1
|
Ma Y, Kong Y, Zhang S, Peng Y, Xu M, Zhang J, Xu H, Hong Z, Xing P, Qian J, Zhang L. The relationship between splenic dose and radiation-induced lymphopenia. JOURNAL OF RADIATION RESEARCH 2024; 65:337-349. [PMID: 38718391 PMCID: PMC11115471 DOI: 10.1093/jrr/rrae023] [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: 01/18/2024] [Revised: 03/11/2024] [Indexed: 05/25/2024]
Abstract
Lymphocytes, which are highly sensitive to radiation, play a crucial role in the body's defense against tumors. Radiation-induced lymphopenia has been associated with poorer outcomes in different cancer types. Despite being the largest secondary lymphoid organ, the spleen has not been officially designated as an organ at risk. This study hypothesizes a connection between spleen irradiation and lymphopenia and seeks to establish evidence-based dosage limits for the spleen. We retrospectively analyzed data from 96 patients with locally advanced gastric cancer who received postoperative chemoradiotherapy (CRT) between May 2010 and May 2017. Complete blood counts were collected before, during and after CRT. We established a model for predicting the minimum absolute lymphocyte count (Min ALC) and to investigate potential associations between spleen dosimetric variables and Min ALC. The median follow-up was 60 months. The 5-year overall survival (OS) and disease-free survival (DFS) were 65.2% and 56.8%, respectively. The median values of pre-treatment ALC, Min ALC and post-treatment ALC were 1.40 × 109, 0.23 × 109 and 0.28 × 109/L, respectively. Regression analysis confirmed that the primary tumor location, number of fractions and spleen V5 were significant predictors of Min ALC during radiation therapy. Changes in ALC (ΔALC) were identified as an independent predictor of both OS and DFS. Spleen V5 is an independent predictor for Min ALC, and the maximum dose of the spleen is associated with an increased risk of severe lymphopenia. Therefore, these doses should be restricted in clinical practice. Additionally, ΔALC can serve as a prognostic indicator for adjuvant radiotherapy in gastric cancer.
Collapse
Affiliation(s)
- Yifu Ma
- PRaG Therapy Center, Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou 215004, China
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou 215004, China
- Institute of Radiotherapy and Oncology, Soochow University, San Xiang Road No. 1055, Suzhou 215004, China
| | - Yuehong Kong
- PRaG Therapy Center, Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou 215004, China
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou 215004, China
- Institute of Radiotherapy and Oncology, Soochow University, San Xiang Road No. 1055, Suzhou 215004, China
| | - Shuying Zhang
- Department of Ultrasound, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou 215004, China
| | - Yong Peng
- PRaG Therapy Center, Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou 215004, China
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou 215004, China
- Institute of Radiotherapy and Oncology, Soochow University, San Xiang Road No. 1055, Suzhou 215004, China
| | - Meiling Xu
- PRaG Therapy Center, Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou 215004, China
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou 215004, China
- Institute of Radiotherapy and Oncology, Soochow University, San Xiang Road No. 1055, Suzhou 215004, China
| | - Junjun Zhang
- PRaG Therapy Center, Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou 215004, China
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou 215004, China
- Institute of Radiotherapy and Oncology, Soochow University, San Xiang Road No. 1055, Suzhou 215004, China
| | - Hong Xu
- PRaG Therapy Center, Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou 215004, China
- Department of Oncology, Changshu Hospital Affiliated to Soochow University, Shu Yuan Road No. 1, Suzhou 215500, China
| | - Zhihui Hong
- Department of Nuclear medicine, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou 215004, China
| | - Pengfei Xing
- PRaG Therapy Center, Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou 215004, China
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou 215004, China
- Institute of Radiotherapy and Oncology, Soochow University, San Xiang Road No. 1055, Suzhou 215004, China
| | - Jianjun Qian
- PRaG Therapy Center, Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou 215004, China
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou 215004, China
- Institute of Radiotherapy and Oncology, Soochow University, San Xiang Road No. 1055, Suzhou 215004, China
| | - Liyuan Zhang
- PRaG Therapy Center, Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou 215004, China
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou 215004, China
- Institute of Radiotherapy and Oncology, Soochow University, San Xiang Road No. 1055, Suzhou 215004, China
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Ren Ai Road No. 199, Suzhou 215004, China
| |
Collapse
|
2
|
Anikina VA, Sorokina SS, Shemyakov AE, Zamyatina EA, Taskaeva IS, Teplova PO, Popova NR. An Experimental Model of Proton-Beam-Induced Radiation Dermatitis In Vivo. Int J Mol Sci 2023; 24:16373. [PMID: 38003561 PMCID: PMC10671732 DOI: 10.3390/ijms242216373] [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: 10/02/2023] [Revised: 11/13/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Radiation dermatitis (RD) is one of the most common side effects of radiation therapy. However, to date, there is a lack of both specific treatments for RD and validated experimental animal models with the use of various sources of ionizing radiation (IR) applied in clinical practice. The aim of this study was to develop and validate a model of acute RD induced using proton radiation in mice. Acute RD (Grade 2-4) was obtained with doses of 30, 40, and 50 Gy, either with or without depilation. The developed model of RD was characterized by typical histological changes in the skin after irradiation. Moreover, the depilation contributed to a skin histology alteration of the irradiated mice. The assessment of animal vital signs indicated that there was no effect of proton irradiation on the well-being or general condition of the animals. This model can be used to develop effective therapeutic agents and study the pathogenesis of radiation-induced skin toxicity, including that caused by proton irradiation.
Collapse
Affiliation(s)
- Viktoriia A. Anikina
- Institute of Theoretical and Experimental Biophysics of the Russian Academy of Sciences, 3 Institutskaya St., Pushchino 142290, Russia; (V.A.A.); (S.S.S.); (A.E.S.); (E.A.Z.)
| | - Svetlana S. Sorokina
- Institute of Theoretical and Experimental Biophysics of the Russian Academy of Sciences, 3 Institutskaya St., Pushchino 142290, Russia; (V.A.A.); (S.S.S.); (A.E.S.); (E.A.Z.)
| | - Alexander E. Shemyakov
- Institute of Theoretical and Experimental Biophysics of the Russian Academy of Sciences, 3 Institutskaya St., Pushchino 142290, Russia; (V.A.A.); (S.S.S.); (A.E.S.); (E.A.Z.)
- Branch “Physical-Technical Center” of P.N. Lebedev Physical Institute of the Russian Academy of Sciences, 2 Akademichesky Proezd, Protvino 142281, Russia
| | - Elizaveta A. Zamyatina
- Institute of Theoretical and Experimental Biophysics of the Russian Academy of Sciences, 3 Institutskaya St., Pushchino 142290, Russia; (V.A.A.); (S.S.S.); (A.E.S.); (E.A.Z.)
| | - Iuliia S. Taskaeva
- Research Institute of Clinical and Experimental Lymphology—Branch of the Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 2 Timakova St., Novosibirsk 630060, Russia;
| | - Polina O. Teplova
- Institute of Cell Biophysics of the Russian Academy of Sciences, 3 Institutskaya St., Pushchino 142290, Russia;
| | - Nelli R. Popova
- Institute of Theoretical and Experimental Biophysics of the Russian Academy of Sciences, 3 Institutskaya St., Pushchino 142290, Russia; (V.A.A.); (S.S.S.); (A.E.S.); (E.A.Z.)
| |
Collapse
|
3
|
Diaz J, Kuhlman BM, Edenhoffer NP, Evans AC, Martin KA, Guida P, Rusek A, Atala A, Coleman MA, Wilson PF, Almeida-Porada G, Porada CD. Immediate effects of acute Mars mission equivalent doses of SEP and GCR radiation on the murine gastrointestinal system-protective effects of curcumin-loaded nanolipoprotein particles (cNLPs). FRONTIERS IN ASTRONOMY AND SPACE SCIENCES 2023; 10:1117811. [PMID: 38741937 PMCID: PMC11089821 DOI: 10.3389/fspas.2023.1117811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Introduction Missions beyond low Earth orbit (LEO) will expose astronauts to ionizing radiation (IR) in the form of solar energetic particles (SEP) and galactic cosmic rays (GCR) including high atomic number and energy (HZE) nuclei. The gastrointestinal (GI) system is documented to be highly radiosensitive with even relatively low dose IR exposures capable of inducing mucosal lesions and disrupting epithelial barrier function. IR is also an established risk factor for colorectal cancer (CRC) with several studies examining long-term GI effects of SEP/GCR exposure using tumor-prone APC mouse models. Studies of acute short-term effects of modeled space radiation exposures in wildtype mouse models are more limited and necessary to better define charged particle-induced GI pathologies and test novel medical countermeasures (MCMs) to promote astronaut safety. Methods In this study, we performed ground-based studies where male and female C57BL/6J mice were exposed to γ-rays, 50 MeV protons, or 1 GeV/n Fe-56 ions at the NASA Space Radiation Laboratory (NSRL) with histology and immunohistochemistry endpoints measured in the first 24 h post-irradiation to define immediate SEP/GCR-induced GI alterations. Results Our data show that unlike matched γ-ray controls, acute exposures to protons and iron ions disrupts intestinal function and induces mucosal lesions, vascular congestion, epithelial barrier breakdown, and marked enlargement of mucosa-associated lymphoid tissue. We also measured kinetics of DNA double-strand break (DSB) repair using gamma-H2AX- specific antibodies and apoptosis via TUNEL labeling, noting the induction and disappearance of extranuclear cytoplasmic DNA marked by gamma-H2AX only in the charged particle-irradiated samples. We show that 18 h pre-treatment with curcumin-loaded nanolipoprotein particles (cNLPs) delivered via IV injection reduces DSB-associated foci levels and apoptosis and restore crypt villi lengths. Discussion These data improve our understanding of physiological alterations in the GI tract immediately following exposures to modeled space radiations and demonstrates effectiveness of a promising space radiation MCM.
