1
|
Fahl WE, Fahl BL, Schult D, Goesch TR. Significant Reduction of Radiation-Induced Death in Mice Treated with PrC-210 and G-CSF after Irradiation. Radiat Res 2024; 202:662-669. [PMID: 39142656 PMCID: PMC11528900 DOI: 10.1667/rade-24-00102.1] [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: 04/03/2024] [Accepted: 07/08/2024] [Indexed: 08/16/2024]
Abstract
The search for single or combined radiation countermeasures that mitigate the development of Acute Radiation Syndrome (ARS) after radiation exposure remains a prominent goal of the U.S. government. This study was undertaken to determine whether PrC-210 and G-CSF, when administered 24-48 h postirradiation, would confer an additive or synergistic survival benefit and mitigate ARS in mice that had received an otherwise 96% lethal radiation dose. Our results show that optimum systemic doses of PrC-210 and G-CSF, when administered 24 h or later after a 96% lethal dose of whole-body irradiation, conferred: 1. strong individual survival benefits (PrC-210 44%, P = 0.003), (G-CSF 48%, P = 0.0002), 2. a profound combined 85% survival benefit (P < 0.0001) when administered together, and on day 14 postirradiation, 3. peripheral white blood cell/lymphocyte counts equal to unirradiated controls, 4. dense bone marrow cell density (>65% of unirradiated controls), 5. jejunal villi density that equaled 90% of unirradiated controls, and 6. spleen weights that equaled 93% of unirradiated controls. Our results show that PrC-210 and G-CSF given together 24 h after irradiation confer strong additive efficacy by protecting the immune system, and enabling recovery of the bone marrow, and they work synergistically to enable recovery of peripheral white blood cells in circulating blood.
Collapse
Affiliation(s)
- William E. Fahl
- Obvia Pharmaceuticals Ltd., Madison, Wisconsin 53719
- Wisconsin Institutes for Medical Research, McArdle Laboratory, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Bryan L. Fahl
- Obvia Pharmaceuticals Ltd., Madison, Wisconsin 53719
- Wisconsin Institutes for Medical Research, McArdle Laboratory, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Devin Schult
- Wisconsin Institutes for Medical Research, McArdle Laboratory, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | | |
Collapse
|
2
|
Pearce LL, Zheng X, Wilen DS, Cronican AA, Frawley KL, Peterson J. Oxidant-Dependent Sensitizing, Protective, and Mitigative Effects in X-Ray-Irradiated Pulmonary Endothelial Cells. J Pharmacol Exp Ther 2024; 388:624-636. [PMID: 38182415 PMCID: PMC10801727 DOI: 10.1124/jpet.123.001714] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 07/10/2023] [Accepted: 07/14/2023] [Indexed: 01/07/2024] Open
Abstract
The primary response of proliferating bovine pulmonary artery endothelial cells (BPAECs) after X-ray irradiation [≤10 gray (Gy)] is shown to be transient cell-cycle arrest. Accompanying oxidant-linked functional changes within the mitochondria are readily measured, but increased autophagy is not. Radiation-induced apoptosis is negligible in this line-important because cells undergoing apoptosis release oxygen-derived species that can overwhelm/mask the radiation-associated species and their effects that we wish to investigate. Cells irradiated and cultured at 3% oxygen exhibited delayed cell-cycle arrest (6-8 hours after 10 Gy irradiation) compared with those maintained at 20% oxygen (2-4 hours after 10 Gy irradiation). At 3% oxygen, either only during or only after irradiation, results intermediate between 20% and 3% oxygen throughout were obtained. No variability in cell-cycle distribution was observed for unirradiated cells cultured under different prevailing oxygen levels. Mitochondrially localized manganese superoxide dismutase delayed the X-ray-induced cell-cycle changes when over-expressed in BPAEC, indicating superoxide to be one of the key oxygen-derived cytotoxic species involved in the radiobiological response. Also, the peroxynitrite biomarker 3-nitrotyrosine was elevated, whereas hydrogen peroxide levels were not. Lastly, the utility of the BPAEC for screening potential countermeasures to ionizing radiation is demonstrated with some quinoline derivatives. Three of the five compounds appeared mitigative, and all were protective. It is suggested that the oxidation-reduction chemistry of these compounds probably offers a reasonable explanation for their observed ameliorative properties. Furthermore, the results suggest a promising new direction in the search for lead compounds as countermeasures to the effects of ionizing radiation. SIGNIFICANCE STATEMENT: The primary radiological response of proliferating bovine pulmonary artery endothelial cells is cell-cycle arrest, starting soon after X-ray irradiation (1-10 Gy) at 20% O2 but delayed by 4 hours at systemic (3%) O2. Oxygen/superoxide is found to be radio-sensitizing in at least two distinct time windows, during and after the irradiation, with both responses antagonized by various hydroxyquinoline derivatives. Similar responses in many other cell lines are likely to be masked by elevated oxidants associated with apoptosis.
Collapse
Affiliation(s)
- Linda L Pearce
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Xi Zheng
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Daniel S Wilen
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Andrea A Cronican
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kristin L Frawley
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jim Peterson
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
3
|
Shrum SA, Nukala U, Shrimali S, Pineda EN, Krager KJ, Thakkar S, Jones DE, Pathak R, Breen PJ, Aykin-Burns N, Compadre CM. Tocotrienols Provide Radioprotection to Multiple Organ Systems through Complementary Mechanisms of Antioxidant and Signaling Effects. Antioxidants (Basel) 2023; 12:1987. [PMID: 38001840 PMCID: PMC10668991 DOI: 10.3390/antiox12111987] [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: 09/23/2023] [Revised: 10/31/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Tocotrienols have powerful radioprotective properties in multiple organ systems and are promising candidates for development as clinically effective radiation countermeasures. To facilitate their development as clinical radiation countermeasures, it is crucial to understand the mechanisms behind their powerful multi-organ radioprotective properties. In this context, their antioxidant effects are recognized for directly preventing oxidative damage to cellular biomolecules from ionizing radiation. However, there is a growing body of evidence indicating that the radioprotective mechanism of action for tocotrienols extends beyond their antioxidant properties. This raises a new pharmacological paradigm that tocotrienols are uniquely efficacious radioprotectors due to a synergistic combination of antioxidant and other signaling effects. In this review, we have covered the wide range of multi-organ radioprotective effects observed for tocotrienols and the mechanisms underlying it. These radioprotective effects for tocotrienols can be characterized as (1) direct cytoprotective effects, characteristic of the classic antioxidant properties, and (2) other effects that modulate a wide array of critical signaling factors involved in radiation injury.
Collapse
Affiliation(s)
- Stephen A. Shrum
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (U.N.); (S.S.); (E.N.P.); (K.J.K.); (S.T.); (D.E.J.); (R.P.); (P.J.B.); (N.A.-B.)
- Tocol Pharmaceuticals, LLC, Little Rock, AR 77205, USA
| | - Ujwani Nukala
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (U.N.); (S.S.); (E.N.P.); (K.J.K.); (S.T.); (D.E.J.); (R.P.); (P.J.B.); (N.A.-B.)
- Joint Bioinformatics Graduate Program, University of Arkansas at Little Rock, Little Rock, AR 72204, USA
| | - Shivangi Shrimali
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (U.N.); (S.S.); (E.N.P.); (K.J.K.); (S.T.); (D.E.J.); (R.P.); (P.J.B.); (N.A.-B.)
- Joint Bioinformatics Graduate Program, University of Arkansas at Little Rock, Little Rock, AR 72204, USA
| | - Edith Nathalie Pineda
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (U.N.); (S.S.); (E.N.P.); (K.J.K.); (S.T.); (D.E.J.); (R.P.); (P.J.B.); (N.A.-B.)
- Joint Bioinformatics Graduate Program, University of Arkansas at Little Rock, Little Rock, AR 72204, USA
| | - Kimberly J. Krager
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (U.N.); (S.S.); (E.N.P.); (K.J.K.); (S.T.); (D.E.J.); (R.P.); (P.J.B.); (N.A.-B.)
| | - Shraddha Thakkar
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (U.N.); (S.S.); (E.N.P.); (K.J.K.); (S.T.); (D.E.J.); (R.P.); (P.J.B.); (N.A.-B.)
| | - Darin E. Jones
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (U.N.); (S.S.); (E.N.P.); (K.J.K.); (S.T.); (D.E.J.); (R.P.); (P.J.B.); (N.A.-B.)
| | - Rupak Pathak
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (U.N.); (S.S.); (E.N.P.); (K.J.K.); (S.T.); (D.E.J.); (R.P.); (P.J.B.); (N.A.-B.)
| | - Philip J. Breen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (U.N.); (S.S.); (E.N.P.); (K.J.K.); (S.T.); (D.E.J.); (R.P.); (P.J.B.); (N.A.-B.)
| | - Nukhet Aykin-Burns
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (U.N.); (S.S.); (E.N.P.); (K.J.K.); (S.T.); (D.E.J.); (R.P.); (P.J.B.); (N.A.-B.)
| | - Cesar M. Compadre
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (U.N.); (S.S.); (E.N.P.); (K.J.K.); (S.T.); (D.E.J.); (R.P.); (P.J.B.); (N.A.-B.)
- Tocol Pharmaceuticals, LLC, Little Rock, AR 77205, USA
| |
Collapse
|
4
|
Xiao M, Li X, Wang L, Lin B, Zhai M, Hull L, Zizzo A, Cui W, Kiang JG. Skin Wound following Irradiation Aggravates Radiation-Induced Brain Injury in a Mouse Model. Int J Mol Sci 2023; 24:10701. [PMID: 37445879 DOI: 10.3390/ijms241310701] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Radiation injury- and radiation combined with skin injury-induced inflammatory responses in the mouse brain were evaluated in this study. Female B6D2F1/J mice were subjected to a sham, a skin wound (SW), 9.5 Gy 60Co total-body gamma irradiation (RI), or 9.5 Gy RI combined with a skin puncture wound (RCI). Survival, body weight, and wound healing were tracked for 30 days, and mouse brain samples were collected on day 30 after SW, RI, RCI, and the sham control. Our results showed that RCI caused more severe animal death and body weight loss compared with RI, and skin wound healing was significantly delayed by RCI compared to SW. RCI and RI increased the chemokines Eotaxin, IP-10, MIG, 6Ckine/Exodus2, MCP-5, and TIMP-1 in the brain compared to SW and the sham control mice, and the Western blot results showed that IP-10 and p21 were significantly upregulated in brain cells post-RI or -RCI. RI and RCI activated both astrocytes and endothelial cells in the mouse brain, subsequently inducing blood-brain barrier (BBB) leakage, as shown by the increased ICAM1 and GFAP proteins in the brain and GFAP in the serum. The Doublecortin (DCX) protein, the "gold standard" for measuring neurogenesis, was significantly downregulated by RI and RCI compared with the sham group. Furthermore, RI and RCI decreased the expression of the neural stem cell marker E-cadherin, the intermediate progenitor marker MASH1, the immature neuron cell marker NeuroD1, and the mature neuron cell marker NeuN, indicating neural cell damage in all development stages after RI and RCI. Immunohistochemistry (IHC) staining further confirmed the significant loss of neural cells in RCI. Our data demonstrated that RI and RCI induced brain injury through inflammatory pathways, and RCI exacerbated neural cell damage more than RI.
Collapse
Affiliation(s)
- Mang Xiao
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
| | - Xianghong Li
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
| | - Li Wang
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Bin Lin
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Min Zhai
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Lisa Hull
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Alex Zizzo
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
| | - Wanchang Cui
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Juliann G Kiang
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| |
Collapse
|
5
|
Fahl WE, Cadarso M, Goesch TR. Significant Reduction of Total-Body Irradiation-Induced Death in Mice Treated with PrC-210 24 Hours Postirradiation. Radiat Res 2022; 198:263-270. [PMID: 35728266 DOI: 10.1667/rade-22-00036.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 06/06/2022] [Indexed: 11/03/2022]
Abstract
The search for radiation countermeasures that can serve as: i. a pre-exposure agent to protect against subsequent irradiation, and/or ii. a post-exposure agent to mitigate the development of Acute Radiation Syndrome after radiation exposure, remains a prominent goal of the U.S. Government. This study was undertaken to determine whether PrC-210, when administered once, 24 h postirradiation, would provide a survival benefit and would mitigate Acute Radiation Syndrome in mice that had received an otherwise 95-100% lethal radiation dose. Our results show that a single intraperitoneal dose of PrC-210 (0.3-0.4 MTD, 151-201 ug/gm body weight) administered 24 h postirradiation, conferred: i. a 45% survival advantage (P = 0.002) in outbred ICR mice and a 25% survival advantage (P = 0.037) in inbred C57Bl/6 mice, ii. a significant increase in body weight in surviving mice (P = 0.012), iii. a discernible protection of intestinal structure by MRI imaging of live mice, iv. visibly denser jejunal villi and surface epithelium and v. visible bone marrow population in PrC-210-treated mice versus saline controls. The ability of PrC-210 to suppress 100% of radiation-induced death when administered minutes before irradiation, or roughly half of this effect (45%) when administered 24 h postirradiation is noteworthy. Determining the multiple paths by which PrC-210 protection is conferred is a process; the results in this report showing protection of two of the major systems central to Acute Radiation Syndrome damage, is a good first step. This was the first study of PrC-210 administered postirradiation; it conferred substantial survival benefit and suppression of Acute Radiation Syndrome. This outcome supports the continued development of PrC-210 to protect humans exposed to ionizing radiation.
Collapse
Affiliation(s)
- William E Fahl
- Wisconsin Institutes for Medical Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin.,Obvia Pharmaceuticals Ltd., Madison, Wisconsin
| | - Michela Cadarso
- Wisconsin Institutes for Medical Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin
| | | |
Collapse
|
6
|
Liu L, Li H, Patterson AM, Plett PA, Sampson CH, Mohammad KS, Capitano ML, Singh P, Yao C, Orschell CM, Pelus LM. Upregulation of SIRT1 Contributes to dmPGE2-dependent Radioprotection of Hematopoietic Stem Cells. Stem Cell Rev Rep 2022; 18:1478-1494. [PMID: 35318613 DOI: 10.1007/s12015-022-10368-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2022] [Indexed: 11/29/2022]
Abstract
Exposure to potentially lethal high-dose ionizing radiation results in bone marrow suppression, known as the hematopoietic acute radiation syndrome (H-ARS), which can lead to pancytopenia and possible death from hemorrhage or infection. Medical countermeasures to protect from or mitigate the effects of radiation exposure are an ongoing medical need. We recently reported that 16,16 dimethyl prostaglandin E2 (dmPGE2) given prior to lethal irradiation protects hematopoietic stem (HSCs) and progenitor (HPCs) cells and accelerates hematopoietic recovery by attenuating mitochondrial compromise, DNA damage, apoptosis, and senescence. However, molecular mechanisms responsible for the radioprotective effects of dmPGE2 on HSCs are not well understood. In this report, we identify a crucial role for the NAD+-dependent histone deacetylase Sirtuin 1 (Sirt1) downstream of PKA and CREB in dmPGE2-dependent radioprotection of hematopoietic cells. We found that dmPGE2 increases Sirt1 expression and activity in hematopoietic cells including HSCs and pharmacologic and genetic suppression of Sirt1 attenuates the radioprotective effects of dmPGE2 on HSC and HPC function and its ability to reduce DNA damage, apoptosis, and senescence and stimulate autophagy in HSCs. DmPGE2-mediated enhancement of Sirt1 activity in irradiated mice is accompanied by epigenetic downregulation of p53 activation and inhibition of H3K9 and H4K16 acetylation at the promoters of the genes involved in DNA repair, apoptosis, and autophagy, including p53, Ku70, Ku80, LC3b, ATG7, and NF-κB. These studies expand our understanding of intracellular events that are induced by IR but prevented/attenuated by dmPGE2 and suggest that modulation of Sirt1 activity may facilitate hematopoietic recovery following hematopoietic stress. Graphical Abstract.
