168 results on '"Grdina DJ"'
Search Results
2. Conformational changes in chromatin structure induced by the radioprotective aminothiol, WR 1065.
- Author
-
Vaughan, ATM, Grdina, DJ, Meechan, PJ, Milner, AE, and Gordon, DJ
- Published
- 1989
- Full Text
- View/download PDF
3. The effect of 2-[(aminopropyl)amino] ethanethiol on fission-neutron-induced DNA damage and repair.
- Author
-
Grdina, DJ, Sigdestad, CP, Dale, PJ, Perrin, JM, Grdina, D J, Sigdestad, C P, Dale, P J, and Perrin, J M
- Published
- 1989
- Full Text
- View/download PDF
4. The effect of 2-[(aminopropyl)amino] ethanethiol (WR-1065) on radiation induced DNA double strand damage and repair in V79 cells.
- Author
-
Sigdestad, CP, Treacy, SH, Knapp, LA, Grdina, DJ, Sigdestad, C P, Treacy, S H, Knapp, L A, and Grdina, D J
- Published
- 1987
- Full Text
- View/download PDF
5. Changes in gene expression associated with radiation exposure
- Author
-
Woloschak, Ge, Tatjana Paunesku, Changliu, Cm, Grdina, Dj, Hagen, U., Harder, D., Jung, H., and Streffer, C.
6. PERIOD 2 regulates low-dose radioprotection via PER2/pGSK3β/β-catenin/Per2 loop.
- Author
-
Alexandrou AT, Duan Y, Xu S, Tepper C, Fan M, Tang J, Berg J, Basheer W, Valicenti T, Wilson PF, Coleman MA, Vaughan AT, Fu L, Grdina DJ, Murley J, Wang A, Woloschak G, and Li JJ
- Abstract
During evolution, humans are acclimatized to the stresses of natural radiation and circadian rhythmicity. Radiosensitivity of mammalian cells varies in the circadian period and adaptive radioprotection can be induced by pre-exposure to low-level radiation (LDR). It is unclear, however, if clock proteins participate in signaling LDR radioprotection. Herein, we demonstrate that radiosensitivity is increased in mice with the deficient Period 2 gene (Per2
def ) due to impaired DNA repair and mitochondrial function in progenitor bone marrow hematopoietic stem cells and monocytes. Per2 induction and radioprotection are also identified in LDR-treated Per2wt mouse cells and in human skin (HK18) and breast (MCF-10A) epithelial cells. LDR-boosted PER2 interacts with pGSK3β(S9) which activates β-catenin and the LEF/TCF mediated gene transcription including Per2 and genes involved in DNA repair and mitochondrial functions. This study demonstrates that PER2 plays an active role in LDR adaptive radioprotection via PER2/pGSK3β/β-catenin/Per2 loop, a potential target for protecting normal cells from radiation injury., Competing Interests: The authors declare no competing interests., (© 2022 The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
7. Dual blockade of CD47 and HER2 eliminates radioresistant breast cancer cells.
- Author
-
Candas-Green D, Xie B, Huang J, Fan M, Wang A, Menaa C, Zhang Y, Zhang L, Jing D, Azghadi S, Zhou W, Liu L, Jiang N, Li T, Gao T, Sweeney C, Shen R, Lin TY, Pan CX, Ozpiskin OM, Woloschak G, Grdina DJ, Vaughan AT, Wang JM, Xia S, Monjazeb AM, Murphy WJ, Sun LQ, Chen HW, Lam KS, Weichselbaum RR, and Li JJ
- Subjects
- Animals, Breast Neoplasms pathology, CD47 Antigen genetics, Cell Proliferation, Clone Cells, Female, Humans, MCF-7 Cells, Macrophages metabolism, Mice, Models, Biological, NF-kappa B metabolism, Phagocytosis, Signal Transduction, Transcription, Genetic, Tumor Burden, Breast Neoplasms metabolism, CD47 Antigen metabolism, Radiation Tolerance, Receptor, ErbB-2 metabolism
- Abstract
Although the efficacy of cancer radiotherapy (RT) can be enhanced by targeted immunotherapy, the immunosuppressive factors induced by radiation on tumor cells remain to be identified. Here, we report that CD47-mediated anti-phagocytosis is concurrently upregulated with HER2 in radioresistant breast cancer (BC) cells and RT-treated mouse syngeneic BC. Co-expression of both receptors is more frequently detected in recurrent BC patients with poor prognosis. CD47 is upregulated preferentially in HER2-expressing cells, and blocking CD47 or HER2 reduces both receptors with diminished clonogenicity and augmented phagocytosis. CRISPR-mediated CD47 and HER2 dual knockouts not only inhibit clonogenicity but also enhance macrophage-mediated attack. Dual antibody of both receptors synergizes with RT in control of syngeneic mouse breast tumor. These results provide the evidence that aggressive behavior of radioresistant BC is caused by CD47-mediated anti-phagocytosis conjugated with HER2-prompted proliferation. Dual blockade of CD47 and HER2 is suggested to eliminate resistant cancer cells in BC radiotherapy.
- Published
- 2020
- Full Text
- View/download PDF
8. ROS modifiers and NOX4 affect the expression of the survivin-associated radio-adaptive response.
- Author
-
Murley JS, Arbiser JL, Weichselbaum RR, and Grdina DJ
- Subjects
- Colonic Neoplasms metabolism, Colonic Neoplasms radiotherapy, Dose-Response Relationship, Radiation, Gamma Rays, Gene Expression Regulation, Neoplastic, Humans, Mutation, NADPH Oxidase 4 genetics, Survivin genetics, Tumor Cells, Cultured, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Cell Survival, Colonic Neoplasms pathology, NADPH Oxidase 4 metabolism, Radiation Tolerance, Reactive Oxygen Species metabolism, Survivin metabolism
- Abstract
The survivin-associated radio-adaptive response can be induced following exposure to ionizing radiation in the dose range from 5 to 100 mGy, and its magnitude of expression is dependent upon the TP53 mutational status of cells and ROS signaling. The purpose of the study was to investigate the potential role of ROS in the development of the survivin-associated adaptive response. Utilizing human colon carcinoma HCT116 TP53 wild type (WT) and HCT116 isogenic TP53 null mutant (Mut) cell cultures, the roles of inter- and intracellular ROS signaling on expression of the adaptive response as evidenced by changes in intracellular translocation of survivin measured by ELISA, and cell survival determined by a standard colony forming assay were investigated using ROS modifying agents that include emodin, N-acetyl-L-cysteine (NAC), fulvene-5, honokiol, metformin and rotenone. The role of NADPH oxidase 4 (NOX4) in the survivin-associated adaptive response was investigated by transfecting HCT116 cells, both WT and Mut, with two different NOX4 siRNA oligomers and Western blotting. A dose of 5 mGy or a 15 min exposure to 50 µM of the ROS producing drug emodin were equally effective in inducing a pro-survival adaptive response in TP53 WT and a radio-sensitization adaptive response in TP53 Mut HCT116 cells. Each response was associated with a corresponding translocation of survivin into the cytoplasm or nucleus, respectively. Exposure to 10 mM NAC completely inhibited both responses. Exposure to 10 µM honokiol induced responses similar to those observed following NAC exposure in TP53 WT and Mut cells. The mitochondrial complex 1 inhibitor rotenone was effective in reducing both cytoplasmic and nuclear survivin levels, but was ineffective in altering the expression of the adaptive response in either TP53 WT or Mut cells. In contrast, both metformin and fulvene-5, inhibitors of NOX4, facilitated the reversal of TP53 WT and Mut adaptive responses from pro-survival to radio-sensitization and vice versa, respectively. These changes were accompanied by corresponding reversals in the translocation of survivin to the nuclei of TP53 WT and to the cytoplasm of TP53 Mut cells. The potential role of NOX4 in the expression of the survivin-associated adaptive response was investigated by transfecting HCT116 cells with NOX4 siRNA oligomers to inhibit NOX4 expression. Under these conditions NOX4 expression was inhibited by about 50%, resulting in a reversal in the expression of the TP53 WT and Mut survivin-associated adaptive responses as was observed following metformin and fulvene-5 treatment. Exposure to 5 mGy resulted in enhanced NOX4 expression by about 40% in both TP53 WT and Mut cells, in contrast to only a 1-2% increase following a 2 Gy only exposure. Utilizing mixed cultures of HCT116 TP53 WT and isogenic null Mut cells, as few as 10% TP53 Mut cells were sufficient to control the expression of the remaining 90% WT cells and resulted in an overall radio-sensitization response accompanied by the nuclear translocation of survivin characteristic of homogeneous TP53 Mut populations., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
9. TP53 Mutational Status and ROS Effect the Expression of the Survivin-Associated Radio-Adaptive Response.
- Author
-
Murley JS, Miller RC, Weichselbaum RR, and Grdina DJ
- Subjects
- Acetylcysteine pharmacology, Dose-Response Relationship, Radiation, Gene Expression Regulation drug effects, Genomics, HCT116 Cells, Humans, Mutation, Radiation Tolerance drug effects, Survivin, Gene Expression Regulation radiation effects, Inhibitor of Apoptosis Proteins metabolism, Radiation Tolerance genetics, Reactive Oxygen Species metabolism, Tumor Suppressor Protein p53 genetics
- Abstract
A survivin-associated radio-adaptive response, characterized by increased radiation resistance or sensitization, was induced by exposure to 5 mGy of ionizing radiation and was correlated to the TP53 mutational status of exposed cells. Ten human cancer lines were investigated: colorectal carcinomas HCT116 and RKO [TP53 wild-type (WT)] and their respective TP53 null isogenic lines; breast adenocarcinomas MCF7 (TP53 WT) and MDA-MB-231 (TP53 Mut); lung carcinomas A549 (TP53 WT) and NCI-H1975 (TP53 Mut); and pancreatic carcinomas Hs766T (TP53 WT) and Panc-1 (TP53 Mut). Radiation induced (5 mGy) changes in the subsequent responses to 2 Gy in a multi-dose paradigm. Effects on radiation sensitivity were associated with changes in survivin's intracellular translocation to the cytoplasm (TP53 WT) or nucleus (TP53 Mut). Survival responses were determined using a colony forming assay. Intracellular localization of survivin was determined by ELISA and correlated with survival response. Two 2 Gy doses had minimal effects on the intracellular translocation of survivin. When preceded 15 min earlier by a 5 mGy exposure, survivin translocated to the cytoplasm in all of the TP53 WT cell lines, and to the nuclei in the TP53 null and Mut cells. All TP53 WT cells were protected (P < 0.001) by 5 mGy exposures, while Mut cells were sensitized (P < 0.001). HCT116 and RKO TP53 WT cells were admixed with their respective isogenic TP53 null counterparts in different proportions: 75% to 25%, 50% to 50% and 25% to 75%, respectively. All mixed confluent cultures expressed enhanced radio-sensitization (P ≤ 0.047) characteristic of TP53 Mut cells, which could be inhibited by their exposure to the antioxidant N-acetyl-l-cysteine (NAC) indicating a role for intercellular signaling by reactive oxygen species (ROS). ROS signaling in propagating the survivin-mediated response is involved in both intra- and intercellular communication processes.
- Published
- 2017
- Full Text
- View/download PDF
10. Altered expression of a metformin-mediated radiation response in SA-NH and FSa tumor cells treated under in vitro and in vivo growth conditions.
- Author
-
Murley JS, Miller RC, Senlik RR, Rademaker AW, and Grdina DJ
- Subjects
- Acetylcysteine administration & dosage, Animals, Cell Line, Tumor, Dose-Response Relationship, Drug, Emodin administration & dosage, Female, Mice, Mice, Inbred C3H, Neoplasms, Experimental pathology, Radiation Dosage, Radiation Tolerance drug effects, Radiation-Sensitizing Agents administration & dosage, Cell Proliferation drug effects, Cell Proliferation radiation effects, Metformin administration & dosage, Neoplasms, Experimental metabolism, Neoplasms, Experimental radiotherapy, Tumor Suppressor Protein p53 metabolism
- Abstract
Purpose: To assess the radiosensitizing effect of the biguanide drug metformin used alone or in combination with reactive oxygen species (ROS) modifying agents N-acetyl-L-cysteine (NAC) or emodin, and contrasted to the mitochondrial complex 1 inhibitor rotenone in altering the radiation responses of the p53 wild-type SA-NH and p53 mutant FSa mouse tumor lines grown either in vitro or in vivo., Materials and Methods: Tumor cells were grown to confluence in vitro and exposed to a single 4 Gy dose in the presence or absence of metformin (5 mM) and ROS modifiers or to 10 Gy with or without metformin as tumors in the flanks of C3H mice using a tumor growth delay assay., Results: Both metformin and rotenone protected SA-NH (p < .001) while sensitizing FSa (p < .001) to 4 Gy. Neither NAC nor emodin altered metformin's action. Metformin was also directly toxic to FSa cells (p = .002). In contrast, in vivo metformin (250 mg/kg) sensitized both SA-NH (9-day growth delay, p < .05) and FSa (4-day growth delay, p < .05) tumors if administered 1 h before irradiation., Conclusion: Metformin effects on tumor cells measured under in vitro conditions may differ from those determined in vivo due to p53 and heterogeneous environmental factors.
