1
|
Singh VK, Srivastava M, Seed TM. Protein biomarkers for radiation injury and testing of medical countermeasure efficacy: promises, pitfalls, and future directions. Expert Rev Proteomics 2023; 20:221-246. [PMID: 37752078 DOI: 10.1080/14789450.2023.2263652] [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: 05/23/2023] [Accepted: 09/11/2023] [Indexed: 09/28/2023]
Abstract
INTRODUCTION Radiological/nuclear accidents, hostile military activity, or terrorist strikes have the potential to expose a large number of civilians and military personnel to high doses of radiation resulting in the development of acute radiation syndrome and delayed effects of exposure. Thus, there is an urgent need for sensitive and specific assays to assess the levels of radiation exposure to individuals. Such radiation exposures are expected to alter primary cellular proteomic processes, resulting in multifaceted biological responses. AREAS COVERED This article covers the application of proteomics, a promising and fast developing technology based on quantitative and qualitative measurements of protein molecules for possible rapid measurement of radiation exposure levels. Recent advancements in high-resolution chromatography, mass spectrometry, high-throughput, and bioinformatics have resulted in comprehensive (relative quantitation) and precise (absolute quantitation) approaches for the discovery and accuracy of key protein biomarkers of radiation exposure. Such proteome biomarkers might prove useful for assessing radiation exposure levels as well as for extrapolating the pharmaceutical dose of countermeasures for humans based on efficacy data generated using animal models. EXPERT OPINION The field of proteomics promises to be a valuable asset in evaluating levels of radiation exposure and characterizing radiation injury biomarkers.
Collapse
Affiliation(s)
- Vijay K Singh
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Meera Srivastava
- Department of Anatomy, Physiology and Genetics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | | |
Collapse
|
2
|
Chen Y, Yang Y, Tang H, Zhang Z, Zhou X, Xu W. ROS-Responsive and pH-Sensitive Aminothiols Dual-Prodrug for Radiation Enteritis. Antioxidants (Basel) 2022; 11:antiox11112145. [PMID: 36358517 PMCID: PMC9686648 DOI: 10.3390/antiox11112145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/20/2022] [Accepted: 10/20/2022] [Indexed: 11/30/2022] Open
Abstract
Radiation exposure can immediately trigger a burst of reactive oxygen species (ROS), which can induce severe cell death and long-term tissue damage. Therefore, instantaneous release of sufficient radioprotective drugs is vital to neutralize those accumulated ROS in IR-exposed areas. To achieve this goal, we designed, synthesized, and evaluated a novel oral ROS-responsive radioprotective compound (M1) with high biocompatibility and efficient ROS-scavenging ability to act as a promising oral drug for radiation protection. The compound is stably present in acidic environments and is hydrolyzed in the intestine to form active molecules rich in thiols. M1 can significantly remove cellular ROS and reduce DNA damage induced by γ-ray radiation. An in vivo experiment showed that oral administration of M1 effectively alleviates acute radiation-induced intestinal injury. Immunohistochemical staining showed that M1 improved cell proliferation, reduced cell apoptosis, and enhanced the epithelial integrity of intestinal crypts. This study provides a promising oral ROS-sensitive agent for acute intestinal radiation syndrome.
Collapse
|
3
|
Checker R, Patwardhan RS, Jayakumar S, Maurya DK, Bandekar M, Sharma D, Sandur SK. Chemical and biological basis for development of novel radioprotective drugs for cancer therapy. Free Radic Res 2021; 55:595-625. [PMID: 34181503 DOI: 10.1080/10715762.2021.1876854] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Ionizing radiation (IR) causes chemical changes in biological systems through direct interaction with the macromolecules or by causing radiolysis of water. This property of IR is harnessed in the clinic for radiotherapy in almost 50% of cancers patients. Despite the advent of stereotactic radiotherapy instruments and other advancements in shielding techniques, the inadvertent deposition of radiation dose in the surrounding normal tissue can cause late effects of radiation injury in normal tissues. Radioprotectors, which are chemical or biological agents, can reduce or mitigate these toxic side-effects of radiotherapy in cancer patients and also during radiation accidents. The desired characteristics of an ideal radioprotector include low chemical toxicity, high risk to benefit ratio and specific protection of normal cells against the harmful effects of radiation without compromising the cytotoxic effects of IR on cancer cells. Since reactive oxygen species (ROS) are the major contributors of IR mediated toxicity, plethora of studies have highlighted the potential role of antioxidants to protect against IR induced damage. However, owing to the lack of any clinically approved radioprotector against whole body radiation, researchers have shifted the focus toward finding alternate targets that could be exploited for the development of novel agents. The present review provides a comprehensive insight in to the different strategies, encompassing prime molecular targets, which have been employed to develop radiation protectors/countermeasures. It is anticipated that understanding such factors will lead to the development of novel strategies for increasing the outcome of radiotherapy by minimizing normal tissue toxicity.
Collapse
Affiliation(s)
- Rahul Checker
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India.,Homi Bhabha National Institute, Anushaktinagar, Mumbai, India
| | - Raghavendra S Patwardhan
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India.,Homi Bhabha National Institute, Anushaktinagar, Mumbai, India
| | - Sundarraj Jayakumar
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India
| | - Dharmendra Kumar Maurya
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India.,Homi Bhabha National Institute, Anushaktinagar, Mumbai, India
| | - Mayuri Bandekar
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India
| | - Deepak Sharma
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India.,Homi Bhabha National Institute, Anushaktinagar, Mumbai, India
| | - Santosh K Sandur
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India.,Homi Bhabha National Institute, Anushaktinagar, Mumbai, India
| |
Collapse
|
4
|
Patterson AM, Wu T, Chua HL, Sampson CH, Fisher A, Singh P, Guise TA, Feng H, Muldoon J, Wright L, Plett PA, Pelus LM, Orschell CM. Optimizing and Profiling Prostaglandin E2 as a Medical Countermeasure for the Hematopoietic Acute Radiation Syndrome. Radiat Res 2021; 195:115-127. [PMID: 33302300 DOI: 10.1667/rade-20-00181.1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/04/2020] [Indexed: 12/18/2022]
Abstract
Identification of medical countermeasures (MCM) to mitigate radiation damage and/or protect first responders is a compelling unmet medical need. The prostaglandin E2 (PGE2) analog, 16,16 dimethyl-PGE2 (dmPGE2), has shown efficacy as a radioprotectant and radiomitigator that can enhance hematopoiesis and ameliorate intestinal mucosal cell damage. In this study, we optimized the time of administration of dmPGE2 for protection and mitigation against mortality from the hematopoietic acute radiation syndrome (H-ARS) in young adult mice, evaluated its activity in pediatric and geriatric populations, and investigated potential mechanisms of action. Windows of 30-day survival efficacy for single administration of dmPGE2 were defined as within 3 h prior to and 6-30 h after total-body γ irradiation (TBI). Radioprotective and radio-mitigating efficacy was also observed in 2-year-old geriatric mice and 6-week-old pediatric mice. PGE2 receptor agonist studies suggest that signaling through EP4 is primarily responsible for the radioprotective effects. DmPGE2 administration prior to TBI attenuated the drop in red blood cells and platelets, accelerated recovery of all peripheral blood cell types, and resulted in higher hematopoietic and mesenchymal stem cells in survivor bone marrow. Multiplex analysis of bone marrow cytokines together with RNA sequencing of hematopoietic stem cells indicated a pro-hematopoiesis cytokine milieu induced by dmPGE2, with IL-6 and G-CSF strongly implicated in dmPGE2-mediated radioprotective activity. In summary, we have identified windows of administration for significant radio-mitigation and radioprotection by dmPGE2 in H-ARS, demonstrated survival efficacy in special populations, and gained insight into radioprotective mechanisms, information useful towards development of dmPGE2 as a MCM for first responders, military personnel, and civilians facing radiation threats.
Collapse
Affiliation(s)
- Andrea M Patterson
- Department of a Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Tong Wu
- Department of a Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Hui Lin Chua
- Department of a Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Carol H Sampson
- Department of a Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Alexa Fisher
- Department of a Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Pratibha Singh
- Department of a Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, Indiana 46202.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Theresa A Guise
- Department of Medicine, Division of Endocrinology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Hailin Feng
- Department of a Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Jessica Muldoon
- Department of a Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Laura Wright
- Department of Medicine, Division of Endocrinology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - P Artur Plett
- Department of a Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Louis M Pelus
- Department of a Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, Indiana 46202.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Christie M Orschell
- Department of a Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| |
Collapse
|
5
|
Yashavarddhan MH, Sharma AK, Chaudhary P, Bajaj S, Singh S, Shukla SK. Development of hematopoietic syndrome mice model for localized radiation exposure. Sci Rep 2021; 11:89. [PMID: 33420217 PMCID: PMC7794306 DOI: 10.1038/s41598-020-80075-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 12/03/2020] [Indexed: 11/09/2022] Open
Abstract
Current models to study the hematopoietic syndrome largely rely on the uniform whole-body exposures. However, in the radio-nuclear accidents or terrorist events, exposure can be non-uniform. The data available on the non-uniform exposures is limited. Thus, we have developed a mice model for studying the hematopoietic syndrome in the non-uniform or partial body exposure scenarios using the localized cobalt60 gamma radiation exposure. Femur region of Strain 'A' male mice was exposed to doses ranging from 7 to 20 Gy. The 30 day survival assay showed 19 Gy as LD100 and 17 Gy as LD50. We measured an array of cytokines and important stem cell markers such as IFN-γ, IL-3, IL-6, GM-CSF, TNF-α, G-CSF, IL-1α, IL-1β, CD 34 and Sca 1. We found significant changes in IL-6, GM-CSF, TNF-α, G-CSF, and IL-1β levels compared to untreated groups and amplified levels of CD 34 and Sca 1 positive population in the irradiated mice compared to the untreated controls. Overall, we have developed a mouse model of the hematopoietic acute radiation syndrome that might be useful for understanding of the non-uniform body exposure scenarios. This may also be helpful in the screening of drugs intended for individuals suffering from radiation induced hematopoietic syndrome.
