1
|
Richter B, Epperly M, Tyurina Y, Shurin G, Johnson C, Korkmaz A, Gao Y, Scott J, Greenberger J, Kagan V, Bayır H. Enhancing survival after ionizing radiation exposure through mitigation of pyroptosis. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167434. [PMID: 39053669 PMCID: PMC11348791 DOI: 10.1016/j.bbadis.2024.167434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/05/2024] [Accepted: 07/16/2024] [Indexed: 07/27/2024]
Abstract
• Pyroptosis, an inflammatory cell death, has been implicated in the pathogenesis of total body irradiation (TBI) so we investigated time course and cell type involvement of key mediators in a murine model. • Pyroptotic mediators were most highly expressed at day 3 post TBI with immune cells from ileum being preferentially activated. • We also investigated the effectiveness of MCC950, a potent pyroptosis inhibitor, in our murine model showing a survival benefit at 50 mg/kg regardless of sex. • Treatment with MCC950 showed elevations in full-length downstream mediator IL-1β in ileum and decreased levels of cleaved IL-1β in splenic tissue.
Collapse
Affiliation(s)
- Brandon Richter
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, 130 Desoto St, Pittsburgh, PA 15261, USA; Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA 15224, USA; Children's Neuroscience Institute, UPMC Children's Hospital of Pittsburgh, 4401 Penn Ave., Pittsburgh, PA 15224, USA
| | - Michael Epperly
- Department of Radiation Oncology, University of Pittsburgh Medical Center, 200 Lothrop St, Pittsburgh, PA 15213, USA
| | - Yulia Tyurina
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, 130 Desoto St, Pittsburgh, PA 15261, USA
| | - Galina Shurin
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, 130 Desoto St, Pittsburgh, PA 15261, USA
| | - Carla Johnson
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA 15224, USA; Children's Neuroscience Institute, UPMC Children's Hospital of Pittsburgh, 4401 Penn Ave., Pittsburgh, PA 15224, USA
| | - Aybike Korkmaz
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, 130 Desoto St, Pittsburgh, PA 15261, USA; Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA 15224, USA; Children's Neuroscience Institute, UPMC Children's Hospital of Pittsburgh, 4401 Penn Ave., Pittsburgh, PA 15224, USA; Department of Pediatrics, Division of Critical Care and Hospital Medicine, Redox Health Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Interdisciplinary Neuroscience, Graduate School of Health Sciences, Ankara University
| | - Yuan Gao
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA 15224, USA; Children's Neuroscience Institute, UPMC Children's Hospital of Pittsburgh, 4401 Penn Ave., Pittsburgh, PA 15224, USA
| | - Julie Scott
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA 15224, USA; Children's Neuroscience Institute, UPMC Children's Hospital of Pittsburgh, 4401 Penn Ave., Pittsburgh, PA 15224, USA
| | - Joel Greenberger
- Department of Radiation Oncology, University of Pittsburgh Medical Center, 200 Lothrop St, Pittsburgh, PA 15213, USA
| | - Valerian Kagan
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, 130 Desoto St, Pittsburgh, PA 15261, USA; Children's Neuroscience Institute, UPMC Children's Hospital of Pittsburgh, 4401 Penn Ave., Pittsburgh, PA 15224, USA; Department of Pediatrics, Division of Critical Care and Hospital Medicine, Redox Health Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Hülya Bayır
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, 130 Desoto St, Pittsburgh, PA 15261, USA; Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh Medical Center, 4401 Penn Ave, Pittsburgh, PA 15224, USA; Children's Neuroscience Institute, UPMC Children's Hospital of Pittsburgh, 4401 Penn Ave., Pittsburgh, PA 15224, USA; Department of Pediatrics, Division of Critical Care and Hospital Medicine, Redox Health Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
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, 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 PMCID: PMC11552446 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
|
3
|
Greenberger JS, Hou W, Shields D, Fisher R, Epperly MW, Sarkaria I, Wipf P, Wang H. SARS-CoV-2 Spike Protein Induces Oxidative Stress and Senescence in Mouse and Human Lung. In Vivo 2024; 38:1546-1556. [PMID: 38936937 PMCID: PMC11215613 DOI: 10.21873/invivo.13605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 03/04/2024] [Accepted: 03/19/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND/AIM There is concern that people who had COVID-19 will develop pulmonary fibrosis. Using mouse models, we compared pulmonary inflammation following injection of the spike protein of SARS-CoV-2 (COVID-19) to radiation-induced inflammation to demonstrate similarities between the two models. SARS-CoV-2 (COVID-19) induces inflammatory cytokines and stress responses, which are also common to ionizing irradiation-induced acute pulmonary damage. Cellular senescence, which is a late effect following exposure to SARS-CoV-2 as well as radiation, was investigated. MATERIALS AND METHODS We evaluated the effect of SARS-CoV-2 spike protein compared to ionizing irradiation in K18-hACE2 mouse lung, human lung cell lines, and in freshly explanted human lung. We measured reactive oxygen species, DNA double-strand breaks, stimulation of transforming growth factor-beta pathways, and cellular senescence following exposure to SARS-CoV-2 spike protein, irradiation or SARS-COV-2 and irradiation. We also measured the effects of the antioxidant radiation mitigator MMS350 following irradiation or exposure to SARS-CoV-2. RESULTS SARS-CoV-2 spike protein induced reactive oxygen species, DNA double-strand breaks, transforming growth factor-β signaling pathways, and senescence, which were exacerbated by prior or subsequent ionizing irradiation. The water-soluble radiation countermeasure, MMS350, reduced spike protein-induced changes. CONCLUSION In both the SARS-Co-2 and the irradiation mouse models, similar responses were seen indicating that irradiation or exposure to SARS-CoV-2 virus may lead to similar lung diseases such as pulmonary fibrosis. Combination of irradiation and SARS-CoV-2 may result in a more severe case of pulmonary fibrosis. Cellular senescence may explain some of the late effects of exposure to SARS-CoV-2 spike protein and to ionizing irradiation.
Collapse
Affiliation(s)
- Joel S Greenberger
- 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
| | - Renee Fisher
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, U.S.A
| | - Michael W Epperly
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, U.S.A
| | - Inderpal Sarkaria
- Department of Thoracic Surgery, UPMC-Shadyside, 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
| |
Collapse
|
4
|
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
|
5
|
Chinnapaka S, Yang KS, Surucu Y, Bengur FB, Arellano JA, Tirmizi Z, Malekzadeh H, Epperly MW, Hou W, Greenberger JS, Rubin JP, Ejaz A. Human adipose ECM alleviates radiation-induced skin fibrosis via endothelial cell-mediated M2 macrophage polarization. iScience 2023; 26:107660. [PMID: 37705953 PMCID: PMC10495661 DOI: 10.1016/j.isci.2023.107660] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/30/2023] [Accepted: 08/15/2023] [Indexed: 09/15/2023] Open
Abstract
Radiation therapy can lead to late radiation-induced skin fibrosis (RISF), causing movement restriction, pain, and organ dysfunction. This study evaluated adipose-derived extracellular matrix (Ad-ECM) as a mitigator of RISF. Female C57BL/6J mice that were irradiated developed fibrosis, which was mitigated by a single local Ad-ECM injection, improving limb movement and reducing epithelium thickness and collagen deposition. Ad-ECM treatment resulted in decreased expression of pro-inflammatory and fibrotic genes, and upregulation of anti-inflammatory cytokines, promoting M2 macrophage polarization. Co-culture of irradiated human fibroblasts with Ad-ECM down-modulated fibrotic gene expression and enhanced bone marrow cell migration. Ad-ECM treatment also increased interleukin (IL)-4, IL-5, and IL-15 expression in endothelial cells, stimulating M2 macrophage polarization and alleviating RISF. Prophylactic use of Ad-ECM showed effectiveness in mitigation. This study suggests Ad-ECM's potential in treating chronic-stage fibrosis.
