1
|
Wu YL, Christodoulou AG, Beumer JH, Rigatti LH, Fisher R, Ross M, Watkins S, Cortes DRE, Ruck C, Manzoor S, Wyman SK, Stapleton MC, Goetzman E, Bharathi S, Wipf P, Wang H, Tan T, Christner SM, Guo J, Lo CWY, Epperly MW, Greenberger JS. Mitigation of Fetal Radiation Injury from Mid-Gestation Total-body Irradiation by Maternal Administration of Mitochondrial-Targeted GS-Nitroxide JP4-039. Radiat Res 2024; 202:565-579. [PMID: 39074819 DOI: 10.1667/rade-24-00095.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 06/11/2024] [Indexed: 07/31/2024]
Abstract
Victims of a radiation terrorist event will include pregnant women and unborn fetuses. Mitochondrial dysfunction and oxidative stress are key pathogenic factors of fetal radiation injury. The goal of this preclinical study is to investigate the efficacy of mitigating fetal radiation injury by maternal administration of the mitochondrial-targeted gramicidin S (GS)-nitroxide radiation mitigator JP4-039. Pregnant female C57BL/6NTac mice received 3 Gy total-body irradiation (TBI) at mid-gestation embryonic day 13.5 (E13.5). Using novel time-and-motion-resolved 4D in utero magnetic resonance imaging (4D-uMRI), we found TBI caused extensive injury to the fetal brain that included cerebral hemorrhage, loss of cerebral tissue, and hydrocephalus with excessive accumulation of cerebrospinal fluid (CSF). Histopathology of the fetal mouse brain showed broken cerebral vessels and elevated apoptosis. Further use of novel 4D Oxy-wavelet MRI capable of probing in vivo mitochondrial function in intact brain revealed a significant reduction of mitochondrial function in the fetal brain after 3 Gy TBI. This was validated by ex vivo Oroboros mitochondrial respirometry. One day after TBI (E14.5) maternal administration of JP4-039, which passes through the placenta, significantly reduced fetal brain radiation injury and improved fetal brain mitochondrial respiration. Treatment also preserved cerebral brain tissue integrity and reduced cerebral hemorrhage and cell death. JP4-039 administration following irradiation resulted in increased survival of pups. These findings indicate that JP4-039 can be deployed as a safe and effective mitigator of fetal radiation injury from mid-gestational in utero ionizing radiation exposure.
Collapse
Affiliation(s)
- Yijen L Wu
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15201
- Rangos Research Center Animal Imaging Core, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | - Anthony G Christodoulou
- Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095
| | - Jan H Beumer
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15232
| | - Lora H Rigatti
- Division of Laboratory Animal Resources (DLAR), University of Pittsburgh, Pittsburgh, Pennsylvania 15213
| | - Renee Fisher
- Department of Radiation Oncology, School of Medicine, UPMC Hillman Cancer Center, Pittsburgh, PA 15232
| | - Mark Ross
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Simon Watkins
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Devin R E Cortes
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15201
- Rangos Research Center Animal Imaging Core, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
- Department of Biomedical Engineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Cody Ruck
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15201
- Rangos Research Center Animal Imaging Core, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | - Shanim Manzoor
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15201
- Rangos Research Center Animal Imaging Core, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | - Samuel K Wyman
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15201
- Rangos Research Center Animal Imaging Core, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | - Margaret C Stapleton
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15201
| | - Eric Goetzman
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15201
| | - Sivakama Bharathi
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15201
| | - Peter Wipf
- Department of Biomedical Engineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
- Department of Chemistry, Kenneth P. Dietrich School of Arts & Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Hong Wang
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Tuantuan Tan
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15201
| | - Susan M Christner
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15232
| | - Jianxia Guo
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15232
| | - Cecilia W Y Lo
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15201
| | - Michael W Epperly
- Department of Radiation Oncology, School of Medicine, UPMC Hillman Cancer Center, Pittsburgh, PA 15232
| | - Joel S Greenberger
- Department of Radiation Oncology, School of Medicine, UPMC Hillman Cancer Center, Pittsburgh, PA 15232
| |
Collapse
|
2
|
Wu YL, Christodoulou AG, Beumer JH, Rigatti LH, Fisher R, Ross M, Watkins S, Cortes DRE, Ruck C, Manzoor S, Wyman SK, Stapleton MC, Goetzman E, Bharathi S, Wipf P, Tan T, Eiseman JL, Christner SM, Guo J, Lo CWY, Epperly MW, Greenberger JS. Mitigation of Fetal Irradiation Injury from Mid-Gestation Total Body Radiation with Mitochondrial-Targeted GS-Nitroxide JP4-039. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.13.580105. [PMID: 38405696 PMCID: PMC10888932 DOI: 10.1101/2024.02.13.580105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Victims of a radiation terrorist event will include pregnant women and unborn fetuses. Mitochondrial dysfunction and oxidative stress are key pathogenic factors of fetal irradiation injury. The goal of this preclinical study is to investigate the efficacy of mitigating fetal irradiation injury by maternal administration of the mitochondrial-targeted gramicidin S (GS)- nitroxide radiation mitigator, JP4-039. Pregnant female C57BL/6NTac mice received 3 Gy total body ionizing irradiation (TBI) at mid-gestation embryonic day 13.5 (E13.5). Using novel time- and-motion-resolved 4D in utero magnetic resonance imaging (4D-uMRI), we found TBI caused extensive injury to the fetal brain that included cerebral hemorrhage, loss of cerebral tissue, and hydrocephalus with excessive accumulation of cerebrospinal fluid (CSF). Histopathology of the fetal mouse brain showed broken cerebral vessels and elevated apoptosis. Further use of novel 4D Oxy-wavelet MRI capable of probing in vivo mitochondrial function in intact brain revealed significant reduction of mitochondrial function in the fetal brain after 3Gy TBI. This was validated by ex vivo Oroboros mitochondrial respirometry. Maternal administration JP4-039 one day after TBI (E14.5), which can pass through the placental barrier, significantly reduced fetal brain radiation injury and improved fetal brain mitochondrial respiration. This also preserved cerebral brain tissue integrity and reduced cerebral hemorrhage and cell death. As JP4-039 administration did not change litter sizes or fetus viability, together these findings indicate JP4-039 can be deployed as a safe and effective mitigator of fetal radiation injury from mid-gestational in utero ionizing radiation exposure. One Sentence Summary Mitochondrial-targeted gramicidin S (GS)-nitroxide JP4-039 is safe and effective radiation mitigator for mid-gestational fetal irradiation injury.
Collapse
|
3
|
Ionizing Radiation Induces Disc Annulus Fibrosus Senescence and Matrix Catabolism via MMP-Mediated Pathways. Int J Mol Sci 2022; 23:ijms23074014. [PMID: 35409374 PMCID: PMC8999232 DOI: 10.3390/ijms23074014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 03/31/2022] [Accepted: 04/02/2022] [Indexed: 01/01/2023] Open
Abstract
Previous research has identified an association between external radiation and disc degeneration, but the mechanism was poorly understood. This study explores the effects of ionizing radiation (IR) on inducing cellular senescence of annulus fibrosus (AF) in cell culture and in an in vivo mouse model. Exposure of AF cell culture to 10–15 Gy IR for 5 min followed by 5 days of culture incubation resulted in almost complete senescence induction as evidenced by SA-βgal positive staining of cells and elevated mRNA expression of the p16 and p21 senescent markers. IR-induced senescent AF cells exhibited increased matrix catabolism, including elevated matrix metalloproteinase (MMP)-1 and -3 protein expression and aggrecanolysis. Analogous results were seen with whole body IR-exposed mice, demonstrating that genotoxic stress also drives disc cellular senescence and matrix catabolism in vivo. These results have important clinical implications in the potential adverse effects of ionizing radiation on spinal health.
