1
|
Giuranno L, Ient J, De Ruysscher D, Vooijs MA. Radiation-Induced Lung Injury (RILI). Front Oncol 2019; 9:877. [PMID: 31555602 PMCID: PMC6743286 DOI: 10.3389/fonc.2019.00877] [Citation(s) in RCA: 236] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Accepted: 08/23/2019] [Indexed: 12/12/2022] Open
Abstract
Radiation pneumonitis (RP) and radiation fibrosis (RF) are two dose-limiting toxicities of radiotherapy (RT), especially for lung, and esophageal cancer. It occurs in 5-20% of patients and limits the maximum dose that can be delivered, reducing tumor control probability (TCP) and may lead to dyspnea, lung fibrosis, and impaired quality of life. Both physical and biological factors determine the normal tissue complication probability (NTCP) by Radiotherapy. A better understanding of the pathophysiological sequence of radiation-induced lung injury (RILI) and the intrinsic, environmental and treatment-related factors may aid in the prevention, and better management of radiation-induced lung damage. In this review, we summarize our current understanding of the pathological and molecular consequences of lung exposure to ionizing radiation, and pharmaceutical interventions that may be beneficial in the prevention or curtailment of RILI, and therefore enable a more durable therapeutic tumor response.
Collapse
Affiliation(s)
- Lorena Giuranno
- Department of Radiotherapy, GROW School for Oncology Maastricht University Medical Centre, Maastricht, Netherlands
| | - Jonathan Ient
- Department of Radiotherapy, GROW School for Oncology Maastricht University Medical Centre, Maastricht, Netherlands
| | - Dirk De Ruysscher
- Department of Radiotherapy, GROW School for Oncology Maastricht University Medical Centre, Maastricht, Netherlands
| | - Marc A Vooijs
- Department of Radiotherapy, GROW School for Oncology Maastricht University Medical Centre, Maastricht, Netherlands
| |
Collapse
|
2
|
Kim JY, Park JH, Seo SM, Park JI, Jeon HY, Lee HK, Yoo RJ, Lee YJ, Woo SK, Lee WJ, Choi CM, Choi YK. Radioprotective effect of newly synthesized toll-like receptor 5 agonist, KMRC011, in mice exposed to total-body irradiation. JOURNAL OF RADIATION RESEARCH 2019; 60:432-441. [PMID: 31165150 PMCID: PMC6640901 DOI: 10.1093/jrr/rrz024] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 02/14/2019] [Indexed: 05/18/2023]
Abstract
Exposure to ionizing radiation leads to severe damages in radiosensitive organs and induces acute radiation syndrome, including effects on the hematopoietic system and gastrointestinal system. In this study, the radioprotective ability of KMRC011, a novel toll-like receptor 5 (TLR5) agonist, was investigated in C57BL6/N mice exposed to lethal total-body gamma-irradiation. In a 30-day survival study, KMRC011-treated mice had a significantly improved survival rate compared with control after 11 Gy total-body irradiation (TBI), and it was found that the radioprotective activity of KMRC011 depended on its dosage and repeated treatment. In a 5-day short-term study, we demonstrated that KMRC011 treatment stimulated cell proliferation and had an anti-apoptotic effect. Furthermore, KMRC011 increased the expressions of genes related to DNA repair, such as Rad21, Gadd45b, Sod2 and Irg1, in the small intestine of lethally irradiated mice. Interestingly, downregulation of NF-κB p65 in the mouse intestine by KMRC011 treatment was observed. This data indicated that KMRC011 exerted a radioprotective activity partially by regulating NF-κB signaling. Finally, peak expression levels of G-CSF, IL-6, IFN-γ, TNF-α and IP-10 induced by KMRC011 treatment were different depending on the route of administration and type of cytokine. These cytokines could be used as candidate biomarkers for the evaluation of KMRC011 clinical efficacy. Our data indicated that KMRC011 has radioprotective activity in lethally irradiated mice and may be developed as a therapeutic agent for radioprotection.
Collapse
Affiliation(s)
- Jun-Young Kim
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, Republic of Korea
| | - Jong-Hyung Park
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, Republic of Korea
- ViroMed Co., Ltd, 1, Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea
| | - Sun-Min Seo
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, Republic of Korea
| | - Jin-Il Park
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, Republic of Korea
- ViroMed Co., Ltd, 1, Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea
| | - Hee-Yeon Jeon
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, Republic of Korea
- Department of Core Research Laboratory, Clinical Research Institute, Kyung Hee University Hospital at Gangdong, 892, Dongnam-ro, Gangdong-gu, Seoul, Republic of Korea
| | - Han-Kyul Lee
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, Republic of Korea
| | - Ran-Ji Yoo
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, 75, Nowon-ro, Nowon-gu, Seoul, Republic of Korea
| | - Yong-Jin Lee
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, 75, Nowon-ro, Nowon-gu, Seoul, Republic of Korea
| | - Sang-Keun Woo
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, 75, Nowon-ro, Nowon-gu, Seoul, Republic of Korea
| | - Woo-Jong Lee
- Biomedical Manufacturing Technology Center, Korea Institute of Industrial Technology, 59, Yangho-gil, Yeongcheon-si, Gyeongsangbuk-do, Republic of Korea
| | - Chi-Min Choi
- Biomedical Manufacturing Technology Center, Korea Institute of Industrial Technology, 59, Yangho-gil, Yeongcheon-si, Gyeongsangbuk-do, Republic of Korea
| | - Yang-Kyu Choi
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, Republic of Korea
- Corresponding author. Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea. Tel: +82-2-2049-6113; Fax: +82-2-450-3037;
| |
Collapse
|
3
|
Popović J, Klajn A, Paunesku T, Ma Q, Chen S, Lai B, Stevanović M, Woloschak GE. Neuroprotective Role of Selected Antioxidant Agents in Preventing Cisplatin-Induced Damage of Human Neurons In Vitro. Cell Mol Neurobiol 2019; 39:619-636. [PMID: 30874981 PMCID: PMC6535150 DOI: 10.1007/s10571-019-00667-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 02/28/2019] [Indexed: 12/17/2022]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a side effect of platinum-based chemotherapy and decreases the quality of life of cancer patients. We compared neuroprotective properties of several agents using an in vitro model of terminally differentiated human cells NT2-N derived from cell line NT2/D1. Sodium azide and an active metabolite of amifostine (WR1065) increase cell viability in simultaneous treatment with cisplatin. In addition, WR1065 protects the non-dividing neurons by decreasing cisplatin caused oxidative stress and apoptosis. Accumulation of Pt in cisplatin-treated cells was heterogeneous, but the frequency and concentration of Pt in cells were lowered in the presence of WR1065 as shown by X-ray fluorescence microscopy (XFM). Transition metals accumulation accompanied Pt increase in cells; this effect was equally diminished in the presence of WR1065. To analyze possible chemical modulation of Pt-DNA bonds, we examined the platinum LIII near edge spectrum by X-ray absorption spectroscopy. The spectrum found in cisplatin-DNA samples is altered differently by the addition of either WR1065 or sodium azide. Importantly, a similar change in Pt edge spectra was noted in cells treated with cisplatin and WR1065. Therefore, amifostine should be reconsidered as a candidate for treatments that reduce or prevent CIPN.
Collapse
Affiliation(s)
- Jelena Popović
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, 11010, Serbia
- Feinberg School of Medicine, Department of Radiation Oncology, Northwestern University, Chicago, IL, 60611, USA
| | - Andrijana Klajn
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, 11010, Serbia
| | - Tatjana Paunesku
- Feinberg School of Medicine, Department of Radiation Oncology, Northwestern University, Chicago, IL, 60611, USA
| | - Qing Ma
- DND CAT, Northwestern Synchrotron Research Center at the Advanced Photon Source, Argonne National Laboratory, 9700 South Cass Avenue, Argonne, IL, 60439, USA
| | - Si Chen
- X-ray Science Division, Advanced Photon Source, Argonne National Laboratory, 9700 South Cass Avenue, Argonne, IL, 60439, USA
| | - Barry Lai
- X-ray Science Division, Advanced Photon Source, Argonne National Laboratory, 9700 South Cass Avenue, Argonne, IL, 60439, USA
| | - Milena Stevanović
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, 11010, Serbia.
- Faculty of Biology, University of Belgrade, Belgrade, 11000, Serbia.