Collapse
Affiliation(s)
- Jonathan Diaz
- Wake Forest Institute for Regenerative Medicine, Winston Salem, NC, United States
| | - Bradford M. Kuhlman
- Wake Forest Institute for Regenerative Medicine, Winston Salem, NC, United States
| | | | - Angela C. Evans
- Department of Radiation Oncology, University of California Davis School of Medicine, Sacramento, CA, United States
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Kelly A. Martin
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Peter Guida
- NASA Space Radiation Laboratory, Brookhaven National Laboratory, Upton, NY, United States
| | - Adam Rusek
- NASA Space Radiation Laboratory, Brookhaven National Laboratory, Upton, NY, United States
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Winston Salem, NC, United States
| | - Matthew A. Coleman
- Department of Radiation Oncology, University of California Davis School of Medicine, Sacramento, CA, United States
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Paul F. Wilson
- Department of Radiation Oncology, University of California Davis School of Medicine, Sacramento, CA, United States
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, United States
| | - Graça Almeida-Porada
- Wake Forest Institute for Regenerative Medicine, Winston Salem, NC, United States
| | | |
Collapse
|
4
|
Sposto R, Sugiyama H, Tsuruyama T, Brenner AV. Effect of radiation exposure on survival after first solid cancer diagnosis in A-bomb survivors. Cancer Epidemiol 2023; 83:102341. [PMID: 36812706 DOI: 10.1016/j.canep.2023.102341] [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: 08/03/2022] [Revised: 11/28/2022] [Accepted: 02/16/2023] [Indexed: 02/22/2023]
Abstract
BACKGROUND Comparison of the estimated effect of atomic bomb radiation exposure on solid cancer incidence and solid cancer mortality in the RERF Life Span Study (LSS) reveals a difference in the magnitude and shape of the excess relative risk dose response. A possible contributing factor to this difference is pre-diagnosis radiation effect on post-diagnosis survival. Pre-diagnosis radiation exposure theoretically could influence post-diagnosis survival by affecting the genetic makeup and possibly aggressiveness of cancer, or by compromising tolerance for aggressive treatment for cancer. METHODS We analyze the radiation effect on post-diagnosis survival in 20,463 LSS subjects diagnosed with first-primary solid cancer between 1958 and 2009 with particular attention to whether death was caused by the first-primary cancer, other cancer, or non-cancer diseases. RESULTS From multivariable Cox regression analysis of cause-specific survival, the excess hazard at 1 Gy (EH1Gy) for death from the first primary cancer was not significantly different from zero - p = 0.23, EH1Gy = 0.038 (95 % CI: -0.023, 0.104). Death from other cancer and death from non-cancer diseases both were significantly associated with radiation dose: other cancer EH1Gy = 0.38 (95 % CI: 0.24, 0.53); non-cancer EH1Gy = 0.24 (95 % CI: 0.13, 0.36), both p < 0.001. CONCLUSION There is no detectable large effect of pre-diagnosis radiation exposure on post-diagnosis death from the first primary cancer in A-bomb survivors. IMPACT A direct effect of pre-diagnosis radiation exposure on cancer prognosis is ruled out as an explanation for the difference in incidence and mortality dose response in A-bomb survivors.
Collapse
Affiliation(s)
- Richard Sposto
- Department of Statistics, Radiation Effects Research Foundation, Japan.
| | - Hiromi Sugiyama
- Department of Epidemiology, Radiation Effects Research Foundation, Japan
| | - Tatsuaki Tsuruyama
- Department of Molecular Biosciences, Radiation Effect Research Foundation, Japan
| | - Alina V Brenner
- Department of Epidemiology, Radiation Effects Research Foundation, Japan
| |
Collapse
|
5
|
Katsube T, Wang B, Tanaka K, Ninomiya Y, Hirakawa H, Liu C, Maruyama K, Vares G, Liu Q, Murakami M, Nakajima T, Fujimori A, Nenoi M. Fluorescence in situ hybridization analysis of chromosomal aberrations in mouse splenocytes at one- and two-months after total body exposure to iron-56 (Fe) ion particles or X-rays. MUTATION RESEARCH. GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2022; 882:503548. [PMID: 36155141 DOI: 10.1016/j.mrgentox.2022.503548] [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: 11/25/2021] [Revised: 08/25/2022] [Accepted: 09/08/2022] [Indexed: 06/16/2023]
Abstract
High atomic number and energy (HZE) particles such as iron-56 (Fe) ions are a major contributor to health risks in long-term manned space exploration. The aim of this study is to understand radiation-induced differential genotoxic effects between HZE particles and low linear energy transfer (LET) photons. C57BL/6J Jms female mice of 8 weeks old were exposed to total body irradiation of accelerated Fe-particles with a dose ranging from 0.1 to 3.0 Gy or of X-rays with a dose ranging from 0.1 to 5.0 Gy. Chromosomal aberrations (CAs) in splenocytes were examined by fluorescence in situ hybridization at 1- and 2-months after exposure. Clonal expansions of cells with CAs were found to be induced only by X-rays but not by Fe-particles. Dose-dependent increase in the frequencies of stable-type CAs was observed at 1- as well as 2-months after exposure to both radiation types. The frequencies of stable-type CAs in average were much higher in mice exposed to X-rays than those to Fe-particles and did not change significantly between 1- and 2-months after exposure to both radiation types. On the other hand, the frequencies of unstable-type CAs induced by X-rays and Fe-particles were not much different, and they appeared to decrease with time from 1- to 2-months after exposure. These results suggested that larger fraction of stable-type CAs induced by Fe-particles might be non-transmissible than those by X-rays because of some associating lethal alterations on themselves or on other chromosomes in the same cells and that these cells might be removed by 1-month after Fe-TBI. We also demonstrated that exposure to Fe-particles induced insertions at relatively higher frequency to other stable-type CAs than X-rays. Our findings suggest that insertions can be used as indicators of past exposure to high-LET particle radiation.
Collapse
Affiliation(s)
- Takanori Katsube
- National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan.
| | - Bing Wang
- National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Kaoru Tanaka
- National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Yasuharu Ninomiya
- National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Hirokazu Hirakawa
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Cuihua Liu
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Kouichi Maruyama
- National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Guillaume Vares
- Experimental Radiotoxicology and Radiobiology Laboratory, Institute for Radioprotection and Nuclear Safety, B.P. 17 - 92262 Fontenay-aux-Roses Cedex, France
| | - Qiang Liu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Masahiro Murakami
- National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Tetsuo Nakajima
- National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Akira Fujimori
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Mitsuru Nenoi
- Human Resources Development Center, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan.
| |
Collapse
|
6
|
Katsube T, Wang B, Tanaka K, Ninomiya Y, Hirakawa H, Liu C, Maruyama K, Vares G, Liu Q, Kito S, Nakajima T, Fujimori A, Nenoi M. Synergistic Effects of Chronic Restraint-Induced Stress and Low-Dose 56Fe-particle Irradiation on Induction of Chromosomal Aberrations in Trp53-Heterozygous Mice. Radiat Res 2021; 196:100-112. [PMID: 33901294 DOI: 10.1667/rade-20-00218.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 03/26/2021] [Indexed: 11/03/2022]
Abstract
Astronauts can develop psychological stress (PS) during space flights due to the enclosed environment, microgravity, altered light-dark cycles, and risks of equipment failure or fatal mishaps. At the same time, they are exposed to cosmic rays including high atomic number and energy (HZE) particles such as iron-56 (Fe) ions. Psychological stress or radiation exposure can cause detrimental effects in humans. An earlier published pioneering study showed that chronic restraint-induced psychological stress (CRIPS) could attenuate Trp53 functions and increase carcinogenesis induced by low-linear energy transfer (LET) γ rays in Trp53-heterozygous (Trp53+/-) mice. To elucidate possible modification effects from CRIPS on high-LET HZE particle-induced health consequences, Trp53+/- mice were received both CRIPS and accelerated Fe ion irradiation. Six-week-old Trp53+/- C57BL/6N male mice were restrained 6 h per day for 28 consecutive days. On day 8, they received total-body Fe-particle irradiation (Fe-TBI, 0.1 or 2 Gy). Metaphase chromosome spreads prepared from splenocytes at the end of the 28-day restraint regimen were painted with the fluorescence in situ hybridization (FISH) probes for chromosomes 1 (green), 2 (red) and 3 (yellow). Induction of psychological stress in our experimental model was confirmed by increase in urinary corticosterone level on day 7 of restraint regimen. Regardless of Fe-TBI, CRIPS reduced splenocyte number per spleen at the end of the 28-day restraint regimen. At 2 Gy, Fe-TBI alone induced many aberrant chromosomes and no modifying effect was detected from CRIPS on induction of aberrant chromosomes. Notably, neither Fe-TBI at 0.1 Gy nor CRIPS alone induced any increase in the frequency of aberrant chromosomes, while simultaneous exposure resulted in a significant increase in the frequency of chromosomal exchanges. These findings clearly showed that CRIPS could enhance the frequency of chromosomal exchanges induced by Fe-TBI at a low dose of 0.1 Gy.
Collapse
Affiliation(s)
- Takanori Katsube
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba
| | - Bing Wang
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba
| | - Kaoru Tanaka
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba
| | - Yasuharu Ninomiya
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba
| | - Hirokazu Hirakawa
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba
| | - Cuihua Liu
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba
| | - Kouichi Maruyama
- Center for Advanced Radiation Emergency Medicine, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Guillaume Vares
- Okinawa Institute of Science and Technology Graduate University, Okinawa 904-0495, Japan
| | - Qiang Liu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Seiji Kito
- Center for Animal Research and Education, Nagoya University, Nagoya 464-8601, Japan
| | - Tetsuo Nakajima
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba
| | - Akira Fujimori
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba
| | - Mitsuru Nenoi
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba
| |
Collapse
|
7
|
Zwart SR, Mulavara AP, Williams TJ, George K, Smith SM. The role of nutrition in space exploration: Implications for sensorimotor, cognition, behavior and the cerebral changes due to the exposure to radiation, altered gravity, and isolation/confinement hazards of spaceflight. Neurosci Biobehav Rev 2021; 127:307-331. [PMID: 33915203 DOI: 10.1016/j.neubiorev.2021.04.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 02/16/2021] [Accepted: 04/23/2021] [Indexed: 12/19/2022]
Abstract
Multi-year crewed space exploration missions are now on the horizon; therefore, it is important that we understand and mitigate the physiological effects of spaceflight. The spaceflight hazards-radiation, isolation, confinement, and altered gravity-have the potential to contribute to neuroinflammation and produce long-term cognitive and behavioral effects-while the fifth hazard, distance from earth, limits capabilities to mitigate these risks. Accumulated evidence suggests that nutrition has an important role in optimizing cognition and reducing the risk of neurodegenerative diseases caused by neuroinflammation. Here we review the nutritional perspective of how these spaceflight hazards affect the astronaut's brain, behavior, performance, and sensorimotor function. We also assess potential nutrient/nutritional countermeasures that could prevent or mitigate spaceflight risks and ensure that crewmembers remain healthy and perform well during their missions. Just as history has taught us the importance of nutrition in terrestrial exploration, we must understand the role of nutrition in the development and mitigation of spaceflight risks before humans can successfully explore beyond low-Earth orbit.
Collapse
Affiliation(s)
- Sara R Zwart
- Univerity of Texas Medical Branch, 301 University Blvd, Galveston, TX, 77555, USA.
| | | | - Thomas J Williams
- NASA Johnson Space Center, Mail Code SK3, 2101 NASA Parkway, Houston, TX, 77058, USA
| | - Kerry George
- KBR, 2400 E NASA Parkway, Houston, TX, 77058, USA
| | - Scott M Smith
- NASA Johnson Space Center, Mail Code SK3, 2101 NASA Parkway, Houston, TX, 77058, USA
| |
Collapse
|
8
|
Laiakis EC, Shuryak I, Deziel A, Wang YW, Barnette BL, Yu Y, Ullrich RL, Fornace AJ, Emmett MR. Effects of Low Dose Space Radiation Exposures on the Splenic Metabolome. Int J Mol Sci 2021; 22:3070. [PMID: 33802822 PMCID: PMC8002539 DOI: 10.3390/ijms22063070] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/14/2021] [Accepted: 03/15/2021] [Indexed: 12/12/2022] Open
Abstract
Future space missions will include a return to the Moon and long duration deep space roundtrip missions to Mars. Leaving the protection that Low Earth Orbit provides will unavoidably expose astronauts to higher cumulative doses of space radiation, in addition to other stressors, e.g., microgravity. Immune regulation is known to be impacted by both radiation and spaceflight and it remains to be seen whether prolonged effects that will be encountered in deep space can have an adverse impact on health. In this study, we investigated the effects in the overall metabolism of three different low dose radiation exposures (γ-rays, 16O, and 56Fe) in spleens from male C57BL/6 mice at 1, 2, and 4 months after exposure. Forty metabolites were identified with significant enrichment in purine metabolism, tricarboxylic acid cycle, fatty acids, acylcarnitines, and amino acids. Early perturbations were more prominent in the γ irradiated samples, while later responses shifted towards more prominent responses in groups with high energy particle irradiations. Regression analysis showed a positive correlation of the abundance of identified fatty acids with time and a negative association with γ-rays, while the degradation pathway of purines was positively associated with time. Taken together, there is a strong suggestion of mitochondrial implication and the possibility of long-term effects on DNA repair and nucleotide pools following radiation exposure.