Collapse
|
4
|
Borg AM, Baker JE. Contemporary biomedical engineering perspective on volitional evolution for human radiotolerance enhancement beyond low-earth orbit. Synth Biol (Oxf) 2021; 6:ysab023. [PMID: 34522784 PMCID: PMC8434797 DOI: 10.1093/synbio/ysab023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 07/15/2021] [Accepted: 09/01/2021] [Indexed: 11/14/2022] Open
Abstract
A primary objective of the National Aeronautics and Space Administration (NASA) is expansion of humankind's presence outside low-Earth orbit, culminating in permanent interplanetary travel and habitation. Having no inherent means of physiological detection or protection against ionizing radiation, humans incur capricious risk when journeying beyond low-Earth orbit for long periods. NASA has made large investments to analyze pathologies from space radiation exposure, emphasizing the importance of characterizing radiation's physiological effects. Because natural evolution would require many generations to confer resistance against space radiation, immediately pragmatic approaches should be considered. Volitional evolution, defined as humans steering their own heredity, may inevitably retrofit the genome to mitigate resultant pathologies from space radiation exposure. Recently, uniquely radioprotective genes have been identified, conferring local or systemic radiotolerance when overexpressed in vitro and in vivo. Aiding in this process, the CRISPR/Cas9 technique is an inexpensive and reproducible instrument capable of making limited additions and deletions to the genome. Although cohorts can be identified and engineered to protect against radiation, alternative and supplemental strategies should be seriously considered. Advanced propulsion and mild synthetic torpor are perhaps the most likely to be integrated. Interfacing artificial intelligence with genetic engineering using predefined boundary conditions may enable the computational modeling of otherwise overly complex biological networks. The ethical context and boundaries of introducing genetically pioneered humans are considered.
Collapse
Affiliation(s)
- Alexander M Borg
- Departments of Biomedical Engineering and Radiation Oncology, Wake Forest University, Winston-Salem, NC, USA
| | - John E Baker
- Radiation Biosciences Laboratory, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
5
|
Epperly MW, Shields D, Fisher R, Hou W, Wang H, Hamade DF, Mukherjee A, Greenberger JS. Radiation-Induced Senescence in p16+/LUC Mouse Lung Compared to Bone Marrow Multilineage Hematopoietic Progenitor Cells. Radiat Res 2021; 196:235-249. [PMID: 34087939 PMCID: PMC8456367 DOI: 10.1667/rade-20-00286.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 05/12/2021] [Indexed: 11/03/2022]
Abstract
We defined the time course of ionizing radiation-induced senescence in lung compared to bone marrow of p16+/LUC mice in which the senescence-induced biomarker (p16) is linked to a luciferase reporter gene. Periodic in situ imaging revealed increased luciferase activity in the lungs of 20 Gy thoracic irradiated, but not 8 Gy total-body irradiated (TBI) mice beginning at day 75 and increasing to day 170. In serial sections of explanted lungs, senescent cells appeared in the same areas as did fibrosis in the 20 Gy thoracic irradiated, but not the 8 Gy TBI group. Lungs from 8 Gy TBI mice at one year did show increased RNA levels for p16, p21, p19 and TGF-β. Individual senescent cells in 20 Gy irradiated mouse lung included those with epithelial, endothelial, fibroblast and hematopoietic cell biomarkers. Rare senescent cells in the lungs of 8 Gy TBI mice at one year were of endothelial phenotype. Long-term bone marrow cultures (LTBMCs) were established at either day 60 or one year after 8 Gy TBI. In freshly removed marrow at both times after irradiation, there were increased senescent cells. In LTBMCs, there were increased senescent cells in both weekly harvested single cells and in colonies of multilineage hematopoietic progenitor cells producing CFU-GEMM (colony forming unit-granulocyte, erythrocyte, monocyte/macrophage, mega-karyocyte) that were formed in secondary cultures when these single cells were plated in semisolid media. LTBMCs from TBI mice produced fewer CFU-GEMM; however, the relative percentage of senescent cell-containing colonies was increased as measured by both p16-luciferase and β-galactosidase. Therefore, 20 Gy thoracic radiation, as well as 8 Gy TBI, induces senescent cells in the lungs. With bone marrow, 8 Gy TBI induced senescence in both hematopoietic cells and in colony-forming progenitors. The p16+/LUC mouse strain provides a valuable system in which to compare the kinetics of radiation-induced senescence between organs in vivo, and to evaluate the potential role of senescent cells in irradiation pulmonary fibrosis.
Collapse
Affiliation(s)
- Michael W. Epperly
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15232
| | - Donna Shields
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15232
| | - Renee Fisher
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15232
| | - Wen Hou
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15232
| | - Hong Wang
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Diala Fatima Hamade
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15232
| | - Amitava Mukherjee
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15232
| | - Joel S. Greenberger
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15232
| |
Collapse
|
6
|
Checker R, Patwardhan RS, Jayakumar S, Maurya DK, Bandekar M, Sharma D, Sandur SK. Chemical and biological basis for development of novel radioprotective drugs for cancer therapy. Free Radic Res 2021; 55:595-625. [PMID: 34181503 DOI: 10.1080/10715762.2021.1876854] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Ionizing radiation (IR) causes chemical changes in biological systems through direct interaction with the macromolecules or by causing radiolysis of water. This property of IR is harnessed in the clinic for radiotherapy in almost 50% of cancers patients. Despite the advent of stereotactic radiotherapy instruments and other advancements in shielding techniques, the inadvertent deposition of radiation dose in the surrounding normal tissue can cause late effects of radiation injury in normal tissues. Radioprotectors, which are chemical or biological agents, can reduce or mitigate these toxic side-effects of radiotherapy in cancer patients and also during radiation accidents. The desired characteristics of an ideal radioprotector include low chemical toxicity, high risk to benefit ratio and specific protection of normal cells against the harmful effects of radiation without compromising the cytotoxic effects of IR on cancer cells. Since reactive oxygen species (ROS) are the major contributors of IR mediated toxicity, plethora of studies have highlighted the potential role of antioxidants to protect against IR induced damage. However, owing to the lack of any clinically approved radioprotector against whole body radiation, researchers have shifted the focus toward finding alternate targets that could be exploited for the development of novel agents. The present review provides a comprehensive insight in to the different strategies, encompassing prime molecular targets, which have been employed to develop radiation protectors/countermeasures. It is anticipated that understanding such factors will lead to the development of novel strategies for increasing the outcome of radiotherapy by minimizing normal tissue toxicity.
Collapse
Affiliation(s)
- Rahul Checker
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India.,Homi Bhabha National Institute, Anushaktinagar, Mumbai, India
| | - Raghavendra S Patwardhan
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India.,Homi Bhabha National Institute, Anushaktinagar, Mumbai, India
| | - Sundarraj Jayakumar
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India
| | - Dharmendra Kumar Maurya
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India.,Homi Bhabha National Institute, Anushaktinagar, Mumbai, India
| | - Mayuri Bandekar
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India
| | - Deepak Sharma
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India.,Homi Bhabha National Institute, Anushaktinagar, Mumbai, India
| | - Santosh K Sandur
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India.,Homi Bhabha National Institute, Anushaktinagar, Mumbai, India
| |
Collapse
|
7
|
van Gisbergen MW, Zwilling E, Dubois LJ. Metabolic Rewiring in Radiation Oncology Toward Improving the Therapeutic Ratio. Front Oncol 2021; 11:653621. [PMID: 34041023 PMCID: PMC8143268 DOI: 10.3389/fonc.2021.653621] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/22/2021] [Indexed: 12/12/2022] Open
Abstract
To meet the anabolic demands of the proliferative potential of tumor cells, malignant cells tend to rewire their metabolic pathways. Although different types of malignant cells share this phenomenon, there is a large intracellular variability how these metabolic patterns are altered. Fortunately, differences in metabolic patterns between normal tissue and malignant cells can be exploited to increase the therapeutic ratio. Modulation of cellular metabolism to improve treatment outcome is an emerging field proposing a variety of promising strategies in primary tumor and metastatic lesion treatment. These strategies, capable of either sensitizing or protecting tissues, target either tumor or normal tissue and are often focused on modulating of tissue oxygenation, hypoxia-inducible factor (HIF) stabilization, glucose metabolism, mitochondrial function and the redox balance. Several compounds or therapies are still in under (pre-)clinical development, while others are already used in clinical practice. Here, we describe different strategies from bench to bedside to optimize the therapeutic ratio through modulation of the cellular metabolism. This review gives an overview of the current state on development and the mechanism of action of modulators affecting cellular metabolism with the aim to improve the radiotherapy response on tumors or to protect the normal tissue and therefore contribute to an improved therapeutic ratio.