Collapse
Affiliation(s)
- Liqiong Liu
- Department of Microbiology & Immunology, Indiana University School of Medicine, 950 West Walnut Street, Indianapolis, IN, 46202, USA
| | - Hongge Li
- Department of Microbiology & Immunology, Indiana University School of Medicine, 950 West Walnut Street, Indianapolis, IN, 46202, USA
| | - Andrea M Patterson
- Department of Microbiology & Immunology, Indiana University School of Medicine, 950 West Walnut Street, Indianapolis, IN, 46202, USA.,Department of Medicine/Hematology Oncology, Indiana University School of Medicine, 980 West Walnut Street, Indianapolis, IN, 46202, USA
| | - P Artur Plett
- Department of Medicine/Hematology Oncology, Indiana University School of Medicine, 980 West Walnut Street, Indianapolis, IN, 46202, USA
| | - Carol H Sampson
- Department of Medicine/Hematology Oncology, Indiana University School of Medicine, 980 West Walnut Street, Indianapolis, IN, 46202, USA
| | - Khalid S Mohammad
- Department of Medicine/Endocrinology, Indiana University School of Medicine, 980 West Walnut Street, Indianapolis, IN, 46202, USA
| | - Maegan L Capitano
- Department of Microbiology & Immunology, Indiana University School of Medicine, 950 West Walnut Street, Indianapolis, IN, 46202, USA
| | - Pratibha Singh
- Department of Microbiology & Immunology, Indiana University School of Medicine, 950 West Walnut Street, Indianapolis, IN, 46202, USA.,Department of Medicine/Hematology Oncology, Indiana University School of Medicine, 980 West Walnut Street, Indianapolis, IN, 46202, USA
| | - Chonghua Yao
- Shanghai Municipal Hospital of Traditional Chinese Medicine, NO.274, middle Zhijiang Road, Shanghai, China
| | - Christie M Orschell
- Department of Medicine/Hematology Oncology, Indiana University School of Medicine, 980 West Walnut Street, Indianapolis, IN, 46202, USA.
| | - Louis M Pelus
- Department of Microbiology & Immunology, Indiana University School of Medicine, 950 West Walnut Street, Indianapolis, IN, 46202, USA. .,Department of Medicine/Hematology Oncology, Indiana University School of Medicine, 980 West Walnut Street, Indianapolis, IN, 46202, USA.
| |
Collapse
|
7
|
Crook A, De Lima Leite A, Payne T, Bhinderwala F, Woods J, Singh VK, Powers R. Radiation exposure induces cross-species temporal metabolic changes that are mitigated in mice by amifostine. Sci Rep 2021; 11:14004. [PMID: 34234212 PMCID: PMC8263605 DOI: 10.1038/s41598-021-93401-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/24/2021] [Indexed: 11/09/2022] Open
Abstract
Exposure to acute, damaging radiation may occur through a variety of events from cancer therapy and industrial accidents to terrorist attacks and military actions. Our understanding of how to protect individuals and mitigate the effects of radiation injury or Acute Radiation Syndrome (ARS) is still limited. There are only a few Food and Drug Administration-approved therapies for ARS; whereas, amifostine is limited to treating low dose (0.7-6 Gy) radiation poisoning arising from cancer radiotherapy. An early intervention is critical to treat ARS, which necessitates identifying diagnostic biomarkers to quickly characterize radiation exposure. Towards this end, a multiplatform metabolomics study was performed to comprehensively characterize the temporal changes in metabolite levels from mice and non-human primate serum samples following γ-irradiation. The metabolomic signature of amifostine was also evaluated in mice as a model for radioprotection. The NMR and mass spectrometry metabolomics analysis identified 23 dysregulated pathways resulting from the radiation exposure. These metabolomic alterations exhibited distinct trajectories within glucose metabolism, phospholipid biosynthesis, and nucleotide metabolism. A return to baseline levels with amifostine treatment occurred for these pathways within a week of radiation exposure. Together, our data suggests a unique physiological change that is independent of radiation dose or species. Furthermore, a metabolic signature of radioprotection was observed through the use of amifostine prophylaxis of ARS.
Collapse
Affiliation(s)
- Alexandra Crook
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304, USA
| | - Aline De Lima Leite
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304, USA
| | - Thomas Payne
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304, USA
| | - Fatema Bhinderwala
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304, USA
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304, USA
| | - Jade Woods
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304, USA
| | - Vijay K Singh
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, USUHS, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
- Armed Forces Radiobiology Research Institute, USUHS, Bethesda, MD, 20814, USA.
| | - Robert Powers
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304, USA.
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304, USA.
| |
Collapse
|
8
|
Wang L, Zhai M, Lin B, Cui W, Hull L, Li X, Anderson MN, Smith JT, Umali MV, Jiang S, Kiang JG, Xiao M. PEG-G-CSF and L-Citrulline Combination Therapy for Mitigating Skin Wound Combined Radiation Injury in a Mouse Model. Radiat Res 2021; 196:113-127. [PMID: 33914884 PMCID: PMC8344563 DOI: 10.1667/rade-20-00151.1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 03/26/2021] [Indexed: 11/03/2022]
Abstract
Radiation combined injury (RCI, radiation exposure coupled with other forms of injury, such as burn, wound, hemorrhage, blast, trauma and/or sepsis) comprises approximately 65% of injuries from a nuclear explosion, and greatly increases the risk of morbidity and mortality when compared to that of radiation injury alone. To date, no U.S. Food and Drug Administration (FDA)-approved countermeasures are available for RCI. Currently, three leukocyte growth factors (Neupogen®, Neulasta® and Leukine®) have been approved by the FDA for mitigating the hematopoietic acute radiation syndrome. However these granulocyte-colony-stimulating factor (G-CSF) and granulocyte-macrophage colony-stimulating factor (GM-CSF) products have failed to increase 30-day survival of mice after RCI, suggesting a more complicated biological mechanism is in play for RCI than for radiation injury. In the current study, the mitigative efficacy of combination therapy using pegylated (PEG)-G-CSF (Neulasta) and -citrulline was evaluated in an RCI mouse model. L-citrulline is a neutral alpha-amino acid shown to improve vascular endothelial function in cardiovascular diseases. Three doses of PEG-G-CSF at 1 mg/kg, subcutaneously administered on days 1, 8 and 15 postirradiation, were supplemented with oral -citrulline (1 g/kg), once daily from day 1 to day 21 postirradiation. The combination treatment significantly improved the 30-day survival of mice after RCI from 15% (vehicle-treated) to 42%, and extended the median survival time by 4 days, as compared to vehicle controls. In addition, the combination therapy significantly increased body weight and bone marrow stem and progenitor cell clonogenicity in RCI mice, and accelerated recovery from RCI-induced intestinal injury, compared to animals treated with vehicle. Treatment with -citrulline alone also accelerated skin wound healing after RCI. In conclusion, these data indicate that the PEG-G-CSF and -citrulline combination therapy is a potentially effective countermeasure for mitigating RCI, likely by enhancing survival of the hematopoietic stem/progenitor cells and accelerating recovery from the RCI-induced intestinal injury and skin wounds.
Collapse
Affiliation(s)
- Li Wang
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| | - Min Zhai
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| | - Bin Lin
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| | - Wanchang Cui
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| | - Lisa Hull
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| | - Xianghong Li
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| | - Marsha N. Anderson
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| | - Joan T. Smith
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| | - Maria Victoria Umali
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| | - Suping Jiang
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| | - Juliann G. Kiang
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
- Department of Pharmacology and Molecular Therapy, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| | - Mang Xiao
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20889
| |
Collapse
|
9
|
Bene BJ, Blakely WF, Burmeister DM, Cary L, Chhetri SJ, Davis CM, Ghosh SP, Holmes-Hampton GP, Iordanskiy S, Kalinich JF, Kiang JG, Kumar VP, Lowy RJ, Miller A, Naeem M, Schauer DA, Senchak L, Singh VK, Stewart AJ, Velazquez EM, Xiao M. Celebrating 60 Years of Accomplishments of the Armed Forces Radiobiology Research Institute1. Radiat Res 2021; 196:129-146. [PMID: 33979439 DOI: 10.1667/21-00064.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 04/20/2021] [Indexed: 11/03/2022]
Abstract
Chartered by the U.S. Congress in 1961, the Armed Forces Radiobiology Research Institute (AFRRI) is a Joint Department of Defense (DoD) entity with the mission of carrying out the Medical Radiological Defense Research Program in support of our military forces around the globe. In the last 60 years, the investigators at AFRRI have conducted exploratory and developmental research with broad application to the field of radiation sciences. As the only DoD facility dedicated to radiation research, AFRRI's Medical Radiobiology Advisory Team provides deployable medical and radiobiological subject matter expertise, advising commanders in the response to a U.S. nuclear weapon incident and other nuclear or radiological material incidents. AFRRI received the DoD Joint Meritorious Unit Award on February 17, 2004, for its exceptionally meritorious achievements from September 11, 2001 to June 20, 2003, in response to acts of terrorism and nuclear/radiological threats at home and abroad. In August 2009, the American Nuclear Society designated the institute a nuclear historic landmark as the U.S.'s primary source of medical nuclear and radiological research, preparedness and training. Since then, research has continued, and core areas of study include prevention, assessment and treatment of radiological injuries that may occur from exposure to a wide range of doses (low to high). AFRRI collaborates with other government entities, academic institutions, civilian laboratories and other countries to research the biological effects of ionizing radiation. Notable early research contributions were the establishment of dose limits for major acute radiation syndromes in primates, applicable to human exposures, followed by the subsequent evolution of radiobiology concepts, particularly the importance of immune collapse and combined injury. In this century, the program has been essential in the development and validation of prophylactic and therapeutic drugs, such as Amifostine, Neupogen®, Neulasta®, Nplate® and Leukine®, all of which are used to prevent and treat radiation injuries. Moreover, AFRRI has helped develop rapid, high-precision, biodosimetry tools ranging from novel assays to software decision support. New drug candidates and biological dose assessment technologies are currently being developed. Such efforts are supported by unique and unmatched radiation sources and generators that allow for comprehensive analyses across the various types and qualities of radiation. These include but are not limited to both 60Co facilities, a TRIGA® reactor providing variable mixed neutron and γ-ray fields, a clinical linear accelerator, and a small animal radiation research platform with low-energy photons. There are five major research areas at AFRRI that encompass the prevention, assessment and treatment of injuries resulting from the effects of ionizing radiation: 1. biodosimetry; 2. low-level and low-dose-rate radiation; 3. internal contamination and metal toxicity; 4. radiation combined injury; and 5. radiation medical countermeasures. These research areas are bolstered by an educational component to broadcast and increase awareness of the medical effects of ionizing radiation, in the mass-casualty scenario after a nuclear detonation or radiological accidents. This work provides a description of the military medical operations as well as the radiation facilities and capabilities present at AFRRI, followed by a review and discussion of each of the research areas.
Collapse
Affiliation(s)
| | | | | | - Lynnette Cary
- Scientific Research Department.,Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | | | - Catherine M Davis
- Scientific Research Department.,Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Sanchita P Ghosh
- Scientific Research Department.,Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Gregory P Holmes-Hampton
- Scientific Research Department.,Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Sergey Iordanskiy
- Scientific Research Department.,Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | | | - Juliann G Kiang
- Scientific Research Department.,Medicine.,Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | | | | | | | | | - David A Schauer
- Radiation Sciences Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | | | - Vijay K Singh
- Scientific Research Department.,Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | | | | | | |
Collapse
|
10
|
Filimonova MV, Makarchuk VM, Shevchenko LI, Saburova AS, Surinova VI, Izmestieva OS, Lychagin AA, Saburov VO, Shegay PV, Kaprin AD, Ivanov SA, Filimonov AS. Radioprotective Activity of the Nitric Oxide Synthase Inhibitor T1023. Toxicological and Biochemical Properties, Cardiovascular and Radioprotective Effects. Radiat Res 2020; 194:532-543. [PMID: 34609510 DOI: 10.1667/rade-20-00046.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 08/03/2020] [Indexed: 11/03/2022]
Abstract
In this work, studies were performed to investigate the toxicological, biochemical, vasotropic and radiomodifying properties of the new nitric oxide synthase (NOS) inhibitor, compound T1023. Toxicological studies included the estimation of acute toxicity in mice after i.p. administration of T1023. Radiometric analysis and electron paramagnetic resonance spectroscopy were used to study NOS-inhibitory properties of T1023 in vitro and in vivo, respectively. T1023 vasoactive properties were studied in rat central hemodynamics. Radiobiological experiments were performed using endogenous and exogenous spleen colony formation as well as 30-day survival tests. The morphological changes in peripheral blood and bone marrow (BM) induced with T1023 were analyzed in mice during hematopoietic acute radiation syndrome (H-ARS). It was shown that T1023 is a sufficiently safe compound (LD10 of 317 mg/kg; LD50 of 410 mg/kg). It is an effective competitive NOS-inhibitor that is 10-to-15-fold selective to endothelial and inducible NOS (IC50 for nNOS, iNOS, eNOS: 52.3, 3.2 and 5.1 µM, respectively). Its NOS-inhibitory activity is realized in vivo and is accompanied by an increase in vascular tone. Its single i.p. administration in doses greater than 1/8 LD10 provides significant (40-50%) and long-lasting (more than 90 min) weakening of cardiac output, which can cause transient hypoxia. In radiobiological studies, T1023 proved to be a hypoxic radioprotector. Its radioprotective effect was observed only when administered prophylactically [single i.p dose, 5-120 min before total-body irradiation (TBI)] and only in doses that reduced cardiac output (1/8 LD10 and more, 40 mg/kg for mice), and was correlated in time with the dynamics of circulatory depression. Its radioprotective effect was not observed when administered in vitro and in the first 4 h after TBI. The optimal radioprotective doses of T1023 are relatively safe (1/ 5-1/4 LD10). In addition, T1023 effectively prevents H-ARS and gastrointestinal acute radiation syndrome (G-ARS) in experimental animals in vivo: dose modifying factor of 1.6-1.9. In the H-ARS mouse model, the prophylactic effect of T1023 (75 mg/kg, single i.p. injection) was accompanied by clinically significant effects. There was an express decrease in the degree of indicators of early BM devastation (by 40%) and maximal neutropenia and thrombocytopenia (2-2.5 times), in addition to a reduction in recovery time (by 30-40%). The obtained experimental results and literature data indicate that NOS inhibitors are an independent class of vasoactive radioprotectors with a specific hypoxic mechanism of action. NOS inhibitors provide new opportunities for developing effective and safe tools for the prevention of ARS.