Collapse
Affiliation(s)
- Evagelia C. Laiakis
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington, DC 20057, USA; (A.D.); (Y.-W.W.); (A.J.F.J.)
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC 20057, USA
| | - Igor Shuryak
- Center for Radiological Research, Columbia University, New York, NY 10032, USA;
| | - Annabella Deziel
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington, DC 20057, USA; (A.D.); (Y.-W.W.); (A.J.F.J.)
| | - Yi-Wen Wang
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington, DC 20057, USA; (A.D.); (Y.-W.W.); (A.J.F.J.)
| | - Brooke L. Barnette
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA; (B.L.B.); (Y.Y.); (M.R.E.)
| | - Yongjia Yu
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA; (B.L.B.); (Y.Y.); (M.R.E.)
| | | | - Albert J. Fornace
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington, DC 20057, USA; (A.D.); (Y.-W.W.); (A.J.F.J.)
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC 20057, USA
| | - Mark R. Emmett
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA; (B.L.B.); (Y.Y.); (M.R.E.)
- Department of Radiation Oncology, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
9
|
A Meta-Analysis of the Effects of High-LET Ionizing Radiations in Human Gene Expression. Life (Basel) 2021; 11:life11020115. [PMID: 33546472 PMCID: PMC7913660 DOI: 10.3390/life11020115] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/31/2021] [Accepted: 01/31/2021] [Indexed: 12/19/2022] Open
Abstract
The use of high linear energy transfer (LET) ionizing radiation (IR) is progressively being incorporated in radiation therapy due to its precise dose localization and high relative biological effectiveness. At the same time, these benefits of particle radiation become a high risk for astronauts in the case of inevitable cosmic radiation exposure. Nonetheless, DNA Damage Response (DDR) activated via complex DNA damage in healthy tissue, occurring from such types of radiation, may be instrumental in the induction of various chronic and late effects. An approach to elucidating the possible underlying mechanisms is studying alterations in gene expression. To this end, we identified differentially expressed genes (DEGs) in high Z and high energy (HZE) particle-, γ-ray- and X-ray-exposed healthy human tissues, utilizing microarray data available in public repositories. Differential gene expression analysis (DGEA) was conducted using the R programming language. Consequently, four separate meta-analyses were conducted, after DEG lists were grouped depending on radiation type, radiation dose and time of collection post-irradiation. To highlight the biological background of each meta-analysis group, functional enrichment analysis and biological network construction were conducted. For HZE particle exposure at 8–24 h post-irradiation, the most interesting finding is the variety of DNA repair mechanisms that were downregulated, a fact that is probably correlated with complex DNA damage formation. Simultaneously, after X-ray exposure during the same hours after irradiation, DNA repair mechanisms continue to take place. Finally, in a further comparison of low- and high-LET radiation effects, the most prominent result is that autophagy mechanisms seem to persist and that adaptive immune induction seems to be present. Such bioinformatics approaches may aid in obtaining an overview of the cellular response to high-LET particles. Understanding these response mechanisms can consequently aid in the development of countermeasures for future space missions and ameliorate heavy ion treatments.
Collapse
|
10
|
Paul AM, Cheng-Campbell M, Blaber EA, Anand S, Bhattacharya S, Zwart SR, Crucian BE, Smith SM, Meller R, Grabham P, Beheshti A. Beyond Low-Earth Orbit: Characterizing Immune and microRNA Differentials following Simulated Deep Spaceflight Conditions in Mice. iScience 2020; 23:101747. [PMID: 33376970 PMCID: PMC7756144 DOI: 10.1016/j.isci.2020.101747] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 10/16/2020] [Accepted: 10/26/2020] [Indexed: 12/21/2022] Open
Abstract
Spaceflight missions can cause immune system dysfunction in astronauts with little understanding of immune outcomes in deep space. This study assessed immune responses in mice following ground-based, simulated deep spaceflight conditions, compared with data from astronauts on International Space Station missions. For ground studies, we simulated microgravity using the hindlimb unloaded mouse model alone or in combination with acute simulated galactic cosmic rays or solar particle events irradiation. Immune profiling results revealed unique immune diversity following each experimental condition, suggesting each stressor results in distinct circulating immune responses, with clear consequences for deep spaceflight. Circulating plasma microRNA sequence analysis revealed involvement in immune system dysregulation. Furthermore, a large astronaut cohort showed elevated inflammation during low-Earth orbit missions, thereby supporting our simulated ground experiments in mice. Herein, circulating immune biomarkers are defined by distinct deep space irradiation types coupled to simulated microgravity and could be targets for future space health initiatives.
Collapse
Affiliation(s)
- Amber M. Paul
- Universities Space Research Association, Columbia, MD 21046, USA
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94043, USA
| | - Margareth Cheng-Campbell
- Department of Biomedical Engineering, Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Elizabeth A. Blaber
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94043, USA
- Department of Biomedical Engineering, Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Sulekha Anand
- Department of Biological Sciences, San Jose State University, San Jose, CA 95112, USA
| | | | - Sara R. Zwart
- Department of Preventive Medicine and Community Health, University of Texas Medical Branch, Galveston, TX 77555, USA
| | | | | | - Robert Meller
- Department of Neurobiology/Pharmacology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Peter Grabham
- Center for Radiological Research, Columbia University, New York, NY 10027, USA
| | - Afshin Beheshti
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94043, USA
| |
Collapse
|
11
|
Kuznetsova EA, Sirota NP, Mitroshina IY, Pikalov VA, Smirnova EN, Rozanova OM, Glukhov SI, Sirota TV, Zaichkina SI. DNA damage in blood leukocytes from mice irradiated with accelerated carbon ions with an energy of 450 MeV/nucleon. Int J Radiat Biol 2020; 96:1245-1253. [DOI: 10.1080/09553002.2020.1807640] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Elena A. Kuznetsova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - Nikolay P. Sirota
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - Irina Yu. Mitroshina
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - Vladimir A. Pikalov
- Institute of High Energy Physics of the National Research Center ‘Kurchatov Institute’, Protvino, Russia
| | - Elena N. Smirnova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - Olga M. Rozanova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - Sergei I. Glukhov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - Tatyana V. Sirota
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - Svetlana I. Zaichkina
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia
| |
Collapse
|
12
|
Akeem S, Lukman O, Eltahir K, Fatai O, Abiola B, Khadijat O. Bone Marrow and Peripheral Blood Cells Toxicity of a Single 2.0 Gy Cobalt 60 Ionizing Radiation: An Animal Model. Ethiop J Health Sci 2019; 29:195-202. [PMID: 31011267 PMCID: PMC6460448 DOI: 10.4314/ejhs.v29i2.6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Background Bone marrow is extremely vulnerable to damage caused by radiation therapy. Hence, bone marrow suppression is an important side effect of radiotherapy. Effective use of radiotherapy is therefore compromised by radiation-related injuries. Material and Methods Six Guinea-pigs were recruited for the study of which three were subjected to total body irradiation with Co60 while the other three served as controls. Bone marrow and peripheral blood samples were collected before and at days 9, 14 and 21, post irradiation. Manual and automated counts were performed for bone marrow nucleated cells and peripheral blood cells respectively. Results Declining bone marrow cellularity was evident immediately post irradiation. Mean ± SD of marrow cell counted per mm3 were 121,924±281, 87,603±772, 121,367±375 and 122,750±1000 pre-irradiation and days 9, 14 and 21, postirradiation (p-values 0.10, 0.27 and 0.29 respectively). Significant drops in counts were noticed on day 9 post-irradiation for all red cell parameters (p-values <0.05), for Total White Blood Cell Count and Neutrophil count (p-values <0.05) and also on days 14 and 21 for Lymphocytes (p-values <0.05) and on day 21 for Eosinophil/Basophil/Monocytes (p-value <0.05). A significant drop in platelets counts was also noticed on day 9 (p-value <0.05) which significantly increased above pre-irradiation value on day 21. Conclusion Total body irrradiation with Co60 significantly affects the bone marrow with maximum reductions in marrow nucleated cells and peripheral blood cells counts on day 9 post irradiation.
Collapse
Affiliation(s)
- Shittu Akeem
- Department of Haematology, University of Ilorin, PMB 1515, Ilorin, Nigeria
| | - Olatunbosun Lukman
- Department of Haematology and Blood Transfusion, University of Ilorin teaching Hospital, PMB 1459, Ilorin, Nigeri
| | - Khalil Eltahir
- Department of Clinical Pathology & Immunology, Institute of Endemic Diseases, University of Khartoum, Sudan
| | - Olalere Fatai
- Department of Haematology and Blood Transfusion, University of Ilorin teaching Hospital, PMB 1459, Ilorin, Nigeri
| | - Babatunde Abiola
- Department of Haematology, University of Ilorin, PMB 1515, Ilorin, Nigeria
| | - Omokanye Khadijat
- Department of Haematology and Blood Transfusion, University of Ilorin teaching Hospital, PMB 1459, Ilorin, Nigeri
| |
Collapse
|
13
|
Deng W, Xu C, Liu A, van Rossum PS, Deng W, Liao Z, Koong AC, Mohan R, Lin SH. The relationship of lymphocyte recovery and prognosis of esophageal cancer patients with severe radiation-induced lymphopenia after chemoradiation therapy. Radiother Oncol 2019; 133:9-15. [DOI: 10.1016/j.radonc.2018.12.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 11/29/2018] [Accepted: 12/02/2018] [Indexed: 12/24/2022]
|
14
|
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
|
15
|
Rudobeck E, Bellone JA, Szücs A, Bonnick K, Mehrotra-Carter S, Badaut J, Nelson GA, Hartman RE, Vlkolinský R. Low-dose proton radiation effects in a transgenic mouse model of Alzheimer's disease - Implications for space travel. PLoS One 2017; 12:e0186168. [PMID: 29186131 PMCID: PMC5706673 DOI: 10.1371/journal.pone.0186168] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 09/26/2017] [Indexed: 12/13/2022] Open
Abstract
Space radiation represents a significant health risk for astronauts. Ground-based animal studies indicate that space radiation affects neuronal functions such as excitability, synaptic transmission, and plasticity, and it may accelerate the onset of Alzheimer's disease (AD). Although protons represent the main constituent in the space radiation spectrum, their effects on AD-related pathology have not been tested. We irradiated 3 month-old APP/PSEN1 transgenic (TG) and wild type (WT) mice with protons (150 MeV; 0.1-1.0 Gy; whole body) and evaluated functional and biochemical hallmarks of AD. We performed behavioral tests in the water maze (WM) before irradiation and in the WM and Barnes maze at 3 and 6 months post-irradiation to evaluate spatial learning and memory. We also performed electrophysiological recordings in vitro in hippocampal slices prepared 6 and 9 months post-irradiation to evaluate excitatory synaptic transmission and plasticity. Next, we evaluated amyloid β (Aβ) deposition in the contralateral hippocampus and adjacent cortex using immunohistochemistry. In cortical homogenates, we analyzed the levels of the presynaptic marker synaptophysin by Western blotting and measured pro-inflammatory cytokine levels (TNFα, IL-1β, IL-6, CXCL10 and CCL2) by bead-based multiplex assay. TG mice performed significantly worse than WT mice in the WM. Irradiation of TG mice did not affect their behavioral performance, but reduced the amplitudes of population spikes and inhibited paired-pulse facilitation in CA1 neurons. These electrophysiological alterations in the TG mice were qualitatively different from those observed in WT mice, in which irradiation increased excitability and synaptic efficacy. Irradiation increased Aβ deposition in the cortex of TG mice without affecting cytokine levels and increased synaptophysin expression in WT mice (but not in the TG mice). Although irradiation with protons increased Aβ deposition, the complex functional and biochemical results indicate that irradiation effects are not synergistic to AD pathology.