Collapse
Affiliation(s)
- Marike W van Gisbergen
- The M-Lab, Department of Precision Medicine, GROW-School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands.,Department of Dermatology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Emma Zwilling
- The M-Lab, Department of Precision Medicine, GROW-School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
| | - Ludwig J Dubois
- The M-Lab, Department of Precision Medicine, GROW-School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
8
|
Late Health Effects of Partial Body Irradiation Injury in a Minipig Model Are Associated with Changes in Systemic and Cardiac IGF-1 Signaling. Int J Mol Sci 2021; 22:ijms22063286. [PMID: 33807089 PMCID: PMC8005067 DOI: 10.3390/ijms22063286] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 12/30/2022] Open
Abstract
Clinical, epidemiological, and experimental evidence demonstrate non-cancer, cardiovascular, and endocrine effects of ionizing radiation exposure including growth hormone deficiency, obesity, metabolic syndrome, diabetes, and hyperinsulinemia. Insulin-like growth factor-1 (IGF-1) signaling perturbations are implicated in development of cardiovascular disease and metabolic syndrome. The minipig is an emerging model for studying radiation effects given its high analogy to human anatomy and physiology. Here we use a minipig model to study late health effects of radiation by exposing male Göttingen minipigs to 1.9–2.0 Gy X-rays (lower limb tibias spared). Animals were monitored for 120 days following irradiation and blood counts, body weight, heart rate, clinical chemistry parameters, and circulating biomarkers were assessed longitudinally. Collagen deposition, histolopathology, IGF-1 signaling, and mRNA sequencing were evaluated in tissues. Our findings indicate a single exposure induced histopathological changes, attenuated circulating IGF-1, and disrupted cardiac IGF-1 signaling. Electrolytes, lipid profiles, liver and kidney markers, and heart rate and rhythm were also affected. In the heart, collagen deposition was significantly increased and transforming growth factor beta-1 (TGF-beta-1) was induced following irradiation; collagen deposition and fibrosis were also observed in the kidney of irradiated animals. Our findings show Göttingen minipigs are a suitable large animal model to study long-term effects of radiation exposure and radiation-induced inhibition of IGF-1 signaling may play a role in development of late organ injuries.
Collapse
|
9
|
Delayed effects of acute whole body lethal radiation exposure in mice pre-treated with BBT-059. Sci Rep 2020; 10:6825. [PMID: 32321983 PMCID: PMC7176697 DOI: 10.1038/s41598-020-63818-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 03/31/2020] [Indexed: 12/20/2022] Open
Abstract
The threat of nuclear exposure is heightened and it is imperative to identify potential countermeasures for acute radiation syndrome. Currently no countermeasures have been approved for prophylactic administration. Effective countermeasures should function to increase survival in the short term as well as to increase the overall prognosis of an exposed individual long term. Here we describe the use of a promising radiation countermeasure, BBT-059, and the results of a long term mouse study (up to 12 months) in the male CD2F1 strain using 60Co gamma irradiation (~0.6 Gy/min, 7.5-12.5 Gy). We report the dose reduction factor of 1.28 for BBT-059 (0.3 mg/kg) compared to control administered 24 h prior to irradiation. In the long term study animals showed accelerated recovery in peripheral blood cell counts, bone marrow colony forming units, sternal cellularity and megakaryocyte numbers in drug treated mice compared to formulation buffer. In addition, increased senescence was observed in the kidneys of animals administered control or drug and exposed to the highest doses of radiation. Decreased levels of E-cadherin, LaminB1 and increased levels of Cyc-D and p21 in spleen lysates were observed in animals administered control. Taken together the results indicate a high level of protection following BBT-059 administration in mice exposed to lethal and supralethal doses of total body gamma-radiation.
Collapse
|
10
|
Epperly MW, Fisher R, Zhang X, Hou W, Shields D, Wipf P, Wang H, Thermozier S, Greenberger JS. Fanconi Anemia Mouse Genotype-specific Mitigation of Total Body Irradiation by GS-Nitroxide JP4-039. In Vivo 2020; 34:33-38. [PMID: 31882460 PMCID: PMC6984088 DOI: 10.21873/invivo.11742] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 10/03/2019] [Accepted: 10/07/2019] [Indexed: 01/06/2023]
Abstract
BACKGROUND/AIM Radiation mitigator, GS-nitroxide, JP4-039, was evaluated for mitigation of total body irradiation (TBI) in Fanconi anemia (FA) Fancd2-/- (129/Sv), Fancg-/- (B6), and Fanca-/- (129/Sv) mice. MATERIALS AND METHODS JP4-039 dissolved in 30% 2-hydroxypropyl-β-cyclodextrin was injected intramuscularly 24 h after total body irradiation (9.25 Gy) into Fanca-/-, Fancd2-/- and Fancg-/- mice. Irradiation survival curves were performed in vitro using bone marrow stromal cell lines derived from Fanca-/-, Fancd2-/- and Fancg-/- mice. RESULTS FA mice demonstrate genotype specific differences in TBI mitigation by JP4-039. Radiation effects in derived bone marrow stromal cell lines in vitro were mitigated by drugs that block apoptosis, but not necroptosis or ferroptosis. CONCLUSION FA mouse models are valuable for elucidating DNA repair pathways in cell and tissue responses to TBI, and the role of drugs that target distinct cell death pathways.
Collapse
Affiliation(s)
- Michael W Epperly
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, U.S.A
| | - Renee Fisher
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, U.S.A
| | - Xichen Zhang
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, U.S.A
| | - Wen Hou
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, U.S.A
| | - Donna Shields
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, U.S.A
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Hong Wang
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Stephanie Thermozier
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, U.S.A
| | - Joel S Greenberger
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, U.S.A.
| |
Collapse
|
11
|
ROS Reduction Does Not Decrease the Anticancer Efficacy of X-Ray in Two Breast Cancer Cell Lines. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3782074. [PMID: 31001373 PMCID: PMC6437742 DOI: 10.1155/2019/3782074] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 01/02/2019] [Indexed: 12/16/2022]
Abstract
Radiotherapy is effective on a large number of cancer types and is one of the most frequently administrated treatments for cancer patients. The anticancer efficacy of X-ray radiotherapy has been frequently correlated with reactive oxygen species (ROS) elevation, which is also a limiting factor for its toxicity on normal tissues. Here, we found that although 4-10 Gy X-rays could significantly reduce cell numbers in both MDA-MB-231 and MCF-7 breast cancer cells, the ROS level changes are less in MCF-7 cells than in MDA-MB-231 cells. Moreover, although both the ROS scavenger N-acetyl-L-cysteine (NAC) and 1 T static magnetic field (SMF) could reduce X-ray-induced ROS elevation, they did not prevent X-ray-induced cell number reduction or cell death increase, which is significantly different from cisplatin. These results demonstrate that although the anticancer efficacy of cisplatin on two breast cancer cell lines is dependent on ROS, the anticancer efficacy of X-ray is not. Moreover, by testing 19 different cell lines, we found that 1 T SMF could effectively reduce ROS levels in multiple cell lines by 10-20%, which encourages further studies to investigate whether SMF could be used as a potential "physical antioxidant" in the future.
