)Ra and (222)Rn. The effects of natural radiation on the inhabitants of these areas and the inhabitant's radiosensitivity or adaptive responses were studied. One group of volunteers from areas with high natural background radiation and another group from areas with normal background radiation were chosen as the case and control group respectively. The frequency of micronuclei, apoptosis, and DNA damage in peripheral blood mononuclear cells were measured following gamma irradiation (4 Gy). The incidence of micronuclei in the case group was significantly lower than that in the control group while their frequency of apoptosis was higher (P < 0.05). However, the rates of induced DNA damage and repair were significantly higher in the case group (P < 0.05). Smaller number of micronuclei and higher levels of apoptosis in the case group could be the result of higher resistance to radiation stress and a more rigorous checkpoint at cell division. However, regarding the alkaline labile sites, the individuals in the case group are more sensitive and susceptible to DNA damage. The results of micronuclei, apoptosis and repair studies suggest that an adaptive response might be induced in people residing in areas with high background radiation.
Collapse
Affiliation(s)
- Shahla Mohammadi
- Radiation Molecular Genetic Lab, National Radiation Protection Department (NRPD), Iranian Nuclear Regulatory Authority (INRA), Tehran.
| | | | | | | | | |
Collapse
|
23
|
Liu G, Gong P, Zhao H, Wang Z, Gong S, Cai L. Effect of low-level radiation on the death of male germ cells. Radiat Res 2006; 165:379-389. [PMID: 16579650 DOI: 10.1667/rr3528.1] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Hormetic and adaptive responses induced by low-level radiation in hematopoietic and immune systems have been observed, as shown by stimulatory effects on cell growth and resistance to subsequent radiation-induced cytogenetic damage. However, in terms of cell death by apoptosis, the effects of low-level radiation are controversial: Some studies showed decreased apoptosis in response to low-level radiation while others showed increased apoptosis. This controversy may be related to the radiation doses or dose rates and also, more importantly, to the cell types. Testes are one of the most radiosensitive organs. The loss of male germ cells after exposure to ionizing radiation has been attributed to apoptosis. In the present study, the effects of low-level radiation at doses up to 200 mGy on mouse male germ cells in terms of apoptosis and the expression of apoptosis-related proteins were examined at different times after whole-body exposure of mice to low-level radiation. In addition, the effect of pre-exposure to low-level radiation on subsequent cell death induced by high doses of radiation was examined to explore the possibility of low-level radiation-induced adaptive response. The results showed that low-level radiation in the dose range of 25-200 mGy induced significant increases in apoptosis in both spermatogonia and spermatocytes, with the maximal effect at 75 mGy. The increased apoptosis is most likely associated with Trp53 protein expression. Furthermore, 75 mGy low-level radiation given pre-irradiation led to an adaptive response of seminiferous germ cells to subsequent high-level radiation-induced apoptosis. These results suggest that low-level radiation induces increased apoptosis in male germ cells but also induces a significant adaptive response that decreases cell death after a subsequent high-dose irradiation.
Collapse
Affiliation(s)
- Guangwei Liu
- Ministry of Health Radiobiology Research Unit, School of Public Health, Jilin University, Changchun, China 130021
| | | | | | | | | | | |
Collapse
|
24
|
Matsumoto H, Takahashi A, Ohnishi T. Radiation-induced adaptive responses and bystander effects. ACTA ACUST UNITED AC 2005; 18:247-54. [PMID: 15858392 DOI: 10.2187/bss.18.247] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
A classical paradigm [correction of paradym] of radiation biology asserts that all radiation effects on cells, tissues and organisms are due to the direct action of radiation. However, there has been a recent growth of interest in the indirect actions of radiation including the radiation-induced adaptive response, the bystander effect, low-dose hypersensitivity, and genomic instability, which are specific modes of stress exhibited in response to low-dose/low-dose rate radiation. This review focuses on the radiation-induced bystander effect and the adaptive response, provides a description of the two phenomena, and discusses the contribution of the former to the latter.
Collapse
Affiliation(s)
- Hideki Matsumoto
- Department of Experimental Radiology and Health Physics, Faculty of Medical Science, University of Fukui, Matsuoka, Fukui, Japan.
| | | | | |
Collapse
|