Collapse
Affiliation(s)
- M H Yashavarddhan
- National Institute of Cancer Prevention & Research, Indian Council of Medical Research, Sector-39, Noida, Uttar Pradesh, 201301, India
| | - Ajay Kumar Sharma
- Institute of Nuclear Medicine and Allied Sciences, Defence Research and Development Organisation, Brig. S K Mazumdar Marg, Timarpur, Delhi, 110054, India
| | - Pankaj Chaudhary
- The Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Sania Bajaj
- Institute of Nuclear Medicine and Allied Sciences, Defence Research and Development Organisation, Brig. S K Mazumdar Marg, Timarpur, Delhi, 110054, India
| | - Sukhvir Singh
- Institute of Nuclear Medicine and Allied Sciences, Defence Research and Development Organisation, Brig. S K Mazumdar Marg, Timarpur, Delhi, 110054, India
| | - Sandeep Kumar Shukla
- Institute of Nuclear Medicine and Allied Sciences, Defence Research and Development Organisation, Brig. S K Mazumdar Marg, Timarpur, Delhi, 110054, India.
| |
Collapse
|
6
|
Ritter GS, Nikolin VP, Popova NA, Proskurina AS, Kisaretova PE, Taranov OS, Dubatolova TD, Dolgova EV, Potter EA, Kirikovich SS, Efremov YR, Bayborodin SI, Romanenko MV, Meschaninova MI, Venyaminova AG, Kolchanov NA, Shurdov MA, Bogachev SS. Characterization of biological peculiarities of the radioprotective activity of double-stranded RNA isolated from Saccharomyces сerevisiae. Int J Radiat Biol 2020; 96:1173-1191. [PMID: 32658564 DOI: 10.1080/09553002.2020.1793020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
THE PURPOSE OF THE ARTICLE Protection from ionizing radiation is the most important component in the curing malignant neoplasms, servicing atomic reactors, and resolving the situations associated with uncontrolled radioactive pollutions. In this regard, discovering new effective radioprotectors as well as novel principles of protecting living organisms from high-dose radiation is the most important factor, determining the new approaches in medical and technical usage of radiation. MATERIALS AND METHODS Experimental animals were irradiated on the γ-emitter (Cs137) with a dose of 9.4 Gy. Radioprotective properties of several agents (total RNA, single-stranded RNA, double-stranded RNA and B-190) were estimated by the survival/death rates of experimental animals within 30-90 d. Pathomorphological examination of internal organs end electron microscope assay was done on days 9-12 after irradiation. Cloning and other molecular procedures were performed accordingly to commonly accepted protocols. For assessment of the internalization of labeled nucleic acid, bone marrow cells were incubated with double-stranded RNA labeled with 6-FAM fluorescent dye. Cells with internalized double-stranded RNA were assayed using Axio Imager M1 microscope. In the other experiment, bone marrow cells after incubation with double-stranded RNA were stained with Cy5-labeled anti-CD34 antibodies and assayed using Axioskop 2 microscope. RESULTS In this study, several biological features of the radioprotective action of double-stranded RNA are characterized. It was shown that 160 µg of the double-stranded RNA per mouse protect experimental animals from the absolutely lethal dose of γ-radiation of 9.4 Gy. In different experiments, 80-100% of irradiated animals survive and live until their natural death. Radioprotective properties of double-stranded RNA were found to be independent on its sequence, but strictly dependent on its double-stranded form. Moreover, double-stranded RNA must have 'open' ends of the molecule to exert its radioprotective activity. CONCLUSIONS Experiments indicate that radioprotective effect of double-stranded RNA is tightly bound to its internalization into hematopoietic stem cells, which further repopulate the spleen parenchyma of irradiated mice. Actively proliferating progenitors form the splenic colonies, which further serve as the basis for restoration of hematopoiesis and immune function and determine the survival of animals received the lethal dose of radiation.
Collapse
Affiliation(s)
- Genrikh S Ritter
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia.,Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| | - Valeriy P Nikolin
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Nelly A Popova
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia.,Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| | - Anastasia S Proskurina
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Polina E Kisaretova
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Oleg S Taranov
- State Research Center of Virology and Biotechnology "Vector", Koltsovo, Russia
| | - Tatiana D Dubatolova
- Institute of Molecular and Cellular Biology, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Evgenia V Dolgova
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Ekaterina A Potter
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Svetlana S Kirikovich
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Yaroslav R Efremov
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia.,Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| | - Sergey I Bayborodin
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia.,Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| | | | - Maria I Meschaninova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Aliya G Venyaminova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Nikolay A Kolchanov
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | | | - Sergey S Bogachev
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
7
|
Dondossola E, Casarin S, Paindelli C, De-Juan-Pardo EM, Hutmacher DW, Logothetis CJ, Friedl P. Radium 223-Mediated Zonal Cytotoxicity of Prostate Cancer in Bone. J Natl Cancer Inst 2020; 111:1042-1050. [PMID: 30657953 DOI: 10.1093/jnci/djz007] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 11/28/2018] [Accepted: 01/09/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Bone-targeting radiotherapy with Radium-223 (Rad-223), a radioisotope emitting genotoxic alpha-radiation with limited tissue penetrance (∼100 µm), prolongs the survival of patients with metastatic prostate cancer (PCa). Confoundingly, the clinical response to Rad-223 is often followed by detrimental relapse and progression, and whether Rad-223 causes tumor-cell directed cytotoxicity in vivo remains unclear. We hypothesized that limited radiation penetrance in situ defines outcome. METHODS We tested Rad-223 overall response by PC3 and C4-2B human PCa cell lines in mouse bones (n = 5-18 tibiae per group). Rad-223 efficacy at subcellular resolution was determined by intravital microscopy analysis of dual-color fluorescent PC3 cells (n = 3-4 mice per group) in tissue-engineered bone constructs. In vivo data were fed into an in silico model to predict Rad-223 effectiveness in lesions of different sizes (1-27, 306 initial cells; n = 10-100 simulations) and the predictions validated in vivo by treating PCa tumors of varying sizes in bones (n = 10-14 tibiae per group). Statistical tests were performed by two-sided Student t test or by one-way ANOVA followed by Tukey's post-hoc test. RESULTS Rad-223 (385 kBq/kg) delayed the growth (means [SD]; comparison with control-treated mice) of PC3 (6.7 × 105[4.2 × 105] vs 2.8 × 106 [2.2 × 106], P = .01) and C4-2B tumors in bone (7.7 × 105 [4.0 × 105] vs 3.5 × 106 [1.3 × 106], P < .001). Cancer cell lethality in response to Rad-223 (385 kBq/kg) was profound but zonally confined along the bone interface compared with the more distant tumor core, which remained unperturbed (day 4; 13.1 [2.3%] apoptotic cells, 0-100 µm distance from bone vs 3.6 [0.2%], >300 µm distance; P = .01).In silico simulations predicted greater efficacy of Rad-223 on single-cell lesions (eradication rate: 88.0%) and minimal effects on larger tumors (no eradication, 16.2% growth reduction in tumors of 27 306 cells), as further confirmed in vivo for PC3 and C4-2B tumors. CONCLUSIONS Micro-tumors showed severe growth delay or eradication in response to Rad-223, whereas macro-tumors persisted and expanded. The relative inefficacy in controlling large tumors points to application of Rad-223 in secondary prevention of early bone-metastatic disease and regimens co-targeting the tumor core.
Collapse
|
8
|
Abstract
The radioprotective effects of naturally occurring sulfur compounds and isothiocyanates such as diallyl sulfide (DAS), diallyl disulfide (DADS), allyl methyl sulfide (AMS), allyl isothiocyanate (AITC) and phenyl isothiocyanate (PITC) have been investigated in whole body irradiated Swiss albino mice. Administration of these sulfur compounds could reduce the serum content of alkaline phosphatase (ALP), which was elevated after irradiation (23.9 ± 1.82 KA units). The elevated liver content of glutamate pyruvate transaminase (GPT) in control animals (76.2 ± 2.2 U/mL) after irradiation was significantly reduced in DAS (58.93 ± 4 U/mL) and AMS (55.7 ± 2.2 U/mL) treated animals. Elevated levels of lipid peroxides in serum and liver of irradiated control animals were also significantly reduced by treatment with these sulfur compounds. The glutathione (GSH) content in liver and intestinal mucosa was drastically reduced after irradiation. All the sulfur compounds and isothiocyanates could effectively enhance the GSH content of intestinal mucosa and liver. Findings at histopathological analysis of the intestine proved to be correlated with the above results.