Collapse
Affiliation(s)
- Emil Rudobeck
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States of America
| | - John A. Bellone
- Department of Psychology, School of Behavioral Health, Loma Linda University, Loma Linda, CA, United States of America
| | - Attila Szücs
- BioCircuits Institute, University of California San Diego, La Jolla, CA, United States of America
| | - Kristine Bonnick
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States of America
| | - Shalini Mehrotra-Carter
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States of America
| | - Jerome Badaut
- Department of Physiology, School of Medicine, Loma Linda University, Loma Linda, CA, United States of America
| | - Gregory A. Nelson
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States of America
| | - Richard E. Hartman
- Department of Psychology, School of Behavioral Health, Loma Linda University, Loma Linda, CA, United States of America
| | - Roman Vlkolinský
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, United States of America
| |
Collapse
|
16
|
Chang J, Feng W, Wang Y, Allen AR, Turner J, Stewart B, Raber J, Hauer-Jensen M, Zhou D, Shao L. 28Si total body irradiation injures bone marrow hematopoietic stem cells via induction of cellular apoptosis. LIFE SCIENCES IN SPACE RESEARCH 2017; 13:39-44. [PMID: 28554508 PMCID: PMC6711775 DOI: 10.1016/j.lssr.2017.03.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Accepted: 03/27/2017] [Indexed: 06/07/2023]
Abstract
Long-term space mission exposes astronauts to a radiation environment with potential health hazards. High-energy charged particles (HZE), including 28Si nuclei in space, have deleterious effects on cells due to their characteristics with high linear energy transfer and dense ionization. The influence of 28Si ions contributes more than 10% to the radiation dose equivalent in the space environment. Understanding the biological effects of 28Si irradiation is important to assess the potential health hazards of long-term space missions. The hematopoietic system is highly sensitive to radiation injury and bone marrow (BM) suppression is the primary life-threatening injuries after exposure to a moderate dose of radiation. Therefore, in the present study we investigated the acute effects of low doses of 28Si irradiation on the hematopoietic system in a mouse model. Specifically, 6-month-old C57BL/6J mice were exposed to 0.3, 0.6 and 0.9Gy 28Si (600MeV) total body irradiation (TBI). The effects of 28Si TBI on BM hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) were examined four weeks after the exposure. The results showed that exposure to 28Si TBI dramatically reduced the frequencies and numbers of HSCs in irradiated mice, compared to non-irradiated controls, in a radiation dose-dependent manner. In contrast, no significant changes were observed in BM HPCs regardless of radiation doses. Furthermore, irradiated HSCs exhibited a significant impairment in clonogenic ability. These acute effects of 28Si irradiation on HSCs may be attributable to radiation-induced apoptosis of HSCs, because HSCs, but not HPCs, from irradiated mice exhibited a significant increase in apoptosis in a radiation dose-dependent manner. However, exposure to low doses of 28Si did not result in an increased production of reactive oxygen species and DNA damage in HSCs and HPCs. These findings indicate that exposure to 28Si irradiation leads to acute HSC damage.
Collapse
Affiliation(s)
- Jianhui Chang
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Wei Feng
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Yingying Wang
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Antiño R Allen
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Jennifer Turner
- Departments of Behavioral Neuroscience, ONPRC, Oregon Health and Science University, Portland, OR, USA
| | - Blair Stewart
- Departments of Behavioral Neuroscience, ONPRC, Oregon Health and Science University, Portland, OR, USA
| | - Jacob Raber
- Departments of Behavioral Neuroscience, ONPRC, Oregon Health and Science University, Portland, OR, USA; Departments of Neurology, and Radiation Medicine, ONPRC, Oregon Health and Science University, Portland, OR, USA; Division of Neuroscience, ONPRC, Oregon Health and Science University, Portland, OR, USA
| | - Martin Hauer-Jensen
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Daohong Zhou
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Lijian Shao
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
17
|
Fernandez-Gonzalo R, Baatout S, Moreels M. Impact of Particle Irradiation on the Immune System: From the Clinic to Mars. Front Immunol 2017; 8:177. [PMID: 28275377 PMCID: PMC5319970 DOI: 10.3389/fimmu.2017.00177] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 02/07/2017] [Indexed: 11/29/2022] Open
Abstract
Despite the generalized use of photon-based radiation (i.e., gamma rays and X-rays) to treat different cancer types, particle radiotherapy (i.e., protons and carbon ions) is becoming a popular, and more effective tool to treat specific tumors due to the improved physical properties and biological effectiveness. Current scientific evidence indicates that conventional radiation therapy affects the tumor immunological profile in a particular manner, which in turn, might induce beneficial effects both at local and systemic (i.e., abscopal effects) levels. The interaction between radiotherapy and the immune system is being explored to combine immune and radiation (including particles) treatments, which in many cases have a greater clinical effect than any of the therapies alone. Contrary to localized, clinical irradiation, astronauts are exposed to whole body, chronic cosmic radiation, where protons and heavy ions are an important component. The effects of this extreme environment during long periods of time, e.g., a potential mission to Mars, will have an impact on the immune system that could jeopardize the health of the astronauts, hence the success of the mission. To this background, the purpose of this mini review is to briefly present the current knowledge in local and systemic immune alterations triggered by particle irradiation and to propose new lines of future research. Immune effects induced by particle radiation relevant to clinical applications will be covered, together with examples of combined radiotherapy and immunotherapy. Then, the focus will move to outer space, where the immune system alterations induced by cosmic radiation during spaceflight will be discussed.
Collapse
Affiliation(s)
- Rodrigo Fernandez-Gonzalo
- Radiobiology Unit, Laboratory of Molecular and Cellular Biology, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre, SCK-CEN , Mol , Belgium
| | - Sarah Baatout
- Radiobiology Unit, Laboratory of Molecular and Cellular Biology, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre, SCK-CEN , Mol , Belgium
| | - Marjan Moreels
- Radiobiology Unit, Laboratory of Molecular and Cellular Biology, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre, SCK-CEN , Mol , Belgium
| |
Collapse
|
18
|
Gridley DS, Pecaut MJ. Changes in the distribution and function of leukocytes after whole-body iron ion irradiation. JOURNAL OF RADIATION RESEARCH 2016; 57:477-491. [PMID: 27380804 PMCID: PMC5045078 DOI: 10.1093/jrr/rrw051] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 01/11/2016] [Accepted: 04/03/2016] [Indexed: 06/06/2023]
Abstract
High-energy particle radiation could have a considerable impact on health during space missions. This study evaluated C57BL/6 mice on Day 40 after total-body 56Fe26+ irradiation at 0, 1, 2 and 3 gray (Gy). Radiation consistently increased thymus mass (one-way ANOVA: P < 0.005); spleen, liver and lung masses were similar among all groups. In the blood, there was no radiation effect on the white blood cell (WBC) count or major leukocyte types. However, the red blood cell count, hemoglobin, hematocrit and the CD8+ T cytotoxic (Tc) cell count and percentage all decreased, while both the CD4:CD8 (Th:Tc) cell ratio and spontaneous blastogenesis increased, in one or more irradiated groups compared with unirradiated controls (P < 0.05 vs 0 Gy). In contrast, splenic WBC, lymphocyte, B cell and T helper (Th) counts, %B cells and the CD4:CD8 ratio were all significantly elevated, while Tc percentages decreased, in one or more of the irradiated groups compared with controls (P < 0.05 vs 0 Gy). Although there were trends for minor, radiation-induced increases in %CD11b+ granulocytes in the spleen, cells double-labeled with adhesion markers (CD11b+CD54+, CD11b+CD62E+) were normal. Splenocyte spontaneous blastogenesis and that induced by mitogens (PHA, ConA, LPS) was equivalent to normal. In bone marrow, the percentage of cells expressing stem cell markers, Sca-1 and CD34/Sca-1, were low in one or more of the irradiated groups (P < 0.05 vs 0 Gy). Collectively, the data indicate that significant immunological abnormalities still exist more than a month after 56Fe irradiation and that there are differences dependent upon body compartment.
Collapse
Affiliation(s)
- Daila S Gridley
- Department of Basic Sciences, Division of Radiation Research, Loma Linda University School of Medicine, Chan Shun Pavilion, 11175 Campus Street, Loma Linda, CA 92354, USA
| | - Michael J Pecaut
- Department of Basic Sciences, Division of Radiation Research, Loma Linda University School of Medicine, Chan Shun Pavilion, 11175 Campus Street, Loma Linda, CA 92354, USA
| |
Collapse
|
19
|
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
|
20
|
Dionet C, Müller-Barthélémy M, Marceau G, Denis JM, Averbeck D, Gueulette J, Sapin V, Pereira B, Tchirkov A, Chautard E, Verrelle P. Different dose rate-dependent responses of human melanoma cells and fibroblasts to low dose fast neutrons. Int J Radiat Biol 2016; 92:527-35. [PMID: 27258624 DOI: 10.1080/09553002.2016.1186300] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
PURPOSE To analyze the dose rate influence in hyper-radiosensitivity (HRS) of human melanoma cells to very low doses of fast neutrons and to compare to the behaviour of normal human skin fibroblasts. MATERIALS AND METHODS We explored different neutron dose rates as well as possible implication of DNA double-strand breaks (DSB), apoptosis, and energy-provider adenosine-triphosphate (ATP) levels during HRS. RESULTS HRS in melanoma cells appears only at a very low dose rate (VLDR), while a high dose rate (HDR) induces an initial cell-radioresistance (ICRR). HRS does not seem to be due either to DSB or to apoptosis. Both phenomena (HRS and ICRR) appear to be related to ATP availability for triggering cell repair. Fibroblast survival after neutron irradiation is also dose rate-dependent but without HRS. CONCLUSIONS Melanoma cells or fibroblasts exert their own survival behaviour at very low doses of neutrons, suggesting that in some cases there is a differential between cancer and normal cells radiation responses. Only the survival of fibroblasts at HDR fits the linear no-threshold model. This new insight into human cell responses to very low doses of neutrons, concerns natural radiations, surroundings of accelerators, proton-therapy devices, flights at high altitude. Furthermore, ATP inhibitors could increase HRS during high-linear energy transfer (high-LET) irradiation.