Collapse
Affiliation(s)
- Somaiah Chinnapaka
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Katherine S. Yang
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yusuf Surucu
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Fuat B. Bengur
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - José A. Arellano
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zayaan Tirmizi
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hamid Malekzadeh
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael W. Epperly
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Wen Hou
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Joel S. Greenberger
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - J. Peter Rubin
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Asim Ejaz
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
6
|
Ren C, Tan P, Gao L, Zeng Y, Hu S, Chen C, Tang N, Chen Y, Zhang W, Qin Y, Zhang X, Du S. Melatonin reduces radiation-induced ferroptosis in hippocampal neurons by activating the PKM2/NRF2/GPX4 signaling pathway. Prog Neuropsychopharmacol Biol Psychiatry 2023; 126:110777. [PMID: 37100272 DOI: 10.1016/j.pnpbp.2023.110777] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 04/19/2023] [Accepted: 04/21/2023] [Indexed: 04/28/2023]
Abstract
Ferroptosis is a type of regulated cell death that is dependent on iron and reactive oxygen species (ROS). Melatonin (N-acetyl-5-methoxytryptamine) reduces hypoxic-ischemic brain damage via mechanisms that involve free radical scavenging. How melatonin regulates radiation-induced ferroptosis of hippocampal neurons is yet to be elucidated. In this study, the mouse hippocampal neuronal cell line HT-22 was treated with 20μM melatonin before being stimulated with a combination of irradiation and 100 μM FeCl3. Furthermore, in vivo experiments were performed in mice treated with melatonin via intraperitoneal injection, which was followed by radiation exposure. A series of functional assays, including CCK-8, DCFH-DA kit, flow cytometry, TUNEL staining, iron estimations, and transmission electron microscopy, were performed on cells as well as hippocampal tissues. The interactions between PKM2 and NRF2 proteins were detected using a coimmunoprecipitation (Co-IP) assay. Moreover, chromatin immunoprecipitation (ChIP), a luciferase reporter assay, and an electrophoretic mobility shift assay (EMSA) were performed to explore the mechanism by which PKM2 regulates the NRF2/GPX4 signaling pathway. The spatial memory of mice was evaluated using the Morris Water Maze test. Hematoxylin-eosin and Nissl staining were performed for histological examination. The results revealed that melatonin protected HT-22 neuronal cells from radiation-induced ferroptosis, as inferred from increased cell viability, decreased ROS production, reduced number of apoptotic cells, and less cristae, higher electron density in mitochondria. In addition, melatonin induced PKM2 nuclear transference, while PKM2 inhibition reversed the effects of melatonin. Further experiments demonstrated that PKM2 bound to and induced the nuclear translocation of NRF2, which regulated GPX4 transcription. Ferroptosis enhanced by PKM2 inhibition was also converted by NRF2 overexpression. In vivo experiments indicated that melatonin alleviated radiation-induced neurological dysfunction and injury in mice. In conclusion, melatonin suppressed ferroptosis to decrease radiation-induced hippocampal neuronal injury by activating the PKM2/NRF2/GPX4 signaling pathway.
Collapse
Affiliation(s)
- Chen Ren
- Department of Radiation Oncology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, Guangdong, China
| | - Peixin Tan
- Department of Radiation Oncology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, Guangdong, China
| | - Lianxuan Gao
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yingying Zeng
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Shushu Hu
- Department of Radiation Oncology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, Guangdong, China
| | - Chen Chen
- Department of Radiation Oncology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, Guangdong, China
| | - Nan Tang
- Department of Radiation Oncology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, Guangdong, China
| | - Yulei Chen
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Wan Zhang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Yue Qin
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Xiaonan Zhang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Shasha Du
- Department of Radiation Oncology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, Guangdong, China.
| |
Collapse
|
7
|
Dar H, Mikulska-Ruminska K, Tyurina Y, Luci D, Yasgar A, Samovich S, Kapralov A, Souryavong A, Tyurin V, Amoscato A, Epperly M, Shurin G, Standley M, Holman T, St. Croix C, Watkins S, VanDemark A, Rana S, Zakharov A, Simeonov A, Marugan J, Mallampalli R, Wenzel S, Greenberger J, Rai G, Bayir H, Bahar I, Kagan V. Discovering selective antiferroptotic inhibitors of the 15LOX/PEBP1 complex noninterfering with biosynthesis of lipid mediators. Proc Natl Acad Sci U S A 2023; 120:e2218896120. [PMID: 37327313 PMCID: PMC10288584 DOI: 10.1073/pnas.2218896120] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 05/12/2023] [Indexed: 06/18/2023] Open
Abstract
Programmed ferroptotic death eliminates cells in all major organs and tissues with imbalanced redox metabolism due to overwhelming iron-catalyzed lipid peroxidation under insufficient control by thiols (Glutathione (GSH)). Ferroptosis has been associated with the pathogenesis of major chronic degenerative diseases and acute injuries of the brain, cardiovascular system, liver, kidneys, and other organs, and its manipulation offers a promising new strategy for anticancer therapy. This explains the high interest in designing new small-molecule-specific inhibitors against ferroptosis. Given the role of 15-lipoxygenase (15LOX) association with phosphatidylethanolamine (PE)-binding protein 1 (PEBP1) in initiating ferroptosis-specific peroxidation of polyunsaturated PE, we propose a strategy of discovering antiferroptotic agents as inhibitors of the 15LOX/PEBP1 catalytic complex rather than 15LOX alone. Here we designed, synthesized, and tested a customized library of 26 compounds using biochemical, molecular, and cell biology models along with redox lipidomic and computational analyses. We selected two lead compounds, FerroLOXIN-1 and 2, which effectively suppressed ferroptosis in vitro and in vivo without affecting the biosynthesis of pro-/anti-inflammatory lipid mediators in vivo. The effectiveness of these lead compounds is not due to radical scavenging or iron-chelation but results from their specific mechanisms of interaction with the 15LOX-2/PEBP1 complex, which either alters the binding pose of the substrate [eicosatetraenoyl-PE (ETE-PE)] in a nonproductive way or blocks the predominant oxygen channel thus preventing the catalysis of ETE-PE peroxidation. Our successful strategy may be adapted to the design of additional chemical libraries to reveal new ferroptosis-targeting therapeutic modalities.
Collapse
Affiliation(s)
- Haider H. Dar
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA15260
| | - Karolina Mikulska-Ruminska
- Department of Biophysics, Faculty of Physics Astronomy and Informatics, Institute of Physics, Nicolaus Copernicus University in Toruń, Toruń, Poland
| | - Yulia Y. Tyurina
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA15260
| | - Diane K. Luci
- National Center for Advancing Translational Sciences, Rockville, MD20892
| | - Adam Yasgar
- National Center for Advancing Translational Sciences, Rockville, MD20892
| | - Svetlana N. Samovich
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA15260
| | - Alexander A. Kapralov
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA15260
| | - Austin B. Souryavong
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA15260
| | - Vladimir A. Tyurin
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA15260
| | - Andrew A. Amoscato
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA15260
| | - Michael W. Epperly
- Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, PA15260
| | - Galina V. Shurin
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA15260
| | - Melissa Standley
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA95064
| | - Theodore R. Holman
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA95064
| | | | - Simon C. Watkins
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA15260
| | - Andrew P. VanDemark
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA15260
| | - Sandeep Rana
- National Center for Advancing Translational Sciences, Rockville, MD20892
| | - Alexey V. Zakharov
- National Center for Advancing Translational Sciences, Rockville, MD20892
| | - Anton Simeonov
- National Center for Advancing Translational Sciences, Rockville, MD20892
| | - Juan Marugan
- National Center for Advancing Translational Sciences, Rockville, MD20892
| | - Rama K. Mallampalli
- Department of Internal Medicine, The Ohio State University, Columbus, OH43210
| | - Sally E. Wenzel
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA15260
| | - Joel S. Greenberger
- Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, PA15260
| | - Ganesha Rai
- National Center for Advancing Translational Sciences, Rockville, MD20892
| | - Hülya Bayir
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA15260
- Department of Pediatrics, Division of Critical Care and Hospital Medicine, Redox Health Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY10032
| | - Ivet Bahar
- Laufer Center for Physical Quantitative Biology and Department of Biochemistry and Cell Biology, School of Medicine, Stony Brook University, NY11794
| | - Valerian E. Kagan
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA15260
| |
Collapse
|
8
|
Hamade DF, Epperly MW, Fisher R, Hou W, Shields D, van Pijkeren JP, Mukherjee A, Yu J, Leibowitz BJ, Vlad AM, Coffman L, Wang H, Huq MS, Huang Z, Rogers CJ, Greenberger JS. Release of Interferon-β (IFN-β) from Probiotic Limosilactobacillus reuteri-IFN-β (LR-IFN-β) Mitigates Gastrointestinal Acute Radiation Syndrome (GI-ARS) following Whole Abdominal Irradiation. Cancers (Basel) 2023; 15:1670. [PMID: 36980556 PMCID: PMC10046795 DOI: 10.3390/cancers15061670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/21/2023] [Accepted: 02/28/2023] [Indexed: 03/11/2023] Open
Abstract
Irradiation can be an effective treatment for ovarian cancer, but its use is limited by intestinal toxicity. Thus, strategies to mitigate toxicity are important and can revitalize the current standard of care. We previously established that LR-IL-22 protects the intestine from WAI. We now hypothesize that LR-IFN-β is an effective radiation protector and mitigator and is rapidly cleared from the digestive tract, making it an option for intestinal radioprotection. We report that the gavage of LR-IFN-β during WAI provides improved intestinal barrier integrity and significantly preserves the numbers of Lgr5+GFP+ intestinal stem cells, improving survival. The rapid clearance of the genetically engineered probiotic from the digestive tract renders it a safe and feasible radiation mitigator. Therefore, the above genetically engineered probiotic is both a feasible and effective radiation mitigator that could potentially revolutionize the management of OC patients. Furthermore, the subsequent addition of platinum/taxane-based chemotherapy to the combination of WAI and LR-IFN-β should reduce tumor volume while protecting the intestine and should improve the overall survival in OC patients.