Collapse
Affiliation(s)
- Marina V Filimonova
- A. Tsyb Medical Radiological Research Center - Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Victoria M Makarchuk
- A. Tsyb Medical Radiological Research Center - Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Ljudmila I Shevchenko
- A. Tsyb Medical Radiological Research Center - Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Alina S Saburova
- A. Tsyb Medical Radiological Research Center - Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Valentina I Surinova
- A. Tsyb Medical Radiological Research Center - Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Olga S Izmestieva
- A. Tsyb Medical Radiological Research Center - Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Anatoly A Lychagin
- A. Tsyb Medical Radiological Research Center - Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Vyacheslav O Saburov
- A. Tsyb Medical Radiological Research Center - Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Petr V Shegay
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Andrey D Kaprin
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Sergey A Ivanov
- A. Tsyb Medical Radiological Research Center - Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Alexander S Filimonov
- A. Tsyb Medical Radiological Research Center - Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| |
Collapse
|
11
|
Tyagi R, Maan K, Khushu S, Rana P. Urine metabolomics based prediction model approach for radiation exposure. Sci Rep 2020; 10:16063. [PMID: 32999294 PMCID: PMC7527994 DOI: 10.1038/s41598-020-72426-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 08/13/2020] [Indexed: 01/21/2023] Open
Abstract
The radiological incidents and terrorism have demanded the need for the development of rapid, precise, and non-invasive technique for detection and quantification of exposed dose of radiation. Though radiation induced metabolic markers have been thoroughly investigated, but reproducibility still needs to be elucidated. The present study aims at assessing the reliability and reproducibility of markers using nuclear magnetic resonance (NMR) spectroscopy and further deriving a logistic regression model based on these markers. C57BL/6 male mice (8-10 weeks) whole body γ-irradiated and sham irradiated controls were used. Urine samples collected at 24 h post dose were investigated using high resolution NMR spectroscopy and the datasets were analyzed using multivariate analysis. Fifteen distinguishable metabolites and 3 metabolic pathways (TCA cycle, taurine and hypotaurine metabolism, primary bile acid biosynthesis) were found to be amended. ROC curve and logistic regression was used to establish a diagnostic model as Logit (p) = log (p/1 - p) = -0.498 + 13.771 (tau) - 3.412 (citrate) - 34.461 (α-KG) + 515.183 (fumarate) with a sensitivity and specificity of 1.00 and 0.964 respectively. The findings demonstrate the proof of concept and the potential of NMR based metabolomics to establish a prediction model that can be implemented as a promising mass screening tool during triage.
Collapse
Affiliation(s)
- Ritu Tyagi
- Metabolomics Research Facility, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, S. K Mazumdar Road, Timarpur, Delhi, 110054, India
| | - Kiran Maan
- Metabolomics Research Facility, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, S. K Mazumdar Road, Timarpur, Delhi, 110054, India
| | - Subash Khushu
- NMR Research Centre, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, Delhi, India
| | - Poonam Rana
- Metabolomics Research Facility, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, S. K Mazumdar Road, Timarpur, Delhi, 110054, India.
| |
Collapse
|
12
|
Singh VK, Seed TM. Pharmacological management of ionizing radiation injuries: current and prospective agents and targeted organ systems. Expert Opin Pharmacother 2020; 21:317-337. [PMID: 31928256 PMCID: PMC6982586 DOI: 10.1080/14656566.2019.1702968] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 12/06/2019] [Indexed: 12/20/2022]
Abstract
Introduction: There is a limited array of currently available medicinals that are useful for either the prevention, mitigation or treatment of bodily injuries arising from ionizing radiation exposure.Area covered: In this brief article, the authors review those pharmacologic agents that either are currently being used to counter the injurious effects of radiation exposure, or those that show promise and are currently under development.Expert opinion: Although significant, but limited progress has been made in the development and fielding of safe and effective pharmacotherapeutics for select types of acute radiation-associated injuries, additional effort is needed to broaden the scope of drug development so that overall health risks associated with both short- and long-term injuries in various organ systems can be reduced and effectively managed. There are several promising radiation countermeasures that may gain regulatory approval from the government in the near future for use in clinical settings and in the aftermath of nuclear/radiological exposure contingencies.
Collapse
Affiliation(s)
- Vijay K. Singh
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Thomas M Seed
- Tech Micro Services, 4417 Maple Avenue, Bethesda, MD 20814, USA
| |
Collapse
|
13
|
Wu T, Liu W, Fan T, Zhong H, Zhou H, Guo W, Zhu X. 5-Androstenediol prevents radiation injury in mice by promoting NF-κB signaling and inhibiting AIM2 inflammasome activation. Biomed Pharmacother 2019; 121:109597. [PMID: 31726369 DOI: 10.1016/j.biopha.2019.109597] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/22/2019] [Accepted: 10/26/2019] [Indexed: 02/06/2023] Open
Abstract
In the present study, the therapeutic effects of 5-androstenediol on radiation-induced myeloid suppression and tissue damage in mice and the possible mechanism were explored. The mice were subjected to whole-body irradiation, and 5-androstenediol was administered subcutaneously at different times and doses. The evaluation of the survival rate showed that the administration of 5-androstenediol every three days post-irradiation was the most effective in decreasing the death of the mice. Additionally, 5-androstenediol dose-dependently reduced the death caused by 9 Gy radiation. The pharmacological mechanism was investigated by blood analysis, western blot analysis, immunofluorescence and immunohistochemistry. 5-Androstenediol significantly ameliorated myeloid suppression, as demonstrated by elevated levels of total white blood cells, including neutrophils and platelets, in the peripheral blood. By H&E staining, we found that radiation-induced myeloid suppression in the bone marrow and spleen, as well as tissue damage in the lung and colon, was significantly ameliorated by treatment with 5-androstenediol. Immunohistochemistry showed elevated phosphorylation of p65 in the bone marrow and spleen, indicating the activation of NF-κB signaling. Moreover, 5-androstenediol markedly hampered the radiation-induced activation of caspase-1 and GSDMD in the colon by decreasing the interaction between AIM2 and ASC. Taken together, our results suggest that, by promoting NF-κB signaling and inhibiting inflammasome-mediated pyroptosis, 5-androstenediol can be used as a radioprotective drug.
Collapse
Affiliation(s)
- Tiancong Wu
- Jinling Hospital, Department of Radiation Oncology, Nanjing University, School Medicine, Nanjing, 210002, PR China
| | - Wen Liu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing, 210093, PR China
| | - Ting Fan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing, 210093, PR China
| | - Haiqing Zhong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing, 210093, PR China
| | - Han Zhou
- Jinling Hospital, Department of Radiation Oncology, Nanjing University, School Medicine, Nanjing, 210002, PR China
| | - Wenjie Guo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing, 210093, PR China.
| | - Xixu Zhu
- Jinling Hospital, Department of Radiation Oncology, Nanjing University, School Medicine, Nanjing, 210002, PR China.
| |
Collapse
|
14
|
Hofer M, Hoferová Z, Falk M. Brief Story on Prostaglandins, Inhibitors of their Synthesis, Hematopoiesis, and Acute Radiation Syndrome. Molecules 2019; 24:molecules24224019. [PMID: 31698831 PMCID: PMC6891503 DOI: 10.3390/molecules24224019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 10/31/2019] [Accepted: 11/04/2019] [Indexed: 01/22/2023] Open
Abstract
Prostaglandins and inhibitors of their synthesis (cyclooxygenase (COX) inhibitors, non-steroidal anti-inflammatory drugs) were shown to play a significant role in the regulation of hematopoiesis. Partly due to their hematopoiesis-modulating effects, both prostaglandins and COX inhibitors were reported to act positively in radiation-exposed mammalian organisms at various pre- and post-irradiation therapeutical settings. Experimental efforts were targeted at finding pharmacological procedures leading to optimization of therapeutical outcomes by minimizing undesirable side effects of the treatments. Progress in these efforts was obtained after discovery of selective inhibitors of inducible selective cyclooxygenase-2 (COX-2) inhibitors. Recent studies have been able to suggest the possibility to find combined therapeutical approaches utilizing joint administration of prostaglandins and inhibitors of their synthesis at optimized timing and dosing of the drugs which could be incorporated into the therapy of patients with acute radiation syndrome.
Collapse
Affiliation(s)
- Michal Hofer
- Correspondence: ; Tel.: +420-541-517-171; Fax: +420-541-211-293
| | | | | |
Collapse
|
15
|
Singh VK, Seed TM. The efficacy and safety of amifostine for the acute radiation syndrome. Expert Opin Drug Saf 2019; 18:1077-1090. [PMID: 31526195 DOI: 10.1080/14740338.2019.1666104] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction: A radiation countermeasure that can be used prior to radiation exposure to protect the population from the harmful effects of radiation exposure remains a major unmet medical need and is recognized as an important area for research. Despite substantial advances in the research and development for finding nontoxic, safe, and effective prophylactic countermeasures for the acute radiation syndrome (ARS), no such agent has been approved by the United States Food and Drug Administration (FDA). Area covered: Despite the progress made to improve the effectiveness of amifostine as a radioprotector for ARS, none of the strategies have resolved the issue of its toxicity/side effects. Thus, the FDA has approved amifostine for limited clinical indications, but not for non-clinical uses. This article reviews recent strategies and progress that have been made to move forward this potentially useful countermeasure for ARS. Expert opinion: Although the recent investigations have been promising for fielding safe and effective radiation countermeasures, additional work is needed to improve and advance drug design and delivery strategies to get FDA approval for broadened, non-clinical use of amifostine during a radiological/nuclear scenario.
Collapse
Affiliation(s)
- Vijay K Singh
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences , Bethesda , MD , USA.,Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences , Bethesda , MD , USA
| | | |
Collapse
|
16
|
Yu L, Meng F, Guo M, Yang Y, He J, Dong J, Guo J, Peng S. Developing a robust LC-MS/MS method to quantify Zn-DTPA, a zinc chelate in human plasma and urine. Biomed Chromatogr 2018; 32:e4298. [DOI: 10.1002/bmc.4298] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 05/10/2018] [Accepted: 05/16/2018] [Indexed: 11/09/2022]
Affiliation(s)
- Lin Yu
- Academy of Military Medicine, Academy of Military Sciences; Beijing People's Republic of China
- Institute of Disease Control and Prevention, People's Liberation Army; Beijing People's Republic of China
| | - Fanhua Meng
- Academy of Military Medicine, Academy of Military Sciences; Beijing People's Republic of China
| | - Miao Guo
- Institute of Disease Control and Prevention, People's Liberation Army; Beijing People's Republic of China
| | - Ying Yang
- Institute of Disease Control and Prevention, People's Liberation Army; Beijing People's Republic of China
| | - Jianchang He
- Kunming General Hospital of Chengdu Military Region; Kunming People's Republic of China
| | - Junxing Dong
- Academy of Military Medicine, Academy of Military Sciences; Beijing People's Republic of China
| | - Jifen Guo
- Wanshu (Beijing) Pharmaceutical Technology Co. Ltd; Beijing People's Republic of China
| | - Shuangqing Peng
- Institute of Disease Control and Prevention, People's Liberation Army; Beijing People's Republic of China
| |
Collapse
|
17
|
Roh C. Metabolomics in Radiation-Induced Biological Dosimetry: A Mini-Review and a Polyamine Study. Biomolecules 2018; 8:biom8020034. [PMID: 29844258 PMCID: PMC6023017 DOI: 10.3390/biom8020034] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/18/2018] [Accepted: 05/25/2018] [Indexed: 01/09/2023] Open
Abstract
In this study, we elucidate that polyamine metabolite is a powerful biomarker to study post-radiation changes. Metabolomics in radiation biodosimetry, the application of a metabolomics analysis to the field of radiobiology, promises to increase the understanding of biological responses by ionizing radiation (IR). Radiation exposure triggers a complex network of molecular and cellular responses that impacts metabolic processes and alters the levels of metabolites. Such metabolites have potential as biomarkers for radiation dosimetry. Among metabolites, polyamine is one of many potential biomarkers to estimate radiation response. In addition, this review provides an opportunity for the understanding of a radiation metabolomics in biodosimetry and a polyamine case study.
Collapse
Affiliation(s)
- Changhyun Roh
- Biotechnology Research Division, Advanced Radiation Technology Institute (ARTI), Korea Atomic Energy Research Institute (KAERI), 29, Geumgu-gil, Jeongeup-si, Jeonbuk 56212, Korea.
- Radiation Biotechnology and Applied Radioisotope Science, University of Science Technology (UST), 217 Gajeong-ro, Daejeon 34113, Korea.
| |
Collapse
|
18
|
Suresh Kumar MA, Laiakis EC, Ghandhi SA, Morton SR, Fornace AJ, Amundson SA. Gene Expression in Parp1 Deficient Mice Exposed to a Median Lethal Dose of Gamma Rays. Radiat Res 2018; 190:53-62. [PMID: 29746213 DOI: 10.1667/rr14990.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
There is a current interest in the development of biodosimetric methods for rapidly assessing radiation exposure in the wake of a large-scale radiological event. This work was initially focused on determining the exposure dose to an individual using biological indicators. Gene expression signatures show promise for biodosimetric application, but little is known about how these signatures might translate for the assessment of radiological injury in radiosensitive individuals, who comprise a significant proportion of the general population, and who would likely require treatment after exposure to lower doses. Using Parp1-/- mice as a model radiation-sensitive genotype, we have investigated the effect of this DNA repair deficiency on the gene expression response to radiation. Although Parp1 is known to play general roles in regulating transcription, the pattern of gene expression changes observed in Parp1-/- mice 24 h postirradiation to a LD50/30 was remarkably similar to that in wild-type mice after exposure to LD50/30. Similar levels of activation of both the p53 and NFκB radiation response pathways were indicated in both strains. In contrast, exposure of wild-type mice to a sublethal dose that was equal to the Parp1-/- LD50/30 resulted in a lower magnitude gene expression response. Thus, Parp1-/- mice displayed a heightened gene expression response to radiation, which was more similar to the wild-type response to an equitoxic dose than to an equal absorbed dose. Gene expression classifiers trained on the wild-type data correctly identified all wild-type samples as unexposed, exposed to a sublethal dose or exposed to an LD50/30. All unexposed samples from Parp1-/- mice were also correctly classified with the same gene set, and 80% of irradiated Parp1-/- samples were identified as exposed to an LD50/30. The results of this study suggest that, at least for some pathways that may influence radiosensitivity in humans, specific gene expression signatures have the potential to accurately detect the extent of radiological injury, rather than serving only as a surrogate of physical radiation dose.