Collapse
Affiliation(s)
- Claude Dionet
- a Centre Jean Perrin , Laboratoire de Radio-Oncologie Expérimentale , Clermont-Ferrand , France
| | - Melanie Müller-Barthélémy
- a Centre Jean Perrin , Laboratoire de Radio-Oncologie Expérimentale , Clermont-Ferrand , France ;,b Clermont Université, Université d'Auvergne, EA7283 CREaT , Clermont-Ferrand , France
| | - Geoffroy Marceau
- c Biochimie et Biologie Moléculaire , CHU Clermont-Ferrand, Centre de Biologie , Clermont-Ferrand , France
| | - Jean-Marc Denis
- d Radiotherapy Department , Cliniques Universitaires Saint Luc-UCL , Bruxelles , Belgique ;,f Université Catholique de Louvain (UCL-IREC-MIRO) , Bruxelles , Belgique
| | - Dietrich Averbeck
- e Institut Curie-Recherche, UMR3348 CNRS/IC, Centre Universitaire, Orsay , France
| | - John Gueulette
- f Université Catholique de Louvain (UCL-IREC-MIRO) , Bruxelles , Belgique
| | - Vincent Sapin
- c Biochimie et Biologie Moléculaire , CHU Clermont-Ferrand, Centre de Biologie , Clermont-Ferrand , France
| | - Bruno Pereira
- g CHU Clermont-Ferrand, Biostatistics unit (DRCI) , Clermont-Ferrand , France
| | - Andrei Tchirkov
- h Cytogénétique médicale, CHU Estaing , Clermont-Ferrand , France
| | - Emmanuel Chautard
- a Centre Jean Perrin , Laboratoire de Radio-Oncologie Expérimentale , Clermont-Ferrand , France ;,b Clermont Université, Université d'Auvergne, EA7283 CREaT , Clermont-Ferrand , France
| | - Pierre Verrelle
- a Centre Jean Perrin , Laboratoire de Radio-Oncologie Expérimentale , Clermont-Ferrand , France ;,b Clermont Université, Université d'Auvergne, EA7283 CREaT , Clermont-Ferrand , France
| |
Collapse
|
21
|
Sridharan DM, Asaithamby A, Blattnig SR, Costes SV, Doetsch PW, Dynan WS, Hahnfeldt P, Hlatky L, Kidane Y, Kronenberg A, Naidu MD, Peterson LE, Plante I, Ponomarev AL, Saha J, Snijders AM, Srinivasan K, Tang J, Werner E, Pluth JM. Evaluating biomarkers to model cancer risk post cosmic ray exposure. LIFE SCIENCES IN SPACE RESEARCH 2016; 9:19-47. [PMID: 27345199 PMCID: PMC5613937 DOI: 10.1016/j.lssr.2016.05.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 05/11/2016] [Indexed: 06/06/2023]
Abstract
Robust predictive models are essential to manage the risk of radiation-induced carcinogenesis. Chronic exposure to cosmic rays in the context of the complex deep space environment may place astronauts at high cancer risk. To estimate this risk, it is critical to understand how radiation-induced cellular stress impacts cell fate decisions and how this in turn alters the risk of carcinogenesis. Exposure to the heavy ion component of cosmic rays triggers a multitude of cellular changes, depending on the rate of exposure, the type of damage incurred and individual susceptibility. Heterogeneity in dose, dose rate, radiation quality, energy and particle flux contribute to the complexity of risk assessment. To unravel the impact of each of these factors, it is critical to identify sensitive biomarkers that can serve as inputs for robust modeling of individual risk of cancer or other long-term health consequences of exposure. Limitations in sensitivity of biomarkers to dose and dose rate, and the complexity of longitudinal monitoring, are some of the factors that increase uncertainties in the output from risk prediction models. Here, we critically evaluate candidate early and late biomarkers of radiation exposure and discuss their usefulness in predicting cell fate decisions. Some of the biomarkers we have reviewed include complex clustered DNA damage, persistent DNA repair foci, reactive oxygen species, chromosome aberrations and inflammation. Other biomarkers discussed, often assayed for at longer points post exposure, include mutations, chromosome aberrations, reactive oxygen species and telomere length changes. We discuss the relationship of biomarkers to different potential cell fates, including proliferation, apoptosis, senescence, and loss of stemness, which can propagate genomic instability and alter tissue composition and the underlying mRNA signatures that contribute to cell fate decisions. Our goal is to highlight factors that are important in choosing biomarkers and to evaluate the potential for biomarkers to inform models of post exposure cancer risk. Because cellular stress response pathways to space radiation and environmental carcinogens share common nodes, biomarker-driven risk models may be broadly applicable for estimating risks for other carcinogens.
Collapse
Affiliation(s)
| | | | - Steve R Blattnig
- Langley Research Center, Langley Research Center (LaRC), VA, United States
| | - Sylvain V Costes
- Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | | | | | | | - Lynn Hlatky
- CCSB-Tufts School of Medicine, Boston, MA, United States
| | - Yared Kidane
- Wyle Science, Technology & Engineering Group, Houston, TX, United States
| | - Amy Kronenberg
- Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Mamta D Naidu
- CCSB-Tufts School of Medicine, Boston, MA, United States
| | - Leif E Peterson
- Houston Methodist Research Institute, Houston, TX, United States
| | - Ianik Plante
- Wyle Science, Technology & Engineering Group, Houston, TX, United States
| | - Artem L Ponomarev
- Wyle Science, Technology & Engineering Group, Houston, TX, United States
| | - Janapriya Saha
- UT Southwestern Medical Center, Dallas, TX, United States
| | | | | | - Jonathan Tang
- Exogen Biotechnology, Inc., Berkeley, CA, United States
| | | | - Janice M Pluth
- Lawrence Berkeley National Laboratory, Berkeley, CA, United States.
| |
Collapse
|
22
|
Kim SC, Im W, Shim JY, Kim SK, Kim BJ. Static magnetic field controls cell cycle in cultured human glioblastoma cells. Cytotechnology 2016; 68:2745-2751. [PMID: 27121019 DOI: 10.1007/s10616-016-9973-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 04/19/2016] [Indexed: 11/29/2022] Open
Abstract
Magnetic field has been widely used in clinical diagnostics or for clinical treatment and is an important biomedical technology. Glioblastoma multiforme U87 and U251 are models of a fast growing malignant cancer. We focused on cellular level drafting of these cell lines as a time-dependent effect indicator of static magnetic fields (2000 ± 600 Gauss) by using their fast-growing properties. Cell viability showed a significant decrease (p < 0.01). The results coincided with the occurrence of apoptotic signals or protein expression of cyclin B1 and cyclin dependent kinase 1 in a non-apoptotic manner. Cdk1 was decreased in proportion to ankyrin G and cyclin B1 (Chi-square test, p = 0.0366). Our findings suggest that static magnetic stimulation creates a specific cyto-proliferative pattern, rather than producing randomized growth impairment.
Collapse
Affiliation(s)
- Seung Chan Kim
- Medical School, College of Medicine, Yonsei University, Yonsei-ro 50, Shinchon-dong, Seodaemoon-gu, Seoul, South Korea.
| | - Wooseok Im
- Seoul National University Hospital, Seoul, South Korea
| | - Jay Yong Shim
- Seoul National University Hospital, Seoul, South Korea
| | - Seung-Ki Kim
- Seoul National University Hospital, Seoul, South Korea
| | - Beom Jin Kim
- Department of Mathematics, Yonsei University, Seoul, South Korea
| |
Collapse
|
23
|
Recovery Profiles of T-Cell Subsets Following Low-Dose Total Body Irradiation and Improvement With Cinnamon. Int J Radiat Oncol Biol Phys 2015; 93:1118-26. [PMID: 26475064 DOI: 10.1016/j.ijrobp.2015.08.034] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 08/09/2015] [Accepted: 08/18/2015] [Indexed: 12/24/2022]
Abstract
PURPOSE Inefficient T-cell reconstitution from x-ray-induced immune damage reduces antitumor response. To understand the profile of T-cell reconstitution after irradiation will overcome the barrier of antitumor immunity. This study aimed to identify the recovery profile of T-cell subsets following x-ray irradiation and to highlight the role of cinnamon on efficient T-cell restoration postexposure in the antitumor response. METHODS AND MATERIALS CD3(+), CD8(+), and CD4(+) T cells and Th1, Th2, Th17, and regulatory T (Treg) cells were evaluated at different time points after single low-dose total body irradiation (SLTBI) with or without cinnamon treatments. T-bet, GATA3, RORγt, and Foxp3 signaling specific for Th1, Th2, Th17, and Treg were also analyzed by RT-PCR assay. The effects of cinnamon on efficient T-cell subset reconstitution was confirmed in a lung melanoma model in irradiated mice. RESULTS Reconstitution of CD4(+) T cells was delayed more than that of CD8(+) T cells in T-cell restoration after SLTBI. The production of IFNγ by Th1 or Tc1 cells was sharply decreased and was accompanied by reduced T-bet mRNA, even when total T-cell numbers had recovered; the frequencies of Th17 and Treg cells and their specific transcription factors (RORγt and Foxp3, respectively) were obviously increased. Irradiation-induced inefficient T-cell reconstitution impaired the antitumor capacities in the lung melanoma model. Pretreatment with cinnamon in irradiated mice accelerated the generation of Th1 and reduced the differentiation of Treg cells by activating T-bet and limiting transcriptions of Foxp3. Improvement resulting from cinnamon pretreatment on the efficient T-cell recovery profile from SLTBI promoted antitumor immunity in the lung melanoma model. CONCLUSIONS T-cell reconstitution from SLTBI was characterized by impaired Th1 and elevated Th17 and Treg cells. Cinnamon effectively improved the imbalance of T-cell subsets by promoting the proliferation of Th1 and by suppressing expansions of Th17 and Tregs. The role of cinnamon in efficient T-cell reconstitution from SLTBI is effective in antitumor immunity.