Collapse
Affiliation(s)
- Diala F. Hamade
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Michael W. Epperly
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Renee Fisher
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Wen Hou
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Donna Shields
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | | | - Amitava Mukherjee
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Jian Yu
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Brian J. Leibowitz
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Anda M. Vlad
- Department of OB/Gyn and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Lan Coffman
- Department of Medicine, University of Pittsburgh, PA 15260, USA
| | - Hong Wang
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - M. Saiful Huq
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Ziyu Huang
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | | | - Joel S. Greenberger
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| |
Collapse
|
9
|
Hamade DF, Espinal A, Yu J, Leibowitz BJ, Fisher R, Hou W, Shields D, van Pijkeren JP, Mukherjee A, Epperly MW, Vlad A, Coffman L, Wang H, Huq MS, Patel R, Huang J, Greenberger JS. Lactobacillus reuteri Releasing IL-22 (LR-IL-22) Facilitates Intestinal Radioprotection for Whole-Abdomen Irradiation (WAI) of Ovarian Cancer. Radiat Res 2022; 198:89-105. [PMID: 35446961 PMCID: PMC9278541 DOI: 10.1667/rade-21-00224.1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 03/28/2022] [Indexed: 12/13/2022]
Abstract
Oral administration (gavage) of a second-generation probiotic, Lactobacillus reuteri (L. reuteri), that releases interleukin-22 (LR-IL-22) at 24 h after total-body irradiation (TBI) mitigates damage to the intestine. We determined that LR-IL-22 also mitigates partial-body irradiation (PBI) and whole-abdomen irradiation (WAI). Irradiation can be an effective treatment for ovarian cancer, but its use is limited by intestinal toxicity. Strategies to mitigate toxicity are important and can revitalize this modality to treat ovarian cancer. In the present studies, we evaluated whether LR-IL-22 facilitates fractionated WAI in female C57BL/6 mice with disseminated ovarian cancer given a single fraction of either 15.75 Gy or 19.75 Gy or 4 daily fractions of 6 Gy or 6.5 Gy. Mice receiving single or multiple administrations of LR-IL-22 during WAI showed improved intestinal barrier integrity (P = 0.0167), reduced levels of radiation-induced intestinal cytokines including KC/CXCL1 (P = 0.002) and IFN-γ (P = 0.0024), and reduced levels of plasma, Eotaxin/CCL11 (P = 0.0088). LR-IL-22 significantly preserved the numbers of Lgr5+GFP+ intestinal stem cells (P = 0.0010) and improved survival (P < 0.0343). Female C57BL/6MUC-1 mice with widespread abdominal syngeneic 2F8cis ovarian cancer that received LR-IL-22 during 6.5 Gy WAI in 4 fractions had reduced tumor burden, less intestinal toxicity, and improved 30-day survival. Furthermore, LR-IL-22 facilitated WAI when added to Paclitaxel and Carboplatin chemotherapy and further increased survival. Oral administration (gavage) of LR-IL-22 is a potentially valuable intestinal radioprotector, which can facilitate therapeutic WAI for widespread intra-abdominal ovarian cancer.
Collapse
Affiliation(s)
- Diala F. Hamade
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232
| | - Alexis Espinal
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232
| | - Jian Yu
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15260
| | | | - Renee Fisher
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232
| | - Wen Hou
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232
| | - Donna Shields
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232
| | | | - Amitava Mukherjee
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232
| | - Michael W. Epperly
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232
| | - Anda Vlad
- Department of OB/Gyn and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA 15260
| | - Lan Coffman
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15260
| | - Hong Wang
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15260
| | - M. Saiful Huq
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232
| | - Ravi Patel
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232
| | - Jason Huang
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232
| | - Joel S. Greenberger
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA 15232
| |
Collapse
|
10
|
Espinal A, Epperly MW, Mukherjee A, Fisher R, Shields D, Wang H, Huq MS, Hamade DF, Vlad AM, Coffman L, Buckanovich R, Yu J, Leibowitz BJ, van Pijkeren JP, Patel RB, Stolz D, Watkins S, Ejaz A, Greenberger JS. Intestinal Radiation Protection and Mitigation by Second-Generation Probiotic Lactobacillus-reuteri Engineered to Deliver Interleukin-22. Int J Mol Sci 2022; 23:5616. [PMID: 35628427 PMCID: PMC9145862 DOI: 10.3390/ijms23105616] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/10/2022] [Accepted: 05/12/2022] [Indexed: 12/13/2022] Open
Abstract
(1) Background: The systemic administration of therapeutic agents to the intestine including cytokines, such as Interleukin-22 (IL-22), is compromised by damage to the microvasculature 24 hrs after total body irradiation (TBI). At that time, there is significant death of intestinal microvascular endothelial cells and destruction of the lamina propria, which limits drug delivery through the circulation, thus reducing the capacity of therapeutics to stabilize the numbers of Lgr5+ intestinal crypt stem cells and their progeny, and improve survival. By its direct action on intestinal stem cells and their villus regeneration capacity, IL-22 is both an ionizing irradiation protector and mitigator. (2) Methods: To improve delivery of IL-22 to the irradiated intestine, we gavaged Lactobacillus-reuteri as a platform for the second-generation probiotic Lactobacillus-reuteri-Interleukin-22 (LR-IL-22). (3) Results: There was effective radiation mitigation by gavage of LR-IL-22 at 24 h after intestinal irradiation. Multiple biomarkers of radiation damage to the intestine, immune system and bone marrow were improved by LR-IL-22 compared to the gavage of control LR or intraperitoneal injection of IL-22 protein. (4) Conclusions: Oral administration of LR-IL-22 is an effective protector and mitigator of intestinal irradiation damage.
Collapse
Affiliation(s)
- Alexis Espinal
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA; (A.E.); (M.W.E.); (A.M.); (R.F.); shieldsd+@pitt.edu (D.S.); (M.S.H.); (D.F.H.); (R.B.P.)
| | - Michael W. Epperly
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA; (A.E.); (M.W.E.); (A.M.); (R.F.); shieldsd+@pitt.edu (D.S.); (M.S.H.); (D.F.H.); (R.B.P.)
| | - Amitava Mukherjee
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA; (A.E.); (M.W.E.); (A.M.); (R.F.); shieldsd+@pitt.edu (D.S.); (M.S.H.); (D.F.H.); (R.B.P.)
| | - Renee Fisher
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA; (A.E.); (M.W.E.); (A.M.); (R.F.); shieldsd+@pitt.edu (D.S.); (M.S.H.); (D.F.H.); (R.B.P.)
| | - Donna Shields
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA; (A.E.); (M.W.E.); (A.M.); (R.F.); shieldsd+@pitt.edu (D.S.); (M.S.H.); (D.F.H.); (R.B.P.)
| | - Hong Wang
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15232, USA;
| | - M. Saiful Huq
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA; (A.E.); (M.W.E.); (A.M.); (R.F.); shieldsd+@pitt.edu (D.S.); (M.S.H.); (D.F.H.); (R.B.P.)
| | - Diala Fatima Hamade
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA; (A.E.); (M.W.E.); (A.M.); (R.F.); shieldsd+@pitt.edu (D.S.); (M.S.H.); (D.F.H.); (R.B.P.)
| | - Anda M. Vlad
- Department of Obstetrics and Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA 15232, USA;
| | - Lan Coffman
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15232, USA; (L.C.); (R.B.)
| | - Ronald Buckanovich
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15232, USA; (L.C.); (R.B.)
| | - Jian Yu
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15232, USA; (J.Y.); (B.J.L.)
| | - Brian J. Leibowitz
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15232, USA; (J.Y.); (B.J.L.)
| | | | - Ravi B. Patel
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA; (A.E.); (M.W.E.); (A.M.); (R.F.); shieldsd+@pitt.edu (D.S.); (M.S.H.); (D.F.H.); (R.B.P.)
| | - Donna Stolz
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15232, USA; (D.S.); (S.W.)
| | - Simon Watkins
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15232, USA; (D.S.); (S.W.)
| | - Asim Ejaz
- Department of Plastic and Reconstructive Surgery, University of Pittsburgh, Pittsburgh, PA 15232, USA;
| | - Joel S. Greenberger
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA; (A.E.); (M.W.E.); (A.M.); (R.F.); shieldsd+@pitt.edu (D.S.); (M.S.H.); (D.F.H.); (R.B.P.)
| |
Collapse
|
11
|
Dar HH, Epperly MW, Tyurin VA, Amoscato AA, Anthonymuthu TS, Souryavong AB, Kapralov AA, Shurin GV, Samovich SN, St. Croix CM, Watkins SC, Wenzel SE, Mallampalli RK, Greenberger JS, Bayır H, Kagan VE, Tyurina YY. P. aeruginosa augments irradiation injury via 15-lipoxygenase-catalyzed generation of 15-HpETE-PE and induction of theft-ferroptosis. JCI Insight 2022; 7:156013. [PMID: 35041620 PMCID: PMC8876480 DOI: 10.1172/jci.insight.156013] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/13/2022] [Indexed: 01/14/2023] Open
Abstract
Total body irradiation (TBI) targets sensitive bone marrow hematopoietic cells and gut epithelial cells, causing their death and inducing a state of immunodeficiency combined with intestinal dysbiosis and nonproductive immune responses. We found enhanced Pseudomonas aeruginosa (PAO1) colonization of the gut leading to host cell death and strikingly decreased survival of irradiated mice. The PAO1-driven pathogenic mechanism includes theft-ferroptosis realized via (a) curbing of the host antiferroptotic system, GSH/GPx4, and (b) employing bacterial 15-lipoxygenase to generate proferroptotic signal - 15-hydroperoxy-arachidonoyl-PE (15-HpETE-PE) - in the intestines of irradiated and PAO1-infected mice. Global redox phospholipidomics of the ileum revealed that lysophospholipids and oxidized phospholipids, particularly oxidized phosphatidylethanolamine (PEox), represented the major factors that contributed to the pathogenic changes induced by total body irradiation and infection by PAO1. A lipoxygenase inhibitor, baicalein, significantly attenuated animal lethality, PAO1 colonization, intestinal epithelial cell death, and generation of ferroptotic PEox signals. Opportunistic PAO1 mechanisms included stimulation of the antiinflammatory lipoxin A4, production and suppression of the proinflammatory hepoxilin A3, and leukotriene B4. Unearthing complex PAO1 pathogenic/virulence mechanisms, including effects on the host anti/proinflammatory responses, lipid metabolism, and ferroptotic cell death, points toward potentially new therapeutic and radiomitigative targets.