- Published
- 2017
- Full Text
- View/download PDF
11. Andrographis paniculata Diterpenoids Protect against Radiation-Induced Transformation in BALB/3T3 Cells.
- Author
-
Nantajit D, Jetawattana S, Suriyo T, Grdina DJ, and Satayavivad J
- Subjects
- Animals, BALB 3T3 Cells, Cell Survival drug effects, Cell Survival radiation effects, Dose-Response Relationship, Drug, Dose-Response Relationship, Radiation, Mice, Plant Extracts chemistry, Radiation Dosage, Radiation Tolerance radiation effects, Andrographis chemistry, Cell Transformation, Neoplastic drug effects, Cell Transformation, Neoplastic radiation effects, Diterpenes administration & dosage, Plant Extracts administration & dosage, Radiation-Protective Agents administration & dosage
- Abstract
One of the most concerning side effects of exposure to radiation are the carcinogenic risks. To reduce the negative effects of radiation, both cytoprotective and radioprotective agents have been developed. However, little is known regarding their potential for suppressing carcinogenesis. Andrographis paniculata , a plant, with multiple medicinal uses that is commonly used in traditional medicine, has three major constituents known to have cellular antioxidant activity: andrographolide (AP1); 14-deoxy-11,12-didehydroandrographolide (AP3); and neoandrographolide (AP4). In our study, we tested these elements for their radioprotective properties as well as their anti-neoplastic effects on transformation using the BALB/3T3 cell model. All three compounds were able to reduce radiation-induced DNA damage. However, AP4 appeared to have superior radioprotective properties compared to the other two compounds, presumably by protecting mitochondrial function. The compound was able to suppress radiation-induced cellular transformation through inhibition of STAT3. Treatment with AP4 also reduced expressions of MMP-2 and MMP-9. These results suggest that AP4 could be further studied and developed into an anti-transformation/carcinogenic drug as well as a radioprotective agent.
- Published
- 2017
- Full Text
- View/download PDF
12. Very low doses of ionizing radiation and redox associated modifiers affect survivin-associated changes in radiation sensitivity.
- Author
-
Miller RC, Murley JS, Rademaker AW, Woloschak GE, Li JJ, Weichselbaum RR, and Grdina DJ
- Subjects
- Acetylcysteine pharmacology, Animals, Antioxidants pharmacology, Cell Line, Tumor, Cell Survival drug effects, Cell Survival radiation effects, Dose-Response Relationship, Radiation, Female, Fibrosarcoma genetics, Fibrosarcoma metabolism, Fibrosarcoma pathology, Gamma Rays, Hindlimb, Inhibitor of Apoptosis Proteins metabolism, Mice, Mice, Inbred C3H, Neoplasm Transplantation, Protein Transport, RNA, Small Interfering genetics, RNA, Small Interfering metabolism, Reactive Oxygen Species metabolism, Repressor Proteins metabolism, Survivin, Tumor Suppressor Protein p53 antagonists & inhibitors, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Emodin pharmacology, Fibrosarcoma therapy, Gene Expression Regulation, Neoplastic, Inhibitor of Apoptosis Proteins genetics, Oxidants pharmacology, Radiation Tolerance drug effects, Repressor Proteins genetics
- Abstract
Exposure of cells to a dose of ionizing radiation as low as 5mGy can induce changes in radiation sensitivity expressed by cells exposed to subsequent higher doses at later times. This is referred to as an adaptive effect. We describe a unique survivin-associated adaptive response in which increased radiation resistance or sensitization of cells can be induced by exposure to 5mGy or to the reactive oxygen species (ROS) generating drug Emodin (1,3,8-trihydroxy-6-methylanthraquinone), a naturally occurring anthraquinone. The purpose of this study was to determine the role of ROS generating processes in affecting both the intracellular localization of the inhibitor of apoptosis protein survivin and its subsequent effect on radiation response in the presence or absence of the anti-oxidant N-acetyl-L-cysteine (NAC). Experiments were performed using two well characterized murine sarcomas: SA-NH p53 wild-type (WT) and FSa p53 mutant (Mut), grown either in culture or as solid tumors in the right hind legs of C3H mice. Doses of 5mGy or 50μM Emodin were used to induce changes in the response of these tumor cells to higher radiation exposures using a multi-dosing paradigm. Effects on radiation sensitivity were determined for SA-NH and FSa cells as a function of survivin translocation either to the cytoplasm or nucleus in the presence or absence of 10mM NAC treatment. In vitro survival assays (2Gy per fraction, two once daily fractions) and tumor growth delay (TGD) (5Gy per fraction, five once daily fractions) studies were performed. Intracellular localization of survivin was determined by enzyme-linked immunosorbent assay (ELISA) and correlated to survival response and treatment conditions. 2Gy alone had no effect on intracellular translocation of survivin. When preceded 15min earlier by 5mGy or Emodin exposures, survivin became elevated in the cytoplasm of p53 WT SA-NH as compared to the nuclei of p53 Mut FSa cells. SA-NH cells transfected with p53 small interfering RNA (siRNA), in contrast, responded similarly to p53 Mut FSa cells by becoming more radiation sensitive if exposed to 5mGy prior to each 2Gy irradiation. In contrast to their respective responses to five once daily 5Gy fractions, SA-NH tumors were protected by 5mGy exposures administered 15min prior to each daily 5Gy dose as evidenced by a more rapid growth (1.9 day decrease in TGD, P=0.032), while FSa tumors were sensitized, growing at a much slower rate (4.5 day increase in TGD, P<0.001). Exposure of SA-NH and FSa tumor cells to 10mM NAC inhibited the ability of 5mGy and Emodin to induce intracellular translocation of survivin and the corresponding altered adaptive survival response. The survivin-associated adaptive response can be induced following a multi-dosing scheme in which very low radiation doses are followed shortly thereafter by higher doses consistent with a standard image guided radiotherapy protocol that is currently widely used in the treatment of cancer. While induced by exposure to ROS generating stresses, the ultimate expression of changes in radiation response is dependent upon the bi-functionality of the tumor associated protein survivin and its intracellular translocation., (Copyright © 2016 Elsevier B.V. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
13. CDK1 Enhances Mitochondrial Bioenergetics for Radiation-Induced DNA Repair.
- Author
-
Qin L, Fan M, Candas D, Jiang G, Papadopoulos S, Tian L, Woloschak G, Grdina DJ, and Li JJ
- Subjects
- Adenosine Triphosphate, Blotting, Western, CDC2 Protein Kinase, Cell Line, Comet Assay, DNA Damage radiation effects, Energy Metabolism physiology, Gene Knockdown Techniques, Humans, Protein Transport radiation effects, RNA, Small Interfering, Radiation Effects, Transfection, Cyclin-Dependent Kinases metabolism, DNA Damage physiology, DNA Repair physiology, Mitochondria metabolism
- Abstract
Nuclear DNA repair capacity is a critical determinant of cell fate under genotoxic stress conditions. DNA repair is a well-defined energy-consuming process. However, it is unclear how DNA repair is fueled and whether mitochondrial energy production contributes to nuclear DNA repair. Here, we report a dynamic enhancement of oxygen consumption and mitochondrial ATP generation in irradiated normal cells, paralleled with increased mitochondrial relocation of the cell-cycle kinase CDK1 and nuclear DNA repair. The basal and radiation-induced mitochondrial ATP generation is reduced significantly in cells harboring CDK1 phosphorylation-deficient mutant complex I subunits. Similarly, mitochondrial ATP generation and nuclear DNA repair are also compromised severely in cells harboring mitochondrially targeted, kinase-deficient CDK1. These results demonstrate a mechanism governing the communication between mitochondria and the nucleus by which CDK1 boosts mitochondrial bioenergetics to meet the increased cellular fuel demand for DNA repair and cell survival under genotoxic stress conditions., (Copyright © 2015 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
14. The Increase in Animal Mortality Risk following Exposure to Sparsely Ionizing Radiation Is Not Linear Quadratic with Dose.
- Author
-
Haley BM, Paunesku T, Grdina DJ, and Woloschak GE
- Subjects
- Animals, Humans, Neoplasms, Radiation-Induced, Risk Factors, Dose-Response Relationship, Radiation, Radiation, Ionizing, Radiobiology, Risk Assessment
- Abstract
Introduction: The US government regulates allowable radiation exposures relying, in large part, on the seventh report from the committee to estimate the Biological Effect of Ionizing Radiation (BEIR VII), which estimated that most contemporary exposures- protracted or low-dose, carry 1.5 fold less risk of carcinogenesis and mortality per Gy than acute exposures of atomic bomb survivors. This correction is known as the dose and dose rate effectiveness factor for the life span study of atomic bomb survivors (DDREFLSS). It was calculated by applying a linear-quadratic dose response model to data from Japanese atomic bomb survivors and a limited number of animal studies., Methods and Results: We argue that the linear-quadratic model does not provide appropriate support to estimate the risk of contemporary exposures. In this work, we re-estimated DDREFLSS using 15 animal studies that were not included in BEIR VII's original analysis. Acute exposure data led to a DDREFLSS estimate from 0.9 to 3.0. By contrast, data that included both acute and protracted exposures led to a DDREFLSS estimate from 4.8 to infinity. These two estimates are significantly different, violating the assumptions of the linear-quadratic model, which predicts that DDREFLSS values calculated in either way should be the same., Conclusions: Therefore, we propose that future estimates of the risk of protracted exposures should be based on direct comparisons of data from acute and protracted exposures, rather than from extrapolations from a linear-quadratic model. The risk of low dose exposures may be extrapolated from these protracted estimates, though we encourage ongoing debate as to whether this is the most valid approach. We also encourage efforts to enlarge the datasets used to estimate the risk of protracted exposures by including both human and animal data, carcinogenesis outcomes, a wider range of exposures, and by making more radiobiology data publicly accessible. We believe that these steps will contribute to better estimates of the risks of contemporary radiation exposures.
- Published
- 2015
- Full Text
- View/download PDF
15. NFκB and Survivin-Mediated Radio-Adaptive Response.
- Author
-
Grdina DJ, Murley JS, Miller RC, Woloschak GE, and Li JJ
- Subjects
- Animals, Apoptosis radiation effects, Cell Line, Tumor, Cell Survival radiation effects, Dose-Response Relationship, Radiation, Mice, Survivin, Adaptation, Physiological radiation effects, Inhibitor of Apoptosis Proteins metabolism, NF-kappa B metabolism, Repressor Proteins metabolism
- 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.
- Published
- 2015
- Full Text
- View/download PDF
16. CDK4-mediated MnSOD activation and mitochondrial homeostasis in radioadaptive protection.
- Author
-
Jin C, Qin L, Shi Y, Candas D, Fan M, Lu CL, Vaughan AT, Shen R, Wu LS, Liu R, Li RF, Murley JS, Woloschak G, Grdina DJ, and Li JJ
- Subjects
- Adaptation, Physiological, Animals, Cell Line, Cyclin D1 genetics, Cyclin D1 metabolism, Cyclin-Dependent Kinase 4 metabolism, Dose-Response Relationship, Radiation, Gene Expression Regulation, Humans, Keratinocytes cytology, Keratinocytes enzymology, Membrane Potential, Mitochondrial radiation effects, Mice, Mice, Inbred BALB C, Mitochondria enzymology, Oxidative Phosphorylation, Phosphorylation drug effects, Radiation Tolerance, Radiation, Ionizing, Signal Transduction, Superoxide Dismutase metabolism, Whole-Body Irradiation, Cyclin-Dependent Kinase 4 genetics, Keratinocytes radiation effects, Mitochondria radiation effects, Superoxide Dismutase genetics
- Abstract
Mammalian cells are able to sense environmental oxidative and genotoxic conditions such as the environmental low-dose ionizing radiation (LDIR) present naturally on the earth's surface. The stressed cells then can induce a so-called radioadaptive response with an enhanced cellular homeostasis and repair capacity against subsequent similar genotoxic conditions such as a high dose radiation. Manganese superoxide dismutase (MnSOD), a primary mitochondrial antioxidant in mammals, has long been known to play a crucial role in radioadaptive protection by detoxifying O2(•-) generated by mitochondrial oxidative phosphorylation. In contrast to the well-studied mechanisms of SOD2 gene regulation, the mechanisms underlying posttranslational regulation of MnSOD for radioprotection remain to be defined. Herein, we demonstrate that cyclin D1/cyclin-dependent kinase 4 (CDK4) serves as the messenger to deliver the stress signal to mitochondria to boost mitochondrial homeostasis in human skin keratinocytes under LDIR-adaptive radioprotection. Cyclin D1/CDK4 relocates to mitochondria at the same time as MnSOD enzymatic activation peaks without significant changes in total MnSOD protein level. The mitochondrial-localized CDK4 directly phosphorylates MnSOD at serine-106 (S106), causing enhanced MnSOD enzymatic activity and mitochondrial respiration. Expression of mitochondria-targeted dominant negative CDK4 or the MnSOD-S106 mutant reverses LDIR-induced mitochondrial enhancement and adaptive protection. The CDK4-mediated MnSOD activation and mitochondrial metabolism boost are also detected in skin tissues of mice receiving in vivo whole-body LDIR. These results demonstrate a unique CDK4-mediated mitochondrial communication that allows cells to sense environmental genotoxic stress and boost mitochondrial homeostasis by enhancing phosphorylation and activation of MnSOD., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
17. Metformin exhibits radiation countermeasures efficacy when used alone or in combination with sulfhydryl containing drugs.