Collapse
|
24
|
Assessment of selected B cells populations in the workers of X-ray departments. Int J Occup Med Environ Health 2014; 27:467-73. [DOI: 10.2478/s13382-014-0242-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 01/21/2014] [Indexed: 11/20/2022] Open
Abstract
Abstract
Collapse
|
25
|
Partial depletion of regulatory T cells does not influence the inflammation caused by high dose hemi-body irradiation. PLoS One 2013; 8:e56607. [PMID: 23409194 PMCID: PMC3569437 DOI: 10.1371/journal.pone.0056607] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 01/14/2013] [Indexed: 11/19/2022] Open
Abstract
There is clinical interest in the modulation of regulatory T cells for cancer therapy. The safety of these therapies in combination with conventional anti-cancer therapies, including radiation therapy, can be studied in animal models. The effects of partial depletion of regulatory T (Treg) cells with an anti-CD25 antibody in conjunction with ionizing radiation on inflammation and tissue injury were analyzed in C57BL/6 mice. An anti-CD25 antibody (PC61) was administered 3 days prior to 13 Gy lower-half hemi-body irradiation (HBI). The blood, spleen, mesenteric lymph nodes (mLNs) and inguinal lymph nodes (iLNs) were harvested at various times thereafter. Alterations in the proportion of leukocyte subsets including CD4+ T cells, CD8+ T cells, Treg cells, B cells, NK cells, NK1.1+ T cells, macrophages and granulocytes were analyzed by FACS. The lungs, liver, pancreas, stomach, jejunum, duodenum, ileum, colon and kidney were harvested and studied by H&E staining. Expression of inflammatory mediators in plasma and tissue were investigated by ELISA. HBI significantly decreased the leukocyte pool though the various leukocyte subsets had different sensitivities to HBI. The administration of PC61 significantly decreased the proportion of Treg cells in spleen, iLN, mLN and blood (reduction of approximately 60%). Irradiation significantly increased the proportion of Treg cells in the spleen, iLN and mLN. HBI induced a systemic inflammatory reaction as demonstrated by increased plasma levels of IL-6, KC/CXCL1 and circulating granulocytes in the blood. Neutrophils also infiltrated the small bowel. The same general patterns were observed whether or not Treg cells were partially depleted with PC61 prior to HBI. These data demonstrate that partial depletion of Treg cells in these mice does not influence HBI-induced inflammatory response and tissue injury, and that combining anti-CD25 therapy with radiation may be safe and well tolerated in a clinical setting.
Collapse
|
26
|
Sonn CH, Choi JR, Kim TJ, Yu YB, Kim K, Shin SC, Park GH, Shirakawa T, Kim HS, Lee KM. Augmentation of natural cytotoxicity by chronic low-dose ionizing radiation in murine natural killer cells primed by IL-2. JOURNAL OF RADIATION RESEARCH 2012; 53:823-9. [PMID: 22915781 PMCID: PMC3483842 DOI: 10.1093/jrr/rrs037] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2011] [Revised: 05/22/2012] [Accepted: 05/23/2012] [Indexed: 05/20/2023]
Abstract
The possible beneficial effects of chronic low-dose irradiation (LDR) and its mechanism of action in a variety of pathophysiological processes such as cancer are a subject of intense investigation. While animal studies involving long-term exposure to LDR have yielded encouraging results, the influence of LDR at the cellular level has been less well defined. We reasoned that since natural killer (NK) cells constitute an early responder to exogenous stress, NK cells may reveal sentinel alterations in function upon exposure to LDR. When purified NK cells received LDR at 4.2 mGy/h for a total of 0.2 Gy in vitro, no significant difference in cell viability was observed. Likewise, no functional changes were detected in LDR-exposed NK cells, demonstrating that LDR alone was insufficient to generate changes at the cellular level. Nonetheless, significant augmentation of cytotoxic, but not proliferative, function was detected when NK cells were stimulated with low-dose IL-2 prior to irradiation. This enhancement of NK cytotoxicity was not due to alterations in NK-activating receptors, NK1.1, NKG2D, CD69 and 2B4, or changes in the rate of early or late apoptosis. Therefore, LDR, in the presence of suboptimal cytokine levels, can facilitate anti-tumor cytotoxicity of NK cells without influencing cellular proliferation or apoptosis. Whether these results translate to in vivo consequences remains to be seen; however, our data provide initial evidence that exposure to LDR can lead to subtle immune-enhancing effects on NK cells and may explain, in part, the functional basis underlying, diverse beneficial effects seen in the animals chronically exposed to LDR.
Collapse
Affiliation(s)
- Chung Hee Sonn
- Global Research Lab, BAERI Institute, Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul 136-705, Korea
| | - Jong Rip Choi
- Global Research Lab, BAERI Institute, Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul 136-705, Korea
| | - Tae-Jin Kim
- Global Research Lab, BAERI Institute, Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul 136-705, Korea
| | - Young-Bin Yu
- Global Research Lab, BAERI Institute, Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul 136-705, Korea
| | - Kwanghee Kim
- Global Research Lab, BAERI Institute, Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul 136-705, Korea
| | - Suk Chul Shin
- Radiation Health Research Institute, Korea Hydro & Nuclear Power Co., Ltd., 388-1, Ssangmun-dong, Dobong-gu, Seoul 132-703, Korea
| | - Gil-Hong Park
- Global Research Lab, BAERI Institute, Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul 136-705, Korea
| | - Toshiro Shirakawa
- Center for Infectious Diseases, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-Cho, Chuo-Ku, Kobe 650-0017, Japan
| | - Hee Sun Kim
- Radiation Health Research Institute, Korea Hydro & Nuclear Power Co., Ltd., 388-1, Ssangmun-dong, Dobong-gu, Seoul 132-703, Korea
- Corresponding authors. Tel: +82-2-3499-6661; Fax: +82-2-3499-6669; (H. S. Kim); Tel: +82-2-920-6251; Fax: +82-2-920-6253; (K. M. Lee)
| | - Kyung-Mi Lee
- Global Research Lab, BAERI Institute, Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul 136-705, Korea
- Corresponding authors. Tel: +82-2-3499-6661; Fax: +82-2-3499-6669; (H. S. Kim); Tel: +82-2-920-6251; Fax: +82-2-920-6253; (K. M. Lee)
| |
Collapse
|
27
|
Kujjo LL, Ronningen R, Ross P, Pereira RJG, Rodriguez R, Beyhan Z, Goissis MD, Baumann T, Kagawa W, Camsari C, Smith GW, Kurumizaka H, Yokoyama S, Cibelli JB, Perez GI. RAD51 plays a crucial role in halting cell death program induced by ionizing radiation in bovine oocytes. Biol Reprod 2012; 86:76. [PMID: 22190703 DOI: 10.1095/biolreprod.111.092064] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Reproductive health of humans and animals exposed to daily irradiants from solar/cosmic particles remains largely understudied. We evaluated the sensitivities of bovine and mouse oocytes to bombardment by krypton-78 (1 Gy) or ultraviolet B (UV-B; 100 microjoules). Mouse oocytes responded to irradiation by undergoing massive activation of caspases, rapid loss of energy without cytochrome-c release, and subsequent necrotic death. In contrast, bovine oocytes became positive for annexin-V, exhibited cytochrome-c release, and displayed mild activation of caspases and downstream DNAses but with the absence of a complete cell death program; therefore, cytoplasmic fragmentation was never observed. However, massive cytoplasmic fragmentation and increased DNA damage were induced experimentally by both inhibiting RAD51 and increasing caspase 3 activity before irradiation. Microinjection of recombinant human RAD51 prior to irradiation markedly decreased both cytoplasmic fragmentation and DNA damage in both bovine and mouse oocytes. RAD51 response to damaged DNA occurred faster in bovine oocytes than in mouse oocytes. Therefore, we conclude that upon exposure to irradiation, bovine oocytes create a physiologically indeterminate state of partial cell death, attributed to rapid induction of DNA repair and low activation of caspases. The persistence of these damaged cells may represent an adaptive mechanism with potential implications for livestock productivity and long-term health risks associated with human activity in space.
Collapse
Affiliation(s)
- Loro L Kujjo
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Datta K, Suman S, Trani D, Doiron K, Rotolo JA, Kallakury BVS, Kolesnick R, Cole MF, Fornace AJ. Accelerated hematopoietic toxicity by high energy (56)Fe radiation. Int J Radiat Biol 2011; 88:213-22. [PMID: 22077279 DOI: 10.3109/09553002.2012.639434] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE There is little information on the relative toxicity of highly charged (Z) high-energy (HZE) radiation in animal models compared to γ or X-rays, and the general assumption based on in vitro studies has been that acute toxicity is substantially greater. METHODS C57BL/6J mice were irradiated with (56)Fe ions (1 GeV/nucleon), and acute (within 30 d) toxicity compared to that of γ rays or protons (1 GeV). To assess relative hematopoietic and gastrointestinal toxicity, the effects of (56)Fe ions were compared to γ rays using complete blood count (CBC), bone marrow granulocyte-macrophage colony forming unit (GM-CFU), terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay for apoptosis in bone marrow, and intestinal crypt survival. RESULTS Although onset was more rapid, (56)Fe ions were only slightly more toxic than γ rays or protons with lethal dose (LD)(50/30) (a radiation dose at which 50% lethality occurs at 30-day) values of 5.8, 7.25, and 6.8 Gy, respectively, with relative biologic effectiveness for (56)Fe ions of 1.25 and 1.06 for protons. CONCLUSIONS (56)Fe radiation caused accelerated and more severe hematopoietic toxicity. Early mortality correlated with more profound leukopenia and subsequent sepsis. Results indicate that there is selective enhanced toxicity to bone marrow progenitor cells, which are typically resistant to γ rays, and bone marrow stem cells, because intestinal crypt cells did not show increased HZE toxicity.
Collapse
Affiliation(s)
- Kamal Datta
- Department of Biochemistry and Molecular & Cell Biology and Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057-1468, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Sanzari JK, Wilson JM, Wagner EB, Kennedy AR. The combined effects of reduced weightbearing and ionizing radiation on splenic lymphocyte population and function. Int J Radiat Biol 2011; 87:1033-8. [PMID: 21770700 DOI: 10.3109/09553002.2011.595875] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
PURPOSE The effects of radiation +/- hypogravity on immunologic function were investigated using the Partial Weight Suspension (PWS) model ( Wagner et al. 2010 ). MATERIALS AND METHODS Mice were exposed to 0.5, 1, or 2 Gray (Gy) dose of gamma radiation and then placed in the PWS system for 4, 24, 48 hours, or 4 days. Spleens were excised and white blood cells were prepared for flow cytometry analyses. RESULTS The combination of PWS + radiation (1 and 2 Gy doses only) resulted in decreased cell viability at the 24 h (∼16% decrease), 48 h (∼20% decrease), and 4 day (∼20% decrease) time points, compared to the PWS (no radiation) and no treatment (non-suspended, non-irradiated) groups. The T lymphocyte (thymus-derived) population increased by ∼10% (24 h, 48 h, and 4 day time points), while the B lymphocyte (bursal or bone marrow-derived) population decreased by ∼10% (at all time points examined), when mice were exposed to PWS + radiation (2 Gy dose only), compared to the PWS or no treatment groups. T cell activation was observed in the PWS group and the 0.5 Gy +/- PWS groups at the 4 and 24 h time points, compared to the no treatment group. However, T cell activation was significantly suppressed (∼85%) at the acute time points in the 2 Gy +/- PWS groups, comparable to the no treatment group. CONCLUSIONS Ionizing radiation in the absence and presence of simulated hypogravity results in acute lymphocyte dysfunction and compromised immune response.