Collapse
Affiliation(s)
- Haider H. Dar
- Department of Environmental and Occupational Health and Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Michael W. Epperly
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Vladimir A. Tyurin
- Department of Environmental and Occupational Health and Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Andrew A. Amoscato
- Department of Environmental and Occupational Health and Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Tamil S. Anthonymuthu
- Department of Environmental and Occupational Health and Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Critical Care Medicine, Safar Center for Resuscitation Research, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Austin B. Souryavong
- Department of Environmental and Occupational Health and Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Alexander A. Kapralov
- Department of Environmental and Occupational Health and Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Galina V. Shurin
- Department of Environmental and Occupational Health and Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Svetlana N. Samovich
- Department of Environmental and Occupational Health and Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Simon C. Watkins
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Sally E. Wenzel
- Department of Environmental and Occupational Health and Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rama K. Mallampalli
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Joel S. Greenberger
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Hülya Bayır
- Department of Environmental and Occupational Health and Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Critical Care Medicine, Safar Center for Resuscitation Research, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.,Children’s Neuroscience Institute, Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Valerian E. Kagan
- Department of Environmental and Occupational Health and Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Institute for Regenerative Medicine, I.M. Sechenov First Moscow State Medical University, Moscow, Russia.,Departments of Pharmacology and Chemical Biology, Chemistry, Radiation Oncology, University of Pittsburgh, Pennsylvania, USA
| | - Yulia Y. Tyurina
- Department of Environmental and Occupational Health and Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
12
|
Rogers CJ, Kyubwa EM, Lukaszewicz AI, Yamada-Hanff J, Starbird MA, Miller TA, Phelps AA, Wallack S, Mahendra S, Thrall K, Menon N. Identification of miRNA Associated with Reduced Survival after Whole-Thorax Lung Irradiation in Non-Human Primates. Radiat Res 2021; 196:510-522. [PMID: 33857299 DOI: 10.1667/rade-20-00031.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 03/19/2021] [Indexed: 11/03/2022]
Abstract
Thoracic exposure to ionizing radiation can lead to delayed injuries to the heart and lung that are serious and even life-threatening. These injuries are difficult to predict since they manifest over many weeks and months. To identify noninvasive, tissue-specific biomarkers for the early detection of late radiation injury, circulating microRNA (miRNA) levels were measured in non-human primates (NHP, Macaca mulatta) that received a single exposure of whole-thorax lung irradiation (WTLI) at a dose likely to result in 20% or 75% mortality within 180 days (9.8 or 10.7 Gy). Animals were observed for 270 days after WTLI. Approximately 58% of 9.8 Gy WTLI animals (7 of 12) and 94% of 10.7 Gy WTLI animals (15 out of 16) did not survive to the primary end point. Evidence of pulmonary fibrosis/pneumonitis was observed in all animals. Animals that received 10.7 Gy WTLI experienced more severe and early-onset pneumonitis, as indicated by reduced aerated lung volume, high non-sedated respiratory rate, earlier and more frequent dexamethasone treatments, and evidence of onset of heart disease. Radiation-induced changes in the circulating miRNA profile were most prominent within the first 30 days postirradiation, before the manifestation of symptoms, and included miRNA sequences known to regulate pathways associated with pulmonary fibrosis (TGF-β/SMAD signaling) and pneumonitis/inflammation (p53 signaling). The abundance of several circulating miRNA differentially expressed at day 6 or 15, such as miR-199a-3p and miR-25-3p, correlated with statistically significant differences in survival. This study supports the hypothesis that it is feasible to use plasma miRNA profiles to identify individuals at high risk of organ-specific late radiation injury. These miRNA profiles could improve radiation oncology clinical practice and serve as biomarkers to predict who might develop late complications in the aftermath of a radiological or nuclear (RAD-NUC) incident.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Seth Wallack
- Veterinary Imaging Center of San Diego, San Diego, California 92111
| | | | - Karla Thrall
- Altasciences Preclinical Seattle LLC, Everett, Washington 98203
| | | |
Collapse
|
13
|
Li K, Epperly MW, Barreto GA, Greenberger JS, Methé BA. "Longitudinal Fecal Microbiome Study of Total Body Irradiated Mice Treated With Radiation Mitigators Identifies Bacterial Associations With Survival". Front Cell Infect Microbiol 2021; 11:715396. [PMID: 34621689 PMCID: PMC8490782 DOI: 10.3389/fcimb.2021.715396] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 08/27/2021] [Indexed: 12/12/2022] Open
Abstract
Total body irradiation (TBI) has been demonstrated to alter the intestinal microbiome, but the effects of successful small molecule ionizing radiation mitigators on the intestinal microbiome are not well-known. Our survival experiments examined the effects of anti-cell death radiation mitigators on and in conjunction with the host's microbiota. Mice received 9.25 Gy TBI and then were administered radiation mitigators 24 hours later. Passed stool were collected pre-irradiation, then on days 1, 3, 5, 7, 10, 14, 21, and 30 post-irradiation for 16S rRNA gene (V4 region) sequencing. The Cox proportional hazards (CPH) model was fit with taxonomic composition (time varying covariates) and treatment as predictors. In the first experiment, mice were administered drugs for "granulocyte stimulation and anti-apoptosis" in four protocol combinations: JP4-039 (anti-apoptosis), granulocyte colony-stimulating factor (G-CSF, granulopoietic precursor cell stimulator), both mitigators, and control. Survival improved relative to control (30.0%) for G-CSF (80%, p-value = 0.025), G-CSF/JP4-039 (70%, p-value = 0.084), but not for JP4-039 (50.0%). In the second experiment, mice were administered mitigation drugs "inhibiting programmed cell death" pathways: JP4-039 (anti-apoptosis), necrostatin-1 (anti-necroptosis), and baicalein (anti-ferroptosis), in eight combinations. The survival of JP4-039/baicalein (60.0%, p-value = 0.010) and JP4-039/baicalein/necrostatin-1 (60.0%, p-value = 0.06) treatment combinations were significantly different from the control (26.7%). The JP4-039/necrostatin-1 (46.7%) and baicalein/necrostatin-1 (40.0%) and singlet treatment combinations (26.7%) were not significantly different from the control. Despite differences between the baseline microbiota compositions of the two experiments, consistent changes in composition after irradiation were found: Lactobacillus decreased post-irradiation, relative to baseline. By day 7, microbiota perturbations had incompletely reversed, and no drug-specific differences were identifiable. The CPH model identified Lactobacillus and members of Ruminococcaceae, including Ruminococcus, as protective and Akkermansia as deleterious. By day 30, the microbiota of surviving mice had not returned to baseline, but the differences between experiments suggest the resultant microbiota composition of the survivors are stochastic or batch specific in nature, rather than a requirement for survival. In conclusion, the study determined that key taxa identified in fecal samples, when applied towards the prediction of TBI survival, improves the survival model relative to treatment information alone.
Collapse
Affiliation(s)
- Kelvin Li
- Center for Medicine and the Microbiome, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Michael W. Epperly
- Department of Radiation Oncology, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| | - Gabriella Acosta Barreto
- Center for Medicine and the Microbiome, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Joel S. Greenberger
- Department of Radiation Oncology, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| | - Barbara A. Methé
- Center for Medicine and the Microbiome, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA, United States
| |
Collapse
|
14
|
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
|
15
|
Montesinos CA, Khalid R, Cristea O, Greenberger JS, Epperly MW, Lemon JA, Boreham DR, Popov D, Gorthi G, Ramkumar N, Jones JA. Space Radiation Protection Countermeasures in Microgravity and Planetary Exploration. Life (Basel) 2021; 11:life11080829. [PMID: 34440577 PMCID: PMC8398261 DOI: 10.3390/life11080829] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/03/2021] [Accepted: 08/06/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Space radiation is one of the principal environmental factors limiting the human tolerance for space travel, and therefore a primary risk in need of mitigation strategies to enable crewed exploration of the solar system. METHODS We summarize the current state of knowledge regarding potential means to reduce the biological effects of space radiation. New countermeasure strategies for exploration-class missions are proposed, based on recent advances in nutrition, pharmacologic, and immune science. RESULTS Radiation protection can be categorized into (1) exposure-limiting: shielding and mission duration; (2) countermeasures: radioprotectors, radiomodulators, radiomitigators, and immune-modulation, and; (3) treatment and supportive care for the effects of radiation. Vehicle and mission design can augment the overall exposure. Testing in terrestrial laboratories and earth-based exposure facilities, as well as on the International Space Station (ISS), has demonstrated that dietary and pharmacologic countermeasures can be safe and effective. Immune system modulators are less robustly tested but show promise. Therapies for radiation prodromal syndrome may include pharmacologic agents; and autologous marrow for acute radiation syndrome (ARS). CONCLUSIONS Current radiation protection technology is not yet optimized, but nevertheless offers substantial protection to crews based on Lunar or Mars design reference missions. With additional research and human testing, the space radiation risk can be further mitigated to allow for long-duration exploration of the solar system.