- Serbian Academy of Sciences and Arts, Belgrade, 11000, Serbia.
| | - Gayle E Woloschak
- Feinberg School of Medicine, Department of Radiation Oncology, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
4
|
Kim JY, An YM, Yoo BR, Kim JM, Han SY, Na Y, Lee YS, Cho J. HSP27 inhibitor attenuates radiation-induced pulmonary inflammation. Sci Rep 2018; 8:4189. [PMID: 29520071 PMCID: PMC5843649 DOI: 10.1038/s41598-018-22635-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 02/27/2018] [Indexed: 01/22/2023] Open
Abstract
Radiation therapy has been used to treat over 70% of thoracic cancer; however, the method usually causes radiation pneumonitis. In the current study, we investigated the radioprotective effects of HSP27 inhibitor (J2) on radiation-induced lung inflammation in comparison to amifostine. In gross and histological findings, J2 treatment significantly inhibited immune cell infiltration in lung tissue, revealing anti-inflammatory potential of J2. Normal lung volume, evaluated by micro-CT analysis, in J2-treated mice was higher compared to that in irradiated mice. J2-treated mice reversed radiation-induced respiratory distress. However, amifostine did not show significant radioprotective effects in comparison to that of J2. In HSP27 transgenic mice, we observed increased immune cells recruitment and decreased volume of normal lung compared to wild type mice. Increased ROS production and oxidative stress after IR were down-regulated by J2 treatment, demonstrating antioxidant property of J2. The entire data of this study collectively showed that J2 may be an effective therapeutic agent for radiation-induced lung injury.
Collapse
Affiliation(s)
- Jee-Youn Kim
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong-Min An
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Byeong Rok Yoo
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jin-Mo Kim
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Song Yee Han
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Younghwa Na
- College of Pharmacy, CHA University, Pocheon, 487-010, Republic of Korea.
| | - Yun-Sil Lee
- College of Pharmacy and Division of Life and Pharmaceutical Science, Ewha Womans University, Seoul, Republic of Korea.
| | - Jaeho Cho
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
5
|
Seshacharyulu P, Baine MJ, Souchek JJ, Menning M, Kaur S, Yan Y, Ouellette MM, Jain M, Lin C, Batra SK. Biological determinants of radioresistance and their remediation in pancreatic cancer. Biochim Biophys Acta Rev Cancer 2017; 1868:69-92. [PMID: 28249796 PMCID: PMC5548591 DOI: 10.1016/j.bbcan.2017.02.003] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 02/16/2017] [Accepted: 02/17/2017] [Indexed: 12/17/2022]
Abstract
Despite recent advances in radiotherapy, a majority of patients diagnosed with pancreatic cancer (PC) do not achieve objective responses due to the existence of intrinsic and acquired radioresistance. Identification of molecular mechanisms that compromise the efficacy of radiation therapy and targeting these pathways is paramount for improving radiation response in PC patients. In this review, we have summarized molecular mechanisms associated with the radio-resistant phenotype of PC. Briefly, we discuss the reversible and irreversible biological consequences of radiotherapy, such as DNA damage and DNA repair, mechanisms of cancer cell survival and radiation-induced apoptosis following radiotherapy. We further describe various small molecule inhibitors and molecular targeting agents currently being tested in preclinical and clinical studies as potential radiosensitizers for PC. Notably, we draw attention towards the confounding effects of cancer stem cells, immune system, and the tumor microenvironment in the context of PC radioresistance and radiosensitization. Finally, we discuss the need for examining selective radioprotectors in light of the emerging evidence on radiation toxicity to non-target tissue associated with PC radiotherapy.
Collapse
Affiliation(s)
- Parthasarathy Seshacharyulu
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Michael J Baine
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Joshua J Souchek
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Melanie Menning
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Sukhwinder Kaur
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Ying Yan
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Michel M. Ouellette
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Chi Lin
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| |
Collapse
|
6
|
Ghorbel I, Chaabane M, Elwej A, Boudawara O, Abdelhedi S, Jamoussi K, Boudawara T, Zeghal N. Expression of metallothioneins I and II related to oxidative stress in the liver of aluminium-treated rats. Arch Physiol Biochem 2016; 122:214-222. [PMID: 27230980 DOI: 10.1080/13813455.2016.1187176] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Hepatotoxicity, induced by aluminium chloride (AlCl3), has been well studied but there are no reports about liver metallothionein (MT) genes induction. Therefore, it is of interest to establish the mechanism involving the relation between MT gene expression levels and the oxidative stress status in hepatic cells of aluminium-treated rats. Aluminium (Al) was administered to rats in their drinking water at a dose of 50 mg/kg body weight for three weeks. AlCl3 provoked hepatotoxicity objectified by an increase in malondialdehyde (MDA), hydrogen peroxide (H2O2), advanced oxidation protein products (AOPP), protein carbonyls (PCO) and a decrease in reduced glutathione (GSH), non-protein thiols (NPSH) and vitamin C. CAT and Glutathione peroxidase (GPx) activities were decreased while Mn-SOD gene expression, total Metallothionein content and MT I and MT II genes induction were increased. There are changes in plasma of some trace elements, albumin levels, transaminases, LDH and ALP activities. All these changes were supported by histopathological observations.
Collapse
Affiliation(s)
| | | | | | - Ons Boudawara
- b Anatomopathology Laboratory, CHU Habib Bourguiba , and
| | - Sameh Abdelhedi
- c Biochemistry Laboratory, CHU Hedi Chaker, University of Sfax , Sfax , Tunisia
| | - Kamel Jamoussi
- c Biochemistry Laboratory, CHU Hedi Chaker, University of Sfax , Sfax , Tunisia
| | | | | |
Collapse
|
7
|
A combination of high dose rate (10X FFF/2400 MU/min/10 MV X-rays) and total low dose (0.5 Gy) induces a higher rate of apoptosis in melanoma cells in vitro and superior preservation of normal melanocytes. Melanoma Res 2016; 25:376-89. [PMID: 26177150 PMCID: PMC4560269 DOI: 10.1097/cmr.0000000000000174] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The aim of this study was to determine the apoptotic effects, toxicity, and radiosensitization of total low dose irradiation delivered at a high dose rate in vitro to melanoma cells, normal human epidermal melanocytes (HEM), or normal human dermal fibroblasts (HDF) and to study the effect of mitochondrial inhibition in combination with radiation to enhance apoptosis in melanoma cells. Cells irradiated using 10X flattening filter-free (FFF) 10 MV X-rays at a dose rate of 400 or 2400 MU/min and a total dose of 0.25–8 Gy were analyzed by cell/colony counting, MitoTracker, MTT, and DNA-damage assays, as well as by quantitative real-time reverse transcriptase PCR in the presence or absence of mitochondrial respiration inhibitors. A dose rate of 2400 MU/min killed on average five-fold more melanoma cells than a dose rate 400 MU/min at a total dose of 0.5 Gy and preserved 80% survival of HEM and 90% survival of HDF. Increased apoptosis at the 2400 MU/min dose rate is mediated by greater DNA damage, reduced cell proliferation, upregulation of apoptotic genes, and downregulation of cell cycle genes. HEM and HDF were relatively unharmed at 2400 MU/min. Radiation induced upregulation of mitochondrial respiration in both normal and cancer cells, and blocking the respiration with inhibitors enhanced apoptosis only in melanoma cells. A high dose rate with a low total dose (2400 MU/min, 0.5 Gy/10X FFF 10 MV X-rays) enhances radiosensitivity of melanoma cells while reducing radiotoxicity toward HEM and HDF. Selective cytotoxicity of melanoma cells is increased by blocking mitochondrial respiration.
Collapse
|
8
|
Quercetin attenuates neuronal death against aluminum-induced neurodegeneration in the rat hippocampus. Neuroscience 2016; 324:163-76. [DOI: 10.1016/j.neuroscience.2016.02.055] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 02/22/2016] [Accepted: 02/23/2016] [Indexed: 11/21/2022]
|
9
|
Chen T, Wang L, Chen K, Qiu S, Cen X, Li H, Hu C. Evaluation of gamma ray-induced gastrointestinal tract morphological and proliferative activity changes in rhesus monkeys. Hum Exp Toxicol 2015; 35:1133-44. [PMID: 26699188 DOI: 10.1177/0960327115622259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
To provide support for future pharmacology and preclinical studies, we have established a stable nonhuman primate animal model to demonstrate the histopathological changes in the gastrointestinal tract following gamma ray irradiation. In this study, 12 healthy rhesus monkeys were divided into 2 groups (control and radiation groups). Animals in the radiation group were exposed to gamma rays (cobalt 60 source) at a dose level of 6.5 Gy total body irradiation bilaterally (i.e. 3.25 Gy on each side). Control animals were sham exposed using identical procedures. After a 5-day in-life observation period, gastrointestinal tract tissues (esophagus, stomach, duodenum, jejunum, ileum, colon, and rectum) were collected and fixed in 10% neutral-buffered formalin for subsequent hematoxylin and eosin and 5-bromo-2-deoxyuridine (BrdU) immunohistochemistry processing. The results showed that the esophagus was undergoing degeneration without obvious inflammatory changes, while the stomach and duodenum exhibited both degeneration and inflammation. From the jejunum to the rectum, late-stage inflammation with glandular regeneration, as well as a high-level BrdU labeling index, was present.
