51
|
Thioredoxin mitigates radiation-induced hematopoietic stem cell injury in mice. Stem Cell Res Ther 2017; 8:263. [PMID: 29141658 PMCID: PMC5688691 DOI: 10.1186/s13287-017-0711-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 10/03/2017] [Accepted: 10/24/2017] [Indexed: 12/17/2022] Open
Abstract
Background Radiation exposure poses a significant threat to public health. Hematopoietic injury is one of the major manifestations of acute radiation sickness. Protection and/or mitigation of hematopoietic stem cells (HSCs) from radiation injury is an important goal in the development of medical countermeasure agents (MCM). We recently identified thioredoxin (TXN) as a novel molecule that has marked protective and proliferative effects on HSCs. In the current study, we investigated the effectiveness of TXN in rescuing mice from a lethal dose of total body radiation (TBI) and in enhancing hematopoietic reconstitution following a lethal dose of irradiation. Methods We used in-vivo and in-vitro methods to understand the biological and molecular mechanisms of TXN on radiation mitigation. BABL/c mice were used for the survival study and a flow cytometer was used to quantify the HSC population and cell senescence. A hematology analyzer was used for the peripheral blood cell count, including white blood cells (WBCs), red blood cells (RBCs), hemoglobin, and platelets. Colony forming unit (CFU) assay was used to study the colongenic function of HSCs. Hematoxylin and eosin staining was used to determine the bone marrow cellularity. Senescence-associated β-galactosidase assay was used for cell senescence. Western blot analysis was used to evaluate the DNA damage and senescence protein expression. Immunofluorescence staining was used to measure the expression of γ-H2AX foci for DNA damage. Results We found that administration of TXN 24 h following irradiation significantly mitigates BALB/c mice from TBI-induced death: 70% of TXN-treated mice survived, whereas only 25% of saline-treated mice survived. TXN administration led to enhanced recovery of peripheral blood cell counts, bone marrow cellularity, and HSC population as measured by c-Kit+Sca-1+Lin– (KSL) cells, SLAM + KSL cells and CFUs. TXN treatment reduced cell senescence and radiation-induced double-strand DNA breaks in both murine bone marrow lineage-negative (Lin–) cells and primary fibroblasts. Furthermore, TXN decreased the expression of p16 and phosphorylated p38. Our data suggest that TXN modulates diverse cellular processes of HSCs. Conclusions Administration of TXN 24 h following irradiation mitigates radiation-induced lethality. To the best of our knowledge, this is the first report demonstrating that TXN reduces radiation-induced lethality. TXN shows potential utility in the mitigation of radiation-induced hematopoietic injury.
Collapse
|
52
|
Kiang JG, Zhai M, Bolduc DL, Smith JT, Anderson MN, Ho C, Lin B, Jiang S. Combined Therapy of Pegylated G-CSF and Alxn4100TPO Improves Survival and Mitigates Acute Radiation Syndrome after Whole-Body Ionizing Irradiation Alone and Followed by Wound Trauma. Radiat Res 2017; 188:476-490. [PMID: 28850300 PMCID: PMC5743055 DOI: 10.1667/rr14647.1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Exposure to ionizing radiation alone or combined with traumatic tissue injury is a crucial life-threatening factor in nuclear and radiological incidents. Radiation injuries occur at the molecular, cellular, tissue and systemic levels; their mechanisms, however, remain largely unclear. Exposure to radiation combined with skin wounding, bacterial infection or burns results in greater mortality than radiation exposure alone in dogs, pigs, rats, guinea pigs and mice. In the current study we observed that B6D2F1/J female mice exposed to 60Co gamma-photon radiation followed by 15% total-body-surface-area skin wounds experienced an increment of 25% higher mortality over a 30-day observation period compared to those subjected to radiation alone. Radiation exposure delayed wound healing by approximately 14 days. On day 30 post-injury, bone marrow and ileum in animals from both groups (radiation alone or combined injury) still displayed low cellularity and structural damage. White blood cell counts, e.g., neutrophils, lymphocytes, monocytes, eosinophils, basophils and platelets, still remained very low in surviving irradiated alone animals, whereas only the lymphocyte count was low in surviving combined injury animals. Likewise, in surviving animals from radiation alone and combined injury groups, the RBCs, hemoglobin, hematocrit and platelets remained low. We observed, that animals treated with both pegylated G-CSF (a cytokine for neutrophil maturation and mobilization) and Alxn4100TPO (a thrombopoietin receptor agonist) at 4 h postirradiation, a 95% survival (vehicle: 60%) over the 30-day period, along with mitigated body-weight loss and significantly reduced acute radiation syndrome. In animals that received combined treatment of radiation and injury that received pegylated G-CSF and Alxn4100TPO, survival was increased from 35% to 55%, but did not accelerate wound healing. Hematopoiesis and ileum showed significant improvement in animals from both groups (irradiation alone and combined injury) when treated with pegylated G-CSF and Alxn4100TPO. Treatment with pegylated G-CSF alone increased survival after irradiation alone and combined injury by 33% and 15%, respectively, and further delayed wound healing, but increased WBC, RBC and platelet counts after irradiation alone, and only RBCs and platelets after combined injury. Treatment with Alxn4100TPO alone increased survival after both irradiation alone and combined injury by 4 and 23%, respectively, and delayed wound healing after combined injury, but increased RBCs, hemoglobin concentrations, hematocrit values and platelets after irradiation alone and only platelets after combined injury. Taken together, the results suggest that combined treatment with pegylated G-CSF and Alxn4100TPO is effective for mitigating effects of both radiation alone and in combination with injury.