Collapse
|
9
|
Feng Y, Xie XY, Yang YQ, Sun YT, Ma WH, Zhou PJ, Li ZY, Liu HQ, Wang YF, Huang YS. Synthesis and evaluation of pyrimidoindole analogs in umbilical cord blood ex vivo expansion. Eur J Med Chem 2019; 174:181-197. [PMID: 31035239 DOI: 10.1016/j.ejmech.2019.04.042] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 04/15/2019] [Accepted: 04/15/2019] [Indexed: 11/28/2022]
Abstract
The scarcity of hematopoietic stem cells (HSCs) significantly hindered their clinical potentials. Umbilical cord blood (UCB) has become the leading source of HSCs for both research and clinical applications. But the low content of HSCs in a single UCB unit limited its use only to pediatric patients. Various cytokines and small molecules have demonstrated strong abilities in promoting HSC ex vivo expansion, of which UM171 is the newest and by far the most potent HSC ex vivo expansion agent. In this study, we synthesized 37 pyrimidoindole analogs and identified 6 compounds to be potent in promoting HSC ex vivo expansion. In particular, analog 11 was found to be the most effective in stimulating ex vivo expansion of UCB CD34+ cells and CD34+CD38- cells. Initial data indicated that compound 11 promoted the absolute number of long term HSCs and inhibited their differentiation. UCB HSCs expanded with 11 retained adequate multi-lineage differentiation capacity. In addition, compound 11 is not cytotoxic at its test concentrations, suggesting that it merits further investigation for potential clinical applications.
Collapse
Affiliation(s)
- Yue Feng
- School of Pharmacy, Guangdong Medical University, 1 Xincheng Ave, Songshan Lake Technology Park, Dongguan, Guangdong 523808, China; Institute of Biomedicine, College of Life Science & Technology, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Xiao-Yang Xie
- School of Pharmacy, Guangdong Medical University, 1 Xincheng Ave, Songshan Lake Technology Park, Dongguan, Guangdong 523808, China
| | - Yi-Qiu Yang
- Department of Pharmacy, Zhaoqing Medical College, 6 Xijiang South Rd, Zhaoqing, Guangdong, 526020, China
| | - Yu-Tong Sun
- School of Pharmacy, Guangdong Medical University, 1 Xincheng Ave, Songshan Lake Technology Park, Dongguan, Guangdong 523808, China
| | - Wen-Hui Ma
- School of Pharmacy, Guangdong Medical University, 1 Xincheng Ave, Songshan Lake Technology Park, Dongguan, Guangdong 523808, China
| | - Peng-Jun Zhou
- Institute of Biomedicine, College of Life Science & Technology, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Zi-Yao Li
- Institute of Biomedicine, College of Life Science & Technology, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Hui-Qiang Liu
- Institute of Biomedicine, College of Life Science & Technology, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Yi-Fei Wang
- Institute of Biomedicine, College of Life Science & Technology, Jinan University, Guangzhou, Guangdong, 510632, China.
| | - Yun-Sheng Huang
- School of Pharmacy, Guangdong Medical University, 1 Xincheng Ave, Songshan Lake Technology Park, Dongguan, Guangdong 523808, China.
| |
Collapse
|
10
|
Mishra K, Alsbeih G. Appraisal of biochemical classes of radioprotectors: evidence, current status and guidelines for future development. 3 Biotech 2017; 7:292. [PMID: 28868219 DOI: 10.1007/s13205-017-0925-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 08/21/2017] [Indexed: 12/13/2022] Open
Abstract
The search for efficient radioprotective agents to protect from radiation-induced toxicity, due to planned or accidental radiation exposure, is still ongoing worldwide. Despite decades of research and development of widely different biochemical classes of natural and derivative compounds, a safe and effective radioprotector is largely unmet. In this comprehensive review, we evaluated the evidence for the radioprotective performance of classical thiols, vitamins, minerals, dietary antioxidants, phytochemicals, botanical and bacterial preparations, DNA-binding agents, cytokines, and chelators including adaptogens. Where radioprotection was demonstrated, the compounds have shown moderate dose modifying factors ranging from 1.1 to 2.7. To date, only few compounds found way to clinic with limited margin of dose prescription due to side effects. Most of these compounds (amifostine, filgratism, pegfilgrastim, sargramostim, palifermin, recombinant salmonella flagellin, Prussian blue, potassium iodide) act primarily via scavenging of free radicals, modulation of oxidative stress, signal transduction, cell proliferation or enhance radionuclide elimination. However, the gain in radioprotection remains hampered with low margin of tolerance. Future development of more effective radioprotectors requires an appropriate nontoxic compound, a model system and biomarkers of radiation exposure. These are important to test the effectiveness of radioprotection on physiological tissues during radiotherapy and field application in cases of nuclear eventualities.
Collapse
Affiliation(s)
- Krishnanand Mishra
- Radiation Biology Section, Biomedical Physics Department, King Faisal Specialist Hospital and Research Centre (KFSH&RC), Riyadh, Saudi Arabia
| | - Ghazi Alsbeih
- Radiation Biology Section, Biomedical Physics Department, King Faisal Specialist Hospital and Research Centre (KFSH&RC), Riyadh, Saudi Arabia
| |
Collapse
|
11
|
Yun KL, Wang ZY. Target/signalling pathways of natural plant-derived radioprotective agents from treatment to potential candidates: A reverse thought on anti-tumour drugs. Biomed Pharmacother 2017; 91:1122-1151. [DOI: 10.1016/j.biopha.2017.05.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 04/15/2017] [Accepted: 05/01/2017] [Indexed: 02/07/2023] Open
|
12
|
Singh VK, Fatanmi OO, Wise SY, Newman VL, Romaine PLP, Seed TM. THE POTENTIATION OF THE RADIOPROTECTIVE EFFICACY OF TWO MEDICAL COUNTERMEASURES, GAMMA-TOCOTRIENOL AND AMIFOSTINE, BY A COMBINATION PROPHYLACTIC MODALITY. RADIATION PROTECTION DOSIMETRY 2016; 172:302-310. [PMID: 27542813 PMCID: PMC5444681 DOI: 10.1093/rpd/ncw223] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
This study was designed to evaluate the possible potentiation of survival protection afforded by relatively low-dose amifostine prophylaxis against total body irradiation in combination with a protective, less toxic agent, gamma-tocotrienol (GT3). Mice were administered amifostine and/or GT3, then exposed to 9.2 Gy 60Co γ-irradiation and monitored for survival for 30 days. To investigate cytokine stimulation, mice were administered amifostine or GT3; serum samples were collected and analyzed for cytokines. Survival studies show single treatments of GT3 or amifostine significantly improved survival, compared to the vehicle, and combination treatments resulted in significantly higher survival compared to single treatments. In vivo studies with GT3 confirmed prior work indicating GT3 induces granulocyte colony-stimulating factor (G-CSF). This approach, the prophylactic combination of amifostine and GT3, which act through different mechanisms, shows promise and should be investigated further as a potential countermeasure for acute radiation syndrome.
Collapse
Affiliation(s)
- Vijay K Singh
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine 'America's Medical School', Services University of the Health Sciences , Bethesda, MD, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences , Bethesda, MD, USA
| | - Oluseyi O Fatanmi
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine 'America's Medical School', Services University of the Health Sciences , Bethesda, MD, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences , Bethesda, MD, USA
| | - Stephen Y Wise
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine 'America's Medical School', Services University of the Health Sciences , Bethesda, MD, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences , Bethesda, MD, USA
| | - Victoria L Newman
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine 'America's Medical School', Services University of the Health Sciences , Bethesda, MD, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences , Bethesda, MD, USA
| | - Patricia L P Romaine
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine 'America's Medical School', Services University of the Health Sciences , Bethesda, MD, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences , Bethesda, MD, USA
| | | |
Collapse
|
13
|
Radiation protective effects of baclofen predicted by a computational drug repurposing strategy. Pharmacol Res 2016; 113:475-483. [PMID: 27664700 DOI: 10.1016/j.phrs.2016.09.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 09/19/2016] [Accepted: 09/20/2016] [Indexed: 11/21/2022]
Abstract
Exposure to ionizing radiation causes damage to living tissues; however, only a small number of agents have been approved for use in radiation injuries. Radioprotector is the primary countermeasure to radiation injury and none radioprotector has indeed reached the drug development stage. Repurposing the long list of approved, non-radioprotective drugs is an attractive strategy to find new radioprotective agents. Here, we applied a computational approach to discover new radioprotectors in silico by comparing publicly available gene expression data of ionizing radiation-treated samples from the Gene Expression Omnibus (GEO) database with gene expression signatures of more than 1309 small-molecule compounds from the Connectivity Map (cmap) dataset. Among the best compounds predicted to be therapeutic for ionizing radiation damage by this approach were some previously reported radioprotectors and baclofen (P<0.01), a chemical that was not previously used as radioprotector. Validation using a cell-based model and a rodent in vivo model demonstrated that treatment with baclofen reduced radiation-induced cytotoxicity in vitro (P<0.01), attenuated bone marrow damage and increased survival in vivo (P<0.05). These findings suggest that baclofen might serve as a radioprotector. The drug repurposing strategy by connecting the GEO data and cmap can be used to identify known drugs as potential radioprotective agents.
Collapse
|
14
|
Protection against Radiotherapy-Induced Toxicity. Antioxidants (Basel) 2016; 5:antiox5030022. [PMID: 27399787 PMCID: PMC5039571 DOI: 10.3390/antiox5030022] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 06/27/2016] [Accepted: 06/28/2016] [Indexed: 01/18/2023] Open
Abstract
Radiation therapy is a highly utilized therapy in the treatment of malignancies with up to 60% of cancer patients receiving radiation therapy as a part of their treatment regimen. Radiation therapy does, however, cause a wide range of adverse effects that can be severe and cause permanent damage to the patient. In an attempt to minimize these effects, a small number of compounds have been identified and are in use clinically for the prevention and treatment of radiation associated toxicities. Furthermore, there are a number of emerging therapies being developed for use as agents that protect against radiation-induced toxicities. The aim of this review was to evaluate and summarise the evidence that exists for both the known radioprotectant agents and the agents that show promise as future radioprotectant agents.