Collapse
Affiliation(s)
- M A Suresh Kumar
- a Center for Radiological Research, Columbia University Medical Center, Columbia University, New York, New York
| | - Evagelia C Laiakis
- b Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, DC
| | - Shanaz A Ghandhi
- a Center for Radiological Research, Columbia University Medical Center, Columbia University, New York, New York
| | - Shad R Morton
- a Center for Radiological Research, Columbia University Medical Center, Columbia University, New York, New York
| | - Albert J Fornace
- b Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, DC
| | - Sally A Amundson
- a Center for Radiological Research, Columbia University Medical Center, Columbia University, New York, New York
| |
Collapse
|
19
|
Rudqvist N, Laiakis EC, Ghandhi SA, Kumar S, Knotts JD, Chowdhury M, Fornace AJ, Amundson SA. Global Gene Expression Response in Mouse Models of DNA Repair Deficiency after Gamma Irradiation. Radiat Res 2018; 189:337-344. [PMID: 29351057 DOI: 10.1667/rr14862.1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
In the event of an improvised nuclear device or "dirty bomb" in a highly populated area, potentially hundreds of thousands of people will require screening to ensure that exposed individuals receive appropriate treatment. For this reason, there is a need to develop tools for high-throughput radiation biodosimetry. Gene expression represents an emerging approach to biodosimetry and could potentially provide an estimate of both absorbed dose and individual radiation-induced injury. Since approximately 2-4% of humans are thought to be radiosensitive, and would suffer greater radiological injury at a given dose than members of the general population, it is of interest to explore the potential impact of such sensitivity on the biodosimetric gene expression signatures being developed. In this study, we used wild-type mice and genetically engineered mouse models deficient in two DNA repair pathways that can contribute to radiation sensitivity to estimate the maximum effect of differences in radiosensitivity. We compared gene expression in response to a roughly equitoxic (LD50/30) dose of gamma rays in wild-type C57BL/6 (8 Gy) and DNA double-strand break repair-deficient Atm-/- (4 Gy) and Prkdcscid (3 Gy) mutants of C57BL/6. Overall, 780 genes were significantly differentially expressed in wild-type mice one day postirradiation, 232 in Atm-/- and 269 in Prkdcscid. Upstream regulators including TP53 and NFκB were predicted to be activated by radiation exposure in the wild-type mice, but not in either of the DNA repair-deficient mutant strains. There was also a significant muting of the apparent inflammatory response triggered by radiation in both mutant strains. These differences impacted the ability of gene expression signatures developed in wild-type mice to detect potentially fatal radiation exposure in the DNA repair-deficient mice, with the greatest impact on Atm-/- mice. However, the inclusion of mutant mice in gene selection vastly improved performance of the classifiers.
Collapse
Affiliation(s)
- Nils Rudqvist
- a Center for Radiological Research, Columbia University Medical Center, Columbia University, New York, New York
| | - Evagelia C Laiakis
- b Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, DC
| | - Shanaz A Ghandhi
- a Center for Radiological Research, Columbia University Medical Center, Columbia University, New York, New York
| | - Suresh Kumar
- a Center for Radiological Research, Columbia University Medical Center, Columbia University, New York, New York
| | - Jeffrey D Knotts
- a Center for Radiological Research, Columbia University Medical Center, Columbia University, New York, New York
| | - Mashkura Chowdhury
- a Center for Radiological Research, Columbia University Medical Center, Columbia University, New York, New York
| | - Albert J Fornace
- b Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, DC
| | - Sally A Amundson
- a Center for Radiological Research, Columbia University Medical Center, Columbia University, New York, New York
| |
Collapse
|
20
|
Cheema AK, Mehta KY, Fatanmi OO, Wise SY, Hinzman CP, Wolff J, Singh VK. A Metabolomic and Lipidomic Serum Signature from Nonhuman Primates Administered with a Promising Radiation Countermeasure, Gamma-Tocotrienol. Int J Mol Sci 2017; 19:E79. [PMID: 29283379 PMCID: PMC5796029 DOI: 10.3390/ijms19010079] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 12/20/2017] [Accepted: 12/27/2017] [Indexed: 12/13/2022] Open
Abstract
The development of radiation countermeasures for acute radiation syndrome (ARS) has been underway for the past six decades, leading to the identification of multiple classes of radiation countermeasures. However, to date, only two growth factors (Neupogen and Neulasta) have been approved by the United States Food and Drug Administration (US FDA) for the mitigation of hematopoietic acute radiation syndrome (H-ARS). No radioprotector for ARS has been approved by the FDA yet. Gamma-tocotrienol (GT3) has been demonstrated to have radioprotective efficacy in murine as well as nonhuman primate (NHP) models. Currently, GT3 is under advanced development as a radioprotector that can be administered prior to radiation exposure. We are studying this agent for its safety profile and efficacy using the NHP model. In this study, we analyzed global metabolomic and lipidomic changes using ultra-performance liquid chromatography (UPLC) quadrupole time-of-flight mass spectrometry (QTOF-MS) in serum samples of NHPs administered GT3. Our study, using 12 NHPs, demonstrates that alterations in metabolites manifest only 24 h after GT3 administration. Furthermore, metabolic changes are associated with transient increase in the bioavailability of antioxidants, including lactic acid and cholic acid and anti-inflammatory metabolites 3 deoxyvitamin D3, and docosahexaenoic acid. Taken together, our results show that the administration of GT3 to NHPs causes metabolic shifts that would provide an overall advantage to combat radiation injury. This initial assessment also highlights the utility of metabolomics and lipidomics to determine the underlying physiological mechanisms involved in the radioprotective efficacy of GT3.
Collapse
Affiliation(s)
- Amrita K Cheema
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA.
- Department of Biochemistry, Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC 20057, USA.
| | - Khyati Y Mehta
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA.
| | - Oluseyi O Fatanmi
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA.
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA.
| | - Stephen Y Wise
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA.
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA.
| | - Charles P Hinzman
- Department of Biochemistry, Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC 20057, USA.
| | - Josh Wolff
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA.
| | - Vijay K Singh
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA.
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA.
| |
Collapse
|
21
|
Kiang JG, Smith JT, Anderson MN, Elliott TB, Gupta P, Balakathiresan NS, Maheshwari RK, Knollmann-Ritschel B. Hemorrhage enhances cytokine, complement component 3, and caspase-3, and regulates microRNAs associated with intestinal damage after whole-body gamma-irradiation in combined injury. PLoS One 2017; 12:e0184393. [PMID: 28934227 PMCID: PMC5608216 DOI: 10.1371/journal.pone.0184393] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 08/23/2017] [Indexed: 12/14/2022] Open
Abstract
Hemorrhage following whole-body γ-irradiation in a combined injury (CI) model increases mortality compared to whole-body γ-irradiation alone (RI). The decreased survival in CI is accompanied by increased bone marrow injury, decreased hematocrit, and alterations of miRNA in the kidney. In this study, our aim was to examine cytokine homeostasis, susceptibility to systemic bacterial infection, and intestinal injury. More specifically, we evaluated the interleukin-6 (IL-6)-induced stress proteins including C-reactive protein (CRP), complement 3 (C3), Flt-3 ligand, and corticosterone. CD2F1 male mice received 8.75 Gy 60Co gamma photons (0.6 Gy/min, bilateral) which was followed by a hemorrhage of 20% of the blood volume. In serum, RI caused an increase of IL-1, IL-2, IL-3, IL-5, IL-6, IL-12, IL-13, IL-15, IL-17A, IL-18, G-CSF, CM-CSF, eotaxin, IFN-γ, MCP-1, MIP, RANTES, and TNF-α, which were all increased by hemorrhage alone, except IL-9, IL-17A, and MCP-1. Nevertheless, CI further elevated RI-induced increases of these cytokines except for G-CSF, IFN- γ and RANTES in serum. In the ileum, hemorrhage in the CI model significantly enhanced RI-induced IL-1β, IL-3, IL-6, IL-10, IL-12p70, IL-13, IL-18, and TNF-α concentrations. In addition, Proteus mirabilis Gram(-) was found in only 1 of 6 surviving RI mice on Day 15, whereas Streptococcus sanguinis Gram(+) and Sphingomonas paucimobilis Gram(-) were detected in 2 of 3 surviving CI mice (with 3 CI mice diseased due to inflammation and infection before day 15) at the same time point. Hemorrhage in the CI model enhanced the RI-induced increases in C3 and decreases in CRP concentrations. However, hemorrhage alone did not alter the basal levels, but hemorrhage in the CI model displayed similar increases in Flt-3 ligand levels as RI did. Hemorrhage alone altered the basal levels of corticosterone early after injury, which then returned to the baseline, but in RI mice and CI mice the increased corticosterone concentration remained elevated throughout the 15 day study. CI increased 8 miRNAs and decreased 10 miRNAs in serum, and increased 16 miRNA and decreased 6 miRNAs in ileum tissue. Among the altered miRNAs, CI increased miR-34 in the serum and ileum which targeted an increased phosphorylation of ERK, p38, and increased NF-κB, thereby leading to increased iNOS expression and activation of caspase-3 in the ileum. Further, let-7g/miR-98 targeted the increased phosphorylation of STAT3 in the ileum, which is known to bind to the iNOS gene. These changes may correlate with cell death in the ileum of CI mice. The histopathology displayed blunted villi and villus edema in RI and CI mice. Based on the in silico analysis, miR-15, miR-99, and miR-100 were predicted to regulate IL-6 and TNF. These results suggest that CI-induced alterations of cytokines/chemokines, CRP, and C3 cause a homeostatic imbalance and may contribute to the pathophysiology of the gastrointestinal injury. Inhibitory intervention in these responses may prove therapeutic for CI and improve recovery of the ileal morphologic damage.
Collapse
Affiliation(s)
- Juliann G. Kiang
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, Maryland, United States of America
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Joan T. Smith
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, Maryland, United States of America
| | - Marsha N. Anderson
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, Maryland, United States of America
| | - Thomas B. Elliott
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, Maryland, United States of America
| | - Paridhi Gupta
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Nagaraja S. Balakathiresan
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Radha K. Maheshwari
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Barbara Knollmann-Ritschel
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| |
Collapse
|
22
|
Guo J, Chen Y, Lei X, Xu Y, Liu Z, Cai J, Gao F, Yang Y. Monophosphoryl lipid a attenuates radiation injury through TLR4 activation. Oncotarget 2017; 8:86031-86042. [PMID: 29156775 PMCID: PMC5689665 DOI: 10.18632/oncotarget.20907] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 08/04/2017] [Indexed: 11/25/2022] Open
Abstract
Ionizing radiation causes severe damage to human body, and normal tissue toxicity in cancer radiotherapy also limits its further application. It is urgently required to develop safe and effective radioprotector. Our previous study has shown that toll like receptor 4 (TLR4) was dispensable for basal radiation resistance. However, severe toxicity of its traditional agonist lipopolysaccharide limits the clinical application. In present study, we demonstrated that monophosphoryl lipid A (MPLA), a potent TLR4 agonist with low toxicity, effectively attenuated radiation injury on in vitro and in vivo. MPLA increased cell survival and inhibited cell apoptosis after irradiation, and cell cycle arrest was also inhibited. Radiosensitive tissues including spleen, intestine, bone marrow and testis were protected from radiation damages in a TLR4 dependent manner. We also found that myeloid differentiation factor 88 (MyD88) accounted more than Toll/IL-1R domain-containing adaptor inducing IFN-β (TRIF) for the radioprotective effects of MPLA. In conclusion, our finding suggests TLR4 agonist MPLA as a safe and effective radioprotector for clinical application.
Collapse
Affiliation(s)
- Jiaming Guo
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, 200433, P.R. China
| | - Yuanyuan Chen
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, 200433, P.R. China
| | - Xiao Lei
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, 200433, P.R. China
| | - Yang Xu
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, 200433, P.R. China
| | - Zhe Liu
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, 200433, P.R. China
| | - Jianming Cai
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, 200433, P.R. China
| | - Fu Gao
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, 200433, P.R. China
| | - Yanyong Yang
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, 200433, P.R. China
| |
Collapse
|
23
|
Hofer M, Hoferová Z, Falk M. Pharmacological Modulation of Radiation Damage. Does It Exist a Chance for Other Substances than Hematopoietic Growth Factors and Cytokines? Int J Mol Sci 2017; 18:E1385. [PMID: 28657605 PMCID: PMC5535878 DOI: 10.3390/ijms18071385] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 06/21/2017] [Accepted: 06/26/2017] [Indexed: 02/03/2023] Open
Abstract
In recent times, cytokines and hematopoietic growth factors have been at the center of attention for many researchers trying to establish pharmacological therapeutic procedures for the treatment of radiation accident victims. Two granulocyte colony-stimulating factor-based radiation countermeasures have been approved for the treatment of the hematopoietic acute radiation syndrome. However, at the same time, many different substances with varying effects have been tested in animal studies as potential radioprotectors and mitigators of radiation damage. A wide spectrum of these substances has been studied, comprising various immunomodulators, prostaglandins, inhibitors of prostaglandin synthesis, agonists of adenosine cell receptors, herbal extracts, flavonoids, vitamins, and others. These agents are often effective, relatively non-toxic, and cheap. This review summarizes the results of animal experiments, which show the potential for some of these untraditional or new radiation countermeasures to become a part of therapeutic procedures applicable in patients with the acute radiation syndrome. The authors consider β-glucan, 5-AED (5-androstenediol), meloxicam, γ-tocotrienol, genistein, IB-MECA (N⁶-(3-iodobezyl)adenosine-5'-N-methyluronamide), Ex-RAD (4-carboxystyryl-4-chlorobenzylsulfone), and entolimod the most promising agents, with regards to their contingent use in clinical practice.
Collapse
Affiliation(s)
- Michal Hofer
- Department of Cell Biology and Radiobiology, Institute of Biophysics, v.v.i., Czech Academy of Sciences, Královopolská 135, 61265 Brno, Czech Republic.
| | - Zuzana Hoferová
- Department of Cell Biology and Radiobiology, Institute of Biophysics, v.v.i., Czech Academy of Sciences, Královopolská 135, 61265 Brno, Czech Republic.
| | - Martin Falk
- Department of Cell Biology and Radiobiology, Institute of Biophysics, v.v.i., Czech Academy of Sciences, Královopolská 135, 61265 Brno, Czech Republic.
| |
Collapse
|
24
|
Singh VK, Seed TM. A review of radiation countermeasures focusing on injury-specific medicinals and regulatory approval status: part I. Radiation sub-syndromes, animal models and FDA-approved countermeasures. Int J Radiat Biol 2017. [PMID: 28650707 DOI: 10.1080/09553002.2017.1332438] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE The increasing global risk of nuclear and radiological accidents or attacks has driven renewed research interest in developing medical countermeasures to potentially injurious exposures to acute irradiation. Clinical symptoms and signs of a developing acute radiation injury, i.e. the acute radiation syndrome, are grouped into three sub-syndromes named after the dominant organ system affected, namely the hematopoietic, gastrointestinal, and neurovascular systems. The availability of safe and effective countermeasures against the above threats currently represents a significant unmet medical need. This is the first article within a three-part series covering the nature of the radiation sub-syndromes, various animal models for radiation countermeasure development, and the agents currently approved by the United States Food and Drug Administration for countering the medical consequences of several of these prominent radiation exposure-associated syndromes. CONCLUSIONS From the U.S. and global perspectives, biomedical research concerning medical countermeasure development is quite robust, largely due to increased government funding following the 9/11 incidence and subsequent rise of terrorist-associated threats. A wide spectrum of radiation countermeasures for specific types of radiation injuries is currently under investigation. However, only a few radiation countermeasures have been fully approved by regulatory agencies for human use during radiological/nuclear contingencies. Additional research effort, with additional funding, clearly will be needed in order to fill this significant, unmet medical health problem.