Collapse
Affiliation(s)
- Juliann G. Kiang
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, Maryland
- Department of Pharmacology and Molecular Pharmacology, Uniformed Services University of the Health Sciences, Bethesda, Maryland
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Min Zhai
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, Maryland
| | - David L. Bolduc
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, Maryland
| | - Joan T. Smith
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, Maryland
| | - Marsha N. Anderson
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, Maryland
| | - Connie Ho
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, Maryland
- College of Letters and Science, University of California, Berkeley, Berkeley, California, 94720
| | - Bin Lin
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, Maryland
| | - Suping Jiang
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, Maryland
| |
Collapse
|
53
|
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: 28] [Impact Index Per Article: 3.5] [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
|
54
|
Ghrelin Therapy Decreases Incidents of Intracranial Hemorrhage in Mice after Whole-Body Ionizing Irradiation Combined with Burn Trauma. Int J Mol Sci 2017; 18:ijms18081693. [PMID: 28771181 PMCID: PMC5578083 DOI: 10.3390/ijms18081693] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 07/17/2017] [Accepted: 07/27/2017] [Indexed: 12/30/2022] Open
Abstract
Nuclear industrial accidents and the detonation of nuclear devices cause a variety of damaging factors which, when their impacts are combined, produce complicated injuries challenging for medical treatment. Thus, trauma following acute ionizing irradiation (IR) can deteriorate the IR-induced secondary reactive metabolic and inflammatory impacts to dose-limiting tissues, such as bone marrow/lymphatic, gastrointestinal tissues, and vascular endothelial tissues, exacerbating the severity of the primary injury and decreasing survival from the exposure. Previously we first reported that ghrelin therapy effectively improved survival by mitigating leukocytopenia, thrombocytopenia, and bone-marrow injury resulting from radiation combined with burn trauma. This study was aimed at investigating whether radiation combined with burn trauma induced the cerebro-vascular impairment and intracranial hemorrhage that could be reversed by ghrelin therapy. When B6D2F1 female mice were exposed to 9.5 Gy Cobalt-60 γ-radiation followed by 15% total skin surface burn, cerebro-vascular impairment and intracranial hemorrhage as well as platelet depletion were observed. Ghrelin treatment after irradiation combined with burn trauma significantly decreased platelet depletion and brain hemorrhage. The results suggest that ghrelin treatment is an effective therapy for ionizing radiation combined with burn trauma.
Collapse
|
55
|
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: 127] [Impact Index Per Article: 15.9] [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
|
56
|
de Andrade CBV, Ramos IPR, de Moraes ACN, do Nascimento ALR, Salata C, Goldenberg RCDS, de Carvalho JJ, de Almeida CEV. Radiotherapy-Induced Skin Reactions Induce Fibrosis Mediated by TGF-β1 Cytokine. Dose Response 2017; 15:1559325817705019. [PMID: 28507463 PMCID: PMC5415163 DOI: 10.1177/1559325817705019] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Purpose: This study aimed to investigate radiation-induced lesions on the skin in an experimental animal model. Methods and Materials: Cutaneous wounds were induced in Wistar rats by 4 MeV energy electron beam irradiation, using a dose rate of 240 cGy/min, for 3 different doses (10 Gy, 40 Gy, and 60 Gy). The skin was observed 5, 10, and 25 days (D) after ionizing radiation exposition. Results: Infiltrate inflammatory process was observed in D5 and D10, for the 40 Gy and 60 Gy groups, and a progressive increase of transforming growth factor β1 is associated with this process. It could also be noted a mischaracterization of collagen fibers at the high-dose groups. Conclusion: It was observed that the lesions caused by ionizing radiation in rats were very similar to radiodermatitis in patients under radiotherapy treatment. Advances in Knowledge: This study is important to develop strategies to prevent radiation-induced skin reactions.
Collapse
Affiliation(s)
- Cherley Borba Vieira de Andrade
- Radiological Sciences Laboratory, Department of Biophysics and Biometry, State University of Rio de Janeiro (UERJ), Rio de Janeiro, RJ, Brazil.,Translational Endocrinology Laboratory, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Isalira Peroba Rezende Ramos
- Center Structural Biology and Bio-imaging (CENABIO), Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Ana Lúcia Rosa do Nascimento
- Ultrastructure Laboratory and Tissue Biology, Department of Histology and Embriology (UERJ), Rio de Janeiro, RJ, Brazil
| | - Camila Salata
- Radiological Sciences Laboratory, Department of Biophysics and Biometry, State University of Rio de Janeiro (UERJ), Rio de Janeiro, RJ, Brazil
| | - Regina Coeli Dos Santos Goldenberg
- Cellular and Molecular Cardiology Laboratory, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Jorge José de Carvalho
- Ultrastructure Laboratory and Tissue Biology, Department of Histology and Embriology (UERJ), Rio de Janeiro, RJ, Brazil
| | - Carlos Eduardo Veloso de Almeida
- Radiological Sciences Laboratory, Department of Biophysics and Biometry, State University of Rio de Janeiro (UERJ), Rio de Janeiro, RJ, Brazil
| |
Collapse
|
57
|
Kiang JG. Exacerbation of Mild Hypoxia on Acute Radiation Syndrome and Subsequent Mortality. ADAPTIVE MEDICINE 2017; 9:28-33. [PMID: 34616568 PMCID: PMC8491646 DOI: 10.4247/am.2017.abg170] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Mild hypoxia induced by 20% hemorrhage results in increases in few cytokine concentrations and sclerostin levels in blood, but shows no changes in bone formation, bone marrow cellularity, and gastrointestinal (GI) integrity and no systemic bacterial infection as well as no subsequent mortality. On the other hand, severe hypoxia induced by 40% hemorrhage causes significant increases in most cytokine concentrations, GI injury, lung injury, systemic bacterial infection, cellular ATP reduction and subsequent mortality. The severe hypoxia drastically damages GI and lung morphology, elevates cytokine concentrations in blood and increases inducible nitric oxide synthase (iNOS) expression in cells that is mediated by transcription factors NF-κB/NF-IL6, subsequently producing free radicals that disrupt mitochondria. ATP depletion, p53 phosphorylation, and caspase-3 activation are found, suggesting cell apoptosis. As a result, mortality occurs. However, when mild hypoxia follows ionizing radiation, the mild hypoxia significantly enhances radiation-induced mortality and acute radiation syndrome, including injury of bone marrow, GI, kidney, and lung. The synergism also occurs at the molecular level, resulting in alteration of microRNAs, amplification of iNOS expression, cytokine increases, sepsis, and ATP depletion. This is the first demonstration of synergistic effects between mild hypoxia and ionizing radiation.