- Author
-
Miller RC, Murley JS, and Grdina DJ
- Subjects
- Acetylcysteine administration & dosage, Acetylcysteine therapeutic use, Acute Radiation Syndrome pathology, Amifostine administration & dosage, Amifostine pharmacology, Amifostine therapeutic use, Animals, Captopril administration & dosage, Captopril pharmacology, Captopril therapeutic use, Cell Line, Transformed, Cell Line, Tumor, Cells, Cultured, Colony-Forming Units Assay, Drug Evaluation, Preclinical, Drug Synergism, Endothelial Cells drug effects, Endothelial Cells radiation effects, Fibroblasts drug effects, Fibroblasts radiation effects, Free Radical Scavengers administration & dosage, Free Radical Scavengers pharmacology, Free Radical Scavengers therapeutic use, Humans, Mesna administration & dosage, Mesna pharmacology, Mesna therapeutic use, Metformin administration & dosage, Metformin pharmacology, Mice, Mice, Inbred C3H, Mice, Inbred C57BL, Radiation Injuries, Experimental pathology, Radiation-Protective Agents administration & dosage, Radiation-Protective Agents pharmacology, Sarcoma pathology, Sulfhydryl Compounds administration & dosage, Sulfhydryl Compounds pharmacology, Acute Radiation Syndrome prevention & control, Metformin therapeutic use, Radiation Injuries, Experimental prevention & control, Radiation-Protective Agents therapeutic use, Sulfhydryl Compounds therapeutic use
- Abstract
Metformin, a biguanide drug used in the treatment of type II diabetes, was evaluated alone and in combination with amifostine, captopril, MESNA or N-acetyl-cysteine (NAC) for its ability to protect when administered 24 h after irradiation. Mouse embryo fibroblasts (MEF), human microvascular endothelial cells (HMEC) and SA-NH mouse sarcoma cells were exposed to 4 Gy in vitro. C3H mice were exposed to 7 Gy and evaluated utilizing an endogenous spleen colony assay system. Amifostine and WR1065, administered 30 min prior to irradiation, were used as positive controls. Treatment of MEF, HMEC and SA-NH cells with metformin elevated survival levels by 1.4-, 1.5- and 1.3-fold compared to 1.9-, 1.8- and 1.6-fold for these same cells treated with WR1065, respectively. Metformin (250 mg/kg) was effective in protecting splenic cells from a 7 Gy dose in vivo (protection factor = 1.8). Amifostine (400 mg/kg), administered 30 min prior to irradiation resulted in a 2.6-fold survival elevation, while metformin administered 24 h after irradiation in combination with NAC (400 mg/kg), MESNA (300 mg/kg) or captopril (200 mg/kg) enhanced survival by 2.6-, 2.8- and 2.4-fold, respectively. Each of these agents has been approved by the FDA for human use and each has a well characterized human safety profile. Metformin alone or in combination with selected sulfhydryl agents possesses radioprotective properties when administered 24 h after radiation exposure comparable to that observed for amifostine administered 30 min prior to irradiation making it a potentially useful agent for radiation countermeasures use.
- Published
- 2014
- Full Text
- View/download PDF
18. Protection from radiation-induced apoptosis by the radioprotector amifostine (WR-2721) is radiation dose dependent.
- Author
-
Ormsby RJ, Lawrence MD, Blyth BJ, Bexis K, Bezak E, Murley JS, Grdina DJ, and Sykes PJ
- Subjects
- Amifostine administration & dosage, Animals, Apoptosis radiation effects, Bone Marrow drug effects, Bone Marrow pathology, Bone Marrow radiation effects, Dose-Response Relationship, Radiation, Female, Male, Mice, Mice, Inbred C57BL, Radiation Injuries, Experimental pathology, Radiation-Protective Agents administration & dosage, Spleen drug effects, Spleen pathology, Spleen radiation effects, Amifostine pharmacology, Apoptosis drug effects, Radiation Injuries, Experimental prevention & control, Radiation-Protective Agents pharmacology
- Abstract
The radioprotective agent amifostine is a free radical scavenger that can protect cells from the damaging effects of ionising radiation when administered prior to radiation exposure. However, amifostine has also been shown to protect cells from chromosomal mutations when administered after radiation exposure. As apoptosis is a common mechanism by which cells with mutations are removed from the cell population, we investigated whether amifostine stimulates apoptosis when administered after radiation exposure. We chose to study a relatively low dose which is the maximum radiation dose for radiation emergency workers (0.25 Gy) and a high dose relevant to radiotherapy exposures (6 Gy). Mice were administered 400 mg/kg amifostine 30 min before, or 3 h after, whole-body irradiation with 0.25 or 6 Gy X-rays and apoptosis was analysed 3 or 7 h later in spleen and bone marrow. We observed a significant increase in radiation-induced apoptosis in the spleen of mice when amifostine was administered before or after 0.25 Gy X-rays. In contrast, when a high dose of radiation was used (6 Gy), amifostine caused a reduction in radiation-induced apoptosis 3 h post-irradiation in spleen and bone marrow similar to previously published studies. This is the first study to investigate the effect of amifostine on radiation-induced apoptosis at a relatively low radiation dose and the first to demonstrate that while amifostine can reduce apoptosis from high doses of radiation, it does not mediate the same effect in response to low-dose exposures. These results suggest that there may be a dose threshold at which amifostine protects from radiation-induced apoptosis and highlight the importance of examining a range of radiation doses and timepoints.
- Published
- 2014
- Full Text
- View/download PDF
19. A survivin-associated adaptive response in radiation therapy.
- Author
-
Grdina DJ, Murley JS, Miller RC, Mauceri HJ, Sutton HG, Li JJ, Woloschak GE, and Weichselbaum RR
- Subjects
- Adaptation, Physiological radiation effects, Animals, Apoptosis radiation effects, Cell Line, Tumor, Cell Survival radiation effects, Female, Humans, Mice, Mice, Inbred C57BL, Mice, Knockout, Pregnancy, Radiation Dosage, Radiation Tolerance, Radiotherapy, Image-Guided methods, Survivin, Xenograft Model Antitumor Assays, Colonic Neoplasms metabolism, Colonic Neoplasms radiotherapy, Inhibitor of Apoptosis Proteins metabolism, Repressor Proteins metabolism, Sarcoma metabolism, Sarcoma radiotherapy
- 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., (©2013 AACR.)
- Published
- 2013
- Full Text
- View/download PDF
20. Comparing radiation toxicities across species: an examination of radiation effects in Mus musculus and Peromyscus leucopus.
- Author
-
Liu W, Haley B, Kwasny MJ, Li JJ, Grdina DJ, Paunesku T, and Woloschak GE
- Subjects
- Animals, Dose-Response Relationship, Radiation, In Vitro Techniques, Incidence, Male, Mice, Mice, Inbred C57BL, Radiation Dosage, Risk Factors, Species Specificity, Mortality, Premature, Radiation Injuries mortality, Survival Rate, Whole-Body Irradiation statistics & numerical data
- Abstract
Purpose: Life shortening and pathological complications in similarly irradiated cohorts of the laboratory mouse Mus musculus and the white-footed mouse Peromyscus leucopus were recorded in the course of the Janus studies conducted at Argonne National Laboratory from 1970-1992. This study examines how lifespan, tumor and non-tumor disease incidence, and tumor multiplicity are differentially affected by gamma-rays and neutron radiation exposure in two different animal species., Materials and Methods: Survival analyses examined differences in lifespan across species, while decision tree analyses examined statistically significant associations between lifespan, radiation exposure, and specific diseases. Logistic regression models were generated to examine the likelihood of disease incidence in these two species following gamma-ray or neutron radiation exposure., Results: Life shortening in response to radiation was more significant in Peromyscus leucopus than in Mus musculus, irrespective of radiation quality. Many types of tumor and non-tumor diseases were found to be consistently species specific. Tumor multiplicity was observed in both species in response to radiation, although more pronounced in Mus musculus., Conclusion: The response to radiation was highly species specific, highlighting the difficulty in extrapolating conclusions from one species to another, irrespective of their phenotypic similarities and ecologic niches.
- Published
- 2013
- Full Text
- View/download PDF
21. CyclinB1/Cdk1 phosphorylates mitochondrial antioxidant MnSOD in cell adaptive response to radiation stress.
- Author
-
Candas D, Fan M, Nantajit D, Vaughan AT, Murley JS, Woloschak GE, Grdina DJ, and Li JJ
- Subjects
- Animals, Antioxidants metabolism, Apoptosis genetics, Apoptosis radiation effects, Cell Cycle genetics, Cell Cycle radiation effects, Cell Line, Tumor, DNA Damage, Female, Humans, Mice, Mice, Inbred BALB C, Mitochondria metabolism, Phosphorylation, Protein Transport, CDC2 Protein Kinase metabolism, Cyclin B1 metabolism, Mitochondria radiation effects, Superoxide Dismutase metabolism
- Abstract
Manganese superoxide dismutase (MnSOD), a major antioxidant enzyme within the mitochondria, is responsible for the detoxification of free radicals generated by cellular metabolism and environmental/therapeutic irradiation. Cell cycle-dependent kinase Cdk1, along with its regulatory partner CyclinB1, plays important roles in the regulation of cell cycle progression as well as in genotoxic stress response. Herein, we identified the presence of the minimal Cdk1 phosphorylation consensus sequence ([S/T]-P; Ser106) in human MnSOD, suggesting Cdk1 as a potential upstream kinase of MnSOD. A substantial amount of CyclinB1/Cdk1 was found to localize in the mitochondrion upon irradiation. The enhanced Cdk1/MnSOD interaction and MnSOD phosphorylation were detected in both the irradiated human cells and mouse tissues. We report that CyclinB1/Cdk1 can regulate MnSOD through reversible Ser106 phosphorylation, both in vivo and in vitro. The CyclinB1/Cdk1-mediated MnSOD Ser106 resulted in increased MnSOD activity and stability, along with improved mitochondrial function and cellular resistance to radiation-induced apoptosis. These results demonstrate a unique pro-survival mechanism by which cells enhance the survival via CyclinB1/Cdk1-mediated MnSOD activation under genotoxic stress conditions.
- Published
- 2013
- Full Text
- View/download PDF
22. A manganese superoxide dismutase (SOD2)-mediated adaptive response.
- Author
-
Grdina DJ, Murley JS, Miller RC, Mauceri HJ, Sutton HG, Thirman MJ, Li JJ, Woloschak GE, and Weichselbaum RR
- Subjects
- Alkylating Agents pharmacology, Animals, Cell Line, Tumor, Cell Survival drug effects, Cell Survival radiation effects, Dose-Response Relationship, Radiation, Ethylnitrosourea pharmacology, Female, Genomic Instability drug effects, Genomic Instability radiation effects, Humans, Mercaptoethylamines pharmacology, Mice, Mice, Inbred C57BL, Pregnancy, Signal Transduction drug effects, Signal Transduction radiation effects, Adaptation, Physiological drug effects, Adaptation, Physiological radiation effects, Superoxide Dismutase metabolism
- 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.