Collapse
Affiliation(s)
- Vijay K Singh
- a Division of Radioprotection, Department of Pharmacology and Molecular Therapeutics , F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences , Bethesda , MD , USA.,b Armed Forces Radiobiology Research Institute , Uniformed Services University of the Health Sciences , Bethesda , MD , USA
| | | |
Collapse
|
25
|
Hofer M, Hoferová Z, Depeš D, Falk M. Combining Pharmacological Countermeasures to Attenuate the Acute Radiation Syndrome-A Concise Review. Molecules 2017; 22:molecules22050834. [PMID: 28534834 PMCID: PMC6154336 DOI: 10.3390/molecules22050834] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 05/16/2017] [Accepted: 05/16/2017] [Indexed: 11/16/2022] Open
Abstract
The goal of combined pharmacological approaches in the treatment of the acute radiation syndrome (ARS) is to obtain an effective therapy producing a minimum of undesirable side effects. This review summarizes important data from studies evaluating the efficacy of combining radioprotective agents developed for administration prior to irradiation and therapeutic agents administered in a post-irradiation treatment regimen. Many of the evaluated results show additivity, or even synergism, of the combined treatments in comparison with the effects of the individual component administrations. It can be deduced from these findings that the research in which combined treatments with radioprotectors/radiomitigators are explored, tested, and evaluated is well-founded. The requirement for studies highly emphasizing the need to minimize undesirable side effects of the radioprotective/radiomitigating therapies is stressed.
Collapse
Affiliation(s)
- Michal Hofer
- Department of Cell Biology and Radiobiology, Institute of Biophysics, v.v.i., Czech Academy of Sciences, Královopolská 135, 61265 Brno, Czech Republic.
| | - Zuzana Hoferová
- Department of Cell Biology and Radiobiology, Institute of Biophysics, v.v.i., Czech Academy of Sciences, Královopolská 135, 61265 Brno, Czech Republic.
| | - Daniel Depeš
- Department of Cell Biology and Radiobiology, Institute of Biophysics, v.v.i., Czech Academy of Sciences, Královopolská 135, 61265 Brno, Czech Republic.
| | - Martin Falk
- Department of Cell Biology and Radiobiology, Institute of Biophysics, v.v.i., Czech Academy of Sciences, Královopolská 135, 61265 Brno, Czech Republic.
| |
Collapse
|
26
|
Dynlacht JR, Garrett J, Joel R, Lane K, Mendonca MS, Orschell CM. Further Characterization of the Mitigation of Radiation Lethality by Protective Wounding. Radiat Res 2017; 187:732-742. [PMID: 28437188 DOI: 10.1667/rr14725.1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
There continues to be a major effort in the United States to develop mitigators for the treatment of mass casualties that received high-intensity acute ionizing radiation exposures from the detonation of an improvised nuclear device during a radiological terrorist attack. The ideal countermeasure should be effective when administered after exposure, and over a wide range of absorbed doses. We have previously shown that the administration of a subcutaneous incision of a defined length, if administered within minutes after irradiation, protected young adult female C57BL/6 mice against radiation-induced lethality, and increased survival after total-body exposure to an LD50/30 X-ray dose from 50% to over 90%. We refer to this approach as "protective wounding". In this article, we report on our efforts to further optimize, characterize and demonstrate the validity of the protective wounding response by comparing the response of female and male mice, varying the radiation dose, the size of the wound, and the timing of wounding with respect to administration of the radiation dose. Both male and female mice that received a subcutaneous incision after irradiation were significantly protected from radiation lethality. We observed that the extent of protection against lethality after an LD50/30 X-ray dose was independent of the size of the subcutaneous cut, and that a 3 mm subcutaneous incision is effective at enhancing the survival of mice exposed to a broad range of radiation doses (LD15-LD100). Over the range of 6.2-6.7 Gy, the increase in survival observed in mice that received an incision was associated with an enhanced recovery of hematopoiesis. The enhanced rate of recovery of hematopoiesis was preceded by an increase in the production of a select group of cytokines. Thus, a thorough knowledge of the timing of the cytokine cascade after wounding could aid in the development of novel pharmacological radiation countermeasures that can be administered several days after the actual radiation exposure.
Collapse
Affiliation(s)
- Joseph R Dynlacht
- a Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Joy Garrett
- a Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Rebecca Joel
- a Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Katharina Lane
- a Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Marc S Mendonca
- a Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Christie M Orschell
- b Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202
| |
Collapse
|
27
|
Zhang Y, Li J, Meng Z, Zhu X, Gan H, Gu R, Wu Z, Zheng Y, Wei J, Dou G. A sharp, robust, and quantitative method by liquid chromatography tandem mass spectrometry for the measurement of EAD for acute radiation syndrome and its application. J Chromatogr B Analyt Technol Biomed Life Sci 2017; 1055-1056:45-50. [PMID: 28445846 DOI: 10.1016/j.jchromb.2017.03.045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 03/22/2017] [Accepted: 03/23/2017] [Indexed: 11/26/2022]
Abstract
17-Ethinyl-3,17-dihydroxyandrost-5-ene (EAD) is an agent designed for the treatment of acute radiation syndrome (ARS). Given its vital role played in the prevention and mitigation of ARS, the development of a sharp, sensitive and robust liquid chromatography tandem mass spectrometry (LC-MS/MS) method to monitor the metabolism of EAD in vivo was crucial. A new method was constructed and validated for the determination of EAD with the internal standard of androst-5-ene-3β,17β-diol (5-AED). The blood samples were precipitated with methanol, centrifuged, from which the supernatant was separated on UPLC with C18 column and eluted in gradient with acetonitrile and Milli-Q water both containing 0.1% formic acid (FA). Quantification was performed by a triple quadrupole mass spectrometer with electro spray ionization (ESI) in multiple reactive monitoring (MRM) positive mode. A good linearity was obtained with R>0.99 for EAD within its calibration range from 5 to 1000ngmL-1 with a lowest limit of quantification (LLOQ) of 5ngmL-1. Inter- and intra-day accuracy and precision of three levels of quality control (QC) samples were within the range of 15%, while the LLOQ was within 20%. Samples were stable under the circumstances of the experiments. The method was simple, accurate and robust applied to determine the concentrations of EAD in Wistar rat after a single administration of EAD orally at the dose of 100mgkg-1.
Collapse
Affiliation(s)
- Yiwei Zhang
- Guangxi Medical University, 22 Shuangyong Road, Nanning 530000, China; Laboratory of Drug Metabolism and Pharmacokinetics, Beijing Institute of Transfusion Medicine, 27 Taiping Road, Beijing 100850, China.
| | - Jian Li
- Laboratory of Drug Metabolism and Pharmacokinetics, Beijing Institute of Transfusion Medicine, 27 Taiping Road, Beijing 100850, China
| | - Zhiyun Meng
- Laboratory of Drug Metabolism and Pharmacokinetics, Beijing Institute of Transfusion Medicine, 27 Taiping Road, Beijing 100850, China
| | - Xiaoxia Zhu
- Laboratory of Drug Metabolism and Pharmacokinetics, Beijing Institute of Transfusion Medicine, 27 Taiping Road, Beijing 100850, China
| | - Hui Gan
- Laboratory of Drug Metabolism and Pharmacokinetics, Beijing Institute of Transfusion Medicine, 27 Taiping Road, Beijing 100850, China
| | - Ruolan Gu
- Laboratory of Drug Metabolism and Pharmacokinetics, Beijing Institute of Transfusion Medicine, 27 Taiping Road, Beijing 100850, China
| | - Zhuona Wu
- Laboratory of Drug Metabolism and Pharmacokinetics, Beijing Institute of Transfusion Medicine, 27 Taiping Road, Beijing 100850, China
| | - Ying Zheng
- Laboratory of Drug Metabolism and Pharmacokinetics, Beijing Institute of Transfusion Medicine, 27 Taiping Road, Beijing 100850, China
| | - Jinbin Wei
- Guangxi Medical University, 22 Shuangyong Road, Nanning 530000, China.
| | - Guifang Dou
- Laboratory of Drug Metabolism and Pharmacokinetics, Beijing Institute of Transfusion Medicine, 27 Taiping Road, Beijing 100850, China.
| |
Collapse
|
28
|
Khan S, Adhikari JS, Rizvi MA, Chaudhury NK. Melatonin attenuates 60 Co γ-ray-induced hematopoietic, immunological and gastrointestinal injuries in C57BL/6 male mice. ENVIRONMENTAL TOXICOLOGY 2017; 32:501-518. [PMID: 26948951 DOI: 10.1002/tox.22254] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Revised: 02/09/2016] [Accepted: 02/14/2016] [Indexed: 06/05/2023]
Abstract
Protection of hematopoietic, immunological, and gastrointestinal injuries from deleterious effects of ionizing radiation is prime rational for developing radioprotector. The objective of this study, therefore, was to evaluate the radioprotective potential of melatonin against damaging effects of radiation-induced hematopoietic, immunological, and gastrointestinal injuries in mice. C57BL/6 male mice were intraperitoneally administered with melatonin (50-150 mg/kg) 30 min prior to whole-body radiation exposure of 5 and 7.5 Gy using 60 Co-teletherapy unit. Thirty-day survival against 7.5 Gy was monitored. Melatonin (100 mg/kg) pretreatment showed 100% survival against 7.5 Gy radiation dose. Melatonin pretreatment expanded femoral HPSCs, and inhibited spleenocyte DNA strands breaks and apoptosis in irradiated mice. At this time, it also protected radiation-induced loss of T cell sub-populations in spleen. In addition, melatonin pretreatment enhanced crypts regeneration and increased villi number and length in irradiated mice. Translocation of gut bacteria to spleen, liver and kidney were controlled in irradiated mice pretreated with melatonin. Radiation-induced gastrointestinal DNA strand breaks, lipid peroxidation, and expression of proapoptotic-p53, Bax, and antiapoptotic-Bcl-xL proteins were reversed in melatonin pretreated mice. This increase of Bcl-xL was associated with the decrease of Bax/Bcl-xL ratio. ABTS and DPPH radical assays revealed that melatonin treatment alleviated total antioxidant capacity in hematopoietic and gastrointestinal tissues. Present study demonstrated that melatonin pretreatment was able to prevent hematopoietic, immunological, and gastrointestinal radiation-induced injury, therefore, overcoming lethality in mice. These results suggest potential of melatonin in developing radioprotector for protection of bone marrow, spleen, and gastrointestine in planned radiation exposure scenarios including radiotherapy. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 501-518, 2017.
Collapse
Affiliation(s)
- Shahanshah Khan
- Division of Radiation Biodosimetry, Institute of Nuclear Medicine and Allied Sciences, Defence Research and Development Organization, Brig. S. K. Mazumdar Marg, Timarpur, Delhi, 110054, India
- Department of Biosciences, Faculty of Natural Sciences, Jamia Millia Islamia-a Central University, Moulana Mohammad Ali Jauhar Marg, New Delhi, 110025, India
| | - Jawahar Singh Adhikari
- Division of Radiation Biodosimetry, Institute of Nuclear Medicine and Allied Sciences, Defence Research and Development Organization, Brig. S. K. Mazumdar Marg, Timarpur, Delhi, 110054, India
| | - Moshahid Alam Rizvi
- Department of Biosciences, Faculty of Natural Sciences, Jamia Millia Islamia-a Central University, Moulana Mohammad Ali Jauhar Marg, New Delhi, 110025, India
| | - Nabo Kumar Chaudhury
- Division of Radiation Biodosimetry, Institute of Nuclear Medicine and Allied Sciences, Defence Research and Development Organization, Brig. S. K. Mazumdar Marg, Timarpur, Delhi, 110054, India
| |
Collapse
|
29
|
Singh VK, Fatanmi OO, Wise SY, Newman VL, Romaine PLP, Seed TM. THE POTENTIATION OF THE RADIOPROTECTIVE EFFICACY OF TWO MEDICAL COUNTERMEASURES, GAMMA-TOCOTRIENOL AND AMIFOSTINE, BY A COMBINATION PROPHYLACTIC MODALITY. RADIATION PROTECTION DOSIMETRY 2016; 172:302-310. [PMID: 27542813 PMCID: PMC5444681 DOI: 10.1093/rpd/ncw223] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
This study was designed to evaluate the possible potentiation of survival protection afforded by relatively low-dose amifostine prophylaxis against total body irradiation in combination with a protective, less toxic agent, gamma-tocotrienol (GT3). Mice were administered amifostine and/or GT3, then exposed to 9.2 Gy 60Co γ-irradiation and monitored for survival for 30 days. To investigate cytokine stimulation, mice were administered amifostine or GT3; serum samples were collected and analyzed for cytokines. Survival studies show single treatments of GT3 or amifostine significantly improved survival, compared to the vehicle, and combination treatments resulted in significantly higher survival compared to single treatments. In vivo studies with GT3 confirmed prior work indicating GT3 induces granulocyte colony-stimulating factor (G-CSF). This approach, the prophylactic combination of amifostine and GT3, which act through different mechanisms, shows promise and should be investigated further as a potential countermeasure for acute radiation syndrome.
Collapse
Affiliation(s)
- Vijay K Singh
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine 'America's Medical School', Services University of the Health Sciences , Bethesda, MD, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences , Bethesda, MD, USA
| | - Oluseyi O Fatanmi
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine 'America's Medical School', Services University of the Health Sciences , Bethesda, MD, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences , Bethesda, MD, USA
| | - Stephen Y Wise
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine 'America's Medical School', Services University of the Health Sciences , Bethesda, MD, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences , Bethesda, MD, USA
| | - Victoria L Newman
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine 'America's Medical School', Services University of the Health Sciences , Bethesda, MD, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences , Bethesda, MD, USA
| | - Patricia L P Romaine
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine 'America's Medical School', Services University of the Health Sciences , Bethesda, MD, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences , Bethesda, MD, USA
| | | |
Collapse
|
30
|
Garty G, Turner HC, Salerno A, Bertucci A, Zhang J, Chen Y, Dutta A, Sharma P, Bian D, Taveras M, Wang H, Bhatla A, Balajee A, Bigelow AW, Repin M, Lyulko OV, Simaan N, Yao YL, Brenner DJ. THE DECADE OF THE RABiT (2005-15). RADIATION PROTECTION DOSIMETRY 2016; 172:201-206. [PMID: 27412510 PMCID: PMC5225976 DOI: 10.1093/rpd/ncw172] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The RABiT (Rapid Automated Biodosimetry Tool) is a dedicated Robotic platform for the automation of cytogenetics-based biodosimetry assays. The RABiT was developed to fulfill the critical requirement for triage following a mass radiological or nuclear event. Starting from well-characterized and accepted assays we developed a custom robotic platform to automate them. We present here a brief historical overview of the RABiT program at Columbia University from its inception in 2005 until the RABiT was dismantled at the end of 2015. The main focus of this paper is to demonstrate how the biological assays drove development of the custom robotic systems and in turn new advances in commercial robotic platforms inspired small modifications in the assays to allow replacing customized robotics with 'off the shelf' systems. Currently, a second-generation, RABiT II, system at Columbia University, consisting of a PerkinElmer cell::explorer, was programmed to perform the RABiT assays and is undergoing testing and optimization studies.