Collapse
|
15
|
Singh VK, Hauer-Jensen M. γ-Tocotrienol as a Promising Countermeasure for Acute Radiation Syndrome: Current Status. Int J Mol Sci 2016; 17:E663. [PMID: 27153057 PMCID: PMC4881489 DOI: 10.3390/ijms17050663] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 04/03/2016] [Accepted: 04/25/2016] [Indexed: 01/13/2023] Open
Abstract
The hazard of ionizing radiation exposure due to nuclear accidents or terrorist attacks is ever increasing. Despite decades of research, still, there is a shortage of non-toxic, safe and effective medical countermeasures for radiological and nuclear emergency. To date, the U.S. Food and Drug Administration (U.S. FDA) has approved only two growth factors, Neupogen (granulocyte colony-stimulating factor (G-CSF), filgrastim) and Neulasta (PEGylated G-CSF, pegfilgrastim) for the treatment of hematopoietic acute radiation syndrome (H-ARS) following the Animal Efficacy Rule. Promising radioprotective efficacy results of γ-tocotrienol (GT3; a member of the vitamin E family) in the mouse model encouraged its further evaluation in the nonhuman primate (NHP) model. These studies demonstrated that GT3 significantly aided the recovery of radiation-induced neutropenia and thrombocytopenia compared to the vehicle controls; these results particularly significant after exposure to 5.8 or 6.5 Gray (Gy) whole body γ-irradiation. The stimulatory effect of GT3 on neutrophils and thrombocytes (platelets) was directly and positively correlated with dose; a 75 mg/kg dose was more effective compared to 37.5 mg/kg. GT3 was also effective against 6.5 Gy whole body γ-irradiation for improving neutrophils and thrombocytes. Moreover, a single administration of GT3 without any supportive care was equivalent, in terms of improving hematopoietic recovery, to multiple doses of Neupogen and two doses of Neulasta with full supportive care (including blood products) in the NHP model. GT3 may serve as an ultimate radioprotector for use in humans, particularly for military personnel and first responders. In brief, GT3 is a promising radiation countermeasure that ought to be further developed for U.S. FDA approval for the ARS indication.
Collapse
Affiliation(s)
- Vijay K Singh
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA.
| | - Martin Hauer-Jensen
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare Systems, Little Rock, AR 72205, USA.
| |
Collapse
|
16
|
Kumar S, Tiku AB. Immunomodulatory potential of acemannan (polysaccharide fromAloe vera) against radiation induced mortality in Swiss albino mice. FOOD AGR IMMUNOL 2015. [DOI: 10.1080/09540105.2015.1079594] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
17
|
Johnke RM, Sattler JA, Allison RR. Radioprotective agents for radiation therapy: future trends. Future Oncol 2015; 10:2345-57. [PMID: 25525844 DOI: 10.2217/fon.14.175] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Only two radioprotective compounds, amifostine and palifermin, currently have the US FDA approval for use in radiation therapy. However, several agents have been reported that show therapeutic promise. Many of these agents are free radical scavengers/antioxidants. Superoxide dismutase and superoxide dismutase mimetics, nitroxides and dietary antioxidants are all being investigated. Recently, alternative strategies of drug development have been evolving, which focus on targeting the series of cellular insult recognition/repair responses initiated following radiation. These agents, which include cytokines/growth factors, angiotensin-converting enzyme inhibitors and apoptotic modulators, show promise of having significant impact on the mitigation of radiation injury. Herein, we review current literature on the development of radioprotectors with emphasis on compounds with proven or potential usefulness in radiation therapy.
Collapse
Affiliation(s)
- Roberta M Johnke
- Department of Radiation Oncology, East Carolina University Brody School of Medicine, Greenville, NC 27834, USA
| | | | | |
Collapse
|
18
|
Mutations of the human interferon alpha-2b (hIFNα-2b) gene in low-dose natural terrestrial ionizing radiation exposed dwellers. Cytokine 2015; 76:294-302. [PMID: 26092410 DOI: 10.1016/j.cyto.2015.05.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 05/07/2015] [Accepted: 05/18/2015] [Indexed: 02/02/2023]
Abstract
Natural terrestrial ionizing radiations emerge from uranium deposits and can impact human tissues by affecting DNA bases which constitute genes. Human interferon alpha-2b (hIFNα-2b) gene synthesizes a protein which exhibits anticancerous, immunomodulatory, anti-proliferative and antiviral properties. This research aimed to find out hIFNα-2b gene mutations for those residents who were chronically exposed to low-dose natural terrestrial ionizing radiations. The gene amplifications was done through PCR technique and gene mutations were identified by bioinformatics in order to conclude as to how mutations identified in hIFNα-2b gene sequences will lead to alterations in the hIFNα-2b protein in radiation exposed residents. The range of radiation dose exposure was 0.4383-4.55832 (mSv/y) for the selected radiation exposed locations which were having uranium mineralization. Mutations (24%) in hIFNα-2b gene shows that some of the radiation exposed inhabitants were having a modulated immune response. The CBC (Complete Blood Count) parameters: WBC (White Blood Cells), MCH (Mean Corpuscular Hemoglobin), MCHC (MCH Concentration) and PLT (Platelets) on average were below the normal range in 24% radiation exposed subjects who were having hIFNα-2b gene mutations. Immunomodulation is observed by the mixed trend of either lymphocytosis or lymphopenia and neutropenia or neutrophilia in the exposed population. Thus, a radioactive exposure from uranium can affect the immune system and can induce mutations.
Collapse
|
19
|
Hsing CH, Wang JJ. Clinical implication of perioperative inflammatory cytokine alteration. ACTA ACUST UNITED AC 2015; 53:23-8. [PMID: 25837846 DOI: 10.1016/j.aat.2015.03.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 03/06/2015] [Indexed: 12/22/2022]
Abstract
Cytokines are key modulators of inflammatory responses, and play an important role in the defense and repair mechanisms following trauma. After traumatic injury, an immuno-inflammatory response is initiated immediately, and cytokines rapidly appear and function as a regulator of immunity. In pathologic conditions, imbalanced cytokines may provide systemic inflammatory responses or immunosuppression. Expression of perioperative cytokines vary by different intensities of surgical trauma and types of anesthesia and anesthetic agents. Inflammatory cytokines play important roles in postoperative organ dysfunction including central nervous system, cardiovascular, lung, liver, and kidney injury. Inhibition of cytokines could protect against traumatic injury in some circumstances, therefore cytokine inhibitors or antagonists might have the potential for reducing postoperative tissue/organ dysfunction. Cytokines are also involved in wound healing and post-traumatic pain. Application of cytokines for the improvement of surgical wound healing has been reported. Anesthesia-related immune response adjustment might reduce perioperative morbidity because it reduces proinflammatory cytokine expression; however, the overall effects of anesthetics on postoperative immune-inflammatory responses needs to be further investigated.
Collapse
Affiliation(s)
- Chung-Hsi Hsing
- Department of Medical Research, Chi-Mei Medical Center, Tainan, Taiwan; Department of Anesthesiology, Chi-Mei Medical Center, Tainan, Taiwan; Department of Anesthesiology, Taipei Medical University, Taipei, Taiwan.
| | - Jhi-Joung Wang
- Department of Medical Research, Chi-Mei Medical Center, Tainan, Taiwan; Department of Anesthesiology, Chi-Mei Medical Center, Tainan, Taiwan
| |
Collapse
|
20
|
Grdina DJ, Murley JS, Miller RC, Woloschak GE, Li JJ. NFκB and Survivin-Mediated Radio-Adaptive Response. Radiat Res 2015; 183:391-7. [PMID: 25763931 DOI: 10.1667/rr14002.1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
A survivin-mediated radio-adaptive response was induced in SA-NH murine sarcoma cells following activation of nuclear transcription factor κB (NFκB) by very low doses of ionizing radiation of 5, 20 or 100 mGy. SA-NH cells and a clone stably transfected with a plasmid containing a mutated IκBα gene that prevents the activation of NFκB (SA-NH+mIκBα1) were used to investigate the role of NFκB activation in the development and expression of the survivin-mediated radio-adaptive response. Tumor cells were exposed to very low doses of radiation 30 min prior to or at times ranging from 30 min to 6 h after the first of two 2 Gy doses separated by 24 h under in vitro conditions. Evidence of very low dose radiation induced a radio-adaptive response only in SA-NH but not SA-NH+mIκBα1 cells was shown by both an increase in SA-NH cell survival of 20-40% using a standard colony forming assay and reduced apoptosis frequencies of 20-40% as determined by the TUNEL assay. Changes in survivin protein levels as a function of irradiation conditions were monitored by Western blot. A 100 mGy exposure 30 min prior to a 2 Gy dose resulted in an elevation in total survivin protein 24 h later in SA-NH but not SA-NH+mIκBα1 cells. Transfection of cells with survivin siRNA inhibited elevation of survivin protein by very low dose radiation and the subsequent radio-adaptive response in SA-NH cells. These data suggest that the survivin-mediated radio-adaptive response is dependent upon the ability of cells to activate NFκB.