Collapse
Affiliation(s)
| | - Radina Khalid
- School of Engineering, Rice University, Houston, TX 77005, USA;
| | - Octav Cristea
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Joel S. Greenberger
- Department of Radiation Oncology, University of Pittsburg Medical Center, Pittsburgh, PA 15213, USA; (J.S.G.); (M.W.E.)
| | - Michael W. Epperly
- Department of Radiation Oncology, University of Pittsburg Medical Center, Pittsburgh, PA 15213, USA; (J.S.G.); (M.W.E.)
| | - Jennifer A. Lemon
- Medical Sciences Division, Northern Ontario School of Medicine, Sudbury, ON P3E 2C6, Canada; (J.A.L.); (D.R.B.)
| | - Douglas R. Boreham
- Medical Sciences Division, Northern Ontario School of Medicine, Sudbury, ON P3E 2C6, Canada; (J.A.L.); (D.R.B.)
| | - Dmitri Popov
- Advanced Medical Technologies and Systems Inc., Richmond Hill, ON L4B 1N1, Canada;
| | | | - Nandita Ramkumar
- Center for Space Medicine, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Jeffrey A. Jones
- Center for Space Medicine, Department of Urology, Baylor College of Medicine, Houston, TX 77030, USA
- Correspondence:
| |
Collapse
|
16
|
Micewicz ED, Damoiseaux RD, Deng G, Gomez A, Iwamoto KS, Jung ME, Nguyen C, Norris AJ, Ratikan JA, Ruchala P, Sayre JW, Schaue D, Whitelegge JP, McBride WH. Classes of Drugs that Mitigate Radiation Syndromes. Front Pharmacol 2021; 12:666776. [PMID: 34084139 PMCID: PMC8167044 DOI: 10.3389/fphar.2021.666776] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/27/2021] [Indexed: 11/13/2022] Open
Abstract
We previously reported several vignettes on types and classes of drugs able to mitigate acute and, in at least one case, late radiation syndromes in mice. Most of these had emerged from high throughput screening (HTS) of bioactive and chemical drug libraries using ionizing radiation-induced lymphocytic apoptosis as a readout. Here we report the full analysis of the HTS screen of libraries with 85,000 small molecule chemicals that identified 220 "hits." Most of these hits could be allocated by maximal common substructure analysis to one of 11 clusters each containing at least three active compounds. Further screening validated 23 compounds as being most active; 15 of these were cherry-picked based on drug availability and tested for their ability to mitigate acute hematopoietic radiation syndrome (H-ARS) in mice. Of these, five bore a 4-nitrophenylsulfonamide motif while 4 had a quinoline scaffold. All but two of the 15 significantly (p < 0.05) mitigated H-ARS in mice. We had previously reported that the lead 4-(nitrophenylsulfonyl)-4-phenylpiperazine compound (NPSP512), was active in mitigating multiple acute and late radiation syndromes in mice of more than one sex and strain. Unfortunately, the formulation of this drug had to be changed for regulatory reasons and we report here on the synthesis and testing of active analogs of NPSP512 (QS1 and 52A1) that have increased solubility in water and in vivo bioavailability while retaining mitigator activity against H-ARS (p < 0.0001) and other radiation syndromes. The lead quinoline 057 was also active in multiple murine models of radiation damage. Taken together, HTS of a total of 150,000 bioactive or chemical substances, combined with maximal common substructure analysis has resulted in the discovery of diverse groups of compounds that can mitigate H-ARS and at least some of which can mitigate multiple radiation syndromes when given starting 24 h after exposure. We discuss what is known about how these agents might work, and the importance of formulation and bioavailability.
Collapse
Affiliation(s)
- Ewa D. Micewicz
- Department of Radiation Oncology, University of California at Los Angeles, Los Angeles, CA, United States
| | - Robert D. Damoiseaux
- California NanoSystems Institute, University of California at Los Angeles, Los Angeles, CA, United States
- Department of Molecular and Medical Pharmacology, University of California at Los Angeles, Los Angeles, CA, United States
- Department of Bioengineering, Henry Samueli School of Engineering, University of California at Los Angeles, Los Angeles, CA, United States
| | - Gang Deng
- Department of Chemistry and Biochemistry, University of California at Los Angeles, Los Angeles, CA, United States
| | - Adrian Gomez
- Pasarow Mass Spectrometry Laboratory, University of California at Los Angeles, Los Angeles, CA, United States
| | - Keisuke S. Iwamoto
- Department of Radiation Oncology, University of California at Los Angeles, Los Angeles, CA, United States
| | - Michael E. Jung
- Department of Chemistry and Biochemistry, University of California at Los Angeles, Los Angeles, CA, United States
| | - Christine Nguyen
- Department of Radiation Oncology, University of California at Los Angeles, Los Angeles, CA, United States
| | | | - Josephine A. Ratikan
- Department of Radiation Oncology, University of California at Los Angeles, Los Angeles, CA, United States
| | - Piotr Ruchala
- Pasarow Mass Spectrometry Laboratory, University of California at Los Angeles, Los Angeles, CA, United States
| | - James W. Sayre
- Department of Biostatistics and Radiology, Fielding School of Public Health, University of California at Los Angeles, Los Angeles, CA, United States
| | - Dörthe Schaue
- Department of Radiation Oncology, University of California at Los Angeles, Los Angeles, CA, United States
| | - Julian P. Whitelegge
- Pasarow Mass Spectrometry Laboratory, University of California at Los Angeles, Los Angeles, CA, United States
| | - William H. McBride
- Department of Radiation Oncology, University of California at Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
17
|
Chinnapaka S, Yang KS, Samadi Y, Epperly MW, Hou W, Greenberger JS, Ejaz A, Rubin JP. Allogeneic adipose-derived stem cells mitigate acute radiation syndrome by the rescue of damaged bone marrow cells from apoptosis. Stem Cells Transl Med 2021; 10:1095-1114. [PMID: 33724714 PMCID: PMC8235137 DOI: 10.1002/sctm.20-0455] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 12/14/2022] Open
Abstract
Acute radiation syndrome (ARS) is the radiation toxicity that can affect the hematopoietic, gastrointestinal, and nervous systems upon accidental radiation exposure within a short time. Currently, there are no effective and safe approaches to treat mass population exposure to ARS. Our study aimed to evaluate the therapeutic potential of allogeneic adipose‐derived stem cells (ASCs) for total body irradiation (TBI)‐induced ARS and understand the underlying mitigation mechanism. We employed 9.25 Gy TBI dose to C57BL/6 mice and studied the effect of allogeneic ASCs on mice survival and regeneration of the hematopoietic system. Our results indicate that intraperitoneal‐injected ASCs migrated to the bone marrow, rescued hematopoiesis, and improved the survival of irradiated mice. Our transwell coculture results confirmed the migration of ASCs to irradiated bone marrow and rescue hematopoietic activity. Furthermore, contact coculture of ASCs improved the survival and hematopoiesis of irradiated bone marrow in vitro. Irradiation results in DNA damage, upregulation of inflammatory signals, and apoptosis in bone marrow cells, while coculture with ASCs reduces apoptosis via activation of DNA repair and the antioxidation system. Upon exposure to irradiated bone marrow cells, ASCs secrete prosurvival and hematopoietic factors, such as GM‐CSF, MIP1α, MIP1β, LIX, KC, 1P‐10, Rantes, IL‐17, MCSF, TNFα, Eotaxin, and IP‐10, which reduces oxidative stress and rescues damaged bone marrow cells from apoptosis. Our findings suggest that allogeneic ASCs therapy is effective in mitigating TBI‐induced ARS in mice and may be beneficial for clinical adaptation to treat TBI‐induced toxicities. Further studies will help to advocate the scale‐up and adaptation of allogeneic ASCs as the radiation countermeasure.