Collapse
Affiliation(s)
- Jenine K Sanzari
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | |
Collapse
|
30
|
Shin SC, Lee KM, Kang YM, Kim K, Lim SA, Yang KH, Kim JY, Nam SY, Kim HS. Differential expression of immune-associated cancer regulatory genes in low- versus high-dose-rate irradiated AKR/J mice. Genomics 2011; 97:358-63. [PMID: 21266193 DOI: 10.1016/j.ygeno.2011.01.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Revised: 11/15/2010] [Accepted: 01/16/2011] [Indexed: 11/26/2022]
Abstract
AKR/J mice carrying leukemia viral inserts develop thymic lymphoma. Recently, we demonstrated that the incidence of thymic lymphoma was decreased when these mice were raised in a low-dose-rate γ-irradiation facility. In contrast, mice irradiated at a high-dose rate developed severe thymic lymphoma and died much earlier. To understand the genetic changes occurred by low- versus high-dose-rate γ-irradiation whole genome microarray was performed. Both groups of mice demonstrated up-regulation of Ifng, Igbp1, and IL7 in their thymuses, however, mice exposed to high-dose-rate γ-irradiation exhibited marked down-regulation of Sp3, Il15, Traf6, IL2ra, Pik3r1, and Hells. In contrast, low-dose-rate irradiated mice demonstrated up-regulation of Il15 and Jag2. These gene expression profiles imply the impaired immune signaling pathways by high-dose-rate γ-irradiation while the facilitation of anti-tumor immune responses by low-dose-rate γ-irradiation. Therefore, our data delineate common and distinct immune-associated pathways downstream of low- versus high-dose-rate irradiation in the process of cancer progression in AKR/J mice.
Collapse
Affiliation(s)
- Suk Chul Shin
- Radiation Health Research Institute, Korea Hydro and Nuclear Power Co., Ltd., SSangmun-dong, Dobong-gu, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Gridley DS, Pecaut MJ. Genetic background and lymphocyte populations after total-body exposure to iron ion radiation. Int J Radiat Biol 2010; 87:8-23. [PMID: 21067301 DOI: 10.3109/09553002.2010.518203] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
PURPOSE Particle radiations could significantly impact astronaut health during space missions. This study quantified the effects of iron ion radiation on lymphocytes in two strains of mice differing in susceptibility to radiation-induced acute myeloid leukemia (AML) and thymic lymphoma (TL): C57BL/6 (AML resistant, TL sensitive) and CBA/Ca (AML sensitive, TL resistant). MATERIALS AND METHODS The animals (n = 60/strain) were irradiated with ⁵⁶Fe(26+) (1 GeV) to total doses of 0, 0.5, 2 and 3 Gray (Gy) at an average dose rate of 1 Gy/min and euthanised on days 4 and 30 thereafter; blood, spleen, and bone marrow were collected for flow cytometry analyses. Cells expressing the following molecules were quantified: Cluster of differentiation (CD) 4, CD8, CD25, CD34, CD71, B220 (isoform of CD45 on B cells), NK1.1 (marker on natural killer or NK cells, C57B mice), panNK (marker on NK cells, CBA mice), and Sca1 (stem cell antigen 1). RESULTS Exposure to radiation resulted in different distribution patterns in lymphocyte populations and leukocytes expressing activation and progenitor markers in the two mouse strains. Significant main effects were dependent upon strain, as well as radiation dose, body compartment, and time of assessment. Especially striking differences were noted on day 4 after 3 Gy irradiation, including in the CD4:CD8 ratio [blood, C57 (2.83 ± 0.25) vs. CBA (6.19 ± 0.24); spleen, C57 (2.29 ± 0.12) vs. CBA (4.98 ± 0.22)], %CD25(+) mononuclear cells in bone marrow [C57 (5.62 ± 1.19) vs. CBA (12.45 ± 0.93)] and %CD34(+)Sca1(+) cells in bone marrow [CD45¹° gate, C57 (2.72 ± 0.74) vs. CBA (21.44 ± 0.73)]. CONCLUSION The results show that genetic background, as well as radiation dose and time post-exposure, had a profound impact on lymphocyte populations, as well as other leukocytes, after exposure to iron ion radiation.
Collapse
Affiliation(s)
- Daila S Gridley
- Department of Radiation Medicine, Radiation Research Laboratories, Divisions of Microbiology & Biochemistry, Loma Linda University and Medical Center, Loma Linda, California 92354, USA.
| | | |
Collapse
|
32
|
Sheikh Sajjadieh MR, Kuznetsova LV, Bojenko VB. Effect of cesium radioisotope on humoral immune status in Ukrainian children with clinical symptoms of irritable bowel syndrome related to Chernobyl disaster. Toxicol Ind Health 2010; 27:51-6. [PMID: 20826551 DOI: 10.1177/0748233710381890] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
UNLABELLED The aim of this study is to determine humoral immune status in Ukrainian children with clinical symptoms of irritable bowel syndrome 23 years after the Chernobyl disaster. METHOD AND MATERIAL The test population consisted of 95 participants: 75 rural patients aged 4-18, who lived in a contaminated area exposed to natural environmental radiation (falling under three groups) and 20 healthy urban participants from Kiev aged 5-15 as a control group. Internal radiation activity has been measured by gamma-ray spectrometry. B-lymphocytes population was analyzed with monoclonal antibody against antigen CD22(+). Serum immunoglobulins were evaluated by enzyme-linked immunosorbent assay (ELISA) method. p < 0.05 was considered significant. RESULT The percentage of CD22(+) in study groups is increased significantly in comparison to control group at p < 0.05. Reduced serum immunoglobulins levels have developed in the majority of the participants. CONCLUSION Humoral immune status of study groups with clinical symptom of irritable bowel syndrome residing in a contaminated area has changed.
Collapse
Affiliation(s)
- M R Sheikh Sajjadieh
- Department of Clinical Immunology and Allergology, National Medical Academy for Post Graduate Education, Kiev, Ukraine.
| | | | | |
Collapse
|
33
|
Sheikh Sajjadieh MR, Kuznetsova LV, Bojenko VB. Low internal radiation alters innate immune status in children with clinical symptom of irritable bowel syndrome. Toxicol Ind Health 2010; 26:525-31. [PMID: 20538707 DOI: 10.1177/0748233710373087] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Adverse health effect of low radiation is clear. The aim of this study was to determine effect of internal low radiation on innate immune status in Ukrainian children with spastic colitis as a result of Chernobyl disaster. The test population consisted of 95 participants: 75 rural participants with clinical symptom of irritable bowel syndrome, aged 4 to 18, who lived in a contaminated area exposed to radio nucleotide due to the disaster in reactor in Chernobyl nuclear power plant (categorized in three groups) and 20 healthy urban participants from Kiev, aged 5 to 15, as the control group. Internal radiation activity has been measured by gamma-ray spectrometry. Peripheral blood leukocytes were analyzed for CD16(+) subset, serum concentration of circulation immune complex was measured by the polyethylene glycol method. Phagocytic activity function was assessed by using latex article and phagocytic index were calculated. p < 0.05 was considered significant. Percent of CD16(+) cell in groups II and III increased significantly in comparison to control group (p < 0.05). Concentration of circulating immune complexes increased significantly in all study groups compared to control group (p < 0.001). Phagocytes activity and phagocyte index decreased significantly in all study groups in comparison to control group (p < 0.001). The innate immune status of study groups has changed. Our data have demonstrated that this change may be related to radioactivity from technogenic pollution due to the disaster in reactor in Chernobyl nuclear power plant.
Collapse
|
34
|
Cai X, Hao J, Zhang X, Yu B, Ren J, Luo C, Li Q, Huang Q, Shi X, Li W, Liu J. The polyhydroxylated fullerene derivative C60(OH)24 protects mice from ionizing-radiation-induced immune and mitochondrial dysfunction. Toxicol Appl Pharmacol 2009; 243:27-34. [PMID: 19914272 DOI: 10.1016/j.taap.2009.11.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Revised: 11/03/2009] [Accepted: 11/06/2009] [Indexed: 11/15/2022]
Abstract
Although the protective effect of the polyhydroxylated fullerene derivative C(60)(OH)(n) against ionizing radiation is an area of much interest, the mechanisms relating to how polyhydroxylated fullerene derivatives improve mitochondrial dysfunction remain unknown. In order to find new and effective radioprotective agents, we synthesized a new polyhydroxylated fullerene molecule with 24 hydroxyl groups of known positions on C(60) and studied its protective effects in mice subjected to irradiation. Mice were pretreated with C(60)(OH)(24) for 2 weeks (daily, 40 mg/kg i. p.), then subjected to a lethal dose of whole body gamma-irradiation (from a (60)Co source). Survival was observed for 30 days after irradiation. Immune and mitochondrial dysfunction and oxidative damage were analyzed in mice with the same C(60)(OH)(24) pretreatment and irradiation except that the animals were euthanized at day 5 after the irradiation. It was found that 2-week C(60)(OH)(24) pretreatment effectively reduced whole body irradiation-induced mortality without apparent toxicity. C(60)(OH)(24) pretreatment also showed significant protective effects against ionizing-radiation-induced decreases in immune and mitochondrial function and antioxidant defense in the liver and spleen. These results suggest that the polyhydroxylated fullerene derivative C(60)(OH)(24) protects against ionizing-radiation-induced mortality, possibly by enhancing immune function, decreasing oxidative damage and improving mitochondrial function.
Collapse
Affiliation(s)
- Xiaoqing Cai
- Laboratory of physical Biology, Shanghai Institure of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Baśkiewicz-Masiuk M, Grymuła K, Hałasa M, Pius E, Boehlke M, Machaliński B. Induction of mixed chimerism in mice by employing different conditioning protocols and bone marrow cell transplantation. Transplant Proc 2009; 41:1894-9. [PMID: 19545752 DOI: 10.1016/j.transproceed.2009.02.084] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2008] [Accepted: 02/09/2009] [Indexed: 10/20/2022]
Abstract
Mixed chimerism has been suggested to produce allograft tolerance. Since this phenomenon is not fully understood, the aim of our study was to evaluate various protocols for chimerism induction in a mouse model. B6.SJL-Ptprc(a)Pep3(b) mice were injected with 20 to 30 x 10(6) bone marrow cells from Balb C mice. Conditioning consisted of total body gamma irradiation with 9.5, 5, and 3 Gy on "-1 day" of the experiment, with 200 mg/kg cyclophosphamide (CP) ("+2 day"). Additionally, one group of mice received blocking antibody against CD40L on days 0, 1, 4, and 7. The presence of mixed chimerism in peripheral blood was assessed at 1, 2, 3, 4, 6, and 8 weeks using flow cytometry to detect CD45.1 or CD45.2 antigen expression. Moreover, the chimerism was examined in CD4, CD8, CD45/B220, Mac-1alpha subpopulations in peripheral blood and bone marrow cells at 8 weeks. We also compared chimerism in peripheral blood, bone marrow, and spleen leukocyte populations. We observed that the most effective conditioning method with relatively low toxicity was based on concomitant use of 5 Gy total body irradiation and CP. The percentage of donor cells differed among peripheral blood subpopulations and bone marrow cells, but was similar in leukocyte populations derived from various sources. Our experiments sought to optimize the induction of stable mixed chimerism.