Collapse
Affiliation(s)
- David J Grdina
- a Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, Illinois 60637
| | | | | | | | | |
Collapse
|
21
|
Melanin Nanoparticles (MNPs) provide protection against whole-body ɣ-irradiation in mice via restoration of hematopoietic tissues. Mol Cell Biochem 2014; 399:59-69. [DOI: 10.1007/s11010-014-2232-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 09/27/2014] [Indexed: 12/11/2022]
|
22
|
Liu T, Pei H, Xu D, Zhang Y, Wan J, Wu X, Zhang X, Sun F, He J, Li P, Tian N, Wang J, Chen W, Zhou G. GANRA-5 protects mice from X-ray irradiation-induced dysfunction of the immune system. Free Radic Res 2014; 48:875-82. [DOI: 10.3109/10715762.2014.919389] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
23
|
Mishra S, Malhotra P, Gupta AK, Singh PK, Javed S, Kumar R. A semiquinone glucoside derivative isolated from Bacillus sp. INM-1 provides protection against 5-fluorouracil-induced immunotoxicity. J Immunotoxicol 2014; 12:56-63. [PMID: 24512327 DOI: 10.3109/1547691x.2014.882448] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
5-Fluorouracil (5-FU) is a widely used anti-cancer agent; however, it induces immunosuppression in patients undergoing a chemotherapy regime. The mode of action by which 5-FU induces immunosuppression is primarily via inhibition of hematopoietic growth factors. In the present study, immunoprotective effects of a semiquinone glucoside derivative (SQGD), a bacterial metabolite isolated from Bacillus sp. INM-1, were evaluated in a model of 5-FU-induced immunotoxicity in C57Bl/6 male mice. The evaluation was done by analyzing G-CSF, GM-CSF, and M-CSF expression in the serum, spleen, and bone marrow cells of the mice at different timepoints after 5-FU treatment. Mice received a single intraperitoneal injection of either 5-FU (75 mg/kg) alone, SQGD (50 mg/kg) alone, or SQGD 2 h prior to the 5-FU treatment. Control mice received saline vehicle only. The results demonstrated that 5-FU treatment significantly inhibited G-CSF, GM-CSF, and M-CSF expression in all three sites at all timepoints from 6-72 h post 5-FU. In SQGD treated mice, up-regulation of factor expression was observed in each compartment, and significantly so most often after 12 h. SQGD treatment prior to 5-FU administration to the mice significantly increased in all sites evaluated - relative to values in both control mice and 5-FU only-treated mice - G-CSF, M-CSF, and GM-CSF expression at almost every timepoint. The present findings suggest that SQGD provides protection against 5-FU-induced immunotoxicity in mice and could protect bone marrow progenitor cells against the effects of cytotoxic drugs used for treatment of cancer. The findings also suggested to us that SQGD is a potential immunomodulator and could protect hematopoiesis against toxic assault caused by anti-cancer drugs in the clinical setting.
Collapse
Affiliation(s)
- Saurabh Mishra
- Radiation Biotechnology Laboratory, Department of Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences , Delhi , India and
| | | | | | | | | | | |
Collapse
|
24
|
Grdina DJ, Murley JS, Miller RC, Mauceri HJ, Sutton HG, Li JJ, Woloschak GE, Weichselbaum RR. A survivin-associated adaptive response in radiation therapy. Cancer Res 2013; 73:4418-28. [PMID: 23651635 DOI: 10.1158/0008-5472.can-12-4640] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Adaptive responses can be induced in cells by very low doses of ionizing radiation resulting in an enhanced resistance to much larger exposures. The inhibitor of apoptosis protein, survivin, has been implicated in many adaptive responses to cellular stress. Computerized axial tomography used in image-guided radiotherapy to position and monitor tumor response uses very low radiation doses ranging from 0.5 to 100 mGy. We investigated the ability of these very low radiation doses administered along with two 2 Gy doses separated by 24 hours, a standard conventional radiotherapy dosing schedule, to initiate adaptive responses resulting in the elevation of radiation resistance in exposed cells. Human colon carcinoma (RKO36), mouse sarcoma (SA-NH), along with transformed mouse embryo fibroblasts, wild type or cells lacking functional tumor necrosis factor receptors 1 and 2 were used to assess their relative ability to express an adaptive response when grown either to confluence in vitro or as tumors in the flank of C57BL/6 mice. The survival of each of these cells was elevated from 5% to 20% (P ≤ 0.05) as compared to cells not receiving a 100 mGy or lesser dose. In addition, the cells exposed to 100 mGy exhibited elevations in survivin levels, reductions in apoptosis frequencies, and loss of an adaptive response if transfected with survivin siRNA. This survivin-mediated adaptive response has the potential for affecting outcomes if regularly induced throughout a course of image guided radiation therapy.
Collapse
Affiliation(s)
- David J Grdina
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL 60637, USA.
| | | | | | | | | | | | | | | |
Collapse
|
25
|
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
|
26
|
Structural characterization and radioprotection of bone marrow hematopoiesis of two novel polysaccharides from the root of Angelica sinensis (Oliv.) Diels. Fitoterapia 2012; 83:1712-20. [DOI: 10.1016/j.fitote.2012.09.029] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Revised: 09/18/2012] [Accepted: 09/26/2012] [Indexed: 11/22/2022]
|
27
|
Ajakaiye MA, Jacob A, Wu R, Yang WL, Nicastro J, Coppa GF, Wang P. Recombinant human MFG-E8 attenuates intestinal injury and mortality in severe whole body irradiation in rats. PLoS One 2012; 7:e46540. [PMID: 23056336 PMCID: PMC3466304 DOI: 10.1371/journal.pone.0046540] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 08/31/2012] [Indexed: 11/19/2022] Open
Abstract
The gastrointestinal (GI) syndrome component of acute radiation syndrome (ARS) results from depletion of immature parenchymal stem cells after high dose irradiation and contributes significantly to early mortality. It is associated with severe, irreparable damage in the GI tract and extremely low survival. There is a need for the development of viable mitigators of whole body irradiation (WBI) due to the possibility of unexpected high level radiation exposure from nuclear accidents or attacks. We therefore examined the effect of recombinant human milk fat globule-EGF factor 8 (rhMFG-E8) in mitigating damage after WBI. Male Sprague-Dawley rats were exposed to 10 Gy WBI using Cesium-137 as the radiation source. The animals in the treatment group received rhMFG-E8 (166 µg/kg BW) subcutaneously once a day with the first dose given 6 h after WBI. Blood and tissue samples from the ileum were collected after 3 days of treatment. A separate cohort of animals was treated for 7 days and the 21 day mortality rate was determined. Treatment with rhMFG-E8 significantly improved the survival from 31% to 75% over 21 days. Furthermore, rhMFG-E8 treatment resulted in a 36% reduction in the radiation injury intestinal mucosal damage score, corresponding to visible histological changes. MFG-E8 gene expression was significantly decreased in WBI-induced animals as compared to sham controls. Treatment with rhMFG-E8 increased p53 and p21 expression by 207% and 84% compared to untreated controls. This was accompanied by an 80% increase in the expression of anti-apoptotic cell regulator Bcl-2. p53 and p21 levels correlate with improved survival after radiation injury. These cell regulators arrest the cell after DNA damage and enable DNA repair as well as optimize cell survival. Taken together, these results indicate that rhMFG-E8 ameliorates the GI syndrome and improves survival after WBI by minimizing intestinal cell damage and optimizing recovery.
Collapse
Affiliation(s)
- Michael A. Ajakaiye
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, and Department of Surgery, Hofstra North Shore-LIJ School of Medicine, Manhasset, New York, United States of America
| | - Asha Jacob
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, and Department of Surgery, Hofstra North Shore-LIJ School of Medicine, Manhasset, New York, United States of America
| | - Rongqian Wu
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, and Department of Surgery, Hofstra North Shore-LIJ School of Medicine, Manhasset, New York, United States of America
| | - Weng Lang Yang
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, and Department of Surgery, Hofstra North Shore-LIJ School of Medicine, Manhasset, New York, United States of America
| | - Jeffrey Nicastro
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, and Department of Surgery, Hofstra North Shore-LIJ School of Medicine, Manhasset, New York, United States of America
| | - Gene F. Coppa
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, and Department of Surgery, Hofstra North Shore-LIJ School of Medicine, Manhasset, New York, United States of America
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, and Department of Surgery, Hofstra North Shore-LIJ School of Medicine, Manhasset, New York, United States of America
- * E-mail:
| |
Collapse
|
28
|
Pernot E, Hall J, Baatout S, Benotmane MA, Blanchardon E, Bouffler S, El Saghire H, Gomolka M, Guertler A, Harms-Ringdahl M, Jeggo P, Kreuzer M, Laurier D, Lindholm C, Mkacher R, Quintens R, Rothkamm K, Sabatier L, Tapio S, de Vathaire F, Cardis E. Ionizing radiation biomarkers for potential use in epidemiological studies. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2012; 751:258-286. [DOI: 10.1016/j.mrrev.2012.05.003] [Citation(s) in RCA: 135] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Revised: 05/04/2012] [Accepted: 05/28/2012] [Indexed: 02/07/2023]
|
29
|
Kulkarni SS, Cary LH, Gambles K, Hauer-Jensen M, Kumar KS, Ghosh SP. Gamma-tocotrienol, a radiation prophylaxis agent, induces high levels of granulocyte colony-stimulating factor. Int Immunopharmacol 2012; 14:495-503. [PMID: 23000517 DOI: 10.1016/j.intimp.2012.09.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Revised: 08/07/2012] [Accepted: 09/07/2012] [Indexed: 01/22/2023]
Abstract
Gamma-tocotrienol (GT3), a promising radioprotectant, is shown to protect CD2F1 mice from radiation-induced neutropenia and thrombocytopenia when given 24h prior to total-body irradiation. GT3 also is shown to increase white blood cells (WBC) and absolute neutrophil counts (ANC) transiently in peripheral blood. We hypothesized that increases in WBC and ANC may involve stimulation of hematopoiesis possibly by cytokines and growth factors. To evaluate the effects of GT3 on hematopoietic system, we measured various cytokines, chemokines and growth factors by cytokine array and Bio-Plex assays. Both showed strong induction of various cytokines and chemokines. GT3 treatment resulted in significant increases in G-CSF, IL-1α, IL-1β, IL-6, IL-12p70, IL-17, MIP-1α, and KC levels. G-CSF levels increased markedly within 12-24h after administration (5441 pg/ml in GT3-treated groups compared to 17 pg/ml in vehicle control). Most of these cytokine levels were elevated in the presence or absence of radiation. Time-course analysis of G-CSF and IL-6 induction showed that both cytokines were induced transiently after GT3 administration, and returned to normal levels by 48 h post-administration. For G-CSF, the peak was observed between 12 and 24h post-administration of GT3; however, the highest levels of IL-6 were obtained between 6 and 12h. These results demonstrate that GT3 induced high levels of G-CSF and other inflammatory cytokines and chemokines within 24h after administration. Survival studies reported showed that the most efficacious time for administering GT3 was 24h prior to irradiation, possibly because it induced key hematopoietic cytokines in that time window. These results also suggest a possible role of GT3-induced G-CSF stimulation in protecting mice from radiation-induced neutropenia and thrombocytopenia.