Collapse
Affiliation(s)
- Juliann G Kiang
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute Department of Pharmacology and Molecular Therapeutics, Department of Medicine Uniformed Services University of the Health Sciences, Bethesda, Maryland, U.S.A
| |
Collapse
|
58
|
Kiang JG, Zhai M, Liao PJ, Ho C, Gorbunov NV, Elliott TB. Thrombopoietin Receptor Agonist Mitigates Hematopoietic Radiation Syndrome and Improves Survival after Whole-Body Ionizing Irradiation Followed by Wound Trauma. Mediators Inflamm 2017; 2017:7582079. [PMID: 28408792 PMCID: PMC5376937 DOI: 10.1155/2017/7582079] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 12/29/2016] [Accepted: 01/29/2017] [Indexed: 01/09/2023] Open
Abstract
Ionizing radiation combined with trauma tissue injury (combined injury, CI) results in greater mortality and H-ARS than radiation alone (radiation injury, RI), which includes thrombocytopenia. The aim of this study was to determine whether increases in numbers of thrombocytes would improve survival and mitigate H-ARS after CI. We observed in mice that WBC and platelets remained very low in surviving RI animals that were given 9.5 Gy 60Co-γ-photon radiation, whereas only lymphocytes and basophils remained low in surviving CI mice that were irradiated and then given skin wounds. Numbers of RBC and platelets, hemoglobin concentrations, and hematocrit values remained low in surviving RI and CI mice. CI induced 30-day mortality higher than RI. Radiation delayed wound healing by approximately 14 days. Treatment with a thrombopoietin receptor agonist, Alxn4100TPO, after CI improved survival, mitigated body-weight loss, and reduced water consumption. Though this therapy delayed wound-healing rate more than in vehicle groups, it greatly increased numbers of platelets in sham, wounded, RI, and CI mice; it significantly mitigated decreases in WBC, spleen weights, and splenocytes in CI mice and decreases in RBC, hemoglobin, hematocrit values, and splenocytes and splenomegaly in RI mice. The results suggest that Alxn4100TPO is effective in mitigating CI.
Collapse
Affiliation(s)
- Juliann G. Kiang
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, 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
| | - Min Zhai
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889, USA
| | - Pei-Jun Liao
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889, USA
| | - Connie Ho
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889, USA
- College of Letters & Science, University of California, Berkeley, CA 94510, USA
| | - Nikolai V. Gorbunov
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889, USA
| | - Thomas B. Elliott
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889, USA
| |
Collapse
|
59
|
Antonic V, Jackson IL, Ganga G, Shea-Donohue T, Vujaskovic Z. Development of A Novel Murine Model of Combined Radiation and Peripheral Tissue Trauma Injuries. Radiat Res 2017; 187:241-250. [PMID: 28118112 DOI: 10.1667/rr14557.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Detonation of a 10-kiloton nuclear bomb in an urban setting would result in >1 million casualties, the majority of which would present with combined injuries. Combined injuries, such as peripheral tissue trauma and radiation exposure, trigger inflammatory events that lead to multiple organ dysfunction (MOD) and death, with gastrointestinal (GI) and pulmonary involvement playing crucial roles. The objective of this study was to develop an animal model of combined injuries, peripheral tissue trauma (TBX animal model) combined with total body irradiation with 5% bone marrow shielding (TBI/BM5) to investigate if peripheral tissue trauma contributes to reduced survival. Male C57BL/6J mice were exposed to TBX10%, irradiation (TBI/BM5), or combined injuries (TBX10% + TBI/BM5). Experiments were conducted to evaluate mortality at day 7 after TBI/BM5. Serial euthanasia was performed at day 1, 3 and 6 or 7 after TBI/BM5 to evaluate the time course of pathophysiologic processes in combined injuries. Functional tests were performed to assess pulmonary function and GI motility. Postmortem samples of lungs and jejunum were collected to assess tissue damage. Results indicated higher lethality and shorter survival in the TBX10% +T BI/BM5 group than in the TBX10% or TBI/BM5 groups (day 1 vs. day 7 and 6, respectively). TBI/BM5 alone had no effects on the lungs but significantly impaired GI function at day 6. As expected, in the animals that received severe trauma (TBX10%), we observed impairment in lung function and delay in GI transit in the first 3 days, effects that decreased at later time points. Trauma combined with radiation (TBX10% + TBI/BM5) significantly augmented impairment of the lung and GI function in comparison to TBX10% and TBI/BM5 groups at 24 h. Histologic evaluation indicated that combined injuries caused greater tissue damage in the intestines in TBX10% + TBI/BM5 group when compared to other groups. We describe here the first combined tissue trauma/radiation injury model that will allow conduction of mechanistic studies to identify new therapeutic targets and serve as a platform for testing novel therapeutic interventions.
Collapse
Affiliation(s)
- Vlado Antonic
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Isabel L Jackson
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Gurung Ganga
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Terez Shea-Donohue
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Zeljko Vujaskovic
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
60
|
Lu Y, Tang G, Lin H, Lin X, Jiang L, Yang GY, Wang Y. A biosafety evaluation of synchrotron radiation X-ray to skin and bone marrow: single dose irradiation study of rats and macaques. Int J Radiat Biol 2017; 93:637-645. [PMID: 28112006 DOI: 10.1080/09553002.2017.1286049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
PURPOSE Very limited experimental data is available regarding the safe dosages related to synchrotron radiation (SR) procedures. We used young rats and macaques to address bone marrow and skin tolerance to various doses of synchrotron radiation. METHODS Rats were subjected to 0, 0.5, 2.5, 5, 25 or 100 Gy local SR X-ray irradiation at left hind limb. Rat blood samples were analyzed at 2-90 days after irradiation. The SR X-ray irradiated skin and tibia were sectioned for morphological examination. For non-human primate study, three male macaques were subjected to 0.5 or 2.5 Gy SR X-ray on crus. Skin responses of macaques were observed. RESULTS All rats that received SR X-ray irradiation doses greater than 2.5 Gy experienced hair loss and bone-growth inhibition, which were accompanied by decreased number of follicles, thickened epidermal layer, and decreased density of bone marrow cells (p < 0.05). Macaque skin could tolerate 0.5 Gy SR X-ray but showed significant hair loss when the dose was raised above 2.5 Gy. CONCLUSION The safety threshold doses of SR X-ray for rat skin, bone marrow and macaque skin are between 0.5 and 2.5 Gy. Our study provided essential information regarding the biosafety of SR X-ray irradiation.