Collapse
Affiliation(s)
- T Chen
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China National Chengdu Center for Safety Evaluation of Drugs, Chengdu, China
| | - L Wang
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China National Chengdu Center for Safety Evaluation of Drugs, Chengdu, China
| | - K Chen
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China National Chengdu Center for Safety Evaluation of Drugs, Chengdu, China
| | - S Qiu
- National Chengdu Center for Safety Evaluation of Drugs, Chengdu, China
| | - X Cen
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China National Chengdu Center for Safety Evaluation of Drugs, Chengdu, China
| | - H Li
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China National Chengdu Center for Safety Evaluation of Drugs, Chengdu, China
| | - C Hu
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China National Chengdu Center for Safety Evaluation of Drugs, Chengdu, China
| |
Collapse
|
10
|
SOD2 Mediates Amifostine-Induced Protection against Glutamate in PC12 Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:4202437. [PMID: 26770652 PMCID: PMC4685138 DOI: 10.1155/2016/4202437] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 08/31/2015] [Accepted: 09/01/2015] [Indexed: 12/31/2022]
Abstract
Background. Cytoprotectant amifostine attenuates radiation-induced oxidative injury by increasing intracellular manganese superoxide dismutase (SOD2) in peripheral tissue. However, whether amifostine could protect neuronal cells against oxidative injury has not been reported. The purpose of this study is to explore the protection of amifostine in PC12 cells. Methods. PC12 cells exposed to glutamate were used to mimic neuronal oxidative injury. SOD assay kit was taken to evaluate intracellular Cu/Zn SOD (SOD1) and SOD2 activities; western blot analysis and immunofluorescence staining were performed to investigate SOD2 protein expression; MTT, lactate dehydrogenase (LDH), release and cell morphology were used to evaluate cell injury degree, and apoptotic rate and cleaved caspase-3 expression were taken to assess apoptosis; mitochondrial superoxide production, intracellular reactive oxygen species (ROS), and glutathione (GSH) and catalase (CAT) levels were evaluated by reagent kits. Results. Amifostine increased SOD2 activity and expression, decreased cell injury and apoptosis, reduced mitochondrial superoxide production and intracellular ROS generation, and restored intracellular GSH and CAT levels in PC12 cells exposed to glutamate. SOD2-siRNA, however, significantly reversed the amifostine-induced cytoprotective and antioxidative actions. Conclusion. SOD2 mediates amifostine-induced protection in PC12 cells exposed to glutamate.
Collapse
|
11
|
Slezak J, Kura B, Ravingerová T, Tribulova N, Okruhlicova L, Barancik M. Mechanisms of cardiac radiation injury and potential preventive approaches. Can J Physiol Pharmacol 2015; 93:737-53. [PMID: 26030720 DOI: 10.1139/cjpp-2015-0006] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
In addition to cytostatic treatment and surgery, the most common cancer treatment is gamma radiation. Despite sophisticated radiological techniques however, in addition to irradiation of the tumor, irradiation of the surrounding healthy tissue also takes place, which results in various side-effects, depending on the absorbed dose of radiation. Radiation either damages the cell DNA directly, or indirectly via the formation of oxygen radicals that in addition to the DNA damage, react with all cell organelles and interfere with their molecular mechanisms. The main features of radiation injury besides DNA damage is inflammation and increased expression of pro-inflammatory genes and cytokines. Endothelial damage and dysfunction of capillaries and small blood vessels plays a particularly important role in radiation injury. This review is focused on summarizing the currently available data concerning the mechanisms of radiation injury, as well as the effectiveness of various antioxidants, anti-inflammatory cytokines, and cytoprotective substances that may be utilized in preventing, mitigating, or treating the toxic effects of ionizing radiation on the heart.
Collapse
Affiliation(s)
- Jan Slezak
- Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic.,Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic
| | - Branislav Kura
- Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic.,Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic
| | - Táňa Ravingerová
- Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic.,Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic
| | - Narcisa Tribulova
- Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic.,Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic
| | - Ludmila Okruhlicova
- Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic.,Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic
| | - Miroslav Barancik
- Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic.,Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic
| |
Collapse
|
12
|
Korpela E, Liu SK. Endothelial perturbations and therapeutic strategies in normal tissue radiation damage. Radiat Oncol 2014; 9:266. [PMID: 25518850 PMCID: PMC4279961 DOI: 10.1186/s13014-014-0266-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 11/18/2014] [Indexed: 02/08/2023] Open
Abstract
Most cancer patients are treated with radiotherapy, but the treatment can also damage the surrounding normal tissue. Radiotherapy side-effects diminish patients’ quality of life, yet effective biological interventions for normal tissue damage are lacking. Protecting microvascular endothelial cells from the effects of irradiation is emerging as a targeted damage-reduction strategy. We illustrate the concept of the microvasculature as a mediator of overall normal tissue radiation toxicity through cell death, vascular inflammation (hemodynamic and molecular changes) and a change in functional capacity. Endothelial cell targeted therapies that protect against such endothelial cell perturbations and the development of acute normal tissue damage are mostly under preclinical development. Since acute radiation toxicity is a common clinical problem in cutaneous, gastrointestinal and mucosal tissues, we also focus on damage in these tissues.
Collapse
Affiliation(s)
- Elina Korpela
- Biological Sciences, Sunnybrook Research Institute and Odette Cancer Centre, Sunnybrook Health Sciences Centre, 2075 Bayview Ave., Toronto, M4N 3M5, Canada. .,Department of Medical Biophysics, University of Toronto, 101 College St., Toronto, M5G 1L7, Canada.
| | - Stanley K Liu
- Biological Sciences, Sunnybrook Research Institute and Odette Cancer Centre, Sunnybrook Health Sciences Centre, 2075 Bayview Ave., Toronto, M4N 3M5, Canada. .,Department of Medical Biophysics, University of Toronto, 101 College St., Toronto, M5G 1L7, Canada. .,Department of Radiation Oncology, University of Toronto, 149 College St., Toronto, M5T 1P5, Canada.
| |
Collapse
|
13
|
Toohey JI, Cooper AJL. Thiosulfoxide (sulfane) sulfur: new chemistry and new regulatory roles in biology. Molecules 2014; 19:12789-813. [PMID: 25153879 PMCID: PMC4170951 DOI: 10.3390/molecules190812789] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 08/11/2014] [Accepted: 08/12/2014] [Indexed: 11/24/2022] Open
Abstract
The understanding of sulfur bonding is undergoing change. Old theories on hypervalency of sulfur and the nature of the chalcogen-chalcogen bond are now questioned. At the same time, there is a rapidly expanding literature on the effects of sulfur in regulating biological systems. The two fields are inter-related because the new understanding of the thiosulfoxide bond helps to explain the newfound roles of sulfur in biology. This review examines the nature of thiosulfoxide (sulfane, S0) sulfur, the history of its regulatory role, its generation in biological systems, and its functions in cells. The functions include synthesis of cofactors (molybdenum cofactor, iron-sulfur clusters), sulfuration of tRNA, modulation of enzyme activities, and regulating the redox environment by several mechanisms (including the enhancement of the reductive capacity of glutathione). A brief review of the analogous form of selenium suggests that the toxicity of selenium may be due to over-reduction caused by the powerful reductive activity of glutathione perselenide.