- Published
- 2013
- Full Text
- View/download PDF
23. The effects of radiation and dose-fractionation on cancer and non-tumor disease development.
- Author
-
Liu W, Haley BM, Kwasny MJ, Li JJ, Grdina DJ, Paunesku T, and Woloschak GE
- Subjects
- Animals, Disease Progression, Dose Fractionation, Radiation, Female, Kaplan-Meier Estimate, Male, Mice, Neoplasms, Radiation-Induced mortality, Neoplasms, Radiation-Induced pathology, Proportional Hazards Models, Radiation Injuries, Experimental mortality, Radiation Injuries, Experimental pathology, Risk Assessment, Dose-Response Relationship, Radiation, Gamma Rays adverse effects, Neoplasms, Radiation-Induced etiology, Neutrons adverse effects, Radiation Injuries, Experimental etiology
- Abstract
The Janus series of radiation experiments, conducted from 1970 to 1992, explored the effects of gamma and neutron radiation on animal lifespan and disease development. Data from these experiments presents an opportunity to conduct a large scale analysis of both tumor and non-tumor disease development. This work was focused on a subset of animals from the Janus series of experiments, comparing acute or fractionated exposures of gamma or neutron radiation on the hazards associated with the development of tumor and non-tumor diseases of the liver, lung, kidney or vascular system. This study also examines how the co-occurrence of non-tumor diseases may affect tumor-associated hazards. While exposure to radiation increases the hazard of dying with tumor and non-tumor diseases, dose fractionation modulates these hazards, which varies across different organ systems. Finally, the effect that concurrent non-cancer diseases have on the hazard of dying with a tumor also differs by organ system. These results highlight the complexity in the effects of radiation on the liver, lung, kidney and vascular system.
- Published
- 2012
- Full Text
- View/download PDF
24. Manganese superoxide dismutase interacts with a large scale of cellular and mitochondrial proteins in low-dose radiation-induced adaptive radioprotection.
- Author
-
Eldridge A, Fan M, Woloschak G, Grdina DJ, Chromy BA, and Li JJ
- Subjects
- Apoptosis, Cell Survival, Cells, Cultured, DNA Repair, Dose-Response Relationship, Radiation, Humans, Mitochondria genetics, Mitochondrial Proteins metabolism, Proteomics, Radiation, Ionizing, Signal Transduction, Superoxide Dismutase genetics, Keratinocytes metabolism, Keratinocytes radiation effects, Mitochondria metabolism, Radiation-Protective Agents metabolism, Superoxide Dismutase metabolism
- Abstract
The cellular adaptive response to certain low-level genotoxic stresses, including exposure to low-dose ionizing radiation (LDIR), shows promise as a tool to enhance radioprotection in normal cells but not in tumor cells. Manganese superoxide dismutase (MnSOD), a fundamental mitochondrial antioxidant in mammalian cells, plays a key role in the LDIR-induced adaptive response. In this study, we aimed to elucidate the signaling network associated with MnSOD-induced radiation protection. A MnSOD-interacting protein profile was established in LDIR-treated human skin cells. Human skin keratinocytes (HK18) were irradiated with a single dose of LDIR (10 cGy X-ray) and the cell lysates were immunoprecipitated using α-MnSOD and applied to two different gel-based proteomic experiments followed by mass spectrometry for protein identification. Analysis of the profiles of MnSOD-interacting partners before and after LDIR detected various patterns of MnSOD protein-protein interactions in response to LDIR. Interestingly, many of the MnSOD-interacting proteins are known to have functions related to mitochondrial regulation of cell metabolism, apoptosis, and DNA repair. These results provide evidence indicating that in addition to the enzymatic action of detoxifying superoxide, the antioxidant MnSOD may function as a signaling regulator in stress-induced adaptive protection through cell survival pathways., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
25. SOD2-mediated adaptive responses induced by low-dose ionizing radiation via TNF signaling and amifostine.
- Author
-
Murley JS, Baker KL, Miller RC, Darga TE, Weichselbaum RR, and Grdina DJ
- Subjects
- Amifostine analogs & derivatives, Amifostine chemistry, Animals, Cell Line, Tumor, Enzyme Activation genetics, Enzyme Activation radiation effects, Mercaptoethylamines chemistry, 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, Mercaptoethylamines pharmacology, Superoxide Dismutase metabolism, Tumor Necrosis Factor-alpha pharmacology
- 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., (Copyright © 2011 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
26. SOD2-mediated effects induced by WR1065 and low-dose ionizing radiation on micronucleus formation in RKO human colon carcinoma cells.
- Author
-
Murley JS, Kataoka Y, Miller RC, Li JJ, Woloschak G, and Grdina DJ
- Subjects
- Adaptation, Physiological, Cell Line, Tumor, Humans, Reactive Oxygen Species metabolism, Superoxide Dismutase antagonists & inhibitors, Superoxide Dismutase radiation effects, Superoxides metabolism, Colorectal Neoplasms genetics, Mercaptoethylamines pharmacology, Micronuclei, Chromosome-Defective, Radiation-Protective Agents pharmacology, Superoxide Dismutase physiology
- Abstract
RKO36 cells exposed to either WR1065 or 10 cGy X rays show elevated SOD2 gene expression and SOD2 enzymatic activity. Cells challenged at this time with 2 Gy exhibit enhanced radiation resistance. This phenomenon has been identified as a delayed radioprotective effect or an adaptive response when induced by thiols or low-dose radiation, respectively. In this study we investigated the relative effectiveness of both WR1065 and low-dose radiation in reducing the incidence of radiation-induced micronucleus formation in binucleated RKO36 human colon carcinoma cells. The role of SOD2 in this process was assessed by measuring changes in enzymatic activity as a function of the inducing agent used, the level of protection afforded, and the inhibitory effects of short interfering RNA (SOD2 siRNA). Both WR1065 and 10 cGy X rays effectively induced a greater than threefold elevation in SOD2 activity 24 h after exposure. Cells irradiated at this time with 2 Gy exhibited a significant resistance to micronucleus formation (P < 0.05; Student's two-tailed t test). This protective effect was significantly inhibited in cells transfected with SOD2 siRNA. SOD2 played an important role in the adaptive/delayed radioprotective response by inhibiting the initiation of a superoxide anion-induced ROS cascade leading to enhanced mitochondrial and nuclear damages.
- Published
- 2011
- Full Text
- View/download PDF
27. Amifostine metabolite WR-1065 disrupts homologous recombination in mammalian cells.
- Author
-
Dziegielewski J, Goetz W, Murley JS, Grdina DJ, Morgan WF, and Baulch JE
- Subjects
- Animals, CHO Cells, Camptothecin pharmacology, Cell Line, Tumor, Cricetinae, Cricetulus, DNA-Binding Proteins metabolism, Genomic Instability, Humans, Hydroxyurea pharmacology, RNA Interference, Rad51 Recombinase metabolism, Reactive Oxygen Species metabolism, Amifostine pharmacology, Mercaptoethylamines pharmacology, Radiation-Protective Agents pharmacology, Recombination, Genetic
- Abstract
Repair of DNA damage through homologous recombination (HR) pathways plays a crucial role in maintaining genome stability. However, overstimulation of HR pathways in response to genotoxic stress may abnormally elevate recombination frequencies, leading to increased mutation rates and delayed genomic instability. Radiation-induced genomic instability has been detected after exposure to both low- and high-linear energy transfer (LET) radiations, but the mechanisms responsible for initiating or propagating genomic instability are not known. We have demonstrated that WR-1065, the active metabolite of amifostine, protects against radiation-induced cell killing and delayed genomic instability. We hypothesize that hyperstimulation of HR pathways plays a mechanistic role in radiation-induced genomic instability and that, in part, WR-1065 exerts it radioprotective effect through suppression of the HR pathway. Results of this study demonstrate that WR-1065 treatment selectively protected against radiation-induced cell killing in HR-proficient cell lines compared to an HR-deficient cell line. Further, WR-1065 treatment decreases HR in response to DNA damage using two different mammalian cell systems. This suppression of hyper-recombination is a previously unrecognized mechanism by which WR-1065 effects radioprotection in mammalian cells.
- Published
- 2010
- Full Text
- View/download PDF
28. Coactivation of ATM/ERK/NF-kappaB in the low-dose radiation-induced radioadaptive response in human skin keratinocytes.
- Author
-
Ahmed KM, Nantajit D, Fan M, Murley JS, Grdina DJ, and Li JJ
- Subjects
- Ataxia Telangiectasia Mutated Proteins, Caffeine pharmacology, Cell Cycle Proteins genetics, Cell Line, Transformed, Cell Survival drug effects, DNA-Binding Proteins genetics, Humans, I-kappa B Proteins genetics, Keratinocytes drug effects, Keratinocytes metabolism, Keratinocytes pathology, Morpholines pharmacology, Mutation, Phosphorylation, Protein Serine-Threonine Kinases genetics, Pyrones pharmacology, RNA, Small Interfering genetics, Radiation, Ionizing, Skin drug effects, Skin metabolism, Skin pathology, Tumor Suppressor Proteins genetics, Cell Cycle Proteins metabolism, DNA-Binding Proteins metabolism, Extracellular Signal-Regulated MAP Kinases metabolism, Keratinocytes radiation effects, NF-kappa B metabolism, Protein Serine-Threonine Kinases metabolism, Skin radiation effects, Tumor Suppressor Proteins metabolism
- Abstract
Elucidating the molecular mechanism of the low-dose radiation (LDR)-mediated radioadaptive response is crucial for inventing potential therapeutic approaches to improving normal tissue protection in radiation therapy. ATM, a DNA-damage sensor, is known to activate the stress-sensitive transcription factor NF-kappaB upon exposure to ionizing radiation. This study provides evidence of the cooperative functions of ATM, ERK, and NF-kappaB in inducing a survival advantage through a radioadaptive response as a result of LDR treatment (10 cGy X-rays). By using p53-inhibited human skin keratinocytes, we show that phosphorylation of ATM, MEK, and ERK (but not JNK or p38) is enhanced along with a twofold increase in NF-kappaB luciferase activity at 24 h post-LDR. However, NF-kappaB reporter gene transactivation without a significant enhancement of p65 or p50 protein level suggests that NF-kappaB is activated as a rapid protein response via ATM without involving the transcriptional activation of NF-kappaB subunit genes. A direct interaction between ATM and NF-kappaB p65 is detected in the resting cells and this interaction is significantly increased with LDR treatment. Inhibition of ATM with caffeine, KU-55933, or siRNA or inhibition of the MEK/ERK pathway can block the LDR-induced NF-kappaB activation and eliminate the LDR-induced survival advantage. Altogether, these results suggest a p53-independent prosurvival network involving the coactivation of the ATM, MEK/ERK, and NF-kappaB pathways in LDR-treated human skin keratinocytes, which is absent from mutant IkappaB cells (HK18/mIkappaB), which fail to express NF-kappaB activity.
- Published
- 2009
- Full Text
- View/download PDF
29. Amifostine induces antioxidant enzymatic activities in normal tissues and a transplantable tumor that can affect radiation response.
- Author
-
Grdina DJ, Murley JS, Kataoka Y, Baker KL, Kunnavakkam R, Coleman MC, and Spitz DR
- Subjects
- Animals, Female, Liver enzymology, Lung enzymology, Mercaptoethylamines pharmacology, Mice, Mice, Inbred C3H, Myocardium enzymology, Pancreas enzymology, Radiation Tolerance drug effects, Sarcoma, Experimental enzymology, Amifostine pharmacology, Catalase metabolism, Glutathione Peroxidase metabolism, Radiation-Protective Agents pharmacology, Superoxide Dismutase metabolism
- Abstract
Purpose: To determine whether amifostine can induce elevated manganese superoxide dismutase (SOD2) in murine tissues and a transplantable SA-NH tumor, resulting in a delayed tumor cell radioprotective effect., Methods and Materials: SA-NH tumor-bearing C3H mice were treated with a single 400 mg/kg or three daily 50 mg/kg doses of amifostine administered intraperitoneally. At selected time intervals after the last injection, the heart, liver, lung, pancreas, small intestine, spleen, and SA-NH tumor were removed and analyzed for SOD2, catalase, and glutathione peroxidase (GPx) enzymatic activity. The effect of elevated SOD2 enzymatic activity on the radiation response of SA-NH cells was determined., Results: SOD2 activity was significantly elevated in selected tissues and a tumor 24 h after amifostine treatment. Catalase and GPx activities remained unchanged except for significant elevations in the spleen. GPx was also elevated in the pancreas. SA-NH tumor cells exhibited a twofold elevation in SOD2 activity and a 27% elevation in radiation resistance. Amifostine administered in three daily fractions of 50 mg/kg each also resulted in significant elevations of these antioxidant enzymes., Conclusions: Amifostine can induce a delayed radioprotective effect that correlates with elevated levels of SOD2 activity in SA-NH tumor. If limited to normal tissues, this delayed radioprotective effect offers an additional potential for overall radiation protection. However, amifostine-induced elevation of SOD2 activity in tumors could have an unanticipated deleterious effect on tumor responses to fractionated radiation therapy, given that the radioprotector is administered daily just before each 2-Gy fractionated dose.