Collapse
Affiliation(s)
- Yifan Lu
- a Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering , Shanghai Jiao Tong University , Shanghai , China
| | - Guanghui Tang
- a Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering , Shanghai Jiao Tong University , Shanghai , China
| | - Hui Lin
- b School of Electronic Science and Application Physics , Hefei University of Technology , Hefei , Anhui , China
| | - Xiaojie Lin
- a Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering , Shanghai Jiao Tong University , Shanghai , China
| | - Lu Jiang
- a Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering , Shanghai Jiao Tong University , Shanghai , China
| | - Guo-Yuan Yang
- a Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering , Shanghai Jiao Tong University , Shanghai , China.,c Department of Neurology, Ruijin Hospital, School of Medicine , Shanghai Jiao Tong University , Shanghai , China
| | - Yongting Wang
- a Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering , Shanghai Jiao Tong University , Shanghai , China
| |
Collapse
|
61
|
Abstract
Recent understanding of the cellular and molecular signaling activations in adult mesenchymal stem cells (MSCs) has provided new insights into their potential clinical applications, particularly for tissue repair and regeneration. This review focuses on these advances, specifically in the context of self-renewal for tissue repair and recovery after radiation injury. Thus far, MSCs have been characterized extensively and shown to be useful in mitigation and therapy for acute radiation syndrome and cognitive dysfunction. Use of MSCs for treating radiation injury alone or in combination with additional trauma is foreseeable.
Collapse
Affiliation(s)
- Juliann G Kiang
- *Scientific Research Department, Armed Forces Radiobiology Research Institute; Department of Radiation Biology, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20889-1076
| |
Collapse
|
62
|
Abstract
In the event of a nuclear disaster, the individuals proximal to the source of radiation will be exposed to combined radiation injury. As irradiation delays cutaneous repair, the purpose of this study was to elucidate the effect of combined radiation and burn injury (CRBI) on apoptosis and inflammation at the site of skin injury. Male C57Bl/6 mice were exposed to no injury, thermal injury only, radiation only (1 and 6 Gy) and CRBI (1 and 6 Gy) and euthanized at various times after for skin collection. TUNEL staining revealed that the CRBI 6 Gy group had a delayed and increased apoptotic response. This correlated with decreased recovery of live cells as compared to the other injuries. Similar response was observed when cleaved-caspase-3 immunohistochemical staining was compared between CRBI 6 Gy and thermal injury. TNFR1, caspase 8, Bax and IL-6 mRNA expression revealed that the higher CRBI group had delayed increase in mRNA expression as compared to thermal injury alone. RIPK1 mRNA expression and necrotic cell counts were delayed in the CRBI 6 Gy group to day 5. TNF-α and NFκB expression peaked in the CRBI 6 Gy group at day 1 and was much higher than the other injuries. Also, inflammatory cell counts in the CRBI 6 Gy group were lower at early time points as compared to thermal injury by itself. These data suggest that CRBI delays and exacerbates apoptosis and inflammation in skin as well as increases necrosis thus resulting in delayed wound healing.
Collapse
|
63
|
Singh VK, Newman VL, Romaine PL, Hauer-Jensen M, Pollard HB. Use of biomarkers for assessing radiation injury and efficacy of countermeasures. Expert Rev Mol Diagn 2015; 16:65-81. [PMID: 26568096 PMCID: PMC4732464 DOI: 10.1586/14737159.2016.1121102] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Several candidate drugs for acute radiation syndrome (ARS) have been identified that have low toxicity and significant radioprotective and radiomitigative efficacy. Inasmuch as exposing healthy human volunteers to injurious levels of radiation is unethical, development and approval of new radiation countermeasures for ARS are therefore presently based on animal studies and Phase I safety study in healthy volunteers. The Animal Efficacy Rule, which underlies the Food and Drug Administration approval pathway, requires a sound understanding of the mechanisms of injury, drug efficacy, and efficacy biomarkers. In this context, it is important to identify biomarkers for radiation injury and drug efficacy that can extrapolate animal efficacy results, and can be used to convert drug doses deduced from animal studies to those that can be efficacious when used in humans. Here, we summarize the progress of studies to identify candidate biomarkers for the extent of radiation injury and for evaluation of countermeasure efficacy.
Collapse
Affiliation(s)
- Vijay K Singh
- a F. Edward Hébert School of Medicine 'America's Medical School' , 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
| | - Victoria L Newman
- a F. Edward Hébert School of Medicine 'America's Medical School' , 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 Lp Romaine
- a F. Edward Hébert School of Medicine 'America's Medical School' , 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
| | - Martin Hauer-Jensen
- c Departments of Pharmaceutical Sciences, Surgery, and Pathology , University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare Systems , Little Rock , AR , USA
| | - Harvey B Pollard
- a F. Edward Hébert School of Medicine 'America's Medical School' , Uniformed Services University of the Health Sciences , Bethesda , MD , USA
| |
Collapse
|
64
|
Swift JM, Swift SN, Smith JT, Kiang JG, Allen MR. Skin wound trauma, following high-dose radiation exposure, amplifies and prolongs skeletal tissue loss. Bone 2015; 81:487-494. [PMID: 26335157 DOI: 10.1016/j.bone.2015.08.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 08/21/2015] [Accepted: 08/24/2015] [Indexed: 12/14/2022]
Abstract
The present study investigated the detrimental effects of non-lethal, high-dose (whole body) γ-irradiation on bone, and the impact that radiation combined with skin trauma (i.e. combined injury) has on long-term skeletal tissue health. Recovery of bone after an acute dose of radiation (RI; 8 Gy), skin wounding (15-20% of total body skin surface), or combined injury (RI+Wound; CI) was determined 3, 7, 30, and 120 days post-irradiation in female B6D2F1 mice and compared to non-irradiated mice (SHAM) at each time-point. CI mice demonstrated long-term (day 120) elevations in serum TRAP 5b (osteoclast number) and sclerostin (bone formation inhibitor), and suppression of osteocalcin levels through 30 days as compared to SHAM (p<0.05). Radiation-induced reductions in distal femur trabecular bone volume fraction and trabecular number through 120 days post-exposure were significantly greater than non-irradiated mice (p<0.05) and were exacerbated in CI mice by day 30 (p<0.05). Negative alterations in trabecular bone microarchitecture were coupled with extended reductions in cancellous bone formation rate in both RI and CI mice as compared to Sham (p<0.05). Increased osteoclast surface in CI animals was observed for 3 days after irradiation and remained elevated through 120 days (p<0.01). These results demonstrate a long-term, exacerbated response of bone to radiation when coupled with non-lethal wound trauma. Changes in cancellous bone after combined trauma were derived from extended reductions in osteoblast-driven bone formation and increases in osteoclast activity.