Collapse
|
12
|
Sivananthan A, Shields D, Fisher R, Hou W, Zhang X, Franicola D, Epperly MW, Wipf P, Greenberger JS. Continuous One Year Oral Administration of the Radiation Mitigator, MMS350, after Total-Body Irradiation, Restores Bone Marrow Stromal Cell Proliferative Capacity and Reduces Senescence in Fanconi Anemia (Fanca -/-) Mice. Radiat Res 2018; 191:139-153. [PMID: 30499383 DOI: 10.1667/rr15199.1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We quantitated age-related accumulation of senescent cells in irradiated Fanconi anemia (FA) (Fanca-/- mouse cell lines in vitro, and monitored the effect of continuous administration (via drinking water) of the water-soluble radiation mitigator, MMS350, on tissues in vivo over one year after 7.5 Gy total-body irradiation (TBI). Irradiated Fanca-/- mouse bone marrow stromal cell lines showed increased numbers of beta-galactosidase- and p21-positive senescent cells compared to Fanca+/+ cell lines, which was reduced by MMS350. One week after 7.5 Gy TBI, Fanca-/- mice showed increased numbers of senescent cells in spleen compared to Fanca+/+ controls, decreased bone marrow cellularity, failure of explanted bone marrow to proliferate in vitro to form a hematopoietic microenvironment and no detectable single stromal cell cloning capacity. There was no detectable amelioration by MMS350 administration at one week. In contrast, one year post-TBI, Fanca-/- mice demonstrated fewer senescent cells in brain and spleen compared to Fanca+/+ controls. While Fanca-/- mouse bone marrow stromal cells explanted one year post-TBI still failed to proliferate in vitro, continuous oral administration of 400 µ M, MMS350 in drinking water restored explanted stromal cell proliferation. The data indicate that continuous administration of MMS350 modulated several properties of TBI-accelerated aging in Fanca-/- mice as well as control mice, and support further study of MMS350 as a modulator of radiation late effects.
Collapse
Affiliation(s)
- Aranee Sivananthan
- a Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15213
| | - Donna Shields
- a Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15213
| | - Renee Fisher
- a Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15213
| | - Wen Hou
- a Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15213
| | - Xichen Zhang
- a Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15213
| | - Darcy Franicola
- a Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15213
| | - Michael W Epperly
- a Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15213
| | - Peter Wipf
- b Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Joel S Greenberger
- a Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15213
| |
Collapse
|
13
|
Singh VK, Hanlon BK, Santiago PT, Seed TM. A review of radiation countermeasures focusing on injury-specific medicinals and regulatory approval status: part III. Countermeasures under early stages of development along with 'standard of care' medicinal and procedures not requiring regulatory approval for use. Int J Radiat Biol 2017; 93:885-906. [PMID: 28657400 DOI: 10.1080/09553002.2017.1332440] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE Terrorist attacks, with their intent to maximize psychological and economic damage as well as inflicting sickness and death on given targeted populations, are an ever-growing worldwide concern in government and public sectors as they become more frequent, violent, and sensational. If given the chance, it is likely that terrorists will use radiological or nuclear weapons. To thwart these sinister efforts, both physical and medical countermeasures against these weapons are currently being researched and developed so that they can be utilized by the first responders, military, and medical providers alike. This is the third article of a three-part series in which we have reviewed additional radiation countermeasures that are currently under early preclinical phases of development using largely animal models and have listed and discussed clinical support measures, including agents used for radiation-induced emesis, as well as countermeasures not requiring Food and Drug Administration approval. CONCLUSIONS Despite the significant progress that has been made in this area during the last several years, additional effort is needed in order to push promising new agents, currently under development, through the regulatory pipeline. This pipeline for new promising drugs appears to be unreasonably slow and cumbersome; possible reasons for this inefficiency are briefly discussed. Significant and continued effort needs to be afforded to this research and development area, as to date, there is no approved radioprotector that can be administered prior to high dose radiation exposure. This represents a very significant, unmet medical need and a significant security issue. A large number of agents with potential to interact with different biological targets are under development. In the next few years, several additional radiation countermeasures will likely receive Food and Drug Administration approval, increasing treatment options for victims exposed to unwanted ionizing irradiation.
Collapse
Affiliation(s)
- Vijay K Singh
- a Division of Radioprotection, Department of Pharmacology and Molecular Therapeutics , F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences , Bethesda , MD , U.S.A.,b Armed Forces Radiobiology Research Institute , Uniformed Services University of the Health Sciences , Bethesda , MD , U.S.A
| | - Briana K Hanlon
- a Division of Radioprotection, Department of Pharmacology and Molecular Therapeutics , F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences , Bethesda , MD , U.S.A.,b Armed Forces Radiobiology Research Institute , Uniformed Services University of the Health Sciences , Bethesda , MD , U.S.A
| | - Paola T Santiago
- a Division of Radioprotection, Department of Pharmacology and Molecular Therapeutics , F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences , Bethesda , MD , U.S.A.,b Armed Forces Radiobiology Research Institute , Uniformed Services University of the Health Sciences , Bethesda , MD , U.S.A
| | | |
Collapse
|
14
|
Radiation protective effects of baclofen predicted by a computational drug repurposing strategy. Pharmacol Res 2016; 113:475-483. [PMID: 27664700 DOI: 10.1016/j.phrs.2016.09.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 09/19/2016] [Accepted: 09/20/2016] [Indexed: 11/21/2022]
Abstract
Exposure to ionizing radiation causes damage to living tissues; however, only a small number of agents have been approved for use in radiation injuries. Radioprotector is the primary countermeasure to radiation injury and none radioprotector has indeed reached the drug development stage. Repurposing the long list of approved, non-radioprotective drugs is an attractive strategy to find new radioprotective agents. Here, we applied a computational approach to discover new radioprotectors in silico by comparing publicly available gene expression data of ionizing radiation-treated samples from the Gene Expression Omnibus (GEO) database with gene expression signatures of more than 1309 small-molecule compounds from the Connectivity Map (cmap) dataset. Among the best compounds predicted to be therapeutic for ionizing radiation damage by this approach were some previously reported radioprotectors and baclofen (P<0.01), a chemical that was not previously used as radioprotector. Validation using a cell-based model and a rodent in vivo model demonstrated that treatment with baclofen reduced radiation-induced cytotoxicity in vitro (P<0.01), attenuated bone marrow damage and increased survival in vivo (P<0.05). These findings suggest that baclofen might serve as a radioprotector. The drug repurposing strategy by connecting the GEO data and cmap can be used to identify known drugs as potential radioprotective agents.
Collapse
|
15
|
Sheshadri P, Kumar A. Managing odds in stem cells: insights into the role of mitochondrial antioxidant enzyme MnSOD. Free Radic Res 2016; 50:570-84. [DOI: 10.3109/10715762.2016.1155708] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
16
|
Gene therapy for radioprotection. Cancer Gene Ther 2015; 22:172-80. [PMID: 25721205 DOI: 10.1038/cgt.2015.8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 08/01/2014] [Accepted: 01/22/2015] [Indexed: 11/08/2022]
Abstract
Radiation therapy is a critical component of cancer treatment with over half of patients receiving radiation during their treatment. Despite advances in image-guided therapy and dose fractionation, patients receiving radiation therapy are still at risk for side effects due to off-target radiation damage of normal tissues. To reduce normal tissue damage, researchers have sought radioprotectors, which are agents capable of protecting tissue against radiation by preventing radiation damage from occurring or by decreasing cell death in the presence of radiation damage. Although much early research focused on small-molecule radioprotectors, there has been a growing interest in gene therapy for radioprotection. The amenability of gene therapy vectors to targeting, as well as the flexibility of gene therapy to accomplish ablation or augmentation of biologically relevant genes, makes gene therapy an excellent strategy for radioprotection. Future improvements to vector targeting and delivery should greatly enhance radioprotection through gene therapy.