Collapse
Affiliation(s)
| | - Arthur J L Cooper
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY 10595, USA.
| |
Collapse
|
14
|
Vasin MV. Comments on the mechanisms of action of radiation protective agents: basis components and their polyvalence. SPRINGERPLUS 2014; 3:414. [PMID: 25133093 PMCID: PMC4132458 DOI: 10.1186/2193-1801-3-414] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 07/31/2014] [Indexed: 12/18/2022]
Abstract
Purpose These comments suggest a division of radiation protective agents on the grounds of their mechanism of action that increase the radio resistance of an organism. Conclusion Given below is the division of radiation protective agents on the basis of their mechanism of action into 3 groups: 1) Radiation protective agents, with the implementation of radiation protective action taking place at the cellular level in the course of rapidly proceeding radiation-chemical reactions. At the same time, when the ionizing radiation energy is absorbed, these agents partially neutralize the “oxygen effect” as a radiobiological phenomenon, especially in the radiolysis of DNA; 2) Radiation protective agents that exert their effect at the system level by accelerating the post-radiation recovery of radiosensitive tissues through activation of a number of pro-inflammatory signaling pathways and an increase in the secretion of hematopoietic growth factors, including their use as mitigators in the early period after irradiation prior to the clinical development of acute radiation syndrome (ARS). 3) Radiomodulators including drugs and nutritional supplements that can elevate the resistance of the organism to adverse environmental factors, including exposure to ionization by means of modulating the gene expression through a hormetic effect of small doses of stressors and a “substrate” maintenance of adaptive changes, resulting in an increased antioxidant protection of the organism. Radiation protective agents having polyvalence in implementation of their action may simultaneously induce radioprotective effect by various routes with a prevalence of basis mechanisms of the action.
Collapse
Affiliation(s)
- Mikhail V Vasin
- Department of Medicine of Catastrophe, Russian Medical Academy of Post-Graduate Education, St. Polikarpova 10, 125284 Moscow, Russia
| |
Collapse
|
15
|
Perona M, Dagrosa MA, Pagotto R, Casal M, Pignataro O, Pisarev MA, Juvenal GJ. Protective effect of an antithyroid compound against γ-radiation-induced damage in human colon cancer cells. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2014; 53:611-619. [PMID: 24811726 DOI: 10.1007/s00411-014-0542-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Accepted: 04/13/2014] [Indexed: 06/03/2023]
Abstract
We have previously reported the radioprotective effect of propylthiouracil (PTU) on thyroid cells. The aim of the present study was to analyze whether tumor cells and normal cells demonstrate the same response to PTU. Human colon carcinoma cells were irradiated with γ-irradiation with or without PTU. We evaluated the clonogenic survival, intracellular reactive oxygen species levels, catalase, superoxide dismutase and glutathione peroxidase activities, and apoptosis by nuclear cell morphology and caspase-3 activity assays. Cyclic AMP (cAMP) levels were measured by radioimmunoassay. PTU treatment increased surviving cell fraction at 2 Gy (SF2) from 56.9 ± 3.6 in controls to 75.0 ± 3.5 (p < 0.05) and diminished radiation-induced apoptosis. In addition, we observed that the level of antioxidant enzymes' activity was increased in cells treated with PTU. Moreover, pretreatment with PTU increased intracellular levels of cAMP. Forskolin (p < 0.01) and dibutyryl cAMP (p < 0.05) mimicked the effect of PTU on SF2. Co-treatment with H89, an inhibitor of protein kinase A, abolished the radioprotective effect of PTU. PTU reduces the toxicity of ionizing radiation by increasing cAMP levels and also possibly through a reduction in apoptosis levels and in radiation-induced oxidative stress damage. We therefore conclude that PTU protects both normal and cancer cells during exposure to radiation in conditions mimicking the radiotherapy.
Collapse
Affiliation(s)
- Marina Perona
- Department of Radiobiology (CAC), National Atomic Energy Commission (CNEA), San Martín, Buenos Aires, Argentina
| | | | | | | | | | | | | |
Collapse
|
16
|
Candas D, Li JJ. MnSOD in oxidative stress response-potential regulation via mitochondrial protein influx. Antioxid Redox Signal 2014; 20:1599-617. [PMID: 23581847 PMCID: PMC3942709 DOI: 10.1089/ars.2013.5305] [Citation(s) in RCA: 466] [Impact Index Per Article: 42.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
SIGNIFICANCE The mitochondrial antioxidant manganese superoxide dismutase (MnSOD) is encoded by genomic DNA and its dismutase function is fully activated in the mitochondria to detoxify free radical O2(•-) generated by mitochondrial respiration. Accumulating evidence shows an extensive communication between the mitochondria and cytoplasm under oxidative stress. Not only is the MnSOD gene upregulated by oxidative stress, but MnSOD activity can be enhanced via the mitochondrial protein influx (MPI). RECENT ADVANCES A cluster of MPI containing cytoplasmic/nuclear proteins, such as cyclins, cyclin-dependent kinases, and p53 interact with and alter MnSOD activity. These proteins modulate MnSOD superoxide scavenging activity via post-translational modifications in the mitochondria. In addition to well-established pathways in gene expression, recent findings suggest that MnSOD enzymatic activity can also be enhanced by phosphorylation of specific motifs in mitochondria. This review attempts to discuss the pre- and post-translational regulation of MnSOD, and how these modifications alter MnSOD activity, which induces a cell adaptive response to oxidative stress. CRITICAL ISSUES MnSOD is biologically significant to aerobic cells. Its role in protecting the cells against the deleterious effects of reactive oxygen species is evident. However, the exact network of MnSOD-associated cellular adaptive reaction to oxidative stress and its post-translational modifications, especially its enzymatic enhancement via phosphorylation, is not yet fully understood. FUTURE DIRECTIONS The broad discussion of the multiple aspects of MnSOD regulation, including gene expression, protein modifications, and enzymatic activity, will shed light onto the unknown mechanisms that govern the prosurvival networks involved in cellular and mitochondrial adaptive response to genotoxic environment.
Collapse
Affiliation(s)
- Demet Candas
- 1 Department of Radiation Oncology, University of California Davis , Sacramento, California
| | | |
Collapse
|
17
|
Radiation protection from whole-body gamma irradiation (6.7 Gy): behavioural effects and brain protein-level changes by an aminothiol compound GL2011 in the Wistar rat. Amino Acids 2014; 46:1681-96. [DOI: 10.1007/s00726-014-1728-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 03/10/2014] [Indexed: 10/25/2022]
|
18
|
Miao L, Holley AK, Zhao Y, St Clair WH, St Clair DK. Redox-mediated and ionizing-radiation-induced inflammatory mediators in prostate cancer development and treatment. Antioxid Redox Signal 2014; 20:1481-500. [PMID: 24093432 PMCID: PMC3936609 DOI: 10.1089/ars.2013.5637] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
SIGNIFICANCE Radiation therapy is widely used for treatment of prostate cancer. Radiation can directly damage biologically important molecules; however, most effects of radiation-mediated cell killing are derived from the generated free radicals that alter cellular redox status. Multiple proinflammatory mediators can also influence redox status in irradiated cells and the surrounding microenvironment, thereby affecting prostate cancer progression and radiotherapy efficiency. RECENT ADVANCES Ionizing radiation (IR)-generated oxidative stress can regulate and be regulated by the production of proinflammatory mediators. Depending on the type and stage of the prostate cancer cells, these proinflammatory mediators may lead to different biological consequences ranging from cell death to development of radioresistance. CRITICAL ISSUES Tumors are heterogeneous and dynamic communication occurs between stromal and prostate cancer cells, and complicated redox-regulated mechanisms exist in the tumor microenvironment. Thus, antioxidant and anti-inflammatory strategies should be carefully evaluated for each patient at different stages of the disease to maximize therapeutic benefits while minimizing unintended side effects. FUTURE DIRECTIONS Compared with normal cells, tumor cells are usually under higher oxidative stress and secrete more proinflammatory mediators. Thus, redox status is often less adaptive in tumor cells than in their normal counterparts. This difference can be exploited in a search for new cancer therapeutics and treatment regimes that selectively activate cell death pathways in tumor cells with minimal unintended consequences in terms of chemo- and radio-resistance in tumor cells and toxicity in normal tissues.