Collapse
Affiliation(s)
- G Garty
- Center for Radiological Research, Columbia University, VC11-230, 630 West 168th Street, New York, NY 10032, USA
| | - H C Turner
- Center for Radiological Research, Columbia University, VC11-230, 630 West 168th Street, New York, NY 10032, USA
| | - A Salerno
- Department of Mechanical Engineering, Columbia University, 500 West 120th Street, New York, NY 10027, USA
- Present address: Pratt & Whitney Canada Corp., 1000 Marie-Victorin, Longueil, QC, Canada J4G 1A1
| | - A Bertucci
- Center for Radiological Research, Columbia University, VC11-230, 630 West 168th Street, New York, NY 10032, USA
| | - J Zhang
- Department of Mechanical Engineering, Columbia University, 500 West 120th Street, New York, NY 10027, USA
- Present address: Auris Surgical Robotics Inc., 125 Shoreway Rd, San Carlos, CA 94070, USA
| | - Y Chen
- Department of Mechanical Engineering, Columbia University, 500 West 120th Street, New York, NY 10027, USA
| | - A Dutta
- Center for Radiological Research, Columbia University, VC11-230, 630 West 168th Street, New York, NY 10032, USA
- Present address: BioReliance Corp., 9630 Medical Center Dr, Rockville, MD 20850, USA
| | - P Sharma
- Center for Radiological Research, Columbia University, VC11-230, 630 West 168th Street, New York, NY 10032, USA
| | - D Bian
- Department of Mechanical Engineering, Columbia University, 500 West 120th Street, New York, NY 10027, USA
| | - M Taveras
- Center for Radiological Research, Columbia University, VC11-230, 630 West 168th Street, New York, NY 10032, USA
| | - H Wang
- Department of Mechanical Engineering, Columbia University, 500 West 120th Street, New York, NY 10027, USA
- Present address: General Motors Co., 30500 Mound Road, Warren, MI 48090, USA
| | - A Bhatla
- Department of Mechanical Engineering, Columbia University, 500 West 120th Street, New York, NY 10027, USA
- Present address: Curiosity Lab Inc., 54 Mallard Pl. Secaucus, NJ, 07094, USA
| | - A Balajee
- Center for Radiological Research, Columbia University, VC11-230, 630 West 168th Street, New York, NY 10032, USA
- Present address: Cytogenetic Biodosimetry Laboratory, Radiation Emergency Assistance Center and Training Site, Oak Ridge Institute for Science and Education, Oak Ridge Associated Universities, Building SC-10, 1299, Bethel Valley Road, Oak Ridge, TN, 37830, USA
| | - A W Bigelow
- Center for Radiological Research, Columbia University, VC11-230, 630 West 168th Street, New York, NY 10032, USA
| | - M Repin
- Center for Radiological Research, Columbia University, VC11-230, 630 West 168th Street, New York, NY 10032, USA
| | - O V Lyulko
- Center for Radiological Research, Columbia University, VC11-230, 630 West 168th Street, New York, NY 10032, USA
| | - N Simaan
- Department of Mechanical Engineering, Columbia University, 500 West 120th Street, New York, NY 10027, USA
- Present address: Department of Mechanical Engineering, Vanderbuilt University, PMB 351592, Nashville, TN, 37235, USA
| | - Y L Yao
- Department of Mechanical Engineering, Columbia University, 500 West 120th Street, New York, NY 10027, USA
| | - D J Brenner
- Center for Radiological Research, Columbia University, VC11-230, 630 West 168th Street, New York, NY 10032, USA
| |
Collapse
|
31
|
Blakely WF, Romanyukha A, Hayes SM, Reyes RA, Stewart HM, Hoefer MH, Williams A, Sharp T, Huff LA. U.S. Department of Defense Multiple-Parameter Biodosimetry Network. RADIATION PROTECTION DOSIMETRY 2016; 172:58-71. [PMID: 27886989 DOI: 10.1093/rpd/ncw295] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/12/2016] [Indexed: 06/06/2023]
Abstract
The U.S. Department of Defense (USDOD) service members are at risk of exposure to ionizing radiation due to radiation accidents, terrorist attacks and national defense activities. The use of biodosimetry is a standard of care for the triage and treatment of radiation injuries. Resources and procedures need to be established to implement a multiple-parameter biodosimetry system coupled with expert medial guidance to provide an integrated radiation diagnostic system to meet USDOD requirements. Current USDOD biodosimetry capabilities were identified and recommendations to fill the identified gaps are provided. A USDOD Multi-parametric Biodosimetry Network, based on the expertise that resides at the Armed Forces Radiobiology Research Institute and the Naval Dosimetry Center, was designed. This network based on the use of multiple biodosimetry modalities would provide diagnostic and triage capabilities needed to meet USDOD requirements. These are not available with sufficient capacity elsewhere but could be needed urgently after a major radiological/nuclear event.
Collapse
Affiliation(s)
- William F Blakely
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, 8901 Wisconsin Avenue, Bethesda, MD 20889-5603, USA
| | | | | | - Ricardo A Reyes
- Defense Health Agency, Walter Reed National Military Medical Command, Bethesda, MD 20889, USA
| | | | - Matthew H Hoefer
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, 8901 Wisconsin Avenue, Bethesda, MD 20889-5603, USA
| | | | - Thad Sharp
- Naval Dosimetry Center, Bethesda, MD 20889, USA
| | - L Andrew Huff
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, 8901 Wisconsin Avenue, Bethesda, MD 20889-5603, USA
| |
Collapse
|
32
|
Raber J, Davis MJ, Pfankuch T, Rosenthal R, Doctrow SR, Moulder JE. Mitigating effect of EUK-207 on radiation-induced cognitive impairments. Behav Brain Res 2016; 320:457-463. [PMID: 27789343 DOI: 10.1016/j.bbr.2016.10.038] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 10/21/2016] [Accepted: 10/23/2016] [Indexed: 10/20/2022]
Abstract
The brain could be exposed to irradiation as part of a nuclear accident, radiological terrorism (dirty bomb scenario) or a medical radiological procedure. In the context of accidents or terrorism, there is considerable interest in compounds that can mitigate radiation-induced injury when treatment is initiated a day or more after the radiation exposure. As it will be challenging to determine the radiation exposure an individual has received within a relatively short time frame, it is also critical that the mitigating agent does not negatively affect individuals, including emergency workers, who might be treated, but who were not exposed. Alterations in hippocampus-dependent cognition often characterize radiation-induced cognitive injury. The catalytic ROS scavenger EUK-207 is a member of the class of metal-containing salen manganese (Mn) complexes that suppress oxidative stress, including in the mitochondria, and have been shown to mitigate radiation dermatitis, promote wound healing in irradiated skin, and mitigate vascular injuries in irradiated lungs. As the effects of EUK-207 against radiation injury in the brain are not known, we assessed the effects of EUK-207 on sham-irradiated animals and the ability of EUK-207 to mitigate radiation-induced cognitive injury. The day following irradiation or sham-irradiation, the mice started to receive EUK-207 and were cognitively tested 3 months following exposure. Mice irradiated at a dose of 15Gy showed cognitive impairments in the water maze probe trial. EUK-207 mitigated these impairments while not affecting cognitive performance of sham-irradiated mice in the water maze probe trial. Thus, EUK-207 has attractive properties and should be considered an ideal candidate to mitigate radiation-induced cognitive injury.
Collapse
Affiliation(s)
- J Raber
- Department of Behavioral Neuroscience, L470, Oregon Health and Science University, Portland, OR 97239, USA; Departments of Neurology and Radiation Medicine, Division of Neuroscience, ONPRC, L470, Oregon Health and Science University, Portland, Oregon 97239, USA.
| | - M J Davis
- Department of Behavioral Neuroscience, L470, Oregon Health and Science University, Portland, OR 97239, USA
| | - T Pfankuch
- Department of Behavioral Neuroscience, L470, Oregon Health and Science University, Portland, OR 97239, USA
| | - R Rosenthal
- Pulmonary Center, Boston University School of Medicine, MA 02215, USA
| | - S R Doctrow
- Pulmonary Center, Boston University School of Medicine, MA 02215, USA
| | - J E Moulder
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
33
|
Singh VK, Romaine PLP, Newman VL, Seed TM. Medical countermeasures for unwanted CBRN exposures: part II radiological and nuclear threats with review of recent countermeasure patents. Expert Opin Ther Pat 2016; 26:1399-1408. [PMID: 27610458 PMCID: PMC5152556 DOI: 10.1080/13543776.2016.1231805] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Introduction: The global threat of a chemical, biological, radiological, or nuclear (CBRN) disaster is an important priority for all government agencies involved in domestic security and public health preparedness. Radiological/nuclear (RN) attacks or accidents have become a larger focus of the United States Food and Drug administration (US FDA) over time because of their increased likeliness. Clinical signs and symptoms of a developing acute radiation syndrome (ARS) are grouped into three sub-syndromes named for the dominant organ system affected, namely the hematopoietic (H-ARS), gastrointestinal (GI-ARS), and neurovascular systems. The availability of safe and effective countermeasures against radiological/nuclear threats currently represents a significant unmet medical need. Areas covered: This article reviews the development of RN threat medical countermeasures and highlights those specific countermeasures that have been recently patented and approved following the FDA Animal Rule. Patents for such agents from 2015 have been presented. Expert opinion: Two granulocyte colony-stimulating factor (G-CSF)-based radiation countermeasures (Neupogen® (Amgen, Thousand Oaks, CA) and Neulasta® (Amgen, Thousand Oaks, CA)) have recently been approved by the FDA for treatment of H-ARS and both these agents are radiomitigators, used after radiation exposure. To date, there are no FDA-approved radioprotectors for ARS.
Collapse
Affiliation(s)
- Vijay K Singh
- a Department of Pharmacology and Molecular Therapeutics , F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences , Bethesda , MD , USA.,b Armed Forces Radiobiology Research Institute , Uniformed Services University of the Health Sciences , Bethesda , MD , USA
| | - Patricia L P Romaine
- b Armed Forces Radiobiology Research Institute , Uniformed Services University of the Health Sciences , Bethesda , MD , USA
| | - Victoria L Newman
- b Armed Forces Radiobiology Research Institute , Uniformed Services University of the Health Sciences , Bethesda , MD , USA
| | | |
Collapse
|
34
|
Abstract
Potential ionising radiation exposure scenarios are varied, but all bring risks beyond the simple issues of short-term survival. Whether accidentally exposed to a single, whole-body dose in an act of terrorism or purposefully exposed to fractionated doses as part of a therapeutic regimen, radiation exposure carries the consequence of elevated cancer risk. The long-term impact of both intentional and unintentional exposure could potentially be mitigated by treatments specifically developed to limit the mutations and precancerous replication that ensue in the wake of irradiation The development of such agents would undoubtedly require a substantial degree of in vitro testing, but in order to accurately recapitulate the complex process of radiation-induced carcinogenesis, well-understood animal models are necessary. Inbred strains of the laboratory mouse, Mus musculus, present the most logical choice due to the high number of molecular and physiological similarities they share with humans. Their small size, high rate of breeding and fully sequenced genome further increase its value for use in cancer research. This chapter will review relevant m. musculus inbred and F1 hybrid animals of radiation-induced myeloid leukemia, thymic lymphoma, breast and lung cancers. Method of cancer induction and associated molecular pathologies will also be described for each model.
Collapse
|
35
|
Li J, Wei Y, Yan L, Wang R, Zhang Y, Su Y, Yang Z, Hu M, Qi R, Tan H, Wu Q, Yin X, Pan X. Multiplacenta derived stem cell/cytokine treatment increases survival time in a mouse model with radiation-induced bone marrow damage. Cytotechnology 2016; 68:2677-2686. [PMID: 27318496 DOI: 10.1007/s10616-016-9993-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 06/06/2016] [Indexed: 02/02/2023] Open
Abstract
Nuclear Warfare and nuclear leakage can result in a large number of patients with radiation-induced bone marrow damage. Based on the fact that hematopoietic stem cells and hematopoietic growth factors are characterized as a novel strategy for therapy, the aim of this study was to explore a safe and routine stem cell/cytokine therapeutic strategy. Allogeneic multiplacenta derived hematopoietic and mesenchymal stem cells/cytokines were intraperitoneally injected into a moderate dose of total body irradiation-induced mouse bone marrow damage model a single time. Then, the mouse posttransplantation survival time, peripheral blood hemoglobin count, bone marrow architecture, and donor cell engraftment were assessed. Each mouse that received placenta-derived stem cells exhibited positive donor hematopoietic and mesenchymal stem cell engraftment both in the bone marrow and peripheral blood after transplantation. The peripheral blood hemoglobin count and survival time were greater in the group with the combined treatment of multiplacenta-derived stem cells and cytokines, compared with model-only controls (both P < 0.001). The blood smear mesenchymal/hematopoietic stem cell count was significantly higher in the combined treatment group than in the mice treated only with placenta-derived cells (28.08 ± 5.824 vs. 20.40 ± 5.989, P < 0.001; 7.74 ± 2.153 vs. 4.23 ± 1.608, P < 0.001, respectively). However, there was no marked change on the bone marrow pathology of any of the experimental mice after the transplantation. These results indicate that for radiation-induced bone marrow damage treatment, multiplacenta-derived stem cells and cytokines can increase the life span of model mice and delay but not abrogate the disease progression. Intraperitoneally transplanted stem cells can survive and engraft into the host body through the blood circulation. Improvement of peripheral blood hemoglobin levels, but not the bone marrow architecture response, probably explains the increase in survival time observed in this study.