Collapse
Affiliation(s)
- M Baśkiewicz-Masiuk
- Department of General Pathology, Pomeranian Medical University, Szczecin, Poland.
| | | | | | | | | | | |
Collapse
|
36
|
Gridley DS, Rizvi A, Luo-Owen X, Makinde AY, Pecaut MJ. Low dose, low dose rate photon radiation modifies leukocyte distribution and gene expression in CD4(+) T cells. JOURNAL OF RADIATION RESEARCH 2009; 50:139-50. [PMID: 19346678 DOI: 10.1269/jrr.08095] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
A better understanding of low dose radiation effects is needed to accurately estimate health risks. In this study, C57BL/6 mice were gamma-irradiated to total doses of 0, 0.01, 0.05, and 0.1 Gy ((57)Co; ~0.02 cGy/h). Subsets per group were euthanized at the end of irradiation (day 0) and on days 4 and 21 thereafter. Relative spleen mass and splenic white blood cell (WBC) counts, major leukocyte populations, and spontaneous DNA synthesis were consistently higher in the irradiated groups on day 0 compared to 0 Gy controls, although significance was not always obtained. In the spleen, all three major leukocyte types were significantly elevated on day 0 (P < 0.05). By day 21 post-irradiation the T, B, and natural killer (NK) cell counts, as well as CD4(+) T cells and CD4:CD8 T cell ratio, were low especially in the 0.01 Gy group. Although blood analyses showed no significant differences in leukocyte counts or red blood cell and platelet characteristics, the total T cells, CD4(+) T cells, and NK cells were increased by day 21 after 0.01 Gy (P < 0.05). Gene analysis of CD4(+) T cells negatively isolated from spleens on day 0 after 0.1 Gy showed significantly enhanced expression of Il27 and Tcfcp2, whereas Inha and Socs5 were down-regulated by 0.01 Gy and 0.1 Gy, respectively (P < 0.05). A trend for enhancement was noted in two additional genes (Il1r1 and Tbx21) in the 0.1 Gy group (P < 0.1). The data show that protracted low dose photons had dose- and time-dependent effects on CD4(+) T cells after whole-body exposure.
Collapse
Affiliation(s)
- Daila S Gridley
- Department of Radiation Medicine, Loma Linda University and Medical Center, CA 92354, USA.
| | | | | | | | | |
Collapse
|
37
|
Gridley DS, Obenaus A, Bateman TA, Pecaut MJ. Long-term changes in rat hematopoietic and other physiological systems after high-energy iron ion irradiation. Int J Radiat Biol 2008; 84:549-59. [PMID: 18661371 DOI: 10.1080/09553000802203614] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
PURPOSE To assess the long-term consequences of high-linear energy transfer (LET) iron ion radiation on immune and other critical body systems in the context of assessing potential effects astronauts may experience during exploratory missions. MATERIALS AND METHODS Sprague-Dawley rats were nearly whole-body irradiated with 56-Fe (5 GeV/n) to total doses of 0, 1, 2, and 4 Gray (Gy) and euthanized 9 months post-exposure for analyses. RESULTS Irradiated groups consistently had low body mass. Numbers of circulating white blood cells (WBC), lymphocytes and monocytes were lower in the 2 Gy group compared to 0 Gy (p < 0.05); a trend for low granulocytes was also noted. Red blood cell counts, hemoglobin, and hematocrit were decreased in irradiated animals (p < 0.05), whereas platelet counts and volume were unaffected. In the spleen, WBC counts and DNA synthesis by T cells were similar among groups and there were no differences in secreted interferon-gamma and interleukin-6. However, trends were noted for increased splenocyte capacity to secrete tumor necrosis factor-alpha and increased level of vascular endothelial cell growth factor in plasma. One or more of the irradiated groups had significant (p < 0.05) aberrations in several blood chemistry parameters associated with liver and kidney function. CONCLUSION The data show that exposure to 56-Fe radiation induced pathological changes in important body systems long after exposure.
Collapse
Affiliation(s)
- Daila S Gridley
- Department of Radiation Medicine, Loma Linda University and Medical Center, Loma Linda, CA 92354, USA.
| | | | | | | |
Collapse
|
38
|
Pecaut MJ, Dutta-Roy R, Smith AL, Jones TA, Nelson GA, Gridley DS. Acute effects of iron-particle radiation on immunity. Part I: Population distributions. Radiat Res 2006; 165:68-77. [PMID: 16392964 DOI: 10.1667/rr3493.1] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Health risks due to exposure to high-linear energy transfer (LET) charged particles remain unclear. The major goal of this study was to confirm and further characterize the acute effects of high-LET radiation ((56)Fe(26)) on erythrocyte, thrombocyte and leukocyte populations in three body compartments after total-body exposure. Adult female C57BL/6 mice were irradiated with total doses of 0, 0.5, 2 and 3 Gy and killed humanely 4 days later. Body and organ masses were determined and blood, spleen and bone marrow leukocytes were evaluated using a hematology analyzer and flow cytometry. Spleen and thymus (but not body, liver and lung) masses were significantly decreased in a dose-dependent manner. In general, red blood cell (RBC) counts and most other RBC parameters were depressed with increasing dose (P < 0.05); the major exception was an increase in cell size at 0.5 Gy. Platelet numbers and volume, total white blood cell counts, and all three major types of leukocytes also decreased (P < 0.05). Lymphocyte populations in blood and spleen exhibited variable degrees of susceptibility to (56)Fe-particle radiation (B > T > NK and T cytotoxic > T helper cells). In the bone marrow, leukocytes with granulocytic, lymphocytic ("dim" and "bright"), and monocytic characteristics exhibited proportional variations at the higher radiation doses in the expression of CD34 and/or Ly-6A/E. The data are discussed in relation to our previous investigations with iron ions, other forms of radiation, and space flight in this same animal model.
Collapse
Affiliation(s)
- Michael J Pecaut
- Department of Radiation Medicine, Radiobiology Program, Loma Linda University and Medical Center, CA 92354, USA.
| | | | | | | | | | | |
Collapse
|
39
|
Gridley DS, Dutta-Roy R, Andres ML, Nelson GA, Pecaut MJ. Acute Effects of Iron-Particle Radiation on Immunity. Part II: Leukocyte Activation, Cytokines and Adhesion. Radiat Res 2006; 165:78-87. [PMID: 16392965 DOI: 10.1667/rr3490.1] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The effects of high-linear energy transfer (LET) radiation on immune function have not been clearly established. The major goal of this study was to evaluate leukocyte responses after whole-body exposure to high-LET radiation. C57BL/6 mice were exposed to 0, 0.5, 2 and 3 Gy (56)Fe(26+) particles (1055 MeV/nucleon, 148.2 keV/microm) and killed humanely 4 days after exposure. Spontaneous synthesis of DNA in blood and spleen cells was increased significantly in groups receiving either 2 or 3 Gy (P < 0.001). In contrast, a significant depression in the response of T lymphocytes to phytohemagglutinin (PHA) and concanavalin A (ConA) was noted (P < 0.005); the response to lipopolysaccharide (LPS), a B-cell mitogen, was similar among groups. A cytometric bead array assay revealed that the level of tumor necrosis factor alpha (Tnfa) secreted by splenocytes increased significantly with increasing (56)Fe-particle dose (P < 0.05); interferon gamma, interleukin2 (Il2), Il4 and Il5 were unaffected. Flow cytometry analysis showed that 2 and 3 Gy markedly reduced splenic mononuclear cells expressing the activation markers CD25 and CD71, both with and without the T-cell marker CD3 (P < 0.05); proportions also varied significantly. Similar patterns were noted in mononuclear and granular cells with adhesion markers CD11b and, to a lesser extent, CD54 (P < 0.05). The results show that a single, acute exposure to high-LET radiation induced changes that can profoundly alter leukocyte functions. The implications of the data are discussed in relation to low-LET radiation, altered gravity, and space flight.
Collapse
Affiliation(s)
- Daila S Gridley
- Department of Radiation Medicine, Radiobiology Program, Loma Linda University and Medical Center, CA 92354, USA.
| | | | | | | | | |
Collapse
|
40
|
Pecaut MJ, Miller GM, Nelson GA, Gridley DS. Hypergravity-induced immunomodulation in a rodent model: hematological and lymphocyte function analyses. J Appl Physiol (1985) 2004; 97:29-38. [PMID: 14978009 DOI: 10.1152/japplphysiol.01304.2003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The major purpose of this study was to quantify hypergravity-induced changes in erythrocyte and thrombocyte characteristics, spontaneous and mitogen-induced lymphoblastogenesis, and capacity of splenocytes to secrete immunoregulatory cytokines. C57BL/6 mice were subjected to chronic 1, 2, and 3 G; subsets were euthanized after 1, 4, 7, 10, and 21 days of centrifugation. Erythrocyte counts, hematocrit, and hemoglobin were significantly reduced by day 21 in both centrifuged groups. Hemoglobin concentration and volume per red blood cell were generally low, but an early, transient spike above normal was noted in thrombocyte counts in the 3-G group. Fluctuations above and below normal in blood and spleen cell spontaneous blastogenesis were dependent on the length of centrifugation time and not on the level of gravity. Depression in splenocyte responses to phytohemagglutinin and lipopolysaccharide due to gravity were noted when the data were expressed as stimulation indexes. Cytokine production by spleen cells was primarily affected during the first week of centrifugation: IL-2, IL-4, and tumor necrosis factor-alpha increased, whereas interferon-gamma decreased. These findings, although not identical to those reported for spaceflight, indicate that altered gravity can influence both hematological and functional variables that may translate into serious health consequences during extended missions.
Collapse
Affiliation(s)
- Michael J Pecaut
- Department of Radiation Medicina, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA.
| | | | | | | |
Collapse
|
41
|
Pecaut MJ, Nelson GA, Peters LL, Kostenuik PJ, Bateman TA, Morony S, Stodieck LS, Lacey DL, Simske SJ, Gridley DS. Genetic models in applied physiology: selected contribution: effects of spaceflight on immunity in the C57BL/6 mouse. I. Immune population distributions. J Appl Physiol (1985) 2003; 94:2085-94. [PMID: 12514166 DOI: 10.1152/japplphysiol.01052.2002] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
There are several aspects of the spaceflight environment that may lead to changes in immunity: mission-related psychological stress, radiation, and changes in gravity. On December 5, 2001, the space shuttle Endeavor launched for a 12-day mission to examine these effects on C57BL/6 mice for the first time. On their return, assays were performed on the spleen, blood, and bone marrow. In response to flight, there were no significant differences in the general circulating leukocyte proportions. In contrast, there was an increase in splenic lymphocyte percentages, with a corresponding decrease in granulocytes. There was an overall shift in splenic lymphocytes away from T cells toward B cells, and a decrease in the CD4-to-CD8 ratios due to a decrease in T helpers. In contrast, there were proportional increases in bone marrow T cells, with decreases in B cells. Although the blast percentage and count were decreased in flight mice, the CD34(+) population was increased. The data were more consistent with a shift in bone marrow populations rather than a response to changes in the periphery. Many of the results are similar to those using other models. Clearly, spaceflight can influence immune parameters ranging from hematopoiesis to mature leukocyte mechanisms.
Collapse
Affiliation(s)
- Michael J Pecaut
- Department of Radiation Medicine, Radiobiology Program, Division of Microbiology and Molecular Genetics, Loma Linda University and Medical Center, Loma Linda, California 92354, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|