Collapse
Affiliation(s)
- Joshua M Swift
- Armed Forces Radiobiology Research Institute, Bethesda, MD 20889-5603, USA; Department of Military and Emergency Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814 USA; Department of Radiation Biology, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814 USA.
| | - Sibyl N Swift
- Armed Forces Radiobiology Research Institute, Bethesda, MD 20889-5603, USA; Department of Military and Emergency Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814 USA.
| | - Joan T Smith
- Armed Forces Radiobiology Research Institute, Bethesda, MD 20889-5603, USA.
| | - Juliann G Kiang
- Armed Forces Radiobiology Research Institute, Bethesda, MD 20889-5603, USA; Department of Radiation Biology, 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.
| | - Matthew R Allen
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, 46202 USA.
| |
Collapse
|
65
|
Kiang JG, Smith JT, Anderson MN, Swift JM, Christensen CL, Gupta P, Balakathiresan N, Maheshwari RK. Hemorrhage Exacerbates Radiation Effects on Survival, Leukocytopenia, Thrombopenia, Erythropenia, Bone Marrow Cell Depletion and Hematopoiesis, and Inflammation-Associated microRNAs Expression in Kidney. PLoS One 2015; 10:e0139271. [PMID: 26422254 PMCID: PMC4589285 DOI: 10.1371/journal.pone.0139271] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 09/09/2015] [Indexed: 12/18/2022] Open
Abstract
Exposure to high-dose radiation results in detrimental effects on survival. The effects of combined trauma, such as radiation in combination with hemorrhage, the typical injury of victims exposed to a radiation blast, on survival and hematopoietic effects have yet to be understood. The purpose of this study was to evaluate the effects of radiation injury (RI) combined with hemorrhage (i.e., combined injury, CI) on survival and hematopoietic effects, and to investigate whether hemorrhage (Hemo) enhanced RI-induced mortality and hematopoietic syndrome. Male CD2F1 mice (10 weeks old) were given one single exposure of γ- radiation (60Co) at various doses (0.6 Gy/min). Within 2 hr after RI, animals under anesthesia were bled 0% (Sham) or 20% (Hemo) of total blood volume via the submandibular vein. In these mice, Hemo reduced the LD50/30 for 30-day survival from 9.1 Gy (RI) to 8.75 Gy (CI) with a DMF of 1.046. RI resulted in leukocytopenia, thrombopenia, erythropenia, and bone marrow cell depletion, but decreased the caspase-3 activation response. RI increased IL-1β, IL-6, IL-17A, and TNF-α concentrations in serum, bone marrow, ileum, spleen, and kidney. Some of these adverse alterations were magnified by CI. Erythropoietin production was increased in kidney and blood more after CI than RI. Furthermore, CI altered the global miRNAs expression in kidney and the ingenuity pathway analysis showed that miRNAs viz., let-7e, miR-30e and miR-29b that were associated with hematopoiesis and inflammation. This study provides preliminary evidence that non-lethal Hemo exacerbates RI-induced mortality and cell losses associated with high-dose γ-radiation. We identified some of the initial changes occurring due to CI which may have facilitated in worsening the injury and hampering the recovery of animals ultimately resulting in higher mortality.
Collapse
Affiliation(s)
- Juliann G. Kiang
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
- Department of Radiation Biology, 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, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Marsha N. Anderson
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Joshua M. Swift
- Department of Radiation Biology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
- Department of Military and Emergency Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
- Undersea Medicine Department, Naval Medical Research Center, Silver Spring, Maryland, United States of America
| | - Christine L. Christensen
- Comparative Pathology Division, Veterinary Sciences Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, 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 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
| |
Collapse
|
66
|
Islam A, Bolduc DL, Zhai M, Kiang JG, Swift JM. Captopril Increases Survival after Whole-Body Ionizing Irradiation but Decreases Survival when Combined with Skin-Burn Trauma in Mice. Radiat Res 2015; 184:273-9. [PMID: 26305295 DOI: 10.1667/rr14113.1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Past and recent radiation events have involved a high incidence of radiation combined injury where victims often succumb to serious infections as a consequence of bacterial translocation and subsequent sepsis. The risk of infection is exacerbated in radiation combined skin-burn injury (RCI), which increase vulnerability. Furthermore, no suitable countermeasures for radiation combined skin-burn injury have been established. In this study, we evaluated captopril as a potential countermeasure to radiation combined skin-burn injury. Captopril is an FDA-approved angiotensin-converting enzyme inhibitor that was previously reported to stimulate hematopoietic recovery after exposure to ionizing radiation. Female B6D2F1/J mice were whole-body bilateral (60)Co gamma-photon irradiated (dose rate of 0.4 Gy/min) with 9.5 Gy (LD70/30 for RCI), followed by nonlethal dorsal skin-burn injury under anesthesia (approximately 15% total-body surface-area burn). Mice were provided with acidified drinking water with or without dissolved captopril (0.55 g/l) for 30 days immediately after injury and were administered topical gentamicin (0.1% cream; day 1-10) and oral levofloxacin (90-100 mg/kg; day 3-16). Surviving mice were euthanized on day 30 after analyses of water consumption, body weight and survival. Our data demonstrate that, while treatment with captopril did mitigate mortality induced by radiation injury (RI) alone (55% captopril vs. 80% vehicle; n = 20, P < 0.05), it also resulted in decreased survival after radiation combined skin-burn injury (22% captopril vs. 41% vehicle; n = 22, P < 0.05). Moreover, captopril administration via drinking water produced an uneven dosage pattern among the different injury groups ranging from 74 ± 5.4 to 115 ± 2.2 mg/kg/day. Captopril treatment also did not counteract the negative alterations in hematology, splenocytes or bone marrow cellularity after either radiation injury or radiation combined skin-burn injury. These data suggest that captopril may exert its actions differently between the two injury models (RI vs. RCI) and that captopril dosing, when combined with topical and systemic antibiotic treatments, may not be a suitable countermeasure for RCI.