Collapse
Affiliation(s)
- Shilpa S Kulkarni
- Armed Forces Radiobiology Research Institute, USUHS, Bethesda, MD 20889-5603, United States
| | | | | | | | | | | |
Collapse
|
30
|
Zhao H, Wang Z, Ma F, Yang X, Cheng C, Yao L. Protective effect of anthocyanin from Lonicera Caerulea var. Edulis on radiation-induced damage in mice. Int J Mol Sci 2012; 13:11773-11782. [PMID: 23109882 PMCID: PMC3472774 DOI: 10.3390/ijms130911773] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Revised: 08/28/2012] [Accepted: 09/03/2012] [Indexed: 11/26/2022] Open
Abstract
The radioprotective effect of anthocyanin extracted from Lonicera caerulea var. edulis (ALC), was studied in ICR mice. Different doses of ALC were intragastrically administered to mice once a day, prior to radiation. After two weeks, the mice received a one-time 5 Gy whole body (60)Coγ radiation. The spleen index, thymus index, activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px), malondialdehyde (MDA) content, and glutathione (GSH) content in liver tissue were measured. Compared with the radiation control group, the levels of MDA in all ALC treated groups decreased significantly (p < 0.05). Moreover, the GSH content, activities of SOD and GSH-Px in liver tissue were enhanced significantly (p < 0.05) in all ALC groups. These results demonstrate that ALC may be a potential radioprotector, and a further study of the molecular mechanism is needed for further application.
Collapse
Affiliation(s)
- Haitian Zhao
- School of Food Science and Engineering, Harbin Institute of Technology, Harbin 150090, China; E-Mails: (H.Z.); (F.M.); (X.Y.); (C.C.); (L.Y.)
| | - Zhenyu Wang
- School of Food Science and Engineering, Harbin Institute of Technology, Harbin 150090, China; E-Mails: (H.Z.); (F.M.); (X.Y.); (C.C.); (L.Y.)
- School of Forestry, Northeast Forestry University, Harbin 150040, China
| | - Fengming Ma
- School of Food Science and Engineering, Harbin Institute of Technology, Harbin 150090, China; E-Mails: (H.Z.); (F.M.); (X.Y.); (C.C.); (L.Y.)
- School of Forestry, Northeast Forestry University, Harbin 150040, China
| | - Xin Yang
- School of Food Science and Engineering, Harbin Institute of Technology, Harbin 150090, China; E-Mails: (H.Z.); (F.M.); (X.Y.); (C.C.); (L.Y.)
| | - Cuilin Cheng
- School of Food Science and Engineering, Harbin Institute of Technology, Harbin 150090, China; E-Mails: (H.Z.); (F.M.); (X.Y.); (C.C.); (L.Y.)
| | - Lei Yao
- School of Food Science and Engineering, Harbin Institute of Technology, Harbin 150090, China; E-Mails: (H.Z.); (F.M.); (X.Y.); (C.C.); (L.Y.)
- National Research Center of Soybean Engineering and Technology, Northeast Agriculture University, Harbin 150030, China
| |
Collapse
|
31
|
Velho-Pereira R, Kumar A, Pandey BN, Mishra KP, Jagtap AG. Radioprotection by Macerated Extract of Nigella sativa in Normal Tissues of Fibrosarcoma Bearing Mice. Indian J Pharm Sci 2012; 74:403-14. [PMID: 23716868 PMCID: PMC3660866 DOI: 10.4103/0250-474x.108415] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Revised: 09/22/2012] [Accepted: 09/28/2012] [Indexed: 11/04/2022] Open
Abstract
The current study was undertaken to study the effect of a macerated extract of Nigella sativa seeds in normal as well as in tumour bearing mice against gamma radiation-induced cellular damage to normal tissues. This was done to mimic the clinical setting where in, normal tissues of cancer patients undergoing radiotherapy are exposed to the deleterious effects of radiation. The protection of cellular DNA was analysed in peripheral blood leucocytes of whole body irradiated mice following pretreatment with macerated extract of Nigella sativa seeds (100 mg/kg), using alkaline comet assay, and also estimating biochemical and blood parameters such as levels of antioxidant enzymes superoxide dismutase and catalase, thiobarbituric acid reactive substances and protein oxidation in organs such as spleen, liver, brain and intestine haemoglobin and total leucocyte count, respectively. The results showed that the macerated extract of Nigella sativa seeds protected the liver, spleen, brain and intestines both in normal as well as tumour bearing mice. This study concludes that macerated extract of Nigella sativa seeds has protective effects against radiation-induced damage and biochemical alterations which could be attributed to the ability to scavenge free radicals and its antioxidant properties. Hence macerated extract of Nigella sativa seeds, could be used in combination with radiation to protect against oxidative stress in normal tissues and improving the quality of life of cancer patients by mitigating unwanted side effects of radiation in normal tissues.
Collapse
Affiliation(s)
- Reelma Velho-Pereira
- Department of Pharmacology, Bombay College of Pharmacy, Mumbai University, Mumbai-400 098, India
| | - A. Kumar
- Radiation and Cancer Biology Section, Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai-400 085, India
| | - B. N. Pandey
- Radiation and Cancer Biology Section, Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai-400 085, India
| | - K. P. Mishra
- Radiation and Cancer Biology Section, Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai-400 085, India
| | - Aarti G. Jagtap
- Department of Pharmacology, Bombay College of Pharmacy, Mumbai University, Mumbai-400 098, India
| |
Collapse
|
32
|
Krivokrysenko VI, Shakhov AN, Singh VK, Bone F, Kononov Y, Shyshynova I, Cheney A, Maitra RK, Purmal A, Whitnall MH, Gudkov AV, Feinstein E. Identification of granulocyte colony-stimulating factor and interleukin-6 as candidate biomarkers of CBLB502 efficacy as a medical radiation countermeasure. J Pharmacol Exp Ther 2012; 343:497-508. [PMID: 22837010 DOI: 10.1124/jpet.112.196071] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Given an ever-increasing risk of nuclear and radiological emergencies, there is a critical need for development of medical radiation countermeasures (MRCs) that are safe, easily administered, and effective in preventing and/or mitigating the potentially lethal tissue damage caused by acute high-dose radiation exposure. Because the efficacy of MRCs for this indication cannot be ethically tested in humans, development of such drugs is guided by the Food and Drug Administration's Animal Efficacy Rule. According to this rule, human efficacious doses can be projected from experimentally established animal efficacious doses based on the equivalence of the drug's effects on efficacy biomarkers in the respective species. Therefore, identification of efficacy biomarkers is critically important for drug development under the Animal Efficacy Rule. CBLB502 is a truncated derivative of the Salmonella flagellin protein that acts by triggering Toll-like receptor 5 (TLR5) signaling and is currently under development as a MRC. Here, we report identification of two cytokines, granulocyte colony-stimulating factor (G-CSF) and interleukin-6 (IL-6), as candidate biomarkers of CBLB502's radioprotective/mitigative efficacy. Induction of both G-CSF and IL-6 by CBLB502 1) is strictly TLR5-dependent, 2) occurs in a CBLB502 dose-dependent manner within its efficacious dose range in both nonirradiated and irradiated mammals, including nonhuman primates, and 3) is critically important for the ability of CBLB502 to rescue irradiated animals from death. After evaluation of CBLB502 effects on G-CSF and IL-6 levels in humans, these biomarkers will be useful for accurate prediction of human efficacious CBLB502 doses, a key step in the development of this prospective radiation countermeasure.