Collapse
Affiliation(s)
- Somaiah Chinnapaka
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Katherine S Yang
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yasamin Samadi
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Michael W Epperly
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, USA
| | - Wen Hou
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, USA
| | - Joel S Greenberger
- Department of Radiation Oncology, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, USA
| | - Asim Ejaz
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - J Peter Rubin
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
18
|
Bao C, Sun Y, Dwarakanath B, Dong Y, Huang Y, Wu X, Guha C, Kong L, Lu JJ. Carbon ion triggered immunogenic necroptosis of nasopharyngeal carcinoma cells involving necroptotic inhibitor BCL-x. J Cancer 2021; 12:1520-1530. [PMID: 33531997 PMCID: PMC7847655 DOI: 10.7150/jca.46316] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 12/06/2020] [Indexed: 01/26/2023] Open
Abstract
To explore the potential and mechanisms of necroptosis, a form of immunogenic cell death, induced by carbon ion as compared to photon beams in established photon resistant- (PR-) and sensitive nasopharyngeal carcinoma (NPC) cells. MLKL is considered a central executor of necroptosis and phosphorylation of MLKL (p-MLKL) was a critical event of necroptosis. The clonogenic survival and DNA microarray demonstrated that after repeated photon irradiation, radiosensitive NPC cells became apoptosis-resistant but could be effectively inhibited by carbon ion irradiation. The relative biologic effectiveness (RBE) at D10 and D37 were 2.15 and 2.78 for PR-NPC cells. Carbon ion induced delayed DNA damage repair, cell cycle arrest, cytogenetic damage, morphological change and cell necrosis, indicating the possibility of necroptosis in both PR- and sensitive NPC cell types. The lower expression of necroptotic inhibitors (caspase-8 and Bcl-x) and higher level of MLKL in PR-NPC cells showed it was relatively more predisposed to necroptosis compared to the sensitive cells. Subsequent experiments demonstrated the significant upregulation of p-MLKL in the PR-NPC cells treated by carbon ion (4 Gy) compared with photon irradiation at both physical (4 Gy) and RBE (10 Gy) doses (P≤0.0001). Moreover, carbon ion induced a robust (up to 28 folds) p-MLKL in the PR-NPC cells as well as sensitive cells (up to 6-fold) coupled with a lower level of BCL-x expression and increased GM-CSF implicated in resculputure of immune system. These results suggested that carbon ion could induce necroptosis of NPC cells, especially in PR-NPC cells, and its mechanisms involve BCL-x.
Collapse
Affiliation(s)
- Cihang Bao
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China.,Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| | - Yun Sun
- Department of Research and Development, Shanghai Proton and Heavy Ion Center, Shanghai, China.,Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| | - Bilikere Dwarakanath
- Department of Research and Development, Shanghai Proton and Heavy Ion Center, Shanghai, China.,Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| | - Yuanli Dong
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China.,Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China.,Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yangle Huang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China.,Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China.,Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiaodong Wu
- Department of Research and Development, Shanghai Proton and Heavy Ion Center, Shanghai, China.,Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| | - Chandan Guha
- Department of Radiation Oncology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, USA
| | - Lin Kong
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China.,Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| | - Jiade J Lu
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China.,Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| |
Collapse
|
19
|
Rogers CJ, Lukaszewicz AI, Yamada-Hanff J, Micewicz ED, Ratikan JA, Starbird MA, Miller TA, Nguyen C, Lee JT, Olafsen T, Iwamoto KS, McBride WH, Schaue D, Menon N. Identification of miRNA signatures associated with radiation-induced late lung injury in mice. PLoS One 2020; 15:e0232411. [PMID: 32392259 PMCID: PMC7213687 DOI: 10.1371/journal.pone.0232411] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/14/2020] [Indexed: 01/03/2023] Open
Abstract
Acute radiation exposure of the thorax can lead to late serious, and even life-threatening, pulmonary and cardiac damage. Sporadic in nature, late complications tend to be difficult to predict, which prompted this investigation into identifying non-invasive, tissue-specific biomarkers for the early detection of late radiation injury. Levels of circulating microRNA (miRNA) were measured in C3H and C57Bl/6 mice after whole thorax irradiation at doses yielding approximately 70% mortality in 120 or 180 days, respectively (LD70/120 or 180). Within the first two weeks after exposure, weight gain slowed compared to sham treated mice along with a temporary drop in white blood cell counts. 52% of C3H (33 of 64) and 72% of C57Bl/6 (46 of 64) irradiated mice died due to late radiation injury. Lung and heart damage, as assessed by computed tomography (CT) and histology at 150 (C3H mice) and 180 (C57Bl/6 mice) days, correlated well with the appearance of a local, miRNA signature in the lung and heart tissue of irradiated animals, consistent with inherent differences in the C3H and C57Bl/6 strains in their propensity for developing radiation-induced pneumonitis or fibrosis, respectively. Radiation-induced changes in the circulating miRNA profile were most prominent within the first 30 days after exposure and included miRNA known to regulate inflammation and fibrosis. Importantly, early changes in plasma miRNA expression predicted survival with reasonable accuracy (88-92%). The miRNA signature that predicted survival in C3H mice, including miR-34a-5p, -100-5p, and -150-5p, were associated with pro-inflammatory NF-κB-mediated signaling pathways, whereas the signature identified in C57Bl/6 mice (miR-34b-3p, -96-5p, and -802-5p) was associated with TGF-β/SMAD signaling. This study supports the hypothesis that plasma miRNA profiles could be used to identify individuals at high risk of organ-specific late radiation damage, with applications for radiation oncology clinical practice or in the context of a radiological incident.
Collapse
Affiliation(s)
| | | | | | - Ewa D. Micewicz
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Josephine A. Ratikan
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, California, United States of America
| | | | | | - Christine Nguyen
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Jason T. Lee
- Crump Institute for Molecular Imaging, University of California Los Angeles, Los Angeles, California, United States of America
| | - Tove Olafsen
- Crump Institute for Molecular Imaging, University of California Los Angeles, Los Angeles, California, United States of America
| | - Keisuke S. Iwamoto
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, California, United States of America
| | - William H. McBride
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Dörthe Schaue
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Naresh Menon
- ChromoLogic LLC, Monrovia, California, United States of America
| |
Collapse
|
20
|
Thermozier S, Hou W, Zhang X, Shields D, Fisher R, Bayir H, Kagan V, Yu J, Liu B, Bahar I, Epperly MW, Wipf P, Wang H, Huq MS, Greenberger JS. Anti-Ferroptosis Drug Enhances Total-Body Irradiation Mitigation by Drugs that Block Apoptosis and Necroptosis. Radiat Res 2020; 193:435-450. [PMID: 32134361 PMCID: PMC7299160 DOI: 10.1667/rr15486.1] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mitigation of total-body irradiation (TBI) in C57BL/6 mice by two drugs, which target apoptosis and necroptosis respectively, increases survival compared to one drug alone. Here we investigated whether the biomarker (signature)directed addition of a third anti-ferroptosis drug further mitigated TBI effects. C57BL/6NTac female mice (30-33 g) received 9.25 Gy TBI, and 24 h or later received JP4-039 (20 mg/kg), necrostatin-1 (1.65 mg/kg) and/or lipoxygenase-15 inhibitor (baicalein) (50 mg/kg) in single-, dual- or three-drug regimens. Some animals were sacrificed at days 0, 1, 2, 3, 4 or 7 postirradiation, while the majority in each group were maintained beyond 30 days. For those mice sacrificed at the early time points, femur bone marrow, intestine (ileum), lung and blood plasma were collected and analyzed for radiation-induced and mitigator-modified levels of 33 pro-inflammatory and stress response proteins. Each single mitigator administered [JP4-039 (24 h), necrostatin-1 (48 h) or baicalein (24 h)] improved survival at day 30 after TBI to 25% (P = 0.0432, 0.2816 or 0.1120, respectively) compared to 5% survival of 9.25 Gy TBI controls. Mice were administered the drug individually based on weight (mg/kg). Drug vehicles comprised 30% cyclodextrin for JP4-039 and baicalein, and 10% Cremphor-EL/10% ethanol/80% water for necrostatin-1; thus, dual-vehicle controls were also tested. The dual-drug combinations further enhanced survival: necrostatin-1 (delayed to 72 h) with baicalein 40% (P = 0.0359); JP4-039 with necrostatin-1 50% (P = 0.0062); and JP4-039 with baicalein 60% (P = 0.0064). The three-drug regimen, timed to signature directed evidence of onset after TBI of each death pathway in marrow and intestine, further increased the 30-day survival to 75% (P = 0.0002), and there was optimal normalization to preirradiation levels of inflammatory cytokine and stress response protein levels in plasma, intestine and marrow. In contrast, lung protein levels were minimally altered by 9.25 Gy TBI or mitigators over 7 days. Significantly, elevated intestinal proteins at day 7 after TBI were reduced by necrostatin-1-containing regimens; however, normalization of plasma protein levels at day 7 required the addition of JP4-039 and baicalein. These findings indicate that mitigator targeting to three distinct cell death pathways increases survival after TBI.