Collapse
Affiliation(s)
- Aminul Islam
- a Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, Maryland
| | - David L Bolduc
- a Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, Maryland
| | - Min Zhai
- a Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, Maryland
| | - Juliann G Kiang
- a Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, Maryland.,b Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland.,c Department of Radiation Biology, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Joshua M Swift
- a Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, Maryland.,c Department of Radiation Biology, Uniformed Services University of the Health Sciences, Bethesda, Maryland.,d Department of Military and Emergency Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| |
Collapse
|
67
|
Pejchal J, Šinkorová Z, Tichý A, Kmochová A, Ďurišová K, Kubelková K, Pohanka M, Bureš J, Tachecí I, Kuča K, Vávrová J. Attenuation of radiation-induced gastrointestinal damage by epidermal growth factor and bone marrow transplantation in mice. Int J Radiat Biol 2015; 91:703-14. [PMID: 25994811 DOI: 10.3109/09553002.2015.1054528] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
PURPOSE We examined the effect of epidermal growth factor (EGF) and bone marrow transplantation (BMT) on gastrointestinal damage after high-dose irradiation of mice. MATERIAL AND METHODS C57Black/6 mice were used. Two survival experiments were performed (12 and 13 Gy; (60)Co, 0.59-0.57 Gy/min). To evaluate BMT and EGF action, five groups were established - 0 Gy, 13 Gy, 13 Gy + EGF (at 2 mg/kg, first dose 24 h after irradiation and then every 48 h), 13 Gy + BMT (5 × 10(6) cells from green fluorescent protein [GFP] syngenic mice, 4 h after irradiation), and 13 Gy + BMT + EGF. Survival data, blood cell counts, gastrointestine and liver parameters and GFP positive cell migration were measured. RESULTS BMT and EGF (three doses, at 2 mg/kg, administered 1, 3 and 5 days after irradiation) significantly increased survival (13 Gy). In blood, progressive cytopenia was observed with BMT, EGF or their combination having no improving effect early after irradiation. In gastrointestinal system, BMT, EGF and their combination attenuated radiation-induced atrophy and increased regeneration during first week after irradiation with the combination being most effective. Signs of systemic inflammatory reaction were observed 30 days after irradiation. CONCLUSIONS Our data indicate that BMT together with EGF is a promising strategy in the treatment of high-dose whole-body irradiation damage.
Collapse
Affiliation(s)
- Jaroslav Pejchal
- a Department of Radiobiology , Faculty of Military Health Sciences, University of Defence , Trebesska, Hradec Kralove , Czech Republic
| | - Zuzana Šinkorová
- a Department of Radiobiology , Faculty of Military Health Sciences, University of Defence , Trebesska, Hradec Kralove , Czech Republic
| | - Aleš Tichý
- a Department of Radiobiology , Faculty of Military Health Sciences, University of Defence , Trebesska, Hradec Kralove , Czech Republic
| | - Adéla Kmochová
- a Department of Radiobiology , Faculty of Military Health Sciences, University of Defence , Trebesska, Hradec Kralove , Czech Republic
| | - Kamila Ďurišová
- a Department of Radiobiology , Faculty of Military Health Sciences, University of Defence , Trebesska, Hradec Kralove , Czech Republic
| | - Klára Kubelková
- b Department of Molecular Pathology and Biology , Faculty of Military Health Sciences, University of Defence , Trebesska, Hradec Kralove , Czech Republic
| | - Miroslav Pohanka
- b Department of Molecular Pathology and Biology , Faculty of Military Health Sciences, University of Defence , Trebesska, Hradec Kralove , Czech Republic
| | - Jan Bureš
- c 2nd Department of Internal Medicine - Gastroenterology , Faculty of Medicine in Hradec Kralove, Charles University in Prague , Simkova, Hradec Kralove , Czech Republic
| | - Ilja Tachecí
- c 2nd Department of Internal Medicine - Gastroenterology , Faculty of Medicine in Hradec Kralove, Charles University in Prague , Simkova, Hradec Kralove , Czech Republic
| | - Kamil Kuča
- d Biomedical Reseach Centre, University Hospital , Sokolska, Hradec Kralove , Czech Republic
| | - Jiřina Vávrová
- a Department of Radiobiology , Faculty of Military Health Sciences, University of Defence , Trebesska, Hradec Kralove , Czech Republic
| |
Collapse
|
68
|
Elliott TB, Bolduc DL, Ledney GD, Kiang JG, Fatanmi OO, Wise SY, Romaine PLP, Newman VL, Singh VK. Combined immunomodulator and antimicrobial therapy eliminates polymicrobial sepsis and modulates cytokine production in combined injured mice. Int J Radiat Biol 2015; 91:690-702. [PMID: 25994812 PMCID: PMC4673550 DOI: 10.3109/09553002.2015.1054526] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Purpose: A combination therapy for combined injury (CI) using a non-specific immunomodulator, synthetic trehalose dicorynomycolate and monophosphoryl lipid A (STDCM-MPL), was evaluated to augment oral antimicrobial agents, levofloxacin (LVX) and amoxicillin (AMX), to eliminate endogenous sepsis and modulate cytokine production. Materials and methods: Female B6D2F1/J mice received 9.75 Gy cobalt-60 gamma-radiation and wound. Bacteria were isolated and identified in three tissues. Incidence of bacteria and cytokines were compared between treatment groups. Results: Results demonstrated that the lethal dose for 50% at 30 days (LD50/30) of B6D2F1/J mice was 9.42 Gy. Antimicrobial therapy increased survival in radiation-injured (RI) mice. Combination therapy increased survival after RI and extended survival time but did not increase survival after CI. Sepsis began five days earlier in CI mice than RI mice with Gram-negative species predominating early and Gram-positive species increasing later. LVX plus AMX eliminated sepsis in CI and RI mice. STDCM-MPL eliminated Gram-positive bacteria in CI and most RI mice but not Gram-negative. Treatments significantly modulated 12 cytokines tested, which pertain to wound healing or elimination of infection. Conclusions: Combination therapy eliminates infection and prolongs survival time but does not assure CI mouse survival, suggesting that additional treatment for proliferative-cell recovery is required.