Collapse
|
33
|
Shakhov AN, Singh VK, Bone F, Cheney A, Kononov Y, Krasnov P, Bratanova-Toshkova TK, Shakhova VV, Young J, Weil MM, Panoskaltsis-Mortari A, Orschell CM, Baker PS, Gudkov A, Feinstein E. Prevention and mitigation of acute radiation syndrome in mice by synthetic lipopeptide agonists of Toll-like receptor 2 (TLR2). PLoS One 2012; 7:e33044. [PMID: 22479357 PMCID: PMC3314012 DOI: 10.1371/journal.pone.0033044] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Accepted: 02/09/2012] [Indexed: 01/28/2023] Open
Abstract
Bacterial lipoproteins (BLP) induce innate immune responses in mammals by activating heterodimeric receptor complexes containing Toll-like receptor 2 (TLR2). TLR2 signaling results in nuclear factor-kappaB (NF-κB)-dependent upregulation of anti-apoptotic factors, anti-oxidants and cytokines, all of which have been implicated in radiation protection. Here we demonstrate that synthetic lipopeptides (sLP) that mimic the structure of naturally occurring mycoplasmal BLP significantly increase mouse survival following lethal total body irradiation (TBI) when administered between 48 hours before and 24 hours after irradiation. The TBI dose ranges against which sLP are effective indicate that sLP primarily impact the hematopoietic (HP) component of acute radiation syndrome. Indeed, sLP treatment accelerated recovery of bone marrow (BM) and spleen cellularity and ameliorated thrombocytopenia of irradiated mice. sLP did not improve survival of irradiated TLR2-knockout mice, confirming that sLP-mediated radioprotection requires TLR2. However, sLP was radioprotective in chimeric mice containing TLR2-null BM on a wild type background, indicating that radioprotection of the HP system by sLP is, at least in part, indirect and initiated in non-BM cells. sLP injection resulted in strong transient induction of multiple cytokines with known roles in hematopoiesis, including granulocyte colony-stimulating factor (G-CSF), keratinocyte chemoattractant (KC) and interleukin-6 (IL-6). sLP-induced cytokines, particularly G-CSF, are likely mediators of the radioprotective/mitigative activity of sLP. This study illustrates the strong potential of LP-based TLR2 agonists for anti-radiation prophylaxis and therapy in defense and medical scenarios.
Collapse
|
34
|
Basile LA, Ellefson D, Gluzman-Poltorak Z, Junes-Gill K, Mar V, Mendonca S, Miller JD, Tom J, Trinh A, Gallaher TK. HemaMax™, a recombinant human interleukin-12, is a potent mitigator of acute radiation injury in mice and non-human primates. PLoS One 2012; 7:e30434. [PMID: 22383962 PMCID: PMC3286478 DOI: 10.1371/journal.pone.0030434] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Accepted: 12/18/2011] [Indexed: 01/10/2023] Open
Abstract
HemaMax, a recombinant human interleukin-12 (IL-12), is under development to address an unmet medical need for effective treatments against acute radiation syndrome due to radiological terrorism or accident when administered at least 24 hours after radiation exposure. This study investigated pharmacokinetics, pharmacodynamics, and efficacy of m-HemaMax (recombinant murine IL-12), and HemaMax to increase survival after total body irradiation (TBI) in mice and rhesus monkeys, respectively, with no supportive care. In mice, m-HemaMax at an optimal 20 ng/mouse dose significantly increased percent survival and survival time when administered 24 hours after TBI between 8–9 Gy (p<0.05 Pearson's chi-square test). This survival benefit was accompanied by increases in plasma interferon-γ (IFN-γ) and erythropoietin levels, recovery of femoral bone hematopoiesis characterized with the presence of IL-12 receptor β2 subunit–expressing myeloid progenitors, megakaryocytes, and osteoblasts. Mitigation of jejunal radiation damage was also examined. At allometrically equivalent doses, HemaMax showed similar pharmacokinetics in rhesus monkeys compared to m-HemaMax in mice, but more robustly increased plasma IFN-γ levels. HemaMax also increased plasma erythropoietin, IL-15, IL-18, and neopterin levels. At non-human primate doses pharmacologically equivalent to murine doses, HemaMax (100 ng/Kg and 250 ng/Kg) administered at 24 hours after TBI (6.7 Gy/LD50/30) significantly increased percent survival of HemaMax groups compared to vehicle (p<0.05 Pearson's chi-square test). This survival benefit was accompanied by a significantly higher leukocyte (neutrophils and lymphocytes), thrombocyte, and reticulocyte counts during nadir (days 12–14) and significantly less weight loss at day 12 compared to vehicle. These findings indicate successful interspecies dose conversion and provide proof of concept that HemaMax increases survival in irradiated rhesus monkeys by promoting hematopoiesis and recovery of immune functions and possibly gastrointestinal functions, likely through a network of interactions involving dendritic cells, osteoblasts, and soluble factors such as IL-12, IFN-γ, and cytoprotectant erythropoietin.
Collapse
Affiliation(s)
- Lena A Basile
- Neumedicines, Inc, Pasadena, California, United States of America.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Singh VK, Ducey EJ, Fatanmi OO, Singh PK, Brown DS, Purmal A, Shakhova VV, Gudkov AV, Feinstein E, Shakhov A. CBLB613: a TLR 2/6 agonist, natural lipopeptide of Mycoplasma arginini , as a novel radiation countermeasure. Radiat Res 2011; 177:628-42. [PMID: 22175300 DOI: 10.1667/rr2657.1] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
To date, there are no safe and effective drugs available for protection against ionizing radiation damage. Therefore, a great need exists to identify and develop non-toxic agents that will be useful as radioprotectors or postirradiation therapies under a variety of operational scenarios. We have developed a new pharmacological agent, CBLB613 (a naturally occurring Mycoplasma-derived lipopeptide ligand for Toll-like receptor 2/6), as a novel radiation countermeasure. Using CD2F1 mice, we investigated CBLB613 for toxicity, immunogenicity, radioprotection, radiomitigation and pharmacokinetics. We also evaluated CBLB613 for its effects on cytokine induction and radiation-induced cytopenia in unirradiated and irradiated mice. The no-observable-adverse-effect level of CBLB613 was 1.79 mg/kg and 1 mg/kg for single and repeated doses, respectively. CBLB613 significantly protected mice against a lethal dose of (60)Co γ radiation. The dose reduction factor of CBLB613 as a radioprotector was 1.25. CBLB613 also mitigated the effects of (60)Co γ radiation on survival in mice. In both irradiated and unirradiated mice, the drug stimulated induction of interleukin-1β (IL-1β), IL-6, IL-10, IL-12, keratinocyte-derived chemokine, granulocyte colony-stimulating factor, granulocyte-macrophage colony-stimulating factor, and tumor necrosis factor-1α. CBLB613 also reduced radiation-induced cytopenia and increased bone marrow cellularity in irradiated mice. Our immunogenicity study demonstrated that CBLB613 is not immunogenic in mice, indicating that it could be developed as a radioprotector and radiomitigator for humans against the potentially lethal effects of radiation exposure.
Collapse
Affiliation(s)
- Vijay K Singh
- Radiation Countermeasures Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889-5603, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
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
|
37
|
Singh PK, Wise SY, Ducey EJ, Brown DS, Singh VK. Radioprotective efficacy of tocopherol succinate is mediated through granulocyte-colony stimulating factor. Cytokine 2011; 56:411-21. [DOI: 10.1016/j.cyto.2011.08.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2011] [Revised: 07/14/2011] [Accepted: 08/05/2011] [Indexed: 12/17/2022]
|
38
|
Yang HJ, Youn H, Seong KM, Yun YJ, Kim W, Kim YH, Lee JY, Kim CS, Jin YW, Youn B. Psoralidin, a dual inhibitor of COX-2 and 5-LOX, regulates ionizing radiation (IR)-induced pulmonary inflammation. Biochem Pharmacol 2011; 82:524-34. [DOI: 10.1016/j.bcp.2011.05.027] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Revised: 05/23/2011] [Accepted: 05/24/2011] [Indexed: 11/28/2022]
|
39
|
Singh VK, Singh PK, Wise SY, Seed TM. Mobilized progenitor cells as a bridging therapy for radiation casualties: A brief review of tocopherol succinate-based approaches. Int Immunopharmacol 2011; 11:842-47. [DOI: 10.1016/j.intimp.2011.01.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Revised: 01/18/2011] [Accepted: 01/19/2011] [Indexed: 12/11/2022]
|
40
|
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
|
41
|
Abstract
IMPORTANCE OF THE FIELD Ionizing radiation (IR) can produce deleterious effects in living tissues, leading to significant morbidity and a potentially fatal illness affecting various organs dose-dependently. As people may be exposed to IR during cancer radiotherapy or as a result of a radiological/nuclear incident or act of terrorism, the danger of irradiation represents a serious public health problem. At present, however, this problem remains largely impervious to medical management. There is, therefore, a pressing need to develop safe and effective radiation countermeasure (RC) agents to prevent, mitigate or treat the harmful consequences of IR exposure. AREAS COVERED IN THIS REVIEW Recent advances in the search for RC agents as reflected by the relevant patent literature of the past five years along with peer-reviewed publications are surveyed. WHAT THE READER WILL GAIN A total of 43 patents, describing approximately 38 chemically diverse compounds with RC potential are analyzed. These include antioxidants capable of scavenging IR-induced free radicals, modulators of cell death signaling or cell cycle progression, cytokines or growth factors promoting tissue repair and inhibitors of inflammatory cytokines. TAKE HOME MESSAGE Several of these RC candidates appear promising, including at least two that are undergoing evaluation for fast-track clinical development.