Collapse
Affiliation(s)
- Lu Miao
- 1 Graduate Center for Toxicology, University of Kentucky , Lexington, Kentucky
| | | | | | | | | |
Collapse
|
19
|
Patwardhan RS, Sharma D, Checker R, Sandur SK. Mitigation of radiation-induced hematopoietic injury via regulation of cellular MAPK/phosphatase levels and increasing hematopoietic stem cells. Free Radic Biol Med 2014; 68:52-64. [PMID: 24287141 DOI: 10.1016/j.freeradbiomed.2013.11.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 10/31/2013] [Accepted: 11/05/2013] [Indexed: 11/22/2022]
Abstract
Here we describe a novel strategy for mitigation of ionizing radiation-induced hematopoietic syndrome by suppressing the activity of MKP3, resulting in ERK activation and enhanced abundance of hematopoietic stem cells, using the antioxidant flavonoid baicalein (5,6,7-trihydroxyflavone). It offered complete protection to mouse splenic lymphocytes against radiation-induced cell death. Inhibitors of ERK and Nrf-2 could significantly abrogate baicalein-mediated radioprotection in lymphocytes. Baicalein inhibited phosphatase MKP3 and thereby enhanced phosphorylation of ERK and its downstream proteins such as Elk and Nrf-2. It also increased the nuclear levels of Nrf-2 and the mRNA levels of its dependent genes. Importantly, baicalein administration to mice before radiation exposure led to significant recovery of loss of bone marrow cellularity and also inhibited cell death. Administration of baicalein increased the hematopoietic stem cell frequency as measured by side-population assay and also by antibody staining. Further, baicalein offered significant protection against whole-body irradiation (WBI; 7.5Gy)-induced mortality in mice. Interestingly, we found that baicalein works by activating the same target molecules ERK and Nrf-2 both in vitro and in vivo. Finally, administration of all-trans-retinoic acid (inhibitor of Nrf-2) significantly abrogated baicalein-mediated protection against WBI-induced mortality in mice. Thus, in contrast to the generalized conception of antioxidants acting as radioprotectors, we provide a rationale that antioxidants exhibit pleiotropic effects through the activation of multiple cellular signaling pathways.
Collapse
Affiliation(s)
- R S Patwardhan
- Radiation Biology and Health Sciences Division, Modular Laboratories, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
| | - Deepak Sharma
- Radiation Biology and Health Sciences Division, Modular Laboratories, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
| | - Rahul Checker
- Radiation Biology and Health Sciences Division, Modular Laboratories, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
| | - Santosh K Sandur
- Radiation Biology and Health Sciences Division, Modular Laboratories, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India.
| |
Collapse
|
20
|
Grdina DJ, Murley JS, Miller RC, Mauceri HJ, Sutton HG, Thirman MJ, Li JJ, Woloschak GE, Weichselbaum RR. A manganese superoxide dismutase (SOD2)-mediated adaptive response. Radiat Res 2012; 179:115-24. [PMID: 23237540 DOI: 10.1667/rr3126.2] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Very low doses of ionizing radiation, 5 to 100 mGy, can induce adaptive responses characterized by elevation in cell survival and reduction in micronuclei formation. Utilizing these end points, RKO human colon carcinoma and transformed mouse embryo fibroblasts (MEF), wild-type or knockout cells missing TNF receptors 1 and 2 (TNFR1(-)R2(-)), and C57BL/6 and TNFR1(-)R2(-) knockout mice, we demonstrate that intact TNF signaling is required for induction of elevated manganese superoxide dismutase (SOD2) activity (P < 0.001) and the subsequent expression of these SOD2-mediated adaptive responses when cells are challenged at a later time with 2 Gy. In contrast, amifostine's free thiol form WR1065 can directly activate NF-κB giving rise to elevated SOD2 activity 24 h later and induce an adaptive response in both MEF wild-type and TNF signaling defective TNFR1(-)R2(-) cells. Transfection of cells with SOD2 siRNA completely abolishes both the elevation in SOD2 activity and expression of the adaptive responses. These results were confirmed in vivo using a micronucleus assay in splenocytes derived from C57BL/6 and TNFR1(-)R2(-) knockout mice that were exposed to 100 mGy or 400 mg/kg amifostine 24 h prior to exposure to a 2 Gy whole-body dose. A dose of 100 mGy also conferred enhanced protection to C57BL/6 mice exposed 24 h later to 100 mg/kg of N-Ethyl-N-nitrosourea (ENU). While very low radiation doses require an intact TNF signaling process to induce a SOD2-mediated adaptive response, amifostine can induce a similar adaptive response in both TNF receptor competent and knockout cells, respectively.
Collapse
Affiliation(s)
- David J Grdina
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL 60637, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Eldridge A, Fan M, Woloschak G, Grdina DJ, Chromy BA, Li JJ. Manganese superoxide dismutase interacts with a large scale of cellular and mitochondrial proteins in low-dose radiation-induced adaptive radioprotection. Free Radic Biol Med 2012; 53:1838-47. [PMID: 23000060 PMCID: PMC3494792 DOI: 10.1016/j.freeradbiomed.2012.08.589] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Revised: 08/24/2012] [Accepted: 08/28/2012] [Indexed: 11/18/2022]
Abstract
The cellular adaptive response to certain low-level genotoxic stresses, including exposure to low-dose ionizing radiation (LDIR), shows promise as a tool to enhance radioprotection in normal cells but not in tumor cells. Manganese superoxide dismutase (MnSOD), a fundamental mitochondrial antioxidant in mammalian cells, plays a key role in the LDIR-induced adaptive response. In this study, we aimed to elucidate the signaling network associated with MnSOD-induced radiation protection. A MnSOD-interacting protein profile was established in LDIR-treated human skin cells. Human skin keratinocytes (HK18) were irradiated with a single dose of LDIR (10 cGy X-ray) and the cell lysates were immunoprecipitated using α-MnSOD and applied to two different gel-based proteomic experiments followed by mass spectrometry for protein identification. Analysis of the profiles of MnSOD-interacting partners before and after LDIR detected various patterns of MnSOD protein-protein interactions in response to LDIR. Interestingly, many of the MnSOD-interacting proteins are known to have functions related to mitochondrial regulation of cell metabolism, apoptosis, and DNA repair. These results provide evidence indicating that in addition to the enzymatic action of detoxifying superoxide, the antioxidant MnSOD may function as a signaling regulator in stress-induced adaptive protection through cell survival pathways.
Collapse
Affiliation(s)
- Angela Eldridge
- Department of Radiation Oncology, University of California at Davis School of Medicine, Sacramento, CA 95817, USA
| | | | | | | | | | | |
Collapse
|
22
|
Koukourakis MI. Radiation damage and radioprotectants: new concepts in the era of molecular medicine. Br J Radiol 2012; 85:313-30. [PMID: 22294702 DOI: 10.1259/bjr/16386034] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Exposure to ionising radiation results in mutagenesis and cell death, and the clinical manifestations depend on the dose and the involved body area. Reducing carcinogenesis in patients treated with radiotherapy, exposed to diagnostic radiation or who are in certain professional groups is mandatory. The prevention or treatment of early and late radiotherapy effects would improve quality of life and increase cancer curability by intensifying therapies. Experimental and clinical data have given rise to new concepts and a large pool of chemical and molecular agents that could be effective in the protection and treatment of radiation damage. To date, amifostine is the only drug recommended as an effective radioprotectant. This review identifies five distinct types of radiation damage (I, cellular depletion; II, reactive gene activation; III, tissue disorganisation; IV, stochastic effects; V, bystander effects) and classifies the radioprotective agents into five relevant categories (A, protectants against all types of radiation effects; B, death pathway modulators; C, blockers of inflammation, chemotaxis and autocrine/paracrine pathways; D, antimutagenic keepers of genomic integrity; E, agents that block bystander effects). The necessity of establishing and funding central committees that guide systematic clinical research into evaluating the novel agents revealed in the era of molecular medicine is stressed.