Collapse
|
17
|
Greenberger J, Kagan V, Bayir H, Wipf P, Epperly M. Antioxidant Approaches to Management of Ionizing Irradiation Injury. Antioxidants (Basel) 2015; 4:82-101. [PMID: 26785339 PMCID: PMC4665573 DOI: 10.3390/antiox4010082] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 01/12/2015] [Indexed: 11/25/2022] Open
Abstract
Ionizing irradiation induces acute and chronic injury to tissues and organs. Applications of antioxidant therapies for the management of ionizing irradiation injury fall into three categories: (1) radiation counter measures against total or partial body irradiation; (2) normal tissue protection against acute organ specific ionizing irradiation injury; and (3) prevention of chronic/late radiation tissue and organ injury. The development of antioxidant therapies to ameliorate ionizing irradiation injury began with initial studies on gene therapy using Manganese Superoxide Dismutase (MnSOD) transgene approaches and evolved into applications of small molecule radiation protectors and mitigators. The understanding of the multiple steps in ionizing radiation-induced cellular, tissue, and organ injury, as well as total body effects is required to optimize the use of antioxidant therapies, and to sequence such approaches with targeted therapies for the multiple steps in the irradiation damage response.
Collapse
Affiliation(s)
- Joel Greenberger
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, 5150 Centre Avenue, Rm. 533, Pittsburgh, PA 15232, USA.
| | - Valerian Kagan
- Department of Environmental/Occupational Health, University of Pittsburgh, Pittsburgh, PA 15219, USA.
| | - Hulya Bayir
- Department of Critical Care Medicine, Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA.
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA 15260, USA.
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA.
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA.
| | - Michael Epperly
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, 5150 Centre Avenue, Rm. 533, Pittsburgh, PA 15232, USA.
| |
Collapse
|
18
|
Barshishat-Kupper M, Tipton AJ, McCart EA, McCue J, Mueller GP, Day RM. Effect of ionizing radiation on liver protein oxidation and metabolic function in C57BL/6J mice. Int J Radiat Biol 2014; 90:1169-78. [PMID: 24899392 DOI: 10.3109/09553002.2014.930536] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE Protein oxidation in response to radiation results in DNA damage, endoplasmic reticulum stress/unfolded protein response, cell cycle arrest, cell death and senescence. The liver, a relatively radiosensitive organ, undergoes measurable alterations in metabolic functions following irradiation. Accordingly, we investigated radiation-induced changes in liver metabolism and alterations in protein oxidation. MATERIALS AND METHODS C57BL/6 mice were sham irradiated or exposed to 8.5 Gy (60)Co (0.6 Gy/min) total body irradiation. Metabolites and metabolic enzymes in the blood and liver tissue were analyzed. Two-dimensional gel electrophoresis and OxyBlot™ were used to detect carbonylated proteins that were then identified by peptide mass fingerprinting. RESULTS Analysis of serum metabolites revealed elevated glucose, bilirubin, lactate dehydrogenase (LDH), high-density lipoprotein, and aspartate aminotransferase within 24-72 h post irradiation. Liver tissue LDH and alkaline phosphatase activities were elevated 24-72 h post irradiation. OxyBlotting revealed that the hepatic proteome contains baseline protein carbonylation. Radiation exposure increased carbonylation of specific liver proteins including carbonic anhydrase 1, α-enolase, and regucalcin. CONCLUSIONS 8.5 Gy irradiation resulted in distinct metabolic alterations in hepatic functions. Coincident with these changes, radiation induced the carbonylation of specific liver enzymes. The oxidation of liver enzymes may underlie some radiation-induced alterations in hepatic function.
Collapse
Affiliation(s)
- Michal Barshishat-Kupper
- Department of Pharmacology, Uniformed Services University of the Health Sciences , Bethesda, MD , USA
| | | | | | | | | | | |
Collapse
|
19
|
Epperly MW, Goff JP, Franicola D, Wang H, Wipf P, Li S, Greenberger JS. Esophageal radioprotection by swallowed JP4-039/F15 in thoracic-irradiated mice with transgenic lung tumors. In Vivo 2014; 28:435-440. [PMID: 24982207 PMCID: PMC6436097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
BACKGROUND/AIM To determine whether Gramicidin S (GS)-nitroxide, JP4-039, esophageal radiation protection protected lung tumors in a transgenic model, LoxP-Stoop-LoxP Kristen Rat Sarcoma Viral oncogene (LSL-K-RAS) mice were administered intra-tracheal- Carbapenem-resistant Enterobacteriaceae (CRE) recombinase, bilateral lung tumors were confirmed at 11 weeks, then thoracic irradiation was delivered. MATERIALS AND METHODS Mice received single-fraction 15 Gy or 24 Gy to both lungs, in subgroups receiving intraesophageal administration 10 min before irradiation of JP4-039 (in F15 emulsion) tumor size reduction and survival were investigated. Mice were followed for survival, and reduction in tumor size. RESULTS There was no evidence of tumor radioprotection in mice receiving JP4-039/F15. CONCLUSION Intraesophageal radioprotective small-molecule antioxidant therapy protects normal tissue but not tumor tissue in mice with transgenic lung tumors.
Collapse
Affiliation(s)
- Michael W Epperly
- Department of Radiation Oncology, UPCI Cancer Institute, Pittsburgh, PA, USA
| | - Julie P Goff
- Department of Radiation Oncology, UPCI Cancer Institute, Pittsburgh, PA, USA
| | - Darcy Franicola
- Department of Radiation Oncology, UPCI Cancer Institute, Pittsburgh, PA, USA
| | - Hong Wang
- Department of Radiation Oncology, UPCI Cancer Institute, Pittsburgh, PA, USA
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, U.S.A. Center for Chemical Methodologies & Library Development, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Song Li
- Department of Pharmaceutical Science, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Joel S Greenberger
- Department of Radiation Oncology, UPCI Cancer Institute, Pittsburgh, PA, USA
| |
Collapse
|
20
|
Epperly MW, Bahary N, Quader M, Dewald V, Greenberger JS. The zebrafish--Danio rerio--is a useful model for measuring the effects of small-molecule mitigators of late effects of ionizing irradiation. In Vivo 2012; 26:889-897. [PMID: 23160669 PMCID: PMC3775014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
BACKGROUND/AIM Use of zebrafish models may decrease the cost of screening new irradiation protectors and mitigators. MATERIALS AND METHODS Zebrafish (Danio rerio) models were tested for screening water-soluble radiation protectors and mitigators. Irradiation of embryos and monitoring survival, and measuring fibrosis of the caudal musculature of adults allowed for testing of acute and late effects, respectively. RESULTS Incubation of zebrafish embryos either before or after irradiation in ethyl pyruvate (1 mM) increased survival. Irradiation of adults to 15 to 75 Gy, delivered in single-fraction at 13 Gy/min, showed dose-dependent fibrosis at 30 days, quantitated as physiological decrease in swimming tail movement, and histopathological detection of collagen deposition in the dorsal musculature. Continuous administration of small-molecule radioprotector drugs in the water after irradiation reduced both acute and chronic injuries. CONCLUSION The zebrafish is cost-effective for screening new radiation countermeasures.