Collapse
Affiliation(s)
- Francis Dumont
- Université de Strasbourg, Centre Régional de Lutte contre le Cancer Paul Strauss, Laboratoire de Radiobiologie EA-3430, 3 rue de la Porte de l'Hôpital, F-67065 Strasbourg, France
| | | | | |
Collapse
|
42
|
Guruvayoorappan C, Kuttan G. Protective effect of Biophytum sensitivum (L.) DC on radiation-induced damage in mice. Immunopharmacol Immunotoxicol 2010; 30:815-35. [PMID: 18951225 DOI: 10.1080/08923970802439480] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The radioprotective effect of Biophytum sensitivum methanol extract was studied using in vivo mice model . Animals were exposed to whole body gamma irradiation (6 Gy/animal) after treatment with B. sensitivum (50mg/kg b.wt.) followed by estimation of cytosolic enzymes, level of antioxidants, hematological parameters, bone marrow cellular progenitors, serum cytokine levels and spleen hematopoietic colonies. Administration of B. sensitivum could reduce the enhanced level of ALP, GPT and LPO levels in irradiated animals. B. sensitivum could significantly enhance the glutathione (GSH) content in liver and intestinal mucosa of irradiated animals. B. sensitivum treatment could enhance the Total WBC count, cellularity of bone marrow, alpha-esterase positive cells, and relative organ weight of spleen as well as thymus. The number of hematopoietic colonies on the surface of the spleen was found to be enhanced after B. sensitivum treatment. B. sensitivum treatment could also stimulate the production of cytokines such as IL-1beta, IFN-gamma and GM-CSF in animals exposed to whole body gamma irradiation. The present investigation suggests that the protective effect of Biophytum sensitivum on Radiation-Induced hemopoietic damage is mediated through immunomodulation as well as sequential induction of IL-1beta, GM-CSF and IFN-gamma.
Collapse
Affiliation(s)
- C Guruvayoorappan
- Department of Immunology, Amala Cancer Research Centre, Kerala, India
| | | |
Collapse
|
43
|
Jiao W, Kiang JG, Cary L, Elliott TB, Pellmar TC, Ledney GD. COX-2 inhibitors are contraindicated for treatment of combined injury. Radiat Res 2009; 172:686-97. [PMID: 19929415 DOI: 10.1667/rr1581.1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Casualties of radiation dispersal devices, nuclear detonation or major ionizing radiation accidents, in addition to radiation exposure, may sustain physical and/or thermal trauma. Radiation exposure plus additional tissue trauma is known as combined injury. There are no definitive therapeutic agents. Cyclooxygenase-2 (COX-2), an inducible enzyme expressed in pathological disorders and radiation injury, plays an important role in inflammation and the production of cytokines and prostaglandin E(2) (PGE(2)) and could therefore affect the outcome for victims of combined injury. The COX-2 inhibitors celecoxib and meloxicam were evaluated for their therapeutic value against combined injury in mice. In survival studies, the COX-2 inhibitors had no beneficial effect on 30-day survival, wound healing or body weight gain after radiation injury alone or after combined injury. Meloxicam accelerated death in both wounded and combined injury mice. These drugs also induced severe hepatic toxicity, exaggerated inflammatory processes, and did not enhance hematopoietic cell regeneration. This study points to potential contraindications for use of COX-2 inhibitors in patients undergoing therapy for radiation injury and combined injury.
Collapse
Affiliation(s)
- W Jiao
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, 8901 Wisconsin Avenue, Bethesda, Maryland 20889-5603, USA
| | | | | | | | | | | |
Collapse
|
44
|
Effects of genistein administration on cytokine induction in whole-body gamma irradiated mice. Int Immunopharmacol 2009; 9:1401-10. [DOI: 10.1016/j.intimp.2009.08.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2009] [Revised: 08/17/2009] [Accepted: 08/18/2009] [Indexed: 12/25/2022]
|
45
|
Tocopherol succinate: A promising radiation countermeasure. Int Immunopharmacol 2009; 9:1423-30. [DOI: 10.1016/j.intimp.2009.08.020] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2009] [Revised: 08/10/2009] [Accepted: 08/20/2009] [Indexed: 11/23/2022]
|
46
|
Tanaka I, Tanaka M, Satoh A, Kurematsu A, Ishiwata A, Suzuki K, Ishihara H. Alteration of radioprotective effects of heat-killed Lactobacillus casei in X-irradiated C3H/He mouse related to blood level of proinflammatory cytokines by corticoids. JOURNAL OF RADIATION RESEARCH 2009; 51:81-86. [PMID: 19851041 DOI: 10.1269/jrr.09095] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
It is well known that a pre-administration of proinflammatory cytokines alters hematopoietic progenitor cells to promote an increase resistance against radiation and increases the survival rate in mice irradiated with lethal doses of radiation. Inflammation stimulators, such as some bacterial constituents, are also reported to have similar radioprotective action. We found that pre-administration of heat-killed Lactobacillus casei (HLC) to mice increases the level of interleukin (IL)-1 beta in circulation as well as the survival rate following lethal dose of radiation. Since HLC stimulates early immune responses, effects by drugs to modify inflammation were studied. The increase of both blood IL-1 beta levels and survival rates by HLC were simultaneously accelerated by coadministration of mineralocorticoid and inhibited by glucocorticoids or corticotropin. Neither parameter was modified by non-steroidal anti-inflammatory or anti-rheumatoid drugs. This suggests that both expected radioprotective action and unexpected systemic action, realized as an increase in plasma cytokines, by inflammation-related radioprotectors can be controlled by the coadministration of drugs at least in C3H/He mice, based on consideration of their pharmacological properties.
Collapse
Affiliation(s)
- Izumi Tanaka
- Treatment Research Team, Medical Treatment for High Dose Exposure Research Group, Research Center for Radiation Emergency Medicine, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-ku, Chiba, Japan
| | | | | | | | | | | | | |
Collapse
|
47
|
Li B, Bailey AS, Jiang S, Liu B, Goldman DC, Fleming WH. Endothelial cells mediate the regeneration of hematopoietic stem cells. Stem Cell Res 2009; 4:17-24. [PMID: 19720572 DOI: 10.1016/j.scr.2009.08.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2009] [Accepted: 08/08/2009] [Indexed: 02/03/2023] Open
Abstract
Recent studies suggest that endothelial cells are a critical component of the normal hematopoietic microenvironment. Therefore, we sought to determine whether primary endothelial cells have the capacity to repair damaged hematopoietic stem cells. Highly purified populations of primary CD31(+) microvascular endothelial cells isolated from the brain or lung did not express the pan hematopoietic marker CD45, most hematopoietic lineage markers, or the progenitor marker c-kit and did not give rise to hematopoietic cells in vitro or in vivo. Remarkably, the transplantation of small numbers of these microvascular endothelial cells consistently restored hematopoiesis following bone marrow lethal doses of irradiation. Analysis of the peripheral blood of rescued recipients demonstrated that both short-term and long-term multilineage hematopoietic reconstitution was exclusively of host origin. Secondary transplantation studies revealed that microvascular endothelial cell-mediated hematopoietic regeneration also occurs at the level of the hematopoietic stem cell. These findings suggest a potential therapeutic role for microvascular endothelial cells in the self-renewal and repair of adult hematopoietic stem cells.
Collapse
Affiliation(s)
- Bei Li
- Division of Hematology and Medical Oncology, Hematologic Malignancies Program, Oregon Health & Science University, Portland, OR 97239, USA
| | | | | | | | | | | |
Collapse
|
48
|
Ghosh SP, Kulkarni S, Hieber K, Toles R, Romanyukha L, Kao TC, Hauer-Jensen M, kumar KS. Gamma-tocotrienol, a tocol antioxidant as a potent radioprotector. Int J Radiat Biol 2009; 85:598-606. [DOI: 10.1080/09553000902985128] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
49
|
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
|
50
|
Gruel G, Voisin P, Vaurijoux A, Roch-Lefevre S, Grégoire E, Maltere P, Petat C, Gidrol X, Voisin P, Roy L. Broad modulation of gene expression in CD4+ lymphocyte subpopulations in response to low doses of ionizing radiation. Radiat Res 2008; 170:335-44. [PMID: 18763857 DOI: 10.1667/rr1147.1] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2007] [Accepted: 04/17/2008] [Indexed: 11/03/2022]
Abstract
To compare the responses of the different lymphocyte subtypes after an exposure of whole blood to low doses of ionizing radiation, we examined variations in gene expression in different lymphocyte subpopulations using microarray technology. Blood samples from five healthy donors were independently exposed to 0 (sham irradiation), 0.05 and 0.5 Gy of ionizing radiation. Three and 24 h after exposure, CD56+, CD4+ and CD8+ cells were negatively isolated. RNA from each set of experimental conditions was competitively hybridized on 25k oligonucleotide microarrays. Modifications of gene expression were measured after both intervals and in all cell types. Twenty-four hours after exposure to 0.5 Gy, we observed an induction of the expression of BAX, PCNA, GADD45, DDB2 and CDKN1A. However, the numbers of modulated genes greatly differed between cell types. In particular, 3 h after exposure to doses as low as 0.05 Gy, the number of down-modulated genes was 10 times greater for CD4+ cells than for all other cell types. Moreover, most of these repressed genes were taking part in the cell processes of protein biosynthesis and oxidative phosphorylation. The results suggest that several biological pathways in CD4+ cells could be sensitive to low doses of radiation. Therefore, specifically studying CD4+ cells could help to understand the mechanisms involved in low-dose response and allow their detection.
Collapse
Affiliation(s)
- Gaëtan Gruel
- Laboratoire de Dosimétrie Biologique, Institut de Radioprotection et de Sûreté Nucléaire (IRSN), 92262 Fontenay-aux-Roses, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|