Collapse
Affiliation(s)
- M I Koukourakis
- Department of Radiotherapy and Oncology, Democritus University of Thrace, Alexandroupolis, Greece.
| |
Collapse
|
23
|
Murley J, Baker K, Miller R, Darga T, Weichselbaum R, Grdina D. SOD2-mediated adaptive responses induced by low-dose ionizing radiation via TNF signaling and amifostine. Free Radic Biol Med 2011; 51:1918-25. [PMID: 21945096 PMCID: PMC3200566 DOI: 10.1016/j.freeradbiomed.2011.08.032] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 08/22/2011] [Accepted: 08/29/2011] [Indexed: 01/27/2023]
Abstract
Manganese superoxide dismutase (SOD2)-mediated adaptive processes that protect against radiation-induced micronucleus formation can be induced in cells after a 2-Gy exposure by previously exposing them to either low-dose ionizing radiation (10cGy) or WR1065 (40μM), the active thiol form of amifostine. Although both adaptive processes culminate in elevated levels of SOD2 enzymatic activity, the underlying pathways differ in complexity, with the tumor necrosis factor α (TNFα) signaling pathway implicated in the low-dose radiation-induced response, but not in the thiol-induced pathway. The goal of this study was the characterization of the effects of TNFα receptors 1 and 2 (TNFR1, TNFR2) on the adaptive responses induced by low-dose irradiation or thiol exposure using micronucleus formation as an endpoint. BFS-1 wild-type cells with functional TNFR1 and 2 were exposed 24h before a 2-Gy dose of ionizing radiation to either 10cGy or a 40μM dose of WR1065. BFS2C-SH02 cells, defective in TNFR1, and BFS2C-SH22 cells, defective in both TNFR1 and TNFR2 and generated from BFS2C-SH02 cells by transfection with a murine TNFR2-targeting vector and confirmed to be TNFR2 defective by quantitative PCR, were also exposed under similar conditions for comparison. A 10-cGy dose of radiation induced a significant elevation in SOD2 activity in BFS-1 (P<0.001) and BFS2C-SH02 (P=0.005) but not BFS2C-SH22 cells (P=0.433), compared to their respective untreated controls. In contrast, WR1065 significantly induced elevations in SOD2 activity in all three cell lines (P=0.001, P=0.007, P=0.020, respectively). A significant reduction in the frequency of radiation-induced micronuclei was observed in each cell line when exposure to a 2-Gy challenge dose of radiation occurred during the period of maximal elevation in SOD2 activity. However, this adaptive effect was completely inhibited if the cells were transfected 24h before low-dose radiation or thiol exposure with SOD2 siRNA. Under the conditions tested, TNFR1 and 2 inhibition negatively affected the low-dose radiation-induced but not the thiol-induced adaptive responses observed to be mediated by elevations in SOD2 activity.
Collapse
MESH Headings
- Amifostine/analogs & derivatives
- Amifostine/chemistry
- Animals
- Cell Line, Tumor
- Enzyme Activation/genetics
- Enzyme Activation/radiation effects
- Mercaptoethylamines/chemistry
- Mercaptoethylamines/pharmacology
- Mice
- Micronuclei, Chromosome-Defective/drug effects
- Micronuclei, Chromosome-Defective/radiation effects
- Micronucleus Tests
- RNA, Small Interfering/genetics
- Radiation, Ionizing
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Receptors, Tumor Necrosis Factor, Type II/genetics
- Receptors, Tumor Necrosis Factor, Type II/metabolism
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Signal Transduction/radiation effects
- Superoxide Dismutase/genetics
- Superoxide Dismutase/metabolism
- Tumor Necrosis Factor-alpha/pharmacology
Collapse
Affiliation(s)
- J.S. Murley
- Department of Radiation and Cellular Oncology, The University of Chicago, 5841 S. Maryland Avenue, Chicago, Illinois, U.S.A. 60637
| | - K.L. Baker
- Department of Radiation and Cellular Oncology, The University of Chicago, 5841 S. Maryland Avenue, Chicago, Illinois, U.S.A. 60637
| | - R.C. Miller
- Department of Radiation and Cellular Oncology, The University of Chicago, 5841 S. Maryland Avenue, Chicago, Illinois, U.S.A. 60637
| | - T.E. Darga
- Department of Radiation and Cellular Oncology, The University of Chicago, 5841 S. Maryland Avenue, Chicago, Illinois, U.S.A. 60637
| | - R.R. Weichselbaum
- Department of Radiation and Cellular Oncology, The University of Chicago, 5841 S. Maryland Avenue, Chicago, Illinois, U.S.A. 60637
| | - D.J. Grdina
- Department of Radiation and Cellular Oncology, The University of Chicago, 5841 S. Maryland Avenue, Chicago, Illinois, U.S.A. 60637
- Corresponding Author. 773-702-5250 (phone); 773-702-5740 (fax);
| |
Collapse
|
24
|
Bogojević D, Poznanović G, Grdović N, Grigorov I, Vidaković M, Dinić S, Mihailović M. Administration of rat acute-phase protein α(2)-macroglobulin before total-body irradiation initiates cytoprotective mechanisms in the liver. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2011; 50:167-179. [PMID: 20848291 DOI: 10.1007/s00411-010-0331-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2009] [Accepted: 09/03/2010] [Indexed: 05/29/2023]
Abstract
Previously, we showed that administration of the acute-phase protein α(2)-macroglobulin (α(2)M) to rats before total-body irradiation with 6.7 Gy (LD(50/30)) of X-rays provides the same level of radioprotection as amifostine. Here, we compare the cytoprotective effects of α(2)M and amifostine on rat liver. The potential of the liver to replenish cells destroyed by ionizing radiation was assessed by immunoblot analysis with antibody to proliferating cell nuclear antigen (PCNA). After irradiation, in unprotected rats PCNA decreased 6-fold from the basal level. In rats pretreated with either α(2)M or amifostine, PCNA was increased throughout a 4 week follow-up period, indicating that hepatocyte proliferation was unaffected. Since PCNA is an important component of the repair machinery, its increased expression was accompanied by significantly lower DNA damage in α(2)M- and amifostine-treated rats. At 2 weeks after irradiation, the Comet assay revealed a 15-fold increase in DNA damage in unprotected rats, while in α(2)M- and amifostine-treated rats we observed 3- and 4-fold rise in damage, respectively. The improved protection to DNA damage was supported by elevated activity of the antioxidant systems. Compared to untreated rats, pretreatments with α(2)M and amifostine led to similar increases in levels of the inflammatory cytokine IL-6 and the redox-sensitive transcription factor NFκB, promoting upregulation of MnSOD, the major component of the cell's antioxidant axis, and subsequent increases in Mn/CuZnSOD and catalase enzymatic activities. The results show that α(2)M induces protein factors whose interplay underlies radioprotection and support the idea that α(2)M is the central effector of natural radioprotection in the rat.
Collapse
Affiliation(s)
- Desanka Bogojević
- Institute for Biological Research Siniša Stanković, University of Belgrade, Despot Stephen Blvd. 142, 11060, Belgrade, Serbia
| | | | | | | | | | | | | |
Collapse
|
25
|
Murley JS, Kataoka Y, Miller RC, Li JJ, Woloschak G, Grdina DJ. SOD2-mediated effects induced by WR1065 and low-dose ionizing radiation on micronucleus formation in RKO human colon carcinoma cells. Radiat Res 2010; 175:57-65. [PMID: 21175348 DOI: 10.1667/rr2349.1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
RKO36 cells exposed to either WR1065 or 10 cGy X rays show elevated SOD2 gene expression and SOD2 enzymatic activity. Cells challenged at this time with 2 Gy exhibit enhanced radiation resistance. This phenomenon has been identified as a delayed radioprotective effect or an adaptive response when induced by thiols or low-dose radiation, respectively. In this study we investigated the relative effectiveness of both WR1065 and low-dose radiation in reducing the incidence of radiation-induced micronucleus formation in binucleated RKO36 human colon carcinoma cells. The role of SOD2 in this process was assessed by measuring changes in enzymatic activity as a function of the inducing agent used, the level of protection afforded, and the inhibitory effects of short interfering RNA (SOD2 siRNA). Both WR1065 and 10 cGy X rays effectively induced a greater than threefold elevation in SOD2 activity 24 h after exposure. Cells irradiated at this time with 2 Gy exhibited a significant resistance to micronucleus formation (P < 0.05; Student's two-tailed t test). This protective effect was significantly inhibited in cells transfected with SOD2 siRNA. SOD2 played an important role in the adaptive/delayed radioprotective response by inhibiting the initiation of a superoxide anion-induced ROS cascade leading to enhanced mitochondrial and nuclear damages.
Collapse
Affiliation(s)
- Jeffrey S Murley
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, Illinois 60637, USA
| | | | | | | | | | | |
Collapse
|
26
|
Abstract
Radiation leaves a fairly characteristic footprint in biological materials, but this is rapidly all but obliterated by the canonical biological responses to the radiation damage. The innate immune recognition systems that sense "danger" through direct radiation damage and through associated collateral damage set in motion a chain of events that, in a tissue compromised by radiation, often unwittingly result in oscillating waves of molecular and cellular responses as tissues attempt to heal. Understanding "nature's whispers" that inform on these processes will lead to novel forms of intervention targeted more precisely towards modifying them in an appropriate and timely fashion so as to improve the healing process and prevent or mitigate the development of acute and late effects of normal tissue radiation damage, whether it be accidental, as a result of a terrorist incident, or of therapeutic treatment of cancer. Here we attempt to discuss some of the non-free radical scavenging mechanisms that modify radiation responses and comment on where we see them within a conceptual framework of an evolving radiation-induced lesion.