- Published
- 2009
- Full Text
- View/download PDF
30. Amifostine reduces lung vascular permeability via suppression of inflammatory signalling.
- Author
-
Fu P, Birukova AA, Xing J, Sammani S, Murley JS, Garcia JG, Grdina DJ, and Birukov KG
- Subjects
- Animals, Antioxidants metabolism, Bronchoalveolar Lavage Fluid, Cytoskeleton metabolism, Inflammation, Interleukin-6 metabolism, Lipopolysaccharides metabolism, Lung metabolism, Male, Mice, Mice, Inbred C57BL, Reactive Oxygen Species, Signal Transduction, Amifostine pharmacology, Capillary Permeability drug effects, Lung drug effects, Radiation-Protective Agents pharmacology
- Abstract
Despite an encouraging outcome of antioxidant therapy in animal models of acute lung injury, effective antioxidant agents for clinical application remain to be developed. The present study investigated the effect of pre-treatment with amifostine, a thiol antioxidant compound, on lung endothelial barrier dysfunction induced by Gram-negative bacteria wall-lipopolysaccharide (LPS). Endothelial permeability was monitored by changes in transendothelial electrical resistance. Cytoskeletal remodelling and reactive oxygen species (ROS) production was examined by immunofluorescence. Cell signalling was assessed by Western blot. Measurements of Evans blue extravasation, cell count and protein content in bronchoalveolar lavage fluid were used as in vivo parameters of lung vascular permeability. Hydrogen peroxide, LPS and interleukin-6 caused cytoskeletal reorganisation and increased permeability in the pulmonary endothelial cells, reflecting endothelial barrier dysfunction. These disruptive effects were inhibited by pre-treatment with amifostine and linked to the amifostine-mediated abrogation of ROS production and redox-sensitive signalling cascades, including p38, extracellular signal regulated kinase 1/2, mitogen-activated protein kinases and the nuclear factor-kappaB pathway. In vivo, concurrent amifostine administration inhibited LPS-induced oxidative stress and p38 mitogen-activated protein kinase activation, which was associated with reduced vascular leak and neutrophil recruitment to the lungs. The present study demonstrates, for the first time, protective effects of amifostine against lipopolysaccharide-induced lung vascular leak in vitro and in animal models of lipopolysaccharide-induced acute lung injury.
- Published
- 2009
- Full Text
- View/download PDF
31. WR-1065, the active metabolite of amifostine, mitigates radiation-induced delayed genomic instability.
- Author
-
Dziegielewski J, Baulch JE, Goetz W, Coleman MC, Spitz DR, Murley JS, Grdina DJ, and Morgan WF
- Subjects
- Cell Line, Tumor, Colorectal Neoplasms pathology, Dose-Response Relationship, Radiation, Green Fluorescent Proteins metabolism, Humans, Micronucleus Tests, Radiation Tolerance, Superoxide Dismutase metabolism, X-Rays, Amifostine pharmacology, Cell Survival drug effects, Cell Survival radiation effects, Genomic Instability drug effects, Mercaptoethylamines pharmacology, Radiation-Protective Agents pharmacology
- 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.
- Published
- 2008
- Full Text
- View/download PDF
32. Incidence of tissue toxicities in gamma ray and fission neutron-exposed mice treated with Amifostine.
- Author
-
Paunesku D, Paunesku T, Wahl A, Kataoka Y, Murley J, Grdina DJ, and Woloschak GE
- Subjects
- Animals, Female, Male, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Organothiophosphorus Compounds pharmacology, Whole-Body Irradiation, Amifostine pharmacology, Gamma Rays adverse effects, Neutrons adverse effects, Radiation-Protective Agents pharmacology
- Abstract
Purpose: To determine the effects of Amifostine or WR-151,327 on the incidence of lethal and non-lethal toxicities in a large cohort of mice exposed to gamma-ray or fission-spectrum neutron radiation., Methods: To analyze data from 4000 B6CF1 mice which received a single whole body irradiation (WBI) with 206 cGy or 417 cGy cobalt-60 gamma rays or 10 cGy or 40 cGy of fission-spectrum neutrons (average energy 0.85 MeV) produced by the Janus reactor at Argonne National Laboratory. In the neutron cohort, Amifostine, WR-151,327, saline or nothing was injected once, intraperitoneally, 30 minutes before irradiation. In the cobalt-60 cohort, WR-151327 was omitted from the same protocol. At the time of natural death, tissue toxicities found in these mice were recorded, and these records were analyzed. While all previous studies focused on the modulation of life shortening effects of WBI by Amifostine, in this study we calculated changes in the frequencies of 59 tissue toxicities and changes in the total number of toxicities per animal., Results: Amifostine protected against specific non-tumor pathological complications (67% of the non-tumor toxicities induced by gamma irradiation, 31% of the neutron induced specific toxicities), as well as specific tumors (56% of the tumor toxicities induced by gamma irradiation, 25% of the neutron induced tumors). Amifostine also reduced the total number of toxicities per animal for both genders in the gamma ray exposed mice and in males in the neutron exposed mice., Conclusions: Amifostine was protective against many, but not all, tissue toxicities caused by WBI gamma and neutron irradiation.
- Published
- 2008
- Full Text
- View/download PDF
33. Maintenance of manganese superoxide dismutase (SOD2)-mediated delayed radioprotection induced by repeated administration of the free thiol form of amifostine.
- Author
-
Murley JS, Nantajit D, Baker KL, Kataoka Y, Li JJ, and Grdina DJ
- Subjects
- Animals, Cells, Cultured, Endothelial Cells drug effects, Endothelial Cells enzymology, Endothelial Cells radiation effects, Humans, Mice, Neoplasms enzymology, Neoplasms pathology, RNA, Small Interfering genetics, Superoxide Dismutase genetics, Amifostine administration & dosage, Sulfhydryl Compounds administration & dosage, Superoxide Dismutase metabolism
- Abstract
Thiol-containing drugs such as WR1065, the free thiol form of amifostine, have been shown to induce a delayed radioprotective effect in both malignant and non-malignant cells. In mammalian cells exposed to a dose as low as 40 microM WR1065, the redox-sensitive nuclear transcription factor kappaB (NFkappaB) is activated, leading to an elevation in the expression of the antioxidant gene manganese superoxide dismutase (SOD2) and a concomitant increase in active SOD2 enzyme levels that peaks 24 to 32 h later. Exposure of cells to ionizing radiation during the period of elevated SOD2 enzymatic activity results in an enhanced radiation resistance. This is seen as an increase in surviving fraction as determined by standard colony formation assays. To determine whether this delayed radioprotection can be maintained over a prolonged period in cells of either malignant or non-malignant origin, both human microvascular endothelial cells (HMEC) and SA-NH mouse sarcoma cells were grown to confluence and exposed to 40 muM WR1065 using three administration protocols: (1) daily drug exposure for 10 days followed each day by irradiation with 2 Gy; (2) drug exposure once every 48 h followed by irradiation with 2 Gy 48 h later for 14 days; and (3) drug exposure every 72 h followed by irradiation with 2 Gy 72 h later for 12 days. As a function of each experimental condition, cell numbers and associated SOD2 enzymatic activities were measured at the time of each irradiation. None of the treatment conditions were toxic to either HMEC or SA-NH cells. SOD2 activity was elevated 5.3- and 1.8-fold over background on average for HMEC exposed to 40 microM WR1065 every 24 or 48 h, respectively. Likewise, SOD2 activity was elevated in SA-NH mouse sarcoma cells 7.8- and 4.9-fold after daily exposure to WR1065 or exposure to WR1065 once every 48 h, respectively. Both HMEC and SA-NH cells exhibited enhanced radiation resistance that correlated with the increase in SOD2 activity. The average respective increases in cell survival were 1.33 +/- 0.01 (SEM), 1.23 +/- 0.01 and 1.04 +/- 0.01 for HMEC exposed to WR1065 every 24, 48 and 72 h, respectively, and 1.27 +/- 0.01, 1.18 +/- 0.02 and 1.02 +/- 0.02 for SA-NH cells exposed to WR1065 every 24, 48 and 72 h, respectively. Both the elevation in WR1065-induced SOD2 enzymatic activity and the corresponding increase in radiation resistance were completely inhibited in HMEC and SA-NH cells transfected with human or mouse SOD2 siRNA oligomers and irradiated 24 h later. These data demonstrate that a delayed radioprotective effect can be induced and maintained over a prolonged period in both non-malignant and malignant cells exposed to thiol-containing drugs such as WR1065. For non-malignant cells this represents a novel paradigm for radiation protection. The ability of WR1065 to induce a persistent elevated radiation resistance in malignant cells, however, suggests a new potential concern regarding the issue of tumor protection in patients exposed to thiol-containing drugs.
- Published
- 2008
- Full Text
- View/download PDF
34. Relationship between phosphorylated histone H2AX formation and cell survival in human microvascular endothelial cells (HMEC) as a function of ionizing radiation exposure in the presence or absence of thiol-containing drugs.
- Author
-
Kataoka Y, Murley JS, Baker KL, and Grdina DJ
- Subjects
- Amifostine analogs & derivatives, Amifostine chemistry, Cell Survival drug effects, Cell Survival radiation effects, Cells, Cultured, Endothelial Cells drug effects, Endothelial Cells radiation effects, Histones chemistry, Humans, Phosphorylation drug effects, Phosphorylation radiation effects, Radiation, Ionizing, Amifostine pharmacology, Endothelial Cells cytology, Endothelial Cells metabolism, Histones metabolism, Sulfhydryl Compounds pharmacology
- Abstract
Human microvascular endothelial cells (HMEC) were exposed to ionizing radiation at doses ranging from 0 to 16 Gy in either the presence or absence of the active thiol forms of amifostine (WR1065), phosphonol (WR255591), N-acetyl-l-cysteine (NAC), captopril or mesna. Each of these clinically relevant thiols, administered to HMEC at a dose of 4 mM for 30 min prior to irradiation, is known to exhibit antioxidant properties. The purpose of this investigation was to determine the relationship(s), if any, between the frequency of radiation-induced histone H2AX phosphorylation at serine 139 (gamma-H2AX) in cells and subsequent survival, as assessed by colony-forming ability, in exposed cell populations as a function of the presence or absence of each of the five thiol compounds during irradiation. gamma-H2AX formation in irradiated cells, as a function of relative DNA content, was quantified by bivariant flow cytometry analysis with FITC-conjugated gamma-H2AX antibody and nuclear DAPI staining. gamma-H2AX formation in cells was measured as the relative fold increase as a function of the treatment conditions. The frequency of gamma-H2AX-positive cells increased with increasing dose of radiation followed by a dose- and time-dependent decay. The most robust response for gamma-H2AX formation occurred 1 h after irradiation with their relative frequencies decreasing as a function of time 4 and 24 h later. To assess the effects of the various thiols on gamma-H2AX formation, all measurements were made 1 h after irradiation. WR1065 was not only effective in protecting HMEC against gamma-H2AX formation across the entire dose range of radiation exposures used, but it was also significantly more cytoprotective than either its prodrug (WR2721) or disulfide (WR33278) analogue. WR1065 had no significant effect on gamma-H2AX formation when administered immediately or up to 30 min after radiation exposure. An inhibitory effect against gamma-H2AX formation induced by 8 Gy of radiation was expressed by each of the thiols tested. NAC, captopril and mesna were equally effective in reducing the frequency of gamma-H2AX formation, with both WR1065 and WR255591 exhibiting a slightly more robust protective effect. Each of the five thiols was effective in reducing the frequency of gamma-H2AX-positive cells across all phases of the cell cycle. In contrast to the relative ability of each of these thiols to inhibit gamma-H2AX formation after irradiation, NAC, captopril and mesna afforded no protection to HMEC as determined using a colony-forming survival assay. Only WR1065 and WR255591 were effective in reducing the frequencies of radiation-induced gamma-H2AX-positive cells as well as protecting against cell death. These results suggest that the use of gamma-H2AX as a biomarker for screening the efficacy of novel antioxidant radioprotective compounds is highly problematic since their formation and disappearance may be linked to processes beyond simply the formation and repair of radiation-induced DSBs.
- Published
- 2007
- Full Text
- View/download PDF
35. Manganese superoxide dismutase (SOD2)-mediated delayed radioprotection induced by the free thiol form of amifostine and tumor necrosis factor alpha.