Collapse
Affiliation(s)
- Michael W Epperly
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15232, USA
| | | | | | | | | |
Collapse
|
21
|
Fullerene nanoparticles and their anti-oxidative effects: a comparison to other radioprotective agents. J Appl Biomed 2012. [DOI: 10.2478/v10136-012-0002-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
22
|
Dysregulated in vitro hematopoiesis, radiosensitivity, proliferation, and osteoblastogenesis with marrow from SAMP6 mice. Exp Hematol 2012; 40:499-509. [PMID: 22326715 DOI: 10.1016/j.exphem.2012.01.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Revised: 01/26/2012] [Accepted: 01/31/2012] [Indexed: 01/12/2023]
Abstract
The senescence accelerated-prone mouse variant 6 (SAMP6) shows normal growth followed by rapid aging, development of osteopenia, and shortened lifespan, compared with control R1 mice. Because oxidative stress is a fundamental mechanism of tissue aging, we tested whether cellular parameters that are associated with oxidative stress are impaired with marrow from SAMP6 mice. We compared in vitro hematopoiesis, irradiation sensitivity, proliferative potential, and osteoblastogenesis with marrow cells from SAMP6 and R1 mice. Marrow cells from SAMP6 mice showed shortened in vitro hematopoiesis; their stromal cells showed greater radiation sensitivity and decreased proliferation. Consistent with those properties, there was constitutive upregulation of transforming growth factor-β(1), an inhibitor of hematopoiesis, and of cell cycle inhibitory genes, p16(INK4A) and p19(ARF). Paradoxically, there was constitutive expression of osteoblast genes in stromal cells from SAMP6 mice, but in vitro matrix mineralization was impaired. These studies and data included in other reports indicate that impaired proliferation of osteoblast progenitors in SAMP6 marrow may be a major factor contributing to accelerated loss of bone mass. In sum, marrow from SAMP6 mice had diminished capacity for long-term hematopoiesis, increased radiosensitivity, and reduced proliferative capacity.
Collapse
|
23
|
Greenberger JS, Clump D, Kagan V, Bayir H, Lazo JS, Wipf P, Li S, Gao X, Epperly MW. Strategies for discovery of small molecule radiation protectors and radiation mitigators. Front Oncol 2012; 1:59. [PMID: 22655254 PMCID: PMC3356036 DOI: 10.3389/fonc.2011.00059] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 12/20/2011] [Indexed: 01/01/2023] Open
Abstract
Mitochondrial targeted radiation damage protectors (delivered prior to irradiation) and mitigators (delivered after irradiation, but before the appearance of symptoms associated with radiation syndrome) have been a recent focus in drug discovery for (1) normal tissue radiation protection during fractionated radiotherapy, and (2) radiation terrorism counter measures. Several categories of such molecules have been discovered: nitroxide-linked hybrid molecules, including GS-nitroxide, GS-nitric oxide synthase inhibitors, p53/mdm2/mdm4 inhibitors, and pharmaceutical agents including inhibitors of the phosphoinositide-3-kinase pathway and the anti-seizure medicine, carbamazepine. Evaluation of potential new radiation dose modifying molecules to protect normal tissue includes: clonogenic radiation survival curves, assays for apoptosis and DNA repair, and irradiation-induced depletion of antioxidant stores. Studies of organ specific radioprotection and in total body irradiation-induced hematopoietic syndrome in the mouse model for protection/mitigation facilitate rational means by which to move candidate small molecule drugs along the drug discovery pipeline into clinical development.
Collapse
Affiliation(s)
- Joel S. Greenberger
- Radiation Oncology Department, University of Pittsburgh Cancer InstitutePittsburgh, PA, USA
| | - David Clump
- Radiation Oncology Department, University of Pittsburgh Cancer InstitutePittsburgh, PA, USA
| | - Valerian Kagan
- Environmental and Occupational Health Department, University of PittsburghPittsburgh, PA, USA
| | - Hülya Bayir
- Critical Care Medicine Department, University of Pittsburgh Medical CenterPittsburgh, PA, USA
| | - John S. Lazo
- Pharmacology Department, University of VirginiaCharlottesville, VA, USA
| | - Peter Wipf
- Department of Chemistry, Accelerated Chemical Discovery Center, University of PittsburghPittsburgh, PA, USA
| | - Song Li
- Pharmaceutical Science Department, University of PittsburghPittsburgh, PA, USA
| | - Xiang Gao
- Pharmaceutical Science Department, University of PittsburghPittsburgh, PA, USA
| | - Michael W. Epperly
- Radiation Oncology Department, University of Pittsburgh Cancer InstitutePittsburgh, PA, USA
| |
Collapse
|
24
|
Delayed Effects of Radiation on Mitochondrial DNA in Radiation-Sensitive Organs. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011. [DOI: 10.1007/978-1-4614-1566-4_21] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register]
|
25
|
Bernard ME, Kim H, Rwigema JC, Epperly MW, Kelley EE, Murdoch GH, Dixon T, Wang H, Greenberger JS. Role of the esophageal vagus neural pathway in ionizing irradiation-induced seizures in nitric oxide synthase-1 homologous recombinant negative NOS1-/- mice. In Vivo 2011; 25:861-869. [PMID: 22021678 PMCID: PMC3593194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
AIM We sought to define the mechanism of total body irradiation (TBI)-induced seizures in NOS1(-/-) mice and amelioration by intra-esophageal manganese superoxide dismutase-plasmid liposomes (MnSOD-PL). MATERIALS AND METHODS We evaluated the role of vagus nerve pathways in irradiation-induced seizures using biochemical, physiologic, and histopathologic techniques. RESULTS Heterozygous NOS1(+/-) mice demonstrated radioresistance similar to wild-type C57BL/6NHsd mice (p=0.9269). Irradiation-induced lipid peroxidation in fetal brain cultures from NOS1(-/-) or wild-type mice was reduced by MnSOD-PL. Right-sided vagotomy did not alter the TBI radiation response of wild-type or reverse the radiosensitivity of NOS1(-/-) mice. Excised esophagus from irradiated NOS1(-/-) mice demonstrated an increased histopathologic inflammatory response compared to C57BL/6NHsd mice. CONCLUSION NOS1(-/-) mice represent a model system for dissecting the developmental abnormalities leading to esophageal-mediated TBI-induced seizures.
Collapse
Affiliation(s)
- Mark E Bernard
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, 5150 Centre Avenue, Pittsburgh, PA 15232, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Williams JP, McBride WH. After the bomb drops: a new look at radiation-induced multiple organ dysfunction syndrome (MODS). Int J Radiat Biol 2011; 87:851-68. [PMID: 21417595 PMCID: PMC3314299 DOI: 10.3109/09553002.2011.560996] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE There is increasing concern that, since the Cold War era, there has been little progress regarding the availability of medical countermeasures in the event of either a radiological or nuclear incident. Fortunately, since much is known about the acute consequences that are likely to be experienced by an exposed population, the probability of survival from the immediate hematological crises after total body irradiation (TBI) has improved in recent years. Therefore focus has begun to shift towards later down-stream effects, seen in such organs as the gastrointestinal tract (GI), skin, and lung. However, the mechanisms underlying therapy-related normal tissue late effects, resulting from localised irradiation, have remained somewhat elusive and even less is known about the development of the delayed syndrome seen in the context of whole body exposures, when it is likely that systemic perturbations may alter tissue microenvironments and homeostasis. CONCLUSIONS The sequence of organ failures observed after near-lethal TBI doses are similar in many ways to that of multiple organ dysfunction syndrome (MODS), leading to multiple organ failure (MOF). In this review, we compare the mechanistic pathways that underlie both MODS and delayed normal tissue effects since these may impact on strategies to identify radiation countermeasures.