Collapse
Affiliation(s)
- Stephanie Thermozier
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15232
| | - Wen Hou
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15232
| | - Xichen Zhang
- 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
| | - Hulya Bayir
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Valerian Kagan
- Departments of Environmental/Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Jian Yu
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Bing Liu
- Departments of Computational and Biology Systems, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Ivet Bahar
- Departments of Computational and Biology Systems, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Michael W. Epperly
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15232
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Hong Wang
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - M. Saiful Huq
- 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
|
21
|
Shi Q, Pei F, Silverman GA, Pak SC, Perlmutter DH, Liu B, Bahar I. Mechanisms of Action of Autophagy Modulators Dissected by Quantitative Systems Pharmacology Analysis. Int J Mol Sci 2020; 21:ijms21082855. [PMID: 32325894 PMCID: PMC7215584 DOI: 10.3390/ijms21082855] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 12/12/2022] Open
Abstract
Autophagy plays an essential role in cell survival/death and functioning. Modulation of autophagy has been recognized as a promising therapeutic strategy against diseases/disorders associated with uncontrolled growth or accumulation of biomolecular aggregates, organelles, or cells including those caused by cancer, aging, neurodegeneration, and liver diseases such as α1-antitrypsin deficiency. Numerous pharmacological agents that enhance or suppress autophagy have been discovered. However, their molecular mechanisms of action are far from clear. Here, we collected a set of 225 autophagy modulators and carried out a comprehensive quantitative systems pharmacology (QSP) analysis of their targets using both existing databases and predictions made by our machine learning algorithm. Autophagy modulators include several highly promiscuous drugs (e.g., artenimol and olanzapine acting as activators, fostamatinib as an inhibitor, or melatonin as a dual-modulator) as well as selected drugs that uniquely target specific proteins (~30% of modulators). They are mediated by three layers of regulation: (i) pathways involving core autophagy-related (ATG) proteins such as mTOR, AKT, and AMPK; (ii) upstream signaling events that regulate the activity of ATG pathways such as calcium-, cAMP-, and MAPK-signaling pathways; and (iii) transcription factors regulating the expression of ATG proteins such as TFEB, TFE3, HIF-1, FoxO, and NF-κB. Our results suggest that PKA serves as a linker, bridging various signal transduction events and autophagy. These new insights contribute to a better assessment of the mechanism of action of autophagy modulators as well as their side effects, development of novel polypharmacological strategies, and identification of drug repurposing opportunities.
Collapse
Affiliation(s)
- Qingya Shi
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (Q.S.); (F.P.)
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Fen Pei
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (Q.S.); (F.P.)
| | - Gary A. Silverman
- Department of Pediatrics, School of Medicine, Washington University in St. Louis, St. Louis, MO 63130, USA; (G.A.S.); (S.C.P.); (D.H.P.)
| | - Stephen C. Pak
- Department of Pediatrics, School of Medicine, Washington University in St. Louis, St. Louis, MO 63130, USA; (G.A.S.); (S.C.P.); (D.H.P.)
| | - David H. Perlmutter
- Department of Pediatrics, School of Medicine, Washington University in St. Louis, St. Louis, MO 63130, USA; (G.A.S.); (S.C.P.); (D.H.P.)
| | - Bing Liu
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (Q.S.); (F.P.)
- Correspondence: (B.L.); (I.B.)
| | - Ivet Bahar
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (Q.S.); (F.P.)
- Correspondence: (B.L.); (I.B.)
| |
Collapse
|
22
|
Zhang X, Fisher R, Hou W, Shields D, Epperly MW, Wang H, Wei L, Leibowitz BJ, Yu J, Alexander LM, VAN Pijkeren JP, Watkins S, Wipf P, Greenberger JS. Second-generation Probiotics Producing IL-22 Increase Survival of Mice After Total Body Irradiation. In Vivo 2020; 34:39-50. [PMID: 31882461 PMCID: PMC6984118 DOI: 10.21873/invivo.11743] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 09/21/2019] [Accepted: 09/27/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND/AIM Intestinal damage induced by total body irradiation (TBI) reduces leucine-rich repeat-containing G-protein-coupled receptor 5 (Lgr5)-expressing stem cells, goblet, and Paneth cells, breaching the epithelial lining, and facilitating bacterial translocation, sepsis, and death. MATERIALS AND METHODS Survival was measured after TBI in animals that received wild-type or recombinant bacteria producing interleukin-22 (IL-22). Changes in survival due to microbially delivered IL-22 were measured. Lactobacillus reuteri producing IL-22, or Escherichia coli-IL-22 were compared to determine which delivery system is better. RESULTS C57BL/6 mice receiving IL-22 probiotics at 24 h after 9.25 Gy TBI, demonstrated green fluorescent protein-positive bacteria in the intestine, doubled the number of Lgr5+ intestinal stem cells, and increased 30-day survival. Bacteria were localized to the jejunum, ileum, and colon. CONCLUSION Second-generation probiotics appear to be valuable for mitigation of TBI, and radiation protection during therapeutic total abdominal irradiation.
Collapse
Affiliation(s)
- Xichen Zhang
- 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
| | - 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
| | - Michael W Epperly
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, U.S.A
| | - Hong Wang
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Liang Wei
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Brian J Leibowitz
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Jian Yu
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Laura M Alexander
- Department of Food Science, University of Wisconsin-Madison, Madison, WI, U.S.A
| | | | - Simon Watkins
- Center for Imaging, Department of Pathology, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Joel S Greenberger
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, U.S.A.
| |
Collapse
|
23
|
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: 1.4] [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
|
24
|
Simader E, Beer L, Laggner M, Vorstandlechner V, Gugerell A, Erb M, Kalinina P, Copic D, Moser D, Spittler A, Tschachler E, Jan Ankersmit H, Mildner M. Tissue-regenerative potential of the secretome of γ-irradiated peripheral blood mononuclear cells is mediated via TNFRSF1B-induced necroptosis. Cell Death Dis 2019; 10:729. [PMID: 31570701 PMCID: PMC6768878 DOI: 10.1038/s41419-019-1974-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 09/05/2019] [Accepted: 09/09/2019] [Indexed: 12/14/2022]
Abstract
Peripheral blood mononuclear cells (PBMCs) have been shown to produce and release a plethora of pro-angiogenetic factors in response to γ-irradiation, partially accounting for their tissue-regenerative capacity. Here, we investigated whether a certain cell subtype of PBMCs is responsible for this effect, and whether the type of cell death affects the pro-angiogenic potential of bioactive molecules released by γ-irradiated PBMCs. PBMCs and PBMC subpopulations, including CD4+ and CD8+ T cells, B cells, monocytes, and natural killer cells, were isolated and subjected to high-dose γ-irradiation. Transcriptome analysis revealed subpopulation-specific responses to γ-irradiation with distinct activation of pro-angiogenic pathways, cytokine production, and death receptor signalling. Analysis of the proteins released showed that interactions of the subsets are important for the generation of a pro-angiogenic secretome. This result was confirmed at the functional level by the finding that the secretome of γ-irradiated PBMCs displayed higher pro-angiogenic activity in an aortic ring assay. Scanning electron microscopy and image stream analysis of γ-irradiated PBMCs revealed distinct morphological changes, indicative for apoptotic and necroptotic cell death. While inhibition of apoptosis had no effect on the pro-angiogenic activity of the secretome, inhibiting necroptosis in stressed PBMCs abolished blood vessel sprouting. Mechanistically, we identified tumor necrosis factor (TNF) receptor superfamily member 1B as the main driver of necroptosis in response to γ-irradiation in PBMCs, which was most likely mediated via membrane-bound TNF-α. In conclusion, our study demonstrates that the pro-angiogenic activity of the secretome of γ-irradiated PBMCs requires interplay of different PBMC subpopulations. Furthermore, we show that TNF-dependent necroptosis is an indispensable molecular process for conferring tissue-regenerative activity and for the pro-angiogenic potential of the PBMC secretome. These findings contribute to a better understanding of secretome-based therapies in regenerative medicine.
Collapse
Affiliation(s)
- Elisabeth Simader
- Department of Internal Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria.,Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria.,FFG Project 852748 "APOSEC", Medical University of Vienna, Vienna, Austria
| | - Lucian Beer
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria.,Department of Radiology and Cancer Research UK Cambridge Center, Cambridge, CB2 0QQ, UK
| | - Maria Laggner
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria.,FFG Project 852748 "APOSEC", Medical University of Vienna, Vienna, Austria.,Vienna Business Agency Project 2343727 "APOSEC to clinic", Medical University Vienna, Vienna, Austria
| | - Vera Vorstandlechner
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria.,FFG Project 852748 "APOSEC", Medical University of Vienna, Vienna, Austria.,Vienna Business Agency Project 2343727 "APOSEC to clinic", Medical University Vienna, Vienna, Austria
| | - Alfred Gugerell
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria.,FFG Project 852748 "APOSEC", Medical University of Vienna, Vienna, Austria.,Vienna Business Agency Project 2343727 "APOSEC to clinic", Medical University Vienna, Vienna, Austria
| | - Michael Erb
- Synlab Analytics and Services Switzerland AG, Birsfelden, Switzerland
| | - Polina Kalinina
- Research Division of Biology and Pathobiology of the SkinDepartment of Dermatology, Research Division of Biology and Pathobiology of the Skin, Medical University of Vienna, Vienna, Austria
| | - Dragan Copic
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria.,FFG Project 852748 "APOSEC", Medical University of Vienna, Vienna, Austria.,Vienna Business Agency Project 2343727 "APOSEC to clinic", Medical University Vienna, Vienna, Austria
| | - Doris Moser
- Division of Oral and Maxillofacial Surgery, Medical University of Vienna, Vienna, Austria
| | - Andreas Spittler
- Research Laboratories, Core Facility Flow Cytometry, Medical University of Vienna, Vienna, Austria
| | - Erwin Tschachler
- Research Division of Biology and Pathobiology of the SkinDepartment of Dermatology, Research Division of Biology and Pathobiology of the Skin, Medical University of Vienna, Vienna, Austria
| | - Hendrik Jan Ankersmit
- Division of Thoracic Surgery, Medical University of Vienna, Vienna, Austria. .,FFG Project 852748 "APOSEC", Medical University of Vienna, Vienna, Austria. .,Vienna Business Agency Project 2343727 "APOSEC to clinic", Medical University Vienna, Vienna, Austria.