Collapse
Affiliation(s)
- Thomas B Elliott
- a Armed Forces Radiobiology Research Institute , Bethesda , MD , USA
| | - David L Bolduc
- a Armed Forces Radiobiology Research Institute , Bethesda , MD , USA
| | - G David Ledney
- a Armed Forces Radiobiology Research Institute , Bethesda , MD , USA
| | - Juliann G Kiang
- a Armed Forces Radiobiology Research Institute , Bethesda , MD , USA.,b Department of Radiation Biology , F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences , Bethesda , MD , USA.,c Department of Medicine , F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences , Bethesda , MD , USA
| | - Oluseyi O Fatanmi
- a Armed Forces Radiobiology Research Institute , Bethesda , MD , USA
| | - Stephen Y Wise
- a Armed Forces Radiobiology Research Institute , Bethesda , MD , USA
| | | | - Victoria L Newman
- a Armed Forces Radiobiology Research Institute , Bethesda , MD , USA
| | - Vijay K Singh
- a Armed Forces Radiobiology Research Institute , Bethesda , MD , USA.,b Department of Radiation Biology , F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences , Bethesda , MD , USA
| |
Collapse
|
69
|
Swift JM, Smith JT, Kiang JG. Ciprofloxacin Therapy Results in Mitigation of ATP Loss after Irradiation Combined with Wound Trauma: Preservation of Pyruvate Dehydrogenase and Inhibition of Pyruvate Dehydrogenase Kinase 1. Radiat Res 2015; 183:684-92. [PMID: 26010714 DOI: 10.1667/rr13853.1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Ionizing radiation exposure combined with wound injury increases animal mortalities than ionizing radiation exposure alone. Ciprofloxacin (CIP) is in the fluroquinolone family of synthetic antibiotic that are available from the strategic national stockpile for emergency use and is known to inhibit bacterial sepsis. The purpose of this study was to evaluate the efficacy of ciprofloxacin as a countermeasure to combined injury mortality and determine the signaling proteins involved in energy machinery. B6D2F1/J female mice were randomly assigned to receive either 9.75 Gy irradiation with Co-60 gamma rays followed by skin wounding (combined injury; CI) or sham procedure (sham). Either ciprofloxacin (90 mg/kg/day) or vehicle (VEH) (water) was administered orally to these mice 2 h after wounding and thereafter daily for 10 days. Determination of tissue adenosine triphosphate (ATP) was conducted, and immunoblotting for signaling proteins involved in ATP machinery was performed. Combined injury resulted in 60% survival after 10 days compared to 100% survival in the sham group. Furthermore, combined injury caused significant reductions of ATP concentrations in ileum, pancreas, brain, spleen, kidney and lung (-25% to -95%) compared to the sham group. Ciprofloxacin administration after combined injury resulted in 100% survival and inhibited reductions in ileum and kidney ATP production. Ileum protein levels of heat-shock protein 70 kDa (HSP-70, a chaperone protein involved in ATP synthesis) and pyruvate dehydrogenase (PDH, an enzyme complex crucial to conversion of pyruvate to acetyl CoA for entrance into TCA cycle) were significantly lower in the CI group (vs. sham group). Using immunoprecipitation and immunoblotting, HSP-70-PDH complex was found to be present in the ileum tissue of CI mice treated with ciprofloxacin. Furthermore, phosphorylation of serine residues of PDH resulting in inactivating PDH enzymatic activity, which occurred after combined injury, was inhibited with ciprofloxacin treatment, thus enabling PDH to increase ATP production. Increased ileum levels of pyruvate dehydrogenase kinase 1 protein (PDK1, an enzyme responsible for PDH phosphorylation) after combined injury were also prevented by ciprofloxacin treatment. Taken together, these data suggest that ciprofloxacin oral administration after combined injury had a role in sustained ileum ATP levels, and may have acted through preservation of PDH by HSP-70 and inhibition of PDK1. These molecular changes in the ileum are simply one of a host of mechanisms working in concert with one another by which ciprofloxacin treatment mitigates body weight loss and drastically enhances subsequent survival after combined injury. To this end, our findings indicate that oral treatment of ciprofloxacin is a valuable therapeutic treatment after irradiation with combined injury and warrants further analyses to elucidate the precise mechanisms involved.
Collapse
Affiliation(s)
- Joshua M Swift
- a Armed Forces Radiobiology Research Institute, Bethesda, Maryland 20889; and.,b Departments of Military and Emergency Medicine;,c Radiation Biology and
| | - Joan T Smith
- a Armed Forces Radiobiology Research Institute, Bethesda, Maryland 20889; and
| | - Juliann G Kiang
- a Armed Forces Radiobiology Research Institute, Bethesda, Maryland 20889; and.,c Radiation Biology and.,d Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814
| |
Collapse
|
70
|
Coniferyl aldehyde reduces radiation damage through increased protein stability of heat shock transcriptional factor 1 by phosphorylation. Int J Radiat Oncol Biol Phys 2015; 91:807-16. [PMID: 25752395 DOI: 10.1016/j.ijrobp.2014.11.031] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 11/10/2014] [Accepted: 11/19/2014] [Indexed: 11/23/2022]
Abstract
PURPOSE We previously screened natural compounds and found that coniferyl aldehyde (CA) was identified as an inducer of HSF1. In this study, we further examined the protective effects of CA against ionizing radiation (IR) in normal cell system. METHODS AND MATERIALS Western blotting and reverse transcription-polymerase chain reaction tests were performed to evaluate expression of HSF1, HSP27, and HSP70 in response to CA. Cell death and cleavage of PARP and caspase-3 were analyzed to determine the protective effects of CA in the presence of IR or taxol. The protective effects of CA were also evaluated using animal models. RESULTS CA increased stability of the HSF1 protein by phosphorylation at Ser326, which was accompanied by increased expression of HSP27 and HSP70. HSF1 phosphorylation at Ser326 by CA was mediated by EKR1/2 activation. Cotreatment of CA with IR or taxol in normal cells induced protective effects with phosphorylation- dependent patterns at Ser326 of HSF1. The decrease in bone marrow (BM) cellularity and increase of terminal deoxynucleotidyl transferase dUTP nick end labeling-positive BM cells by IR were also significantly inhibited by CA in mice (30.6% and 56.0%, respectively). A549 lung orthotopic lung tumor model indicated that CA did not affect the IR-mediated reduction of lung tumor nodules, whereas CA protected normal lung tissues from the therapeutic irradiation. CONCLUSIONS These results suggest that CA may be useful for inducing HSF1 to protect against normal cell damage after IR or chemotherapeutic agents.