Collapse
Affiliation(s)
- Jun Li
- Medical School of Kunming University, Kunming, 650214, China
- State Local Joint Engineering Laboratory of Stem Cell and Immunocyte Biomedical Technology, Kunming General Hospital of Chengdu Military Command, Kunming, 650032, China
| | - Yunfang Wei
- Medical School of Kunming University, Kunming, 650214, China
| | - Lei Yan
- Anesthesiology Department, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830000, China
| | - Rui Wang
- Medical School of Kunming University, Kunming, 650214, China
| | - Ying Zhang
- Medical School of Kunming University, Kunming, 650214, China
| | - Yingzhen Su
- Medical School of Kunming University, Kunming, 650214, China
| | - Zhaoyu Yang
- Medical School of Kunming University, Kunming, 650214, China
| | - Min Hu
- Research Center for Molecular Medicine, Kunming University, Kunming, 650214, China
| | - Rui Qi
- Medical School of Kunming University, Kunming, 650214, China
| | - Hongbo Tan
- Orthopedics Department, Kunming General Hospital of Chengdu Military Command, Kunming, 650032, China
| | - Qiong Wu
- Department of Chemical Science and Technology, Kunming University, Kunming, 650214, China
| | - Xudong Yin
- Department of Life Science and Technology, Kunming University, Kunming, 650214, China
| | - Xinghua Pan
- State Local Joint Engineering Laboratory of Stem Cell and Immunocyte Biomedical Technology, Kunming General Hospital of Chengdu Military Command, Kunming, 650032, China.
| |
Collapse
|
36
|
Singh VK, Hauer-Jensen M. γ-Tocotrienol as a Promising Countermeasure for Acute Radiation Syndrome: Current Status. Int J Mol Sci 2016; 17:E663. [PMID: 27153057 PMCID: PMC4881489 DOI: 10.3390/ijms17050663] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 04/03/2016] [Accepted: 04/25/2016] [Indexed: 01/13/2023] Open
Abstract
The hazard of ionizing radiation exposure due to nuclear accidents or terrorist attacks is ever increasing. Despite decades of research, still, there is a shortage of non-toxic, safe and effective medical countermeasures for radiological and nuclear emergency. To date, the U.S. Food and Drug Administration (U.S. FDA) has approved only two growth factors, Neupogen (granulocyte colony-stimulating factor (G-CSF), filgrastim) and Neulasta (PEGylated G-CSF, pegfilgrastim) for the treatment of hematopoietic acute radiation syndrome (H-ARS) following the Animal Efficacy Rule. Promising radioprotective efficacy results of γ-tocotrienol (GT3; a member of the vitamin E family) in the mouse model encouraged its further evaluation in the nonhuman primate (NHP) model. These studies demonstrated that GT3 significantly aided the recovery of radiation-induced neutropenia and thrombocytopenia compared to the vehicle controls; these results particularly significant after exposure to 5.8 or 6.5 Gray (Gy) whole body γ-irradiation. The stimulatory effect of GT3 on neutrophils and thrombocytes (platelets) was directly and positively correlated with dose; a 75 mg/kg dose was more effective compared to 37.5 mg/kg. GT3 was also effective against 6.5 Gy whole body γ-irradiation for improving neutrophils and thrombocytes. Moreover, a single administration of GT3 without any supportive care was equivalent, in terms of improving hematopoietic recovery, to multiple doses of Neupogen and two doses of Neulasta with full supportive care (including blood products) in the NHP model. GT3 may serve as an ultimate radioprotector for use in humans, particularly for military personnel and first responders. In brief, GT3 is a promising radiation countermeasure that ought to be further developed for U.S. FDA approval for the ARS indication.
Collapse
Affiliation(s)
- Vijay K Singh
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA.
| | - Martin Hauer-Jensen
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare Systems, Little Rock, AR 72205, USA.
| |
Collapse
|
37
|
Milner EE, Daxon EG, Anastasio MT, Nesler JT, Miller RL, Blakely WF. Concepts of Operations (CONOPS) for Biodosimetry Tools Employed in Operational Environments. HEALTH PHYSICS 2016; 110:370-379. [PMID: 26910029 DOI: 10.1097/hp.0000000000000470] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
It is essential to identify improved capabilities to accurately identify, confirm, and/or quantify radiological exposure and injury in order to inform critical triage, diagnosis, and treatment decisions. Herein the authors report characteristic requirements and potential Concepts of Operations (CONOPS) for biodosimetry tools employed in operational environments. While similar significant efforts have been completed in this area for the U.S. civilian sector, limited perspectives are published in the peer-reviewed literature regarding the use of radiological diagnostic technologies in deployed military medical treatment settings. Two radiological exposure scenarios were developed to clarify the diagnostic performance criteria and identify capability gaps. The emerging technology areas associated with radiation exposure diagnostics were reviewed and assessed to gauge their suitability in supporting triage, treatment, and return to duty decisions within the military medical support system.
Collapse
Affiliation(s)
- Erin E Milner
- *Medical Countermeasure Systems, Department of Defense, Ft. Detrick, MD, USA; †Battelle Memorial Institute, Medical Readiness and Response, Columbus, OH, USA; ‡Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Department of Defense, Bethesda, MD, USA
| | | | | | | | | | | |
Collapse
|
38
|
Bertucci A, Smilenov LB, Turner HC, Amundson SA, Brenner DJ. In vitro RABiT measurement of dose rate effects on radiation induction of micronuclei in human peripheral blood lymphocytes. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2016; 55:53-59. [PMID: 26791381 PMCID: PMC4792265 DOI: 10.1007/s00411-015-0628-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 11/28/2015] [Indexed: 05/29/2023]
Abstract
Developing new methods for radiation biodosimetry has been identified as a high-priority need in case of a radiological accident or nuclear terrorist attacks. A large-scale radiological incident would result in an immediate critical need to assess the radiation doses received by thousands of individuals. Casualties will be exposed to different doses and dose rates due to their geographical position and sheltering conditions, and dose rate is one of the principal factors that determine the biological consequences of a given absorbed dose. In these scenarios, high-throughput platforms are required to identify the biological dose in a large number of exposed individuals for clinical monitoring and medical treatment. The Rapid Automated Biodosimetry Tool (RABiT) is designed to be completely automated from the input of blood sample into the machine to the output of a dose estimate. The primary goal of this paper was to quantify the dose rate effects for RABiT-measured micronuclei in vitro in human lymphocytes. Blood samples from healthy volunteers were exposed in vitro to different doses of X-rays to acute and protracted doses over a period up to 24 h. The acute dose was delivered at ~1.03 Gy/min and the low dose rate exposure at ~0.31 Gy/min. The results showed that the yield of micronuclei decreases with decreasing dose rate starting at 2 Gy, whereas response was indistinguishable from that of acute exposure in the low dose region, up to 0.5 Gy. The results showed a linear-quadratic dose-response relationship for the occurrence of micronuclei for the acute exposure and a linear dose-response relationship for the low dose rate exposure.
Collapse
Affiliation(s)
- Antonella Bertucci
- Center for Radiological Research, Columbia University Medical Center, 630 W. 168th St., New York, NY, 10032, USA.
| | - Lubomir B Smilenov
- Center for Radiological Research, Columbia University Medical Center, 630 W. 168th St., New York, NY, 10032, USA
| | - Helen C Turner
- Center for Radiological Research, Columbia University Medical Center, 630 W. 168th St., New York, NY, 10032, USA
| | - Sally A Amundson
- Center for Radiological Research, Columbia University Medical Center, 630 W. 168th St., New York, NY, 10032, USA
| | - David J Brenner
- Center for Radiological Research, Columbia University Medical Center, 630 W. 168th St., New York, NY, 10032, USA
| |
Collapse
|
39
|
Vitamin E Analogs as Radiation Response Modifiers. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:741301. [PMID: 26366184 PMCID: PMC4558447 DOI: 10.1155/2015/741301] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 05/06/2015] [Accepted: 07/22/2015] [Indexed: 02/07/2023]
Abstract
The potentially life-threatening effects of total body ionizing radiation exposure have been known for more than a century. Despite considerable advances in our understanding of the effects of radiation over the past six decades, efforts to identify effective radiation countermeasures for use in case of a radiological/nuclear emergency have been largely unsuccessful. Vitamin E is known to have antioxidant properties capable of scavenging free radicals, which have critical roles in radiation injuries. Tocopherols and tocotrienols, vitamin E analogs together known as tocols, have shown promise as radioprotectors. Although the pivotal mechanisms of action of tocols have long been thought to be their antioxidant properties and free radical scavenging activities, other alternative mechanisms have been proposed to drive their activity as radioprotectors. Here we provide a brief overview of the effects of ionizing radiation, the mechanistic mediators of radiation-induced damage, and the need for radiation countermeasures. We further outline the role for, efficacy of, and mechanisms of action of tocols as radioprotectors, and we compare and contrast their efficacy and mode of action with that of another well-studied chemical radioprotector, amifostine.
Collapse
|
40
|
Khan S, Kumar A, Adhikari JS, Rizvi MA, Chaudhury NK. Protective effect of sesamol against60Co γ-ray-induced hematopoietic and gastrointestinal injury in C57BL/6 male mice. Free Radic Res 2015; 49:1344-61. [DOI: 10.3109/10715762.2015.1071485] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
41
|
Siderocalin-mediated recognition, sensitization, and cellular uptake of actinides. Proc Natl Acad Sci U S A 2015; 112:10342-7. [PMID: 26240330 DOI: 10.1073/pnas.1508902112] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Synthetic radionuclides, such as the transuranic actinides plutonium, americium, and curium, present severe health threats as contaminants, and understanding the scope of the biochemical interactions involved in actinide transport is instrumental in managing human contamination. Here we show that siderocalin, a mammalian siderophore-binding protein from the lipocalin family, specifically binds lanthanide and actinide complexes through molecular recognition of the ligands chelating the metal ions. Using crystallography, we structurally characterized the resulting siderocalin-transuranic actinide complexes, providing unprecedented insights into the biological coordination of heavy radioelements. In controlled in vitro assays, we found that intracellular plutonium uptake can occur through siderocalin-mediated endocytosis. We also demonstrated that siderocalin can act as a synergistic antenna to sensitize the luminescence of trivalent lanthanide and actinide ions in ternary protein-ligand complexes, dramatically increasing the brightness and efficiency of intramolecular energy transfer processes that give rise to metal luminescence. Our results identify siderocalin as a potential player in the biological trafficking of f elements, but through a secondary ligand-based metal sequestration mechanism. Beyond elucidating contamination pathways, this work is a starting point for the design of two-stage biomimetic platforms for photoluminescence, separation, and transport applications.
Collapse
|
42
|
Ohtsuru A, Tanigawa K, Kumagai A, Niwa O, Takamura N, Midorikawa S, Nollet K, Yamashita S, Ohto H, Chhem RK, Clarke M. Nuclear disasters and health: lessons learned, challenges, and proposals. Lancet 2015; 386:489-97. [PMID: 26251394 DOI: 10.1016/s0140-6736(15)60994-1] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Past nuclear disasters, such as the atomic bombings in 1945 and major accidents at nuclear power plants, have highlighted similarities in potential public health effects of radiation in both circumstances, including health issues unrelated to radiation exposure. Although the rarity of nuclear disasters limits opportunities to undertake rigorous research of evidence-based interventions and strategies, identification of lessons learned and development of an effective plan to protect the public, minimise negative effects, and protect emergency workers from exposure to high-dose radiation is important. Additionally, research is needed to help decision makers to avoid premature deaths among patients already in hospitals and other vulnerable groups during evacuation. Since nuclear disasters can affect hundreds of thousands of people, a substantial number of people are at risk of physical and mental harm in each disaster. During the recovery period after a nuclear disaster, physicians might need to screen for psychological burdens and provide general physical and mental health care for many affected residents who might experience long-term displacement. Reliable communication of personalised risks has emerged as a challenge for health-care professionals beyond the need to explain radiation protection. To overcome difficulties of risk communication and provide decision aids to protect workers, vulnerable people, and residents after a nuclear disaster, physicians should receive training in nuclear disaster response. This training should include evidence-based interventions, support decisions to balance potential harms and benefits, and take account of scientific uncertainty in provision of community health care. An open and joint learning process is essential to prepare for, and minimise the effects of, future nuclear disasters.
Collapse
Affiliation(s)
- Akira Ohtsuru
- Department of Radiation Health Management, Fukushima Medical University, Fukushima, Japan.
| | - Koichi Tanigawa
- Fukushima Global Medical Science Center, Fukushima Medical University, Fukushima, Japan
| | - Atsushi Kumagai
- Education Center of Disaster Medicine, Fukushima Medical University, Fukushima, Japan
| | - Ohtsura Niwa
- Fukushima Global Medical Science Center, Fukushima Medical University, Fukushima, Japan
| | - Noboru Takamura
- Department of Global Health, Medicine and Welfare, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Sanae Midorikawa
- Department of Radiation Health Management, Fukushima Medical University, Fukushima, Japan
| | - Kenneth Nollet
- Fukushima Global Medical Science Center, Fukushima Medical University, Fukushima, Japan
| | - Shunichi Yamashita
- Department of Radiation Medical Sciences, Nagasaki University, Nagasaki, Japan
| | - Hitoshi Ohto
- Radiation Medical Science Center, Fukushima Medical University, Fukushima, Japan
| | - Rethy K Chhem
- Cambodia Development Resource Institute, Phnom Penh, Cambodia
| | - Mike Clarke
- Centre for Public Health, Queens University Belfast, Royal Hospitals, Belfast, UK
| |
Collapse
|
43
|
Ramakumar A, Subramanian U, Prasanna PGS. High-throughput sample processing and sample management; the functional evolution of classical cytogenetic assay towards automation. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2015; 793:132-41. [PMID: 26520383 DOI: 10.1016/j.mrgentox.2015.07.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 07/23/2015] [Indexed: 10/23/2022]
Abstract
High-throughput individual diagnostic dose assessment is essential for medical management of radiation-exposed subjects after a mass casualty. Cytogenetic assays such as the Dicentric Chromosome Assay (DCA) are recognized as the gold standard by international regulatory authorities. DCA is a multi-step and multi-day bioassay. DCA, as described in the IAEA manual, can be used to assess dose up to 4-6 weeks post-exposure quite accurately but throughput is still a major issue and automation is very essential. The throughput is limited, both in terms of sample preparation as well as analysis of chromosome aberrations. Thus, there is a need to design and develop novel solutions that could utilize extensive laboratory automation for sample preparation, and bioinformatics approaches for chromosome-aberration analysis to overcome throughput issues. We have transitioned the bench-based cytogenetic DCA to a coherent process performing high-throughput automated biodosimetry for individual dose assessment ensuring quality control (QC) and quality assurance (QA) aspects in accordance with international harmonized protocols. A Laboratory Information Management System (LIMS) is designed, implemented and adapted to manage increased sample processing capacity, develop and maintain standard operating procedures (SOP) for robotic instruments, avoid data transcription errors during processing, and automate analysis of chromosome-aberrations using an image analysis platform. Our efforts described in this paper intend to bridge the current technological gaps and enhance the potential application of DCA for a dose-based stratification of subjects following a mass casualty. This paper describes one such potential integrated automated laboratory system and functional evolution of the classical DCA towards increasing critically needed throughput.