- Author
-
Murley JS, Kataoka Y, Baker KL, Diamond AM, Morgan WF, and Grdina DJ
- Subjects
- Amifostine administration & dosage, Cell Line, Tumor, Dose-Response Relationship, Drug, Humans, Radiation Tolerance, Cell Survival radiation effects, Colorectal Neoplasms enzymology, Colorectal Neoplasms pathology, Mercaptoethylamines administration & dosage, Radiation-Protective Agents administration & dosage, Superoxide Dismutase metabolism, Tumor Necrosis Factor-alpha administration & dosage
- Abstract
RKO36 cells, a subclone of RKO colorectal carcinoma cells that have been stably transfected with the pCMV-EGFP2Xho vector, were grown to confluence and then exposed to either the radioprotector WR-1065, i.e. the active thiol form of amifostine, for 30 min at doses of 40 microM and 4 mM or the cytokine tumor necrosis factor alpha (TNFalpha, TNFA) for 30 min at a concentration of 10 ng/ml and then washed. Total protein was isolated as a function of time up to 32 h after these treatments. Both doses of WR-1065 as well as the concentration of TNFalpha used were effective in elevating intracellular levels of the antioxidant protein SOD2 (also known as MnSOD) at least 15-fold over background levels as determined by Western blot analysis, while measured SOD2 activity was elevated between 5.5- and 6.9-fold. SOD2 reached a maximal level 24 h and 20 h after WR-1065 and TNFalpha treatments, respectively. The antioxidant proteins catalase and glutathione peroxidase (GPX) were also monitored over the 32-h period. In contrast to the robust changes observed in intracellular levels of SOD2 as a function of time after exposure of cells to WR-1065, catalase levels were elevated only 2.6-fold over background as determined by Western blot analysis, while GPX activity was unaffected by WR-1065 exposure. GPX protein levels were extremely low in cells, and analysis of GPX activity using a spectrophotometric method based on the consumption of reduced NADPH also revealed no measurable change as a function of WR-1065 or TNFalpha exposure. RKO36 cells either were irradiated with X rays in the presence of either 40 microM or 4 mM WR-1065 or 10 ng/ml TNFalpha or were irradiated 24 or 20 h later, respectively, when SOD2 protein levels were most elevated. The concentrations and exposure conditions used for WR-1065 and TNFalpha were not cytotoxic and had no effect on plating efficiencies or cell survival compared to untreated controls. No protection or sensitization was observed for cells irradiated in the presence of 40 microM WR-1065 or TNFalpha. Survival was elevated 1.90-fold for cells irradiated in the presence of 4 mM WR-1065. When RKO36 cells were irradiated with 2 Gy 24 h after 40 microM or 4 mM WR-1065 and 20 h after TNFalpha treatments when SOD2 levels were the most increased, survival was elevated 1.42-, 1.48- and 1.36-fold, respectively. This increased survival represents a SOD2-mediated delayed radioprotective effect. SOD2 appears to be an important antioxidant gene whose inducible expression is an important element in adaptive cellular responses in general, and the delayed radioprotective effect in particular. It can be induced by a range of agents including cytoprotective nonprotein thiols such as WR-1065 and pleiotropic cytokines such as TNFalpha.
- Published
- 2007
- Full Text
- View/download PDF
36. Flow cytometric analysis of phosphorylated histone H2AX following exposure to ionizing radiation in human microvascular endothelial cells.
- Author
-
Kataoka Y, Bindokas VP, Duggan RC, Murley JS, and Grdina DJ
- Subjects
- Cells, Cultured, Dose-Response Relationship, Radiation, Histones ultrastructure, Humans, Microcirculation cytology, Microcirculation physiology, Microcirculation radiation effects, Radiation Dosage, Radiation, Ionizing, DNA radiation effects, DNA Breaks, Endothelial Cells physiology, Endothelial Cells radiation effects, Flow Cytometry methods, Histones genetics, Histones radiation effects
- Abstract
We applied a flow cytometric method to quantify IR-induced histone H2AX phosphorylation at serine 139 (gammaH2AX) and compared those values to those obtained using a standard microscopy based foci counting method. After PFA fixation, methanol permeabilization was suitable for both FITC- or Alexa647-gammaH2AX. In contrast, Alexa647-gammaH2AX was not suitable for ethanol permeabilization. Antibody concentrations at 1-2 microg/ml yielded the highest gammaH2AX positive percentage for both antibodies. Without DAPI staining, gammaH2AX formation can be measured as a relative fold increase. Values determined by bivariant flow cytometric analysis and those obtained using microscopic foci formation exhibited a good quantitative correlation. Values obtained by both methods could vary according to the gating or threshold setting used. gammaH2AX positive cells increased as a function of radiation dose (2-16 Gy) followed by a dose-dependent decay. The free radical scavenger N-acetyl-L-cysteine (NAC), if administered at a concentration of 4 mM 30 min before IR, was effective in reducing IR-induced gammaH2AX formation in all phases of the cell cycle. We have developed a simplified and quantitative flow cytometry based method to measure IR-induced gammaH2AX in cells and demonstrated strong correlation to values obtained by a standard automated digital microscopic foci analysis along with NIH ImageJ custom macro software.
- Published
- 2006
- Full Text
- View/download PDF
37. Delayed radioprotection by nuclear transcription factor kappaB -mediated induction of manganese superoxide dismutase in human microvascular endothelial cells after exposure to the free radical scavenger WR1065.
- Author
-
Murley JS, Kataoka Y, Weydert CJ, Oberley LW, and Grdina DJ
- Subjects
- Blotting, Western, Cells, Cultured, Endothelium, Vascular radiation effects, Enzyme Induction, Humans, Sesquiterpenes pharmacology, Sesquiterpenes, Guaiane, X-Rays, Free Radical Scavengers pharmacology, Mercaptoethylamines pharmacology, NF-kappa B physiology, Radiation-Protective Agents pharmacology, Superoxide Dismutase biosynthesis
- Abstract
The free radical scavenger WR1065 (SH) is the active thiol form of the clinically approved cytoprotector amifostine. At doses of 40 microM and 4 mM it can activate the redox-sensitive nuclear transcription factor kappaB (NFkappaB) and elevate the expression of the antioxidant gene manganese superoxide dismutase (MnSOD) in human microvascular endothelial cells (HMEC). MnSOD contains binding motifs for a number of transcription factors other than NFkappaB and codes for a potent antioxidant enzyme localized in the mitochondria that is known to confer enhanced radiation resistance to cells. The purpose of this study was to determine the effect of WR1065 exposure on the various transcription factors known to affect MnSOD expression along with the subsequent kinetics of intracellular elevation of MnSOD protein levels and associated change in sensitivity to ionizing radiation in HMEC. Cells were grown to confluence and exposed to WR1065 for 30 min. Affects on the transcription factors AP1, AP2, CREB, NFkappaB, and Sp1 were monitored as a function of time ranging from 30 min to 4 h after drug exposure using a gel-shift assay. Only NFkappaB exhibited a marked activation and that occurred 30 min following the cessation of drug exposure. MnSOD protein levels, as determined by Western blot analysis, increased up to 16-fold over background control levels by 16 h following drug treatment, and remained at 10-fold or higher levels for an additional 32 h. MnSOD activity was evaluated using a gel-based assay and was found to be active throughout this time period. HMEC were irradiated with X-rays either in the presence of 40 microM or 4 mM WR1065 or 24 h after its removal when MnSOD levels were most elevated. No protection was observed for cells irradiated in the presence of 40 microM WR1065. In contrast, a 4 mM dose of WR1065 afforded an increase in cell survival by a factor of 2. A "delayed radioprotective" effect was, however, observed when cells were irradiated 24 h later, regardless of the concentration of WR1065 used. This effect is characterized as an increase in survival at the 2 Gy dose point, i.e., a 40% increase in survival, and an increase in the initial slope of the survival curve by a factor of 2. The anti-inflammatory sesquiterpene lactone, Helenalin, is an effective inhibitor of NFkappaB activation. HMEC were exposed to Helenalin for 2 h at a nontoxic concentration of 5 microM prior to exposure to WR1065. This treatment not only inhibited activation of NFkappaB by WR1065, but also inhibited the subsequent elevation of MnSOD and the delayed radioprotective effect. A persistent marked elevation of MnSOD in cells following their exposure to a thiol-containing reducing agent such as WR1065 can result in an elevated resistance to the cytotoxic effects of ionizing radiation and represents a novel radioprotection paradigm.
- Published
- 2006
- Full Text
- View/download PDF
38. Radioprotectors: current status and new directions.
- Author
-
Grdina DJ, Murley JS, Kataoka Y, Zhou D, and Seed TM
- Subjects
- Humans, Radiation Injuries etiology, Radiation Injuries prevention & control, Radiation Protection methods, Radiation Tolerance drug effects, Radiation Tolerance physiology, Radiation-Protective Agents administration & dosage, Radiotherapy adverse effects
- Published
- 2005
39. Models for evaluating agents intended for the prophylaxis, mitigation and treatment of radiation injuries. Report of an NCI Workshop, December 3-4, 2003.
- Author
-
Stone HB, Moulder JE, Coleman CN, Ang KK, Anscher MS, Barcellos-Hoff MH, Dynan WS, Fike JR, Grdina DJ, Greenberger JS, Hauer-Jensen M, Hill RP, Kolesnick RN, Macvittie TJ, Marks C, McBride WH, Metting N, Pellmar T, Purucker M, Robbins ME, Schiestl RH, Seed TM, Tomaszewski JE, Travis EL, Wallner PE, Wolpert M, and Zaharevitz D
- Subjects
- Animals, Central Nervous System radiation effects, Gastrointestinal Tract radiation effects, Hematopoietic System radiation effects, Humans, Kidney radiation effects, Lung radiation effects, Radiation Injuries drug therapy, Skin radiation effects, Zebrafish, Radiation Injuries prevention & control
- Abstract
To develop approaches to prophylaxis/protection, mitigation and treatment of radiation injuries, appropriate models are needed that integrate the complex events that occur in the radiation-exposed organism. While the spectrum of agents in clinical use or preclinical development is limited, new research findings promise improvements in survival after whole-body irradiation and reductions in the risk of adverse effects of radiotherapy. Approaches include agents that act on the initial radiochemical events, agents that prevent or reduce progression of radiation damage, and agents that facilitate recovery from radiation injuries. While the mechanisms of action for most of the agents with known efficacy are yet to be fully determined, many seem to be operating at the tissue, organ or whole animal level as well as the cellular level. Thus research on prophylaxis/protection, mitigation and treatment of radiation injuries will require studies in whole animal models. Discovery, development and delivery of effective radiation modulators will also require collaboration among researchers in diverse fields such as radiation biology, inflammation, physiology, toxicology, immunology, tissue injury, drug development and radiation oncology. Additional investment in training more scientists in radiation biology and in the research portfolio addressing radiological and nuclear terrorism would benefit the general population in case of a radiological terrorism event or a large-scale accidental event as well as benefit patients treated with radiation.
- Published
- 2004
- Full Text
- View/download PDF
40. Delayed radioprotection by NFkappaB-mediated induction of Sod2 (MnSOD) in SA-NH tumor cells after exposure to clinically used thiol-containing drugs.
- Author
-
Murley JS, Kataoka Y, Cao D, Li JJ, Oberley LW, and Grdina DJ
- Subjects
- Acetylcysteine pharmacology, Amifostine pharmacology, Angiotensin-Converting Enzyme Inhibitors pharmacology, Animals, Blotting, Western, Captopril pharmacology, Cell Line, Tumor, Cell Survival, DNA Damage, Dose-Response Relationship, Drug, Dose-Response Relationship, Radiation, I-kappa B Proteins metabolism, Mercaptoethylamines pharmacology, Mesna pharmacology, Mice, Mutation, NF-KappaB Inhibitor alpha, Phosphorylation, Plasmids metabolism, Protective Agents pharmacology, Radiation-Protective Agents pharmacology, Serine chemistry, Sulfhydryl Compounds metabolism, Time Factors, Transfection, NF-kappa B metabolism, Superoxide Dismutase metabolism
- Abstract
The ability of thiol-containing reducing agents to activate transcription factors leading to changes in gene expression and enzyme activities provides an additional mechanism to potentially protect against radiation-induced cell killing. Manganese superoxide dismutase (Sod2) is one such gene whose expression levels have been shown to be elevated after exposure to the thiol compounds WR-1065 and N-acetyl-L-cysteine (NAC), resulting in an increase in radiation resistance. To further characterize this effect, SA-NH sarcoma cells, both wild-type and a clone stably transfected with a plasmid containing an IkappaBalpha gene mutated at serines 32 and 36, which prevents the inducible phosphorylation of these residues and the subsequent activation of NFkappaB (SA-NH+mIkappaBalpha1), were grown to confluence and then exposed to amifostine's free thiol WR-1065 at a concentration of 4 mM for 30 min. Effects of thiol exposure on NFKB activation in SA-NH+mIkappaBalpha1 cells were determined by a gel shift assay, and changes in Sod2 protein levels in these cells 24 h after exposure to 40 microM or 4 mM WR-1065 were measured by Western blot analysis and compared with wild-type cells exposed to the NFkappaB inhibitor BAY 11-7082. Changes in radiation response, measured immediately after thiol exposure or 24 h later, were determined using a colony-forming assay and were correlated with NFKB activation and Sod2 protein levels. The effects of captopril, mesna and NAC, each at a dose of 4 mM, on radiation response were also determined and contrasted with those of WR-1065. Only WR-1065 and captopril protected SA-NH cells when present during irradiation, i.e. 1.57 and 1.31 times increase in survival at 2 Gy, respectively. All four thiols were protective if irradiation with 2 Gy occurred 24 h later; i.e. increases in survival of 1.40, 1.22, 1.35, and 1.25 times were found for WR-1065, captopril, mesna and NAC, respectively. This delayed radioprotective effect correlated with elevated Sod2 protein levels in wild-type SA-NH tumor cells but was not observed in SA-NH+mIkappaBalpha1 cells, indicating that interference with thiol-induced NFKB activation abrogates this delayed radioprotective effect. Because the delayed radioprotective effect is readily demonstrable at a radiation dose of 2 Gy 24 h after exposure to clinically approved thiol-containing drugs such as amifostine, captopril, mesna and NAC, it suggests a new potential concern regarding the issue of tumor protection and the use of these agents in cancer therapy.