Collapse
Affiliation(s)
- Kwanghee Kim
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - William H. McBride
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90095, USA
| |
Collapse
|
27
|
Colon J, Hsieh N, Ferguson A, Kupelian P, Seal S, Jenkins DW, Baker CH. Cerium oxide nanoparticles protect gastrointestinal epithelium from radiation-induced damage by reduction of reactive oxygen species and upregulation of superoxide dismutase 2. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2010; 6:698-705. [DOI: 10.1016/j.nano.2010.01.010] [Citation(s) in RCA: 241] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2009] [Revised: 01/20/2010] [Accepted: 01/25/2010] [Indexed: 10/19/2022]
|
28
|
Dziegielewski J, Goetz W, Baulch JE. Heavy ions, radioprotectors and genomic instability: implications for human space exploration. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2010; 49:303-316. [PMID: 20035342 DOI: 10.1007/s00411-009-0261-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2009] [Accepted: 12/09/2009] [Indexed: 05/28/2023]
Abstract
The risk associated with space radiation exposure is unique from terrestrial radiation exposures due to differences in radiation quality, including linear energy transfer (LET). Both high- and low-LET radiations are capable of inducing genomic instability in mammalian cells, and this instability is thought to be a driving force underlying radiation carcinogenesis. Unfortunately, during space exploration, flight crews cannot entirely avoid radiation exposure. As a result, chemical and biological countermeasures will be an important component of successful extended missions such as the exploration of Mars. There are currently several radioprotective agents (radioprotectors) in use; however, scientists continue to search for ideal radioprotective compounds-safe to use and effective in preventing and/or reducing acute and delayed effects of irradiation. This review discusses the agents that are currently available or being evaluated for their potential as radioprotectors. Further, this review discusses some implications of radioprotection for the induction and/or propagation of genomic instability in the progeny of irradiated cells.
Collapse
|
29
|
Brown AP, Chung EJ, Urick ME, Shield WP, Sowers AL, Thetford A, Shankavaram UT, Mitchell JB, Citrin DE. Evaluation of the fullerene compound DF-1 as a radiation protector. Radiat Oncol 2010; 5:34. [PMID: 20459795 PMCID: PMC2877563 DOI: 10.1186/1748-717x-5-34] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2010] [Accepted: 05/11/2010] [Indexed: 01/21/2023] Open
Abstract
Background Fullerene compounds are known to possess antioxidant properties, a common property of chemical radioprotectors. DF-1 is a dendrofullerene nanoparticle with antioxidant properties previously found to be radioprotective in a zebrafish model. The purpose of this study was to evaluate the radioprotective effects of DF-1 in a murine model of lethal total body irradiation and to assess for selective radioprotection of normal cells versus tumor cells. Methods In vitro radioresponse was evaluated with clonogenic assays with human tumor cells and fibroblast lines in the presence of varying concentrations of DF-1 or vehicle. DNA double strand break induction and repair was evaluated with immunocytochemistry for γH2AX. Lethal total body irradiation was delivered with 137Cs after intraperitoneal delivery of DF-1 or vehicle control. Bone marrow hypoxia was evaluated with piminidazole uptake assessed by flow cytometry. Results DF-1 provided modest radioprotection of human cancer cell lines and fibroblast cell lines when delivered prior to irradiation (dose modifying factor or 1.1). There was no evidence of selective protection of fibroblasts versus tumor cells. Cells treated with DF-1 at radioprotective doses were found to have fewer γH2AX foci at 1 and 6 hours after irradiation compared to vehicle treated controls. The LD50/30 for C57Bl6/Ncr mice treated with a single 300 mg/kg dose of DF-1 pre-irradiation was 10.09 Gy (95% CI 9.58-10.26) versus 8.29 Gy (95% CI, 8.21-8.32) for control mice. No protective effects were seen with a single 200 mg/kg dose. No increase in pimonidazole uptake was appreciated in bone marrow of mice treated with DF-1 compared to vehicle controls. Conclusions DF-1 has modest activity as a radiation protector in vivo. There was no evidence of selective protection from irradiation of normal versus tumor cells with DF-1.
Collapse
Affiliation(s)
- Aaron P Brown
- Radiation Oncology Branch, National Cancer Institute, Building 10 CRC/B2-3500, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Dziegielewski J, Goetz W, Murley JS, Grdina DJ, Morgan WF, Baulch JE. Amifostine metabolite WR-1065 disrupts homologous recombination in mammalian cells. Radiat Res 2010; 173:175-83. [PMID: 20095849 DOI: 10.1667/rr1982.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Repair of DNA damage through homologous recombination (HR) pathways plays a crucial role in maintaining genome stability. However, overstimulation of HR pathways in response to genotoxic stress may abnormally elevate recombination frequencies, leading to increased mutation rates and delayed genomic instability. Radiation-induced genomic instability has been detected after exposure to both low- and high-linear energy transfer (LET) radiations, but the mechanisms responsible for initiating or propagating genomic instability are not known. We have demonstrated that WR-1065, the active metabolite of amifostine, protects against radiation-induced cell killing and delayed genomic instability. We hypothesize that hyperstimulation of HR pathways plays a mechanistic role in radiation-induced genomic instability and that, in part, WR-1065 exerts it radioprotective effect through suppression of the HR pathway. Results of this study demonstrate that WR-1065 treatment selectively protected against radiation-induced cell killing in HR-proficient cell lines compared to an HR-deficient cell line. Further, WR-1065 treatment decreases HR in response to DNA damage using two different mammalian cell systems. This suppression of hyper-recombination is a previously unrecognized mechanism by which WR-1065 effects radioprotection in mammalian cells.
Collapse
Affiliation(s)
- Jaroslaw Dziegielewski
- Department of Radiation Oncology, Radiation Oncology Research Laboratory, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | |
Collapse
|
31
|
Pardo M, Tirosh O. Protective signalling effect of manganese superoxide dismutase in hypoxia-reoxygenation of hepatocytes. Free Radic Res 2010; 43:1225-39. [PMID: 19905985 DOI: 10.3109/10715760903271256] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
This study investigates the mechanism by which MnSOD exerts its protective effect in hypoxia-reoxygenation (H/R) injury in hepatocytes. Following induction of H/R, MnSOD expression and activity levels increased and remained high for over 24 h. Hepatocytes silenced for MnSOD (siMnSOD) demonstrated increased susceptibility to H/R-induced apoptotic cell death and a lower capacity to generate mitochondrial reactive oxygen species. Microarray and real time PCR analysis of gene expression from siMnSOD cells revealed a number of down-regulated protective genes, including hemeoxygenase-1, glutamate-cysteine ligase and Nrf2, a master regulator of cellular adaptation to stress. Decreased Nrf2 protein expression and nuclear translocation were also confirmed in siMnSOD cells. siMnSOD cells showed low glutathione (GSH) content with no oxidation to GSSG, lower lipid peroxidation levels than their controls and lower mitochondrial membrane potential, which all were even more salient after H/R. Therefore, MnSOD appears to act as a signalling mediator for the activation of survival genes following H/R injury in hepatocytes.