Collapse
Affiliation(s)
- Adarsh Ramakumar
- Scientific Research Department, Armed Forces Radiobiology Research Institute, 8901 Wisconsin Avenue, Bldg. 42, Bethesda, MD 20889, USA.
| | - Uma Subramanian
- Scientific Research Department, Armed Forces Radiobiology Research Institute, 8901 Wisconsin Avenue, Bldg. 42, Bethesda, MD 20889, USA
| | - Pataje G S Prasanna
- Scientific Research Department, Armed Forces Radiobiology Research Institute, 8901 Wisconsin Avenue, Bldg. 42, Bethesda, MD 20889, USA
| |
Collapse
|
44
|
Khan S, Adhikari JS, Rizvi MA, Chaudhury NK. Radioprotective potential of melatonin against ⁶⁰Co γ-ray-induced testicular injury in male C57BL/6 mice. J Biomed Sci 2015. [PMID: 26205951 PMCID: PMC4514449 DOI: 10.1186/s12929-015-0156-9] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Background Melatonin, the chief secretary product of pineal gland, is a strong free radical scavenger and antioxidant molecule. The radioprotective efficacy and underlying mechanisms refer to its antioxidant role in somatic cells. The purpose of this study, therefore, was to investigate the prophylactic implications of melatonin against γ-ray-induced injury in germinal cells (testes). C57BL/6 male mice were administered melatonin (100 mg/kg) intra-peritoneally 30 min prior to a single dose of whole-body γ-irradiation (5 Gy, 1 Gy/minute) using 60Co teletherapy unit. Animals were sacrificed at 2h, 4h and 8h post-irradiation and their testes along with its spermatozoa taken out and used for total antioxidant capacity (TAC), lipid peroxidation, comet assay, western blotting and sperm motility and viability. In another set of experiment, animals were similarly treated were sacrificed on 1st, 3rd, 7th, 15th and 30th day post-irradiation and evaluated for sperm abnormalities and histopathological analysis. Results Whole-body γ-radiation exposure (5 Gy) drastically depleted the populations of spermatogenic cells in seminiferous tubules on day three, which were significantly protected by melatonin. In addition, radiation-induced sperm abnormalities, motility and viability in cauda-epididymes were significantly reduced by melatonin. Melatonin pre-treatment significantly inhibited radiation-induced DNA strands breaks and lipid peroxidation. At this time, radiation-induces activation of ATM-dependent p53 apoptotic proteins-ATM, p53, p21, Bax, cytochrome C, active caspase-3 and caspases-9 expression, which were significantly reversed in melatonin pre-treated mice. This reduced apoptotic proteins by melatonin pre-treatment was associated with the increase of anti-apoptotic-Bcl-x and DNA repair-PCNA proteins in irradiated mice. Further, radiation-induced decline in the TAC was significantly reversed in melatonin pre-treated mice. Conclusions The present results indicated that melatonin as prophylactic agent protected male reproductive system against radiation-induced injury in mice. The detailed study will benefit in understanding the role of melatonin in modulation of radiation-induced ATM-dependent p53-mediated pro-vs.-anti apoptotic proteins in testicular injury. These results can be further exploited for use of melatonin for protection of male reproductive system in radiotherapy applications involving hemibody abdominal exposures. Electronic supplementary material The online version of this article (doi:10.1186/s12929-015-0156-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shahanshah Khan
- Chemical Radioprotector and Radiation Dosimetry Research Group, Division of Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences, Defence Research & Development Organization, Brig. S. K. Mazumdar Road, New Delhi, Delhi, 110054, India. .,Genome Biology Laboratory, Department of Biosciences, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi, 110025, India.
| | - Jawahar Singh Adhikari
- Chemical Radioprotector and Radiation Dosimetry Research Group, Division of Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences, Defence Research & Development Organization, Brig. S. K. Mazumdar Road, New Delhi, Delhi, 110054, India.
| | - Moshahid Alam Rizvi
- Genome Biology Laboratory, Department of Biosciences, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi, 110025, India.
| | - Nabo Kumar Chaudhury
- Chemical Radioprotector and Radiation Dosimetry Research Group, Division of Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences, Defence Research & Development Organization, Brig. S. K. Mazumdar Road, New Delhi, Delhi, 110054, India.
| |
Collapse
|
45
|
Role of thrombocytopenia in radiation-induced mortality and review of therapeutic approaches targeting platelet regeneration after radiation exposure. ACTA ACUST UNITED AC 2015. [DOI: 10.1007/s13566-015-0201-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
46
|
Paul S, Smilenov LB, Elliston CD, Amundson SA. Radiation Dose-Rate Effects on Gene Expression in a Mouse Biodosimetry Model. Radiat Res 2015; 184:24-32. [PMID: 26114327 DOI: 10.1667/rr14044.1] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
In the event of a nuclear accident or radiological terrorist attack, there will be a pressing need for biodosimetry to triage a large, potentially exposed population and to assign individuals to appropriate treatment. Exposures from fallout are likely, resulting in protracted dose delivery that would, in turn, impact the extent of injury. Biodosimetry approaches that can distinguish such low-dose-rate (LDR) exposures from acute exposures have not yet been developed. In this study, we used the C57BL/6 mouse model in an initial investigation of the impact of low-dose-rate delivery on the transcriptomic response in blood. While a large number of the same genes responded to LDR and acute radiation exposures, for many genes the magnitude of response was lower after LDR exposures. Some genes, however, were differentially expressed (P < 0.001, false discovery rate <5%) in mice exposed to LDR compared with mice exposed to acute radiation. We identified a set of 164 genes that correctly classified 97% of the samples in this experiment as exposed to acute or LDR radiation using a support vector machine algorithm. Gene expression is a promising approach to radiation biodosimetry, enhanced greatly by this first demonstration of its potential for distinguishing between acute and LDR exposures. Further development of this aspect of radiation biodosimetry, either as part of a complete gene expression biodosimetry test or as an adjunct to other methods, could provide vital triage information in a mass radiological casualty event.
Collapse
Affiliation(s)
- Sunirmal Paul
- a Center for Radiological Research, Columbia University Medical Center, New York, New York 10032;,b Rutgers University, Newark, New Jersey 07103; and
| | - Lubomir B Smilenov
- a Center for Radiological Research, Columbia University Medical Center, New York, New York 10032
| | - Carl D Elliston
- a Center for Radiological Research, Columbia University Medical Center, New York, New York 10032;,c Maimonides Medical Center, Brooklyn, New York 11219
| | - Sally A Amundson
- a Center for Radiological Research, Columbia University Medical Center, New York, New York 10032
| |
Collapse
|
47
|
Singh V, Gupta D, Arora R. NF-kB as a key player in regulation of cellular radiation responses and identification of radiation countermeasures. Discoveries (Craiova) 2015; 3:e35. [PMID: 32309561 PMCID: PMC7159829 DOI: 10.15190/d.2015.27] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Nuclear factor (NF)-κB is a transcription factor that plays significant role in immunity, cellular survival and inhibition of apoptosis, through the induction of genetic networks. Depending on the stimulus and the cell type, the members of NF-κB related family (RelA, c-Rel, RelB, p50, and p52), forms different combinations of homo and hetero-dimers. The activated complexes (Es) translocate into the nucleus and bind to the 10bp κB site of promoter region of target genes in stimulus specific manner. In response to radiation, NF-κB is known to reduce cell death by promoting the expression of anti-apoptotic proteins and activation of cellular antioxidant defense system. Constitutive activation of NF-κB associated genes in tumour cells are known to enhance radiation resistance, whereas deletion in mice results in hypersensitivity to IR-induced GI damage. NF-κB is also known to regulate the production of a wide variety of cytokines and chemokines, which contribute in enhancing cell proliferation and tissue regeneration in various organs, such as the GI crypts stem cells, bone marrow etc., following exposure to IR. Several other cytokines are also known to exert potent pro-inflammatory effects that may contribute to the increase of tissue damage following exposure to ionizing radiation. Till date there are a series of molecules or group of compounds that have been evaluated for their radio-protective potential, and very few have reached clinical trials. The failure or less success of identified agents in humans could be due to their reduced radiation protection efficacy.
In this review we have considered activation of NF-κB as a potential marker in screening of radiation countermeasure agents (RCAs) and cellular radiation responses. Moreover, we have also focused on associated mechanisms of activation of NF-κB signaling and their specified family member activation with respect to stimuli. Furthermore, we have categorized their regulated gene expressions and their function in radiation response or modulation. In addition, we have discussed some recently developed radiation countermeasures in relation to NF-κB activation
Collapse
Affiliation(s)
- Vijay Singh
- Division of Radiation Biosciences, Institute of Nuclear Medicine & Allied Sciences, Brig SK Mazumdar Marg, Timarpur, Delhi, India
| | - Damodar Gupta
- Division of Radiation Biosciences, Institute of Nuclear Medicine & Allied Sciences, Brig SK Mazumdar Marg, Timarpur, Delhi, India
| | - Rajesh Arora
- Division of Radiation Biosciences, Institute of Nuclear Medicine & Allied Sciences, Brig SK Mazumdar Marg, Timarpur, Delhi, India
| |
Collapse
|
48
|
Wilson JP, Cobb RR, Dungan NW, Matthews LL, Eppler B, Aiello KV, Curtis S, Boger T, Guilmette RA, Weber W, Doyle-Eisele M, Talton JD. Decorporation of systemically distributed americium by a novel orally administered diethylenetriaminepentaacetic acid (DTPA) formulation in beagle dogs. HEALTH PHYSICS 2015; 108:308-318. [PMID: 25627942 DOI: 10.1097/hp.0000000000000199] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Novel decorporation agents are being developed to protect against radiological accidents and terrorists attacks. Radioactive americium is a significant component of nuclear fallout. Removal of large radioactive materials, such as 241Am, from exposed persons is a subject of significant interest due to the hazards they pose. The objective of this study was to evaluate the dose-related efficacy of daily doses of NanoDTPA™ Capsules for decorporating Am administered intravenously as a soluble citrate complex to male and female beagle dogs. In addition, the efficacy of the NanoDTPA™ Capsules for decorporating 241Am was directly compared to intravenously administered saline and DTPA. Animals received a single IV administration of 241Am(III)-citrate on Day 0. One day after radionuclide administration, one of four different doses of NanoDTPA™ Capsules [1, 2, or 6 capsules d(-1) (30 mg, 60 mg, or 180 mg DTPA) or 2 capsules BID], IV Zn-DTPA (5 mg kg(-1) pentetate zinc trisodium) as a positive control, or IV saline as a placebo were administered. NanoDTPA™ Capsules, IV Zn-DTPA, or IV saline was administered on study days 1-14. Animals were euthanized on day 21. A full necropsy was conducted, and liver, spleen, kidneys, lungs and trachea, tracheobronchial lymph nodes (TBLN), muscle samples (right and left quadriceps), gastrointestinal (GI) tract (stomach plus esophagus, upper and lower intestine), gonads, two femurs, lumbar vertebrae (L1-L4), and all other soft tissue remains were collected. Urinary and fecal excretion profiles were increased approximately 10-fold compared to those for untreated animals. Tissue contents were decreased compared to untreated controls. In particular, liver content was decreased by approximately eightfold compared to untreated animals. The results from this study further demonstrate that oral NanoDTPA™ Capsules are equally efficient compared to IV Zn-DTPA in decorporation of actinides.
Collapse
Affiliation(s)
- James P Wilson
- *Nanotherapeutics, Inc., Alachua, FL 32615; †Lovelace Biomedical and Environmental Research Institute, Albuquerque, NM
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Choi TA, Furimsky AM, Swezey R, Bunin DI, Byrge P, Iyer LV, Chang PY, Abergel RJ. In vitro metabolism and stability of the actinide chelating agent 3,4,3-LI(1,2-HOPO). J Pharm Sci 2015; 104:1832-8. [PMID: 25727482 DOI: 10.1002/jps.24394] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 01/26/2015] [Accepted: 01/28/2015] [Indexed: 11/11/2022]
Abstract
The hydroxypyridinonate ligand 3,4,3-LI(1,2-HOPO) is currently under development for radionuclide chelation therapy. The preclinical characterization of this highly promising ligand comprised the evaluation of its in vitro properties, including microsomal, plasma, and gastrointestinal fluid stability, cytochrome P450 inhibition, plasma protein binding, and intestinal absorption using the Caco-2 cell line. When mixed with active human liver microsomes, no loss of parent compound was observed after 60 min, indicating compound stability in the presence of liver microsomal P450. At the tested concentrations, 3,4,3-LI(1,2-HOPO) did not significantly influence the activities of any of the cytochromal isoforms screened. Thus, 3,4,3-LI(1,2-HOPO) is unlikely to cause drug-drug interactions by inhibiting the metabolic clearance of coadministered drugs metabolized by these enzymes. Plasma protein-binding assays revealed that the compound is protein-bound in dogs and less extensively in rats and humans. In the plasma stability study, the compound was stable after 1 h at 37°C in mouse, rat, dog, and human plasma samples. Finally, a bidirectional permeability assay demonstrated that 3,4,3-LI(1,2-HOPO) is not permeable across the Caco-2 monolayer, highlighting the need to further evaluate the effects of various compounds with known permeability enhancement properties on the permeability of the ligand in future studies.
Collapse
Affiliation(s)
- Taylor A Choi
- Chemical Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Singh VK, Wise SY, Fatanmi OO, Scott J, Romaine PLP, Newman VL, Verma A, Elliott TB, Seed TM. Progenitors mobilized by gamma-tocotrienol as an effective radiation countermeasure. PLoS One 2014; 9:e114078. [PMID: 25423021 PMCID: PMC4244184 DOI: 10.1371/journal.pone.0114078] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 11/03/2014] [Indexed: 11/18/2022] Open
Abstract
The purpose of this study was to elucidate the role of gamma-tocotrienol (GT3)-mobilized progenitors in mitigating damage to mice exposed to a supralethal dose of cobalt-60 gamma-radiation. CD2F1 mice were transfused 24 h post-irradiation with whole blood or isolated peripheral blood mononuclear cells (PBMC) from donors that had received GT3 72 h prior to blood collection and recipient mice were monitored for 30 days. To understand the role of GT3-induced granulocyte colony-stimulating factor (G-CSF) in mobilizing progenitors, donor mice were administered a neutralizing antibody specific to G-CSF or its isotype before blood collection. Bacterial translocation from gut to heart, spleen and liver of irradiated recipient mice was evaluated by bacterial culture on enriched and selective agar media. Endotoxin in serum samples also was measured. We also analyzed the colony-forming units in the spleens of irradiated mice. Our results demonstrate that whole blood or PBMC from GT3-administered mice mitigated radiation injury when administered 24 h post-irradiation. Furthermore, administration of a G-CSF antibody to GT3-injected mice abrogated the efficacy of blood or PBMC obtained from such donors. Additionally, GT3-mobilized PBMC inhibited the translocation of intestinal bacteria to the heart, spleen, and liver, and increased colony forming unit-spleen (CFU-S) numbers in irradiated mice. Our data suggests that GT3 induces G-CSF, which mobilizes progenitors and these progenitors mitigate radiation injury in recipient mice. This approach using mobilized progenitor cells from GT3-injected donors could be a potential treatment for humans exposed to high doses of radiation.
Collapse
Affiliation(s)
- Vijay K. Singh
- Armed Forces Radiobiology Research Institute, Bethesda, Maryland, United States of America
- Department of Radiation Biology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
- * E-mail:
| | - Stephen Y. Wise
- Armed Forces Radiobiology Research Institute, Bethesda, Maryland, United States of America
| | - Oluseyi O. Fatanmi
- Armed Forces Radiobiology Research Institute, Bethesda, Maryland, United States of America
| | - Jessica Scott
- Armed Forces Radiobiology Research Institute, Bethesda, Maryland, United States of America
| | - Patricia L. P. Romaine
- Armed Forces Radiobiology Research Institute, Bethesda, Maryland, United States of America
| | - Victoria L. Newman
- Armed Forces Radiobiology Research Institute, Bethesda, Maryland, United States of America
| | - Amit Verma
- Armed Forces Radiobiology Research Institute, Bethesda, Maryland, United States of America
| | - Thomas B. Elliott
- Armed Forces Radiobiology Research Institute, Bethesda, Maryland, United States of America
| | - Thomas M. Seed
- Tech Micro Services, Bethesda, Maryland, United States of America
| |
Collapse
|