- Published
- 2004
- Full Text
- View/download PDF
41. Interaction of amifostine and ionizing radiation on transcriptional patterns of apoptotic genes expressed in human microvascular endothelial cells (HMEC).
- Author
-
Khodarev NN, Kataoka Y, Murley JS, Weichselbaum RR, and Grdina DJ
- Subjects
- Amifostine pharmacology, Apoptosis genetics, Cell Cycle drug effects, Cell Cycle genetics, Cell Cycle radiation effects, Cell Survival drug effects, Cell Survival genetics, Cell Survival radiation effects, Endothelial Cells drug effects, Endothelial Cells radiation effects, Endothelium, Vascular cytology, Gene Expression Profiling methods, Humans, ROC Curve, Transcription, Genetic genetics, Apoptosis drug effects, Apoptosis radiation effects, Mercaptoethylamines pharmacology, Radiation-Protective Agents pharmacology, Transcription, Genetic drug effects, Transcription, Genetic radiation effects
- Abstract
Purpose: Amifostine is a prodrug that requires dephosphorylation by alkaline phosphatase to become activated. This process occurs rapidly within the bloodstream after its i.v. administration to patients undergoing cancer treatment with selected radiation and chemotherapies. Vascular endothelial cells will, therefore, represent a normal cell system that is among the first to experience the radioprotective effects of this agent. Amifostine's active free thiol WR-1065 was investigated to determine its effect on radiation-induced changes in transcriptional patterns and subsequent apoptosis in human microvascular endothelial cells (HMEC) growing in vitro., Methods and Materials: Human microvascular endothelial cells were grown to confluency and then exposed to WR-1065 at a concentration of 4 mM for 30 min, radiation doses that ranged from 0 to 6 Gy, and WR-1065 at a concentration of 4 mM for 30 min before exposure to ionizing radiation. Cell survival was assessed by clonogenic assay, cell cycle phase was analyzed by flow cytometry, apoptosis was also assessed by flow cytometry in which Anexin V staining and sub-G1 fraction analysis were applied, and gene expression was analyzed by the Clontech Atlas Human cDNA array to identify synergistic and antagonistic effects as a function of amifostine and radiation exposure conditions with a focus on apoptotic-related factors., Results: Exposure of HMEC to 4 mM WR-1065 30 min before irradiation resulted in a protection enhancement factor of 2.0; that is, D(O-IRR) of 1.25 Gy and D(O-IRR+WR) of 2.56 Gy. Expression profiling revealed 29 genes that were synergistically activated by the combined action of WR-1065 and ionizing radiation, and an additional 12 genes were synergistically or additively suppressed. In particular, a subset of apoptosis-related genes that included caspases 2, 4, and 9 and different members of the bcl family, along with apoptosis-related receptors, were identified as being significantly affected by the combined treatment of WR-1065 and radiation exposure. In addition, a number of cell cycle-related genes that express cyclins A, G1, G2, and D3 and DNA damage/check point proteins ATM, DNA-PK and RAD23B were also found to be significantly affected. Functional assays of apoptosis were also performed that demonstrated the ability of WR-1065 to protect against radiation-induced apoptosis., Conclusions: WR-1065, the active thiol form of amifostine, is an effective radioprotector of HMEC as determined by use of clonogenic and apoptotic assays for cell survival. Expression profiling successfully defined the transcriptional response of HMEC to both WR-1065 and ionizing radiation exposure, either alone or in combination, and demonstrated both synergistic and antagonistic effects on the expression of different cellular genes, along with corresponding functional responses. The radioprotective effects of amifostine are not limited to its well-characterized physiochemical properties, which include free-radical scavenging, auto-oxidation leading to intracellular hypoxia, and chemical repair by hydrogen atom donation, but include its ability to modulate the complex transcriptional regulation of genes that are involved in apoptosis, cell cycle, and DNA repair.
- Published
- 2004
- Full Text
- View/download PDF
42. Oral administration is as effective as intraperitoneal administration of amifostine in decreasing nitroxide EPR signal decay in vivo.
- Author
-
Elas M, Parasca A, Grdina DJ, and Halpern HJ
- Subjects
- Administration, Oral, Amifostine pharmacokinetics, Animals, Biological Availability, Female, Half-Life, Injections, Intraperitoneal, Mice, Mice, Inbred C3H, Models, Animal, Amifostine administration & dosage, Electron Spin Resonance Spectroscopy methods, Radiation-Protective Agents administration & dosage
- Abstract
A rapid method to determine the systemic incorporation of amifostine has been sought in order to determine the effectiveness of different administration routes without the delay inherent in awaiting therapeutic results. Consistent changes in animal measurements of nitroxide signal decay were monitored using in vivo EPR at frequencies low enough to ensure uniform sensitivity to organs deep in 20-g C3H mice. Conditions included both co-administration of the amifostine with the carbamoyl-proxyl spin probe (CP) via i.p. injection (n=6) and oral administration (n=8) of the amifostine. These decreased the first order rate of decay of the CP EPR signal after a dose of 13.5 Gy radiation, by 23% and 18%, respectively. These changes were significantly different from the rate of decay of the CP EPR signal without amifostine, but were statistically indistinguishable from each other. These data demonstrate: (1) condition-dependent exponential decay of CP EPR signal allowing its use to determine systemic availability of a drug, and (2) that oral administration and i.p. injection of amifostine are both effective in affecting the CP EPR signal decay rate in a mouse model. This is a strong indicator of similar bioavailability in mice from both routes of administration.
- Published
- 2003
- Full Text
- View/download PDF
43. Antimetastatic effectiveness of amifostine therapy following surgical removal of Sa-NH tumors in mice.
- Author
-
Grdina DJ, Kataoka Y, Murley JS, Swedberg K, Lee JY, Hunter N, Weichselbaum RR, and Milas L
- Subjects
- Amifostine administration & dosage, Angiostatins, Animals, Antineoplastic Agents administration & dosage, Mice, Mice, Inbred C3H, Peptide Fragments blood, Plasminogen, Radiation-Protective Agents administration & dosage, Sarcoma, Experimental blood, Sarcoma, Experimental pathology, Sarcoma, Experimental surgery, Amifostine pharmacology, Antineoplastic Agents pharmacology, Cytoprotection, Neoplasm Metastasis, Radiation-Protective Agents pharmacology, Sarcoma, Experimental drug therapy
- Abstract
The effects of dose per fraction on the ability of amifostine exposure to elevate angiostatin levels in the serum of mice and to inhibit spontaneous metastases formation using the well-characterized murine Sa-NH sarcoma were investigated. Amifostine was administered intraperitoneally at doses of 50, 100, or 200 mg/kg every other day for 6 days to C3Hf/Kam mice until tumors reached an average size of 8 mm in diameter. Amifostine was again administered immediately following surgical removal of the tumor-bearing limbs by amputation, and then once more 2 days later. Nontumor-bearing control animals were treated using the same dosing and surgery schedules. The average number of pulmonary metastases per animal was determined for each experimental group. A significant reduction (P <.05) in the average number of pulmonary metastases was observed only in the group of animals exposed to a dose per fraction of 50 mg/kg. A dose of 100 mg/kg was less effective while 200 mg/kg had no effect on metastases formation in this study. The effects of amifostine exposure on serum levels of the angiogenesis inhibitor angiostatin were also determined using Western analysis. Correlating with the antimetastatic effect measured, exposure of animals to 50 mg/kg of amifostine resulted in a four-fold enhanced serum level of angiostatin above control levels. This phenomenon occurred in both tumor-bearing as well as nontumor-bearing animals. In contrast, a dose of 200-mg/kg amifostine administered intraperitoneally under these conditions had no measurable effect on angiostatin serum levels in this animal system. The enhanced ability of relatively low doses of amifostine to inhibit spontaneous metastases formation suggests that effective antimetastatic therapies with amifostine can be designed with minimal toxic side effects. While the dose responses for angiostatin production and metastases inhibition by amifostine are well correlated, the precise mechanism of action underlying these phenomena is unclear but is suggestive of a redox driven process(es)., (Copyright 2002, Elsevier Science (USA). All rights reserved.)
- Published
- 2002
- Full Text
- View/download PDF
44. Delayed cytoprotection after enhancement of Sod2 (MnSOD) gene expression in SA-NH mouse sarcoma cells exposed to WR-1065, the active metabolite of amifostine.
- Author
-
Murley JS, Kataoka Y, Weydert CJ, Oberley LW, and Grdina DJ
- Subjects
- Animals, Cell Survival drug effects, Cell Survival radiation effects, Electrophoresis, Polyacrylamide Gel, Mice, Sarcoma, Experimental genetics, Tumor Cells, Cultured, X-Rays, Gene Expression Regulation, Enzymologic, Mercaptoethylamines pharmacology, Radiation-Protective Agents pharmacology, Sarcoma, Experimental enzymology, Superoxide Dismutase genetics
- Abstract
SA-NH mouse sarcoma cells were grown to confluence and then exposed to either 40 microM or 4 mM of WR-1065, i.e. the active thiol form of amifostine, for 30 min and then washed. Total RNA and protein were isolated at various times up to 24 h after exposure. Both concentrations of WR-1065 were equally effective in affecting Sod2 (also known as MnSOD) gene expression and protein levels. Northern blot analysis using a mouse cDNA probe revealed three Sod2 transcripts of 1, 4 and 6 kb. Expression of both the 4- and 6-kb transcripts increased by 20 and 60%, respectively, and remained elevated over a period of 4 to 20 h. Sod2 protein levels, as determined by Western blot analysis, increased 15-fold over background control levels over the same interval. Sod2 protein was evaluated using activity gels and was found to be active. SA-NH cells were irradiated with X rays either in the presence of 40 microM or 4 mM WR-1065 or 24 h later after its removal, when Sod2 protein levels were most elevated. No protection was observed for cells irradiated in the presence of 40 microM WR-1065. In contrast, survival after a dose of 2 Gy was elevated 1.27-, 1.14- and 1.20-fold in SA-NH cells irradiated in the presence of 4 mM WR-1065 or 24 h after exposure of the cells to 40 microM and 4 mM WR-1065, respectively. The increased survival levels observed 24 h after exposure to WR-1065 represents a delayed radioprotective effect of WR-1065 and corresponds to the time at which Sod2 protein levels are most elevated. These data demonstrate a novel mechanism for radioprotection by WR-1065 and suggest a new potential concern regarding the issue of tumor protection.
- Published
- 2002
- Full Text
- View/download PDF
45. Activation of the nuclear transcription factor kappaB (NFkappaB) and differential gene expression in U87 glioma cells after exposure to the cytoprotector amifostine.