Collapse
Affiliation(s)
- Michal Pardo
- Department of Biochemistry and Nutrition, Institute of Biochemistry, Food Science and Nutrition, Robert H Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel.
| | | |
Collapse
|
32
|
Grdina DJ, Murley JS, Kataoka Y, Baker KL, Kunnavakkam R, Coleman MC, Spitz DR. Amifostine induces antioxidant enzymatic activities in normal tissues and a transplantable tumor that can affect radiation response. Int J Radiat Oncol Biol Phys 2009; 73:886-96. [PMID: 19215822 DOI: 10.1016/j.ijrobp.2008.10.061] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2008] [Revised: 10/03/2008] [Accepted: 10/08/2008] [Indexed: 01/24/2023]
Abstract
PURPOSE To determine whether amifostine can induce elevated manganese superoxide dismutase (SOD2) in murine tissues and a transplantable SA-NH tumor, resulting in a delayed tumor cell radioprotective effect. METHODS AND MATERIALS SA-NH tumor-bearing C3H mice were treated with a single 400 mg/kg or three daily 50 mg/kg doses of amifostine administered intraperitoneally. At selected time intervals after the last injection, the heart, liver, lung, pancreas, small intestine, spleen, and SA-NH tumor were removed and analyzed for SOD2, catalase, and glutathione peroxidase (GPx) enzymatic activity. The effect of elevated SOD2 enzymatic activity on the radiation response of SA-NH cells was determined. RESULTS SOD2 activity was significantly elevated in selected tissues and a tumor 24 h after amifostine treatment. Catalase and GPx activities remained unchanged except for significant elevations in the spleen. GPx was also elevated in the pancreas. SA-NH tumor cells exhibited a twofold elevation in SOD2 activity and a 27% elevation in radiation resistance. Amifostine administered in three daily fractions of 50 mg/kg each also resulted in significant elevations of these antioxidant enzymes. CONCLUSIONS Amifostine can induce a delayed radioprotective effect that correlates with elevated levels of SOD2 activity in SA-NH tumor. If limited to normal tissues, this delayed radioprotective effect offers an additional potential for overall radiation protection. However, amifostine-induced elevation of SOD2 activity in tumors could have an unanticipated deleterious effect on tumor responses to fractionated radiation therapy, given that the radioprotector is administered daily just before each 2-Gy fractionated dose.
Collapse
Affiliation(s)
- David J Grdina
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL 60637, USA.
| | | | | | | | | | | | | |
Collapse
|
33
|
Dziegielewski J, Baulch JE, Goetz W, Coleman MC, Spitz DR, Murley JS, Grdina DJ, Morgan WF. WR-1065, the active metabolite of amifostine, mitigates radiation-induced delayed genomic instability. Free Radic Biol Med 2008; 45:1674-81. [PMID: 18845240 PMCID: PMC2629584 DOI: 10.1016/j.freeradbiomed.2008.09.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2008] [Revised: 08/14/2008] [Accepted: 09/02/2008] [Indexed: 11/24/2022]
Abstract
Compounds that can protect cells from the effects of radiation are important for clinical use, in the event of an accidental or terrorist-generated radiation event, and for astronauts traveling in space. One of the major concerns regarding the use of radio-protective agents is that they may protect cells initially, but predispose surviving cells to increased genomic instability later. In this study we used WR-1065, the active metabolite of amifostine, to determine how protection from direct effects of high- and low-LET radiation exposure influences genomic stability. When added 30 min before irradiation and in high concentrations, WR-1065 protected cells from immediate radiation-induced effects as well as from delayed genomic instability. Lower, nontoxic concentrations of WR-1065 did not protect cells from death; however, it was effective in significantly decreasing delayed genomic instability in the progeny of irradiated cells. The observed increase in manganese superoxide dismutase protein levels and activity may provide an explanation for this effect. These results confirm that WR-1065 is protective against both low- and high-LET radiation-induced genomic instability in surviving cells.
Collapse
Affiliation(s)
- Jaroslaw Dziegielewski
- Department of Radiation Oncology, Radiation Oncology Research Laboratory, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Paunesku D, Paunesku T, Wahl A, Kataoka Y, Murley J, Grdina DJ, Woloschak GE. Incidence of tissue toxicities in gamma ray and fission neutron-exposed mice treated with Amifostine. Int J Radiat Biol 2008; 84:623-34. [PMID: 18661379 DOI: 10.1080/09553000802241762] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
PURPOSE To determine the effects of Amifostine or WR-151,327 on the incidence of lethal and non-lethal toxicities in a large cohort of mice exposed to gamma-ray or fission-spectrum neutron radiation. METHODS To analyze data from 4000 B6CF1 mice which received a single whole body irradiation (WBI) with 206 cGy or 417 cGy cobalt-60 gamma rays or 10 cGy or 40 cGy of fission-spectrum neutrons (average energy 0.85 MeV) produced by the Janus reactor at Argonne National Laboratory. In the neutron cohort, Amifostine, WR-151,327, saline or nothing was injected once, intraperitoneally, 30 minutes before irradiation. In the cobalt-60 cohort, WR-151327 was omitted from the same protocol. At the time of natural death, tissue toxicities found in these mice were recorded, and these records were analyzed. While all previous studies focused on the modulation of life shortening effects of WBI by Amifostine, in this study we calculated changes in the frequencies of 59 tissue toxicities and changes in the total number of toxicities per animal. RESULTS Amifostine protected against specific non-tumor pathological complications (67% of the non-tumor toxicities induced by gamma irradiation, 31% of the neutron induced specific toxicities), as well as specific tumors (56% of the tumor toxicities induced by gamma irradiation, 25% of the neutron induced tumors). Amifostine also reduced the total number of toxicities per animal for both genders in the gamma ray exposed mice and in males in the neutron exposed mice. CONCLUSIONS Amifostine was protective against many, but not all, tissue toxicities caused by WBI gamma and neutron irradiation.
Collapse
Affiliation(s)
- David Paunesku
- Department of Radiation Oncology, University of Chicago, Chicago, Illinois, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
Murley JS, Nantajit D, Baker KL, Kataoka Y, Li JJ, Grdina DJ. Maintenance of manganese superoxide dismutase (SOD2)-mediated delayed radioprotection induced by repeated administration of the free thiol form of amifostine. Radiat Res 2008; 169:495-505. [PMID: 18439041 DOI: 10.1667/rr1194.1] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2007] [Accepted: 01/30/2008] [Indexed: 01/21/2023]
Abstract
Thiol-containing drugs such as WR1065, the free thiol form of amifostine, have been shown to induce a delayed radioprotective effect in both malignant and non-malignant cells. In mammalian cells exposed to a dose as low as 40 microM WR1065, the redox-sensitive nuclear transcription factor kappaB (NFkappaB) is activated, leading to an elevation in the expression of the antioxidant gene manganese superoxide dismutase (SOD2) and a concomitant increase in active SOD2 enzyme levels that peaks 24 to 32 h later. Exposure of cells to ionizing radiation during the period of elevated SOD2 enzymatic activity results in an enhanced radiation resistance. This is seen as an increase in surviving fraction as determined by standard colony formation assays. To determine whether this delayed radioprotection can be maintained over a prolonged period in cells of either malignant or non-malignant origin, both human microvascular endothelial cells (HMEC) and SA-NH mouse sarcoma cells were grown to confluence and exposed to 40 muM WR1065 using three administration protocols: (1) daily drug exposure for 10 days followed each day by irradiation with 2 Gy; (2) drug exposure once every 48 h followed by irradiation with 2 Gy 48 h later for 14 days; and (3) drug exposure every 72 h followed by irradiation with 2 Gy 72 h later for 12 days. As a function of each experimental condition, cell numbers and associated SOD2 enzymatic activities were measured at the time of each irradiation. None of the treatment conditions were toxic to either HMEC or SA-NH cells. SOD2 activity was elevated 5.3- and 1.8-fold over background on average for HMEC exposed to 40 microM WR1065 every 24 or 48 h, respectively. Likewise, SOD2 activity was elevated in SA-NH mouse sarcoma cells 7.8- and 4.9-fold after daily exposure to WR1065 or exposure to WR1065 once every 48 h, respectively. Both HMEC and SA-NH cells exhibited enhanced radiation resistance that correlated with the increase in SOD2 activity. The average respective increases in cell survival were 1.33 +/- 0.01 (SEM), 1.23 +/- 0.01 and 1.04 +/- 0.01 for HMEC exposed to WR1065 every 24, 48 and 72 h, respectively, and 1.27 +/- 0.01, 1.18 +/- 0.02 and 1.02 +/- 0.02 for SA-NH cells exposed to WR1065 every 24, 48 and 72 h, respectively. Both the elevation in WR1065-induced SOD2 enzymatic activity and the corresponding increase in radiation resistance were completely inhibited in HMEC and SA-NH cells transfected with human or mouse SOD2 siRNA oligomers and irradiated 24 h later. These data demonstrate that a delayed radioprotective effect can be induced and maintained over a prolonged period in both non-malignant and malignant cells exposed to thiol-containing drugs such as WR1065. For non-malignant cells this represents a novel paradigm for radiation protection. The ability of WR1065 to induce a persistent elevated radiation resistance in malignant cells, however, suggests a new potential concern regarding the issue of tumor protection in patients exposed to thiol-containing drugs.
Collapse
Affiliation(s)
- Jeffrey S Murley
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL 60637, USA
| | | | | | | | | | | |
Collapse
|