Collapse
Affiliation(s)
- Jacqueline P Williams
- Department of Radiation Oncology, University of Rochester Medical Center Rochester, NY 14642, USA.
| | | |
Collapse
|
27
|
Epperly MW, Wang H, Jones JA, Dixon T, Montesinos CA, Greenberger JS. Antioxidant-chemoprevention diet ameliorates late effects of total-body irradiation and supplements radioprotection by MnSOD-plasmid liposome administration. Radiat Res 2011; 175:759-65. [PMID: 21466381 DOI: 10.1667/rr2398.1] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Abstract Many acute and chronic effects of ionizing radiation are mediated by reactive oxygen species and reactive nitrogen species, which deplete antioxidant stores, leading to cellular apoptosis, stem cell depletion and accelerated aging. C57BL/6NHsd mice receiving intravenous MnSOD-PL prior to 9.5 Gy total-body irradiation (TBI) show increased survival from the acute hematopoietic syndrome, and males demonstrated improved long-term survival (Epperly et al., Radiat. Res. 170, 437-444, 2008). We evaluated the effect of an antioxidant-chemopreventive diet compared to a regular diet on long-term survival in female mice. Twenty-four hours before the LD(50/30) dose of 9.5 Gy TBI, subgroups of mice were injected intravenously with MnSOD-PL (100 μg plasmid DNA in 100 μl of liposomes). Mice on either diet treated with MnSOD-PL showed decreased death after irradiation compared to irradiated mice on the house diet alone (P = 0.031 for the house diet plus MnSOD-PL or 0.015 for antioxidant diet plus MnSOD-PL). The mice on the antioxidant-chemoprevention diet alone or with MnSOD-PL that survived 30 days after irradiation had a significant increase in survival compared to mice on the regular diet (P = 0.04 or 0.01, respectively). In addition, mice treated with MnSOD-PL only and surviving 30 days after radiation also had increased survival compared to those on the regular diet alone (P = 0.02). Survivors of acute ionizing radiation damage have ameliorated life shortening if they are fed an antioxidant-chemopreventive diet.
Collapse
Affiliation(s)
- Michael W Epperly
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania 15232, USA
| | | | | | | | | | | |
Collapse
|
28
|
Epperly MW, Smith T, Zhang X, Goff JP, Franicola D, Greenberger B, Komanduri P, Wang H, Greenberger JS. Modulation of in utero total body irradiation induced newborn mouse growth retardation by maternal manganese superoxide dismutase-plasmid liposome (MnSOD-PL) gene therapy. Gene Ther 2011; 18:579-83. [PMID: 21248791 PMCID: PMC3111807 DOI: 10.1038/gt.2010.178] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
To determine the effects of manganese superoxide dismutase (MnSOD) plasmid liposome (PL) maternal radioprotection on fetal mice, timed pregnant female mice (E14 gestation) were irradiated to 3.0 Gy total body irradiation (TBI) dose, and the number, weight and growth and development over 6 months after birth of newborn mice was quantitated compared with irradiated controls. Maternal MnSOD-PL treatment at E13 improved pup survival at birth (5.4±0.9 per litter) compared with non-irradiated 3.0 Gy controls 4.9±1.1. There was no statistically significant difference in newborn abnormalities, male to female ratio in newborn litters, or other evidence of teratogenesis in surviving newborn mice from MnSOD-PL treated compared with irradiated controls. However, E14 3 Gy irradiated pups from gene therapy-treated mothers showed a significant increase in both growth and overall survival over 6 months after birth (P=0.0022). To determine if transgene product crossed the placenta pregnant E13 mice were injected intravenously with hemagglutinin-epitope-tagged MnSOD (100 μg plasmid in 100 μl liposomes), then after 24 h, fetal mice, placentas and maternal tissues were removed and tested by both immunohistochemistry and reverse transcriptase-PCR for transgene and product. There was no evidence of transgene or product in placenta or any fetal tissue while maternal liver was positive by both assays. The data provide evidence for fetal radioprotection by maternal MnSOD-PL gene therapy before irradiation, which is mediated by an indirect bystander effect and is associated with a significant improvement in both survival at birth and growth and development of newborn mice.
Collapse
Affiliation(s)
- M W Epperly
- Department of Radiation Oncology, UPCI Cancer Institute, Pittsburgh, PA 15232, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Rajagopalan MS, Stone B, Rwigema JC, Salimi U, Epperly MW, Goff J, Franicola D, Dixon T, Cao S, Zhang X, Buchholz BM, Bauer AJ, Choi S, Bakkenist C, Wang H, Greenberger JS. Intraesophageal manganese superoxide dismutase-plasmid liposomes ameliorates novel total-body and thoracic radiation sensitivity of NOS1-/- mice. Radiat Res 2010; 174:297-312. [PMID: 20726721 DOI: 10.1667/rr2019.1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The effect of deletion of the nitric oxide synthase 1 gene (NOS1(-/-)) on radiosensitivity was determined. In vitro, long-term cultures of bone marrow stromal cells derived from NOS1(-/-) were more radioresistant than cells from C57BL/6NHsd (wild-type), NOS2(-/-) or NOS3(-/-) mice. Mice from each strain received 20 Gy thoracic irradiation or 9.5 Gy total-body irradiation (TBI), and NOS1(-/-) mice were more sensitive to both. To determine the etiology of radiosensitivity, studies of histopathology, lower esophageal contractility, gastrointestinal transit, blood counts, electrolytes and inflammatory markers were performed; no significant differences between irradiated NOS1(-/-) and control mice were found. Video camera surveillance revealed the cause of death in NOS1(-/-) mice to be grand mal seizures; control mice died with fatigue and listlessness associated with low blood counts after TBI. NOS1(-/-) mice were not sensitive to brain-only irradiation. MnSOD-PL therapy delivered to the esophagus of wild-type and NOS1(-/-) mice resulted in equivalent biochemical levels in both; however, in NOS1(-/-) mice, MnSOD-PL significantly increased survival after both thoracic and total-body irradiation. The mechanism of radiosensitivity of NOS1(-/-) mice and its reversal by MnSOD-PL may be related to the developmental esophageal enteric neuronal innervation abnormalities described in these mice.
Collapse
Affiliation(s)
- Malolan S Rajagopalan
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania 15232, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Shuvaev VV, Han J, Yu KJ, Huang S, Hawkins BJ, Madesh M, Nakada M, Muzykantov VR. PECAM-targeted delivery of SOD inhibits endothelial inflammatory response. FASEB J 2010; 25:348-57. [PMID: 20876216 DOI: 10.1096/fj.10-169789] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Elevated generation of reactive oxygen species (ROS) by endothelial enzymes, including NADPH-oxidase, is implicated in vascular oxidative stress and endothelial proinflammatory activation involving exposure of vascular cell adhesion molecule-1 (VCAM-1). Catalase and superoxide dismutase (SOD) conjugated with antibodies to platelet/endothelial cell adhesion molecule 1 (PECAM-1) bind specifically to endothelium and inhibit effects of corresponding ROS, H(2)O(2), and superoxide anion. In this study, anti-PECAM/SOD, but not anti-PECAM/catalase or nontargeted enzymes, including polyethylene glycol (PEG)-SOD, inhibited 2- to 3-fold VCAM expression caused by tumor necrosis factor (TNF), interleukin-1β, and lipopolysaccharide. Anti- PECAM/SOD, but not nontargeted counterparts, accumulated in vascular endothelium after intravenous injection, localized in endothelial endosomes, and inhibited by 70% lipopolysaccharide-caused VCAM-1 expression in mice. Anti-PECAM/SOD colocalized with EEA-1-positive endothelial vesicles and quenched ROS produced in response to TNF. Inhibitors of NADPH oxidase and anion channel ClC3 blocked TNF-induced VCAM expression, affirming that superoxide produced and transported by these proteins, respectively, mediates inflammatory signaling. Anti-PECAM/SOD abolished VCAM expression caused by poly(I:C)-induced activation of toll-like receptor 3 localized in intracellular vesicles. These results directly implicate endosomal influx of superoxide in endothelial inflammatory response and suggest that site-specific interception of this signal attained by targeted delivery of anti-PECAM/SOD into endothelial endosomes may have anti-inflammatory effects.