- Author
-
Kataoka Y, Murley JS, Khodarev NN, Weichselbaum RR, and Grdina DJ
- Subjects
- Chemokine CCL5 metabolism, Gene Expression Regulation, Genes, myb drug effects, Genes, myc drug effects, Glioma genetics, Glutathione Transferase drug effects, Glutathione Transferase genetics, Humans, Interleukin-2 Receptor alpha Subunit, Mercaptoethylamines pharmacology, NF-kappa B metabolism, Oxidation-Reduction, Radiobiology, Receptors, Interleukin drug effects, Receptors, Interleukin genetics, Superoxide Dismutase drug effects, Superoxide Dismutase metabolism, Tumor Cells, Cultured, Amifostine pharmacology, Glioma metabolism, NF-kappa B drug effects, Radiation-Protective Agents pharmacology
- Abstract
Purpose: Amifostine has been approved as a therapy to decrease the incidence of moderate-to-severe xerostomia in patients undergoing postoperative radiation treatment for head-and-neck cancer. As a reducing agent capable of participating in intracellular reductive/oxidative processes, it has the potential to affect redox-sensitive transcription factors and gene expression. Amifostine's active free thiol WR-1065 was investigated to determine its effect on nuclear transcription factor kappaB (NFkappaB) activation and subsequent gene expression in U87 glioma cells., Methods and Materials: The human glioma cell line U87 was grown to confluency and then exposed to WR-1065 at a concentration of 40 microM for times ranging from 30 min to 24 h. Changes in cell cycle were monitored by flow cytometry. The effect of WR-1065 on NFkappaB activation was determined by a gel shift assay. Changes in gene expression as a function of time of exposure to WR-1065 were determined by Northern blot and the Atlas Human cDNA Expression Array (Clontech, Palo Alto, CA). Changes in gene expression using the Atlas Array were verified by reverse transcriptase-polymerase chain reaction (RT-PCR) with gene-specific primers., Results: Exposure of U87 cells to 40 microM WR-1065 resulted in a marked activation of NFkappaB between 30 min and 1 h after treatment. Expression of MnSOD, an NFkappaB-responsive gene, was enhanced by over 2-fold after 16 h of treatment and remained elevated at 24 h. During this period of time, no changes in cell cycle distribution were observed. To assess changes in the expression levels of NFkappaB-responsive genes as a function of WR-1065 exposure, cDNA arrays containing 49 genes identified as having DNA-binding motifs for NFkappaB were used. Only five genes were found to be significantly affected at 1, 4, and/or 16 h of treatment. GST-3 and c-myc were repressed up to 2- and 4-fold, respectively. The expression levels of IL-2Ra, RANTES, and c-myb, in contrast, were enhanced up to 14-, 3-, and 2-fold, respectively. The remaining genes having NFkappaB-responsive elements in their promoter regions were either not expressed (20 genes) or were not affected (24 genes) by exposure to WR-1065., Conclusions: The redox-sensitive transcription factor NFkappaB can be activated in U87 glioma cells by the active thiol form of the cytoprotector amifostine. Activation of NFkappaB by the antioxidant WR-1065 is accompanied by a reduced expression of the oncogene c-myc and an enhanced expression of the antioxidant gene MnSOD, a gene whose expression in tumor cells is relatively low, but when overexpressed has been correlated with a suppression of the malignant phenotype. Activation of NFkappaB by WR-1065, however, results in selective rather than global changes in the expression of genes containing NFkappaB-responsive elements.
- Published
- 2002
- Full Text
- View/download PDF
46. Method of RNA purification from endothelial cells for DNA array experiments.
- Author
-
Khodarev NN, Yu J, Nodzenski E, Murley JS, Kataoka Y, Brown CK, Grdina DJ, and Weichselbaum RR
- Subjects
- Animals, Guanidines, Humans, Phenols, Endothelium, Vascular chemistry, Oligonucleotide Array Sequence Analysis, RNA isolation & purification
- Published
- 2002
- Full Text
- View/download PDF
47. Relationships between cytoprotection and mutation prevention by WR-1065.
- Author
-
Grdina DJ, Murley JS, Kataoka Y, and Epperly W
- Subjects
- Amifostine pharmacology, Animals, CHO Cells, Cricetinae, Humans, Mercaptoethylamines pharmacology, Mutation, Radiation-Protective Agents pharmacology
- Abstract
WR-2721, the leading compound that has emerged from the U.S. Army's Drug Synthesis and Development Program, has become the first clinically used cytoprotector in cancer therapy. The ability of its active thiol, WR-1065, to act as an antioxidant to scavenge free radicals, donate hydrogen atoms, and induce auto-oxidation is related to its cytoprotective effectiveness. However, at concentrations 10 to 20 times lower than those required for cytoprotection, this nonprotein thiol also affects gene expression, transcription factor binding, protein phosphorylation levels, and mutagenesis. For example, WR-1065 treatment activates binding of the redox-sensitive transcription factor NF-kappa B to DNA and affects expression of a number of genes, including thymidine kinase, c-myc, and manganese superoxide dismutase. At these lower drug concentrations, phosphorylation levels and subsequent activities on intracellular enzymes are also inhibited. These observations suggest that the mechanisms of action of WR-2721 and its metabolites to protect against cell killing are different from those that protect against mutagenesis and cancer formation. The ability to affect these postexposure processes makes WR-2721 an important agent in the reduction of the carcinogenic risks of environmental exposure to radiation or deleterious chemicals.
- Published
- 2002
48. Inhibition of spontaneous metastases formation by amifostine.
- Author
-
Grdina DJ, Kataoka Y, Murley JS, Hunter N, Weichselbaum RR, and Milas L
- Subjects
- Angiostatins, Animals, Blotting, Western, Cell Division drug effects, Dose-Response Relationship, Drug, Lung Neoplasms enzymology, Lung Neoplasms secondary, Male, Matrix Metalloproteinase 2 metabolism, Matrix Metalloproteinase 9 metabolism, Mercaptoethylamines therapeutic use, Mice, Mice, Inbred C3H, Muscle Neoplasms enzymology, Muscle Neoplasms pathology, Neoplasm Invasiveness, Peptide Fragments metabolism, Plasminogen metabolism, Sarcoma, Experimental enzymology, Sarcoma, Experimental secondary, Xenograft Model Antitumor Assays, Amifostine therapeutic use, Lung Neoplasms prevention & control, Muscle Neoplasms drug therapy, Radiation-Protective Agents therapeutic use, Sarcoma, Experimental prevention & control
- Abstract
Amifostine was investigated for its ability to inhibit spontaneous metastases formation using the well-characterized murine sarcoma, Sa-NH. Amifostine was administered intraperitoneally at a dose of 50 mg/kg every other day for 6 days to C3Hf/Kam mice until tumors reached an average size of 8-8.5 mm in diameter. Amifostine was again administered immediately after surgical removal of the tumor-bearing limbs by amputation, and then once more 2 days later. Twenty-one days later, animals were evaluated for the presence of spontaneously developed pulmonary metastases. Nontumor-bearing control animals were sham treated using the same dosing and surgery schedules. Treatment with amifostine appeared to slightly delay tumor growth, that is, 13 vs. 12 days for tumors to reach an average diameter of 8 mm. Amifostine reduced both the incidence of pulmonary metastases formed in experimental animals from 77% to 57% (p < 0.05), and their average number per animal from 12.8 +/- 5.4 (SEM) to 2.9 +/- 1.1 (SEM). The effect of amifostine exposure on serum levels of the angiogenesis inhibitor angiostatin was also determined using Western blot analysis. Consistent with the antimetastatic effect, exposure of animals to 50 mg/kg of amifostine resulted in a 4-fold enhanced serum level of angiostatin above control levels. This phenomenon occurred in tumor-bearing and nontumor-bearing animals. The effects of amifostine on matrix metalloproteinase (MMP) enzymatic activity was also determined using gelatin zymography. Conditioned growth medium collected from Sa-NH cells grown to confluency was exposed to various concentrations of SH, i.e., 2-[(aminopropyl)amino]ethane-thiol (WR-1065), the active thiol form of amifostine, for either 30 min or 18 hr. WR-1065, as a function of increasing dose and time, inhibited the enzymatic activities of MMP-2 and MMP-9. At a concentration and time of exposure likely to be achieved in vivo, that is, 40 microM and 30 min, MMP-2 and MMP-9 activities were reduced to between 30% and 40% of control values. Consistent with these affects, WR-1065 was also found to be effective in inhibiting the ability of Sa-NH cells to migrate through Matrigel membranes. After an 18-hr exposure under in vitro conditions, WR-1065 at concentrations of 4, 40 and 400 microM, and 4 mM, inhibited Sa-NH migration to 11%, 44%, 81% and 97% of control values, respectively. The abilities of amifostine and its active thiol WR-1065 to stimulate angiostatin production in mice, and to inhibit the MMP enzymatic activities and invasion ability of Sa-NH cells under in vitro conditions, are consistent with the observed antimetastatic effects exhibited against Sa-NH tumors growing in vivo., (Copyright 2002 Wiley-Liss, Inc.)
- Published
- 2002
- Full Text
- View/download PDF
49. Radioprotectants: current status and new directions.
- Author
-
Grdina DJ, Murley JS, and Kataoka Y
- Subjects
- Acrolein adverse effects, Antibiotics, Antineoplastic adverse effects, Antineoplastic Agents, Alkylating adverse effects, Clinical Trials as Topic, Dose-Response Relationship, Drug, Dose-Response Relationship, Radiation, Heart drug effects, Humans, Mesna pharmacology, Oxazines adverse effects, Razoxane pharmacology, Amifostine pharmacology, Mutagenesis drug effects, Radiation-Protective Agents pharmacology, Radiotherapy adverse effects
- Abstract
The ability to prevent radiotherapy-induced toxicity without affecting antitumor efficacy has the potential to enhance the therapeutic benefit for cancer patients without increasing their risk of serious adverse effects. Among the currently available cytoprotective agents capable of protecting normal tissue against damage caused by either chemo- or radiotherapy, only amifostine has been shown in clinical trials to reduce radiation-induced toxicity. Most notably, it reduces the incidence of xerostomia, which is a clinically significant long-term toxicity arising in patients undergoing irradiation of head and neck cancers. In vitro studies with the active metabolite of amifostine (WR-1065) have shown it to prevent both radiation-induced cell death and radiation-induced mutagenesis. The potential of this agent to prevent secondary tumors, as well as other radiation-induced toxicities is now the focus of ongoing research. Among other novel approaches to radioprotection being explored are methods to increase levels of the antioxidant mitochondrial enzyme manganese superoxide dismutase (MnSOD). In addition, the use of epoetin alfa, alone or in combination with cytoprotectants (e.g., amifostine), to treat radiation-induced anemia is also being investigated. The objective of developing newer cytoprotective therapies is to improve the therapeutic ratio by reducing the acute and chronic toxicities associated with more intensive and more effective anticancer therapies., (Copyright 2002 S. Karger AG, Basel)
- Published
- 2002
- Full Text
- View/download PDF
50. Differential activation of nuclear transcription factor kappaB, gene expression, and proteins by amifostine's free thiol in human microvascular endothelial and glioma cells.
- Author
-
Grdina DJ, Murley JS, Kataoka Y, and Calvin DP
- Subjects
- Cell Adhesion Molecules genetics, Cell Adhesion Molecules metabolism, Cells, Cultured, Endothelium, Vascular radiation effects, Glioma radiotherapy, Growth Substances genetics, Growth Substances metabolism, Humans, Monokines genetics, Monokines metabolism, NF-kappa B metabolism, Radiation Dosage, Superoxide Dismutase genetics, Superoxide Dismutase metabolism, Tumor Cells, Cultured radiation effects, Amifostine pharmacology, Endothelium, Vascular metabolism, Gene Expression drug effects, Glioma metabolism, Mercaptoethylamines pharmacology, NF-kappa B genetics, Radiation-Protective Agents pharmacology, Transcriptional Activation drug effects
- Abstract
The effects of WR1065 (SH), the free thiol form of amifostine, on nuclear transcription factor kappaB (NFkappaB) activation, manganese superoxide dismutase (MnSOD) gene expression, and secretion of human vascular endothelial cell growth factor (hVEGF), basic fibroblast growth factor (bFGF), tumor necrosis factor-alpha (TNF-alpha), vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1 (ICAM-1), E-selectin, P-selectin, and interleukins IL-1alpha, IL-6, and IL-8 were investigated and compared in human microvascular endothelial (HMEC) and human glioma cells. WR1065 was evaluated at 2 concentrations, 4 mmol/L, ie, its most effective cytoprotective dose, and 40 micromol/L, a noncytoprotective but highly effective dose capable of preventing radiation and chemotherapeutic drug-induced mutations in exposed cells. A 30-minute exposure of HMEC and glioma cell lines U87 and U251 to WR1065 at either of the concentrations resulted in a marked activation of NFkappaB as determined by a gel shift assay, with the maximum effect observed between 30 minutes and 1 hour after treatment. Using a supershift assay, WR1065 exposure was observed to affect only the p50-p65 heterodimer, and not the homodimers or heterodimers containing p52 or c-Rel subunits of NFkappaB. WR1065 was also found to enhance MnSOD gene expression in both HMEC and glioma cells. Gene expression was enhanced 1.8-fold over control levels in HMEC over a period ranging from 12 to 24 hours after the time of maximum activation of NFkappaB. In contrast, MnSOD gene expression in U87 cells rose 3.5 times above control levels over this same period. WR1065 had no effect on the levels of adhesion molecules, cytokines, and growth factors secreted by cells exposed for up to 24 hours as measured by enzyme-linked immunosorbent assay., (Copyright 2002, Elsevier Science (USA). All rights reserved.)
- Published
- 2002
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.