| | - Michael Mildner
- Research Division of Biology and Pathobiology of the SkinDepartment of Dermatology, Research Division of Biology and Pathobiology of the Skin, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
25
|
Thermozier S, Zhang X, Hou W, Fisher R, Epperly MW, Liu B, Bahar I, Wang H, Greenberger JS. Radioresistance of Serpinb3a-/- Mice and Derived Hematopoietic and Marrow Stromal Cell Lines. Radiat Res 2019; 192:267-281. [PMID: 31295086 PMCID: PMC6759811 DOI: 10.1667/rr15379.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Serpins are a group of serine-proteases involved in multiple signal transduction pathways in mammalian cells. In particular, Serpinb3a is involved in the lysosomal necrosis cell death pathway with components that overlap with radiation-induced apoptosis. We investigated the radiation response of Serpinb3a-/- mice compared to Serpinb3a+/+ mice on the Balb/c background. Serpinb3a-/- mice showed significant radioresistance to a dose of 8.0 Gy total-body irradiation, compared to Serpinb3a+/+ Balb/c mice. Long-term bone marrow cultures from Serpinb3a-/- mice showed increased longevity. In clonogenic survival assays, fresh bone marrow hematopoietic progenitors, as well as clonal interleukin-3 (IL-3)-dependent hematopoietic progenitor and bone marrow stromal cell lines from Serpinb3a-/- mice were radioresistant. Serpinb3a-/- mouse bone marrow-derived stromal cell lines had increased baseline and postirradiation antioxidant capacity. Serpinb3a-/- bone marrow stromal cells showed increased radiation-induced RNA transcripts for MnSOD and p21, and decreased levels of p53 and TGF-b. Both irradiated Serpinb3a-/- mouse bone marrow stromal cell lines and plasma removed from total-body irradiated mice had decreased levels of expression of stress response and inflammation-associated proteins. Abrogation of Serpinb3a may be a potential new target for mitigation of radiation effects.
Collapse
Affiliation(s)
- Stephanie Thermozier
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15213
| | - Xichen Zhang
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15213
| | - Wen Hou
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15213
| | - Renee Fisher
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15213
| | - Michael W. Epperly
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15213
| | - Bing Liu
- Department of Computational Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Ivet Bahar
- Department of Computational Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Hong Wang
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Joel S. Greenberger
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania 15213
| |
Collapse
|
26
|
Epperly MW, Fisher R, Rigatti L, Watkins S, Zhang X, Hou W, Shields D, Franicola D, Bayir H, Wang H, Thermozier S, Henderson A, Donnelly C, Wipf P, Greenberger JS. Amelioration of Amyotrophic Lateral Sclerosis in SOD1 G93A Mice by M 2 Microglia from Transplanted Marrow. In Vivo 2019; 33:675-688. [PMID: 31028184 PMCID: PMC6559904 DOI: 10.21873/invivo.11526] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 03/29/2019] [Accepted: 04/02/2019] [Indexed: 12/11/2022]
Abstract
Background/Aim: The cause of fatal neuromuscular amyotrophic lateral sclerosis (ALS) is not known. Materials and Methods: Ninety-day-old superoxide-dismutase-1 G93A (SOD1 G93A ) mice demonstrating level 1 paralysis, received 9.0 Gy total body irradiation (TBI) from a cesium source at 340 cGy per minute, and intravenous transplantation with 1×10 6 C57BL/6 green fluorescent protein (GFP)+ donor bone marrow cells. Results: Paralysis-free survival was prolonged in TBI and bone marrow-transplanted SOD1 G93A mice from 100 to over 250 days (p=0.0018). Other mice transplanted with SOD1 G93A marrow or marrow treated with the free-radical scavenger MMS350 showed no therapeutic effect. GFP+ macrophage-2 (M2) microglial cells of bone marrow origin, were seen at sites of degenerating anterior horn motor neurons. SOD1 G93A mice had a disruption in the blood-brain barrier permeability which was reversed by marrow transplant from C57BL/6 mice. SOD1 G93A marrow showed unexpected robust hematopoiesis in vitro, and radioresistance. Conclusion: After TBI, M2 microglial cells from transplanted donor marrow extended the paralysis-free interval in SOD1 G93A mice.
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
| | - Lora Rigatti
- Division of Laboratory Animal Resources (DLAR), University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Simon Watkins
- Department of Cell Biology and Center for Images, University of Pittsburgh, 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
| | - Darcy Franicola
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, U.S.A
| | - Hulya Bayir
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Hong Wang
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, U.S.A
| | - Stephanie Thermozier
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, U.S.A
| | - Andrew Henderson
- 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
| | - Joel S Greenberger
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, U.S.A.
| |
Collapse
|
27
|
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: 1.3] [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
|
28
|
Epperly MW, Wipf P, Fisher R, Franicola D, Beumer J, Li S, Brand RM, Falo LD, Erdos G, Greenberger JS. Evaluation of Different Formulations and Routes for the Delivery of the Ionizing Radiation Mitigator GS-Nitroxide (JP4-039). In Vivo 2018; 32:1009-1023. [PMID: 30150422 PMCID: PMC6199586 DOI: 10.21873/invivo.11341] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 05/10/2018] [Accepted: 05/17/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND/AIM The mitochondrial targeted GS-nitroxide, JP4-039, is an effective total body irradiation (TBI) mitigator when delivered intravenously (IV) up to 72 h after exposure. Effective systemic and localized administration to oral cavity/oropharynx and esophagus has been demonstrated. The objective of the study was to establish alternatives to IV administration suitable for JP4-039 delivery to mass casualties. MATERIALS AND METHODS JP4-039 was administered to C57BL/6 mice by topically applied carboxy-methyl-cellulose microneedle arrays (MNAs) or by intramuscular (IM) injection. Three different formulations that have passed Food and Drug Administration review, namely Captisol, 2-hydroxypropyl-β-cyclodextrin (cyclodextrin), and Miglyol-812-N, were used for drug delivery. Intraoral (IO) administration with each formulation was also evaluated. RESULTS All tested formulations and MNAs successfully delivered JP4-039. However, IM delivery of the Miglyol-812-N displayed very efficient and highly reproducible radiation mitigation. CONCLUSION Effective IM delivery of JP4-039 in animal models after TBI or partial-body irradiation suggested the use of the Miglyol-812-N formulation in both medical indications and radiation countermeasures.
Collapse
Affiliation(s)
- Michael W Epperly
- 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
- Department of Pharmaceutical Science, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Renee Fisher
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, U.S.A
| | - Darcy Franicola
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, U.S.A
| | - Jan Beumer
- Department of Pharmaceutical Science, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Song Li
- Department of Pharmaceutical Science, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Rhonda M Brand
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Louis D Falo
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Geza Erdos
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Joel S Greenberger
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, U.S.A.
| |
Collapse
|
29
|
Castle KD, Daniel AR, Moding EJ, Luo L, Lee CL, Kirsch DG. Mice Lacking RIP3 Kinase are not Protected from Acute Radiation Syndrome. Radiat Res 2018; 189:627-633. [PMID: 29634408 DOI: 10.1667/rr15001.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Exposure to high doses of ionizing radiation can cause lethal injury to normal tissue, thus inducing acute radiation syndrome. Acute radiation syndrome is caused by depletion of bone marrow cells (hematopoietic syndrome) and irreparable damage to the epithelial cells in the gastrointestinal tract (gastrointestinal syndrome). Although radiation initiates apoptosis in the hematopoietic and gastrointestinal compartments within the first few hours after exposure, alternative mechanisms of cell death may contribute to injury in these radiosensitive tissues. In this study, we utilized mice lacking a critical regulator of necroptosis, receptor interacting protein 3 (RIP3) kinase, to characterize the role of RIP3 in normal tissue toxicity after irradiation. Our results suggest that RIP3-mediated signaling is not a critical driver of acute radiation syndrome.
Collapse
Affiliation(s)
- Katherine D Castle
- a Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710
| | - Andrea R Daniel
- b Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina 27710
| | - Everett J Moding
- a Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710
| | - Lixia Luo
- b Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina 27710
| | - Chang-Lung Lee
- b Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina 27710
| | - David G Kirsch
- a Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710.,b Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina 27710
| |
Collapse
|