Collapse
Affiliation(s)
- Vladimir V Shuvaev
- Institute for Environmental Medicine, Department of Pharmacology, University of Pennsylvania School of Medicine, 1 John Morgan Bldg., 3620 Hamilton Walk, Philadelphia, PA 19104-6068, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Pandey BN, Kumar A, Tiwari P, Mishra KP. Radiobiological basis in management of accidental radiation exposure. Int J Radiat Biol 2010; 86:613-35. [DOI: 10.3109/09553001003746059] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
32
|
Niu Y, Wang H, Wiktor-Brown D, Rugo R, Shen H, Huq MS, Engelward B, Epperly M, Greenberger JS. Irradiated esophageal cells are protected from radiation-induced recombination by MnSOD gene therapy. Radiat Res 2010; 173:453-61. [PMID: 20334517 DOI: 10.1667/rr1763.1] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Radiation-induced DNA damage is a precursor to mutagenesis and cytotoxicity. During radiotherapy, exposure of healthy tissues can lead to severe side effects. We explored the potential of mitochondrial SOD (MnSOD) gene therapy to protect esophageal, pancreatic and bone marrow cells from radiation-induced genomic instability. Specifically, we measured the frequency of homologous recombination (HR) at an integrated transgene in the Fluorescent Yellow Direct Repeat (FYDR) mice, in which an HR event can give rise to a fluorescent signal. Mitochondrial SOD plasmid/liposome complex (MnSOD-PL) was administered to esophageal cells 24 h prior to 29 Gy upper-body irradiation. Single cell suspensions from FYDR, positive control FYDR-REC, and negative control C57BL/6NHsd (wild-type) mouse esophagus, pancreas and bone marrow were evaluated by flow cytometry. Radiation induced a statistically significant increase in HR 7 days after irradiation compared to unirradiated FYDR mice. MnSOD-PL significantly reduced the induction of HR by radiation at day 7 and also reduced the level of HR in the pancreas. Irradiation of the femur and tibial marrow with 8 Gy also induced a significant increase in HR at 7 days. Radioprotection by intraesophageal administration of MnSOD-PL was correlated with a reduced level of radiation-induced HR in esophageal cells. These results demonstrate the efficacy of MnSOD-PL for suppressing radiation-induced HR in vivo.
Collapse
Affiliation(s)
- Yunyun Niu
- Departments of Radiation Oncology and Surgery, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Kagan VE, Wipf P, Stoyanovsky D, Greenberger JS, Borisenko G, Belikova NA, Yanamala N, Samhan Arias AK, Tungekar MA, Jiang J, Tyurina YY, Ji J, Klein-Seetharaman J, Pitt BR, Shvedova AA, Bayir H. Mitochondrial targeting of electron scavenging antioxidants: Regulation of selective oxidation vs random chain reactions. Adv Drug Deliv Rev 2009; 61:1375-85. [PMID: 19695225 PMCID: PMC2784017 DOI: 10.1016/j.addr.2009.06.008] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2009] [Accepted: 06/08/2009] [Indexed: 10/20/2022]
Abstract
Effective regulation of highly compartmentalized production of reactive oxygen species and peroxidation reactions in mitochondria requires targeting of small molecule antioxidants and antioxidant enzymes into the organelles. This review describes recently developed approaches to mitochondrial targeting of small biologically active molecules based on: (i) preferential accumulation in mitochondria because of their hydrophobicity and positive charge (hydrophobic cations), (ii) binding with high affinity to an intra-mitochondrial constituent, and (iii) metabolic conversions by specific mitochondrial enzymes to reveal an active entity. In addition, targeted delivery of antioxidant enzymes via expression of leader sequences directing the proteins into mitochondria is considered. Examples of successful antioxidant and anti-apoptotic protection based on the ability of targeted cargoes to inhibit cytochrome c-catalyzed peroxidation of a mitochondria-specific phospholipid cardiolipin, in vitro and in vivo are presented. Particular emphasis is placed on the employment of triphenylphosphonium- and hemi-gramicidin S-moieties as two effective vehicles for mitochondrial delivery of antioxidants.
Collapse
Affiliation(s)
- Valerian E Kagan
- Center for Free Radical and Antioxidant Health, University of Pittsburgh, PA 15219, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Belikova NA, Glumac A, Rafikov R, Jiang J, Greenberger JS, Kagan VE, Bayir H. Radioprotection by short-term oxidative preconditioning: role of manganese superoxide dismutase. FEBS Lett 2009; 583:3437-42. [PMID: 19822147 DOI: 10.1016/j.febslet.2009.10.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2009] [Revised: 09/03/2009] [Accepted: 10/06/2009] [Indexed: 10/20/2022]
Abstract
Manganese superoxide dismutase (MnSOD) is vital to the protection of mitochondria and cells against oxidative stress. Earlier, we demonstrated that catalytically active homo-tetramer of MnSOD can be stabilized by oxidative cross-linking. Here we report that this effect may be translated into increased radioresistance of mouse embryonic cells (MECs) by pre-exposure to oxidative stress. Pre-treatment of MECs with antimycin A, rotenone or H(2)O(2) increased their survival after irradiation. Using MnSOD siRNA, we show that MECs with decreased MnSOD levels displayed a lowered ability to preconditioning. Thus oxidative preconditioning may be used for targeted regulation of MnSOD.
Collapse
Affiliation(s)
- Natalia A Belikova
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
The recovery of tissues and organs from ionizing irradiation is critically dependent on the repopulation of resident stem cells, defined as the subset of cells with capacity for both self-renewal and differentiation. Stem cells of both hematopoietic and epithelial origin reside in defined areas of the cellular microenvironment (recently defined as the stem cell "niche"). Experiments using serial repopulation assays in serial generations of total body irradiated mice receiving transplanted marrow and in continuous bone marrow cultures both identified specific microanatomic sites that comprise the bone marrow stem cell niche. Supportive cells of the hematopoietic microenvironment not only contribute to stem cell repopulation capacity but also to the maintenance of their quiescent or nonproliferative state, which allows the most primitive hematopoietic stem cells to stay in a noncycling state protected from both direct ionizing radiation-induced cell-cycle phase-specific killing and indirect cytokine and free radical mediated killing. Recent evidence has defined both cell contact and humoral mechanisms of protection of hematopoietic stem cells by stromal cells. There is also recent evidence for multilineage differentiation capacity of cells of the hematopoietic microenvironment termed bone marrow stromal cells (mesenchymal stem cells). Both hematopoietic stem cells and mesenchymal stem cell populations have been shown to be involved in the repair of ionizing irradiation damage of distant epithelial as well as other hematopoietic sites through their capacity to migrate through the circulation. The radiobiology of these 2 bone marrow stem cell populations is the subject of intense investigation. This review defines the status of research in the areas of stem cell quiescence, niche contact, and migratory responses to ionizing irradiation.
Collapse
Affiliation(s)
- Joel S Greenberger
- Radiation Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA.
| | | |
Collapse
|
36
|
Greenberger JS. Radioprotection. In Vivo 2009; 23:323-36. [PMID: 19414422 PMCID: PMC2981866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Over 40% of cancer patients will require radiation therapy during management of their disease. Although radiation therapy improves the survival of a significant number of cancer patients, both acute radiation toxicity (which manifests during a course of clinical radiotherapy or shortly thereafter), and late toxicity (developing months to years after completion of radiotherapy) compromise overall outcomes for successfully treated cancer patients.
Collapse
Affiliation(s)
- Joel S Greenberger
- Department of Radiation Oncology, University of Pittsburgh Medical Center, 200 Lothrop Street, Pittsburgh, PA 15213, USA.
| |
Collapse
|