Collapse
|
71
|
Swift JM, Smith JT, Kiang JG. Hemorrhage trauma increases radiation-induced trabecular bone loss and marrow cell depletion in mice. Radiat Res 2015; 183:578-83. [PMID: 25897554 DOI: 10.1667/rr13960.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Exposure to high-dose radiation results in deleterious effects on skeletal tissue. However, the effects of combined trauma such as radiation and hemorrhage on skeletal properties have yet to be elucidated. The purpose of this study was to evaluate the effects of radiation injury combined with hemorrhage on trabecular bone properties and biomarkers of bone metabolism, and to determine whether hemorrhage enhances radiation-associated bone loss. Male CD2F1 mice (10 weeks old) were exposed to one single dose of gamma radiation ((60)Co): 0 or 7.25 Gy. Two hours after irradiation, animals were bled 0% (n = 8) or 20% (n = 8) of total blood volume via the submandibular vein. Mice were euthanized 30 days after irradiation, and distal femora were analyzed using standard histomorphometry to determine changes in trabecular bone volume (BV/TV), thickness (Tb.Th), spacing (Tb.Sp), number (Tb.N) and marrow adipocyte density. Femurs from mice euthanized 1, 7 and 15 days post injury were flushed and total bone marrow cells were counted. Radiation exposure resulted in deleterious effects on distal femur BV/TV (-63%), Tb.Th (-34%), Tb.N (-45%), Tb.Sp (+125%) and adipocyte density (+286%) compared with the sham-irradiated mice (0 Gy; P < 0.05). Hemorrhage after irradiation resulted in greater deleterious effects on the distal femur with BV/TV (-13%), Tb.Th (-44%), Tb.N (-26%), Tb.Sp (+29%) and marrow adipocyte density (+33%) compared with radiation exposure only (P < 0.05). Analysis of the biomarkers of bone metabolism in serum from irradiated and hemorrhaged mice revealed significantly lower levels of osteocalcin (-60%) and procollagen type 1 amino-terminal propeptide (-36%; P1NP, biomarkers of bone formation activity), as well as elevations in sclerostin (+56%; SOST, an inhibitor of bone formation) compared with serum from irradiated only mice (P < 0.05). Additionally, the onset of bone marrow cell depletion in irradiated and hemorrhaged mice occurred earlier and to a greater extent compared to that in irradiated only mice. This study provides definitive, preliminary evidence that hemorrhage further exacerbates trabecular bone loss associated with nonlethal high-dose gamma radiation.
Collapse
Affiliation(s)
- Joshua M Swift
- a Armed Forces Radiobiology Research Institute, Bethesda, Maryland
| | | | | |
Collapse
|
72
|
Gorbunov NV, McDaniel DP, Zhai M, Liao PJ, Garrison BR, Kiang JG. Autophagy and mitochondrial remodelling in mouse mesenchymal stromal cells challenged with Staphylococcus epidermidis. J Cell Mol Med 2015; 19:1133-50. [PMID: 25721260 PMCID: PMC4420615 DOI: 10.1111/jcmm.12518] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 11/07/2014] [Indexed: 12/01/2022] Open
Abstract
The bone marrow stroma constitutes the marrow-blood barrier, which sustains immunochemical homoeostasis and protection of the haematopoietic tissue in sequelae of systemic bacterial infections. Under these conditions, the bone marrow stromal cells affected by circulating bacterial pathogens shall elicit the adaptive stress-response mechanisms to maintain integrity of the barrier. The objective of this communication was to demonstrate (i) that in vitro challenge of mesenchymal stromal cells, i.e. colony-forming unit fibroblasts (CFU-F), with Staphylococcus epidermidis can activate the autophagy pathway to execute antibacterial defence response, and (ii) that homoeostatic shift because of the bacteria-induced stress includes the mitochondrial remodelling and sequestration of compromised organelles via mitophagy. Implication of Drp1 and PINK1–PARK2-dependent mechanisms in the mitophagy turnover of the aberrant mitochondria in mesenchymal stromal cells is investigated and discussed.
Collapse
Affiliation(s)
- Nikolai V Gorbunov
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD, USA
| | | | | | | | | | | |
Collapse
|
73
|
Zawaski JA, Yates CR, Miller DD, Kaffes CC, Sabek OM, Afshar SF, Young DA, Yang Y, Gaber MW. Radiation Combined Injury Models to Study the Effects of Interventions and Wound Biomechanics. Radiat Res 2014; 182:640-52. [DOI: 10.1667/rr13751.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Janice A. Zawaski
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Charles R. Yates
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Duane D. Miller
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee
| | | | - Omaima M. Sabek
- Department of Surgery, The Methodist Health System, Houston, Texas
| | - Solmaz F. Afshar
- Department of Surgery, The Methodist Health System, Houston, Texas
| | - Daniel A. Young
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois
| | - Yunzhi Yang
- Department of Orthopedic Surgery, Stanford University, Stanford, California
| | - M. Waleed Gaber
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
74
|
Kiang JG, Zhai M, Liao PJ, Elliott TB, Gorbunov NV. Ghrelin therapy improves survival after whole-body ionizing irradiation or combined with burn or wound: amelioration of leukocytopenia, thrombocytopenia, splenomegaly, and bone marrow injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:215858. [PMID: 25374650 PMCID: PMC4211157 DOI: 10.1155/2014/215858] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 09/19/2014] [Indexed: 12/21/2022]
Abstract
Exposure to ionizing radiation alone (RI) or combined with traumatic tissue injury (CI) is a crucial life-threatening factor in nuclear and radiological events. In our laboratory, mice exposed to (60)Co-γ-photon radiation (9.5 Gy, 0.4 Gy/min, bilateral) followed by 15% total-body-surface-area skin wounds (R-W CI) or burns (R-B CI) experienced an increment of ≥18% higher mortality over a 30-day observation period compared to RI alone. CI was accompanied by severe leukocytopenia, thrombocytopenia, erythropenia, and anemia. At the 30th day after injury, numbers of WBC and platelets still remained very low in surviving RI and CI mice. In contrast, their RBC, hemoglobin, and hematocrit were recovered towards preirradiation levels. Only RI induced splenomegaly. RI and CI resulted in bone-marrow cell depletion. In R-W CI mice, ghrelin (a hunger-stimulating peptide) therapy increased survival, mitigated body-weight loss, accelerated wound healing, and increased hematocrit. In R-B CI mice, ghrelin therapy increased survival and numbers of neutrophils, lymphocytes, and platelets and ameliorated bone-marrow cell depletion. In RI mice, this treatment increased survival, hemoglobin, and hematocrit and inhibited splenomegaly. Our novel results are the first to suggest that ghrelin therapy effectively improved survival by mitigating CI-induced leukocytopenia, thrombocytopenia, and bone-marrow injury or the RI-induced decreased hemoglobin and hematocrit.
Collapse
Affiliation(s)
- Juliann G. Kiang
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889, USA
- Department of Radiation Biology, 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
| | - Min Zhai
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889, USA
| | - Pei-Jyun Liao
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889, USA
| | - Thomas B. Elliott
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889, USA
| | - Nikolai V. Gorbunov
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889, USA
| |
Collapse
|
75
|
Chen Q, Xia X, Wu S, Wu A, Qi D, Liu W, Cui F, Jiao Y, Zhu W, Gu Y, Gao H, Zhang X, Cao J. Apoptosis, necrosis, and autophagy in mouse intestinal damage after 15-Gy whole body irradiation. Cell Biochem Funct 2014; 32:647-56. [DOI: 10.1002/cbf.3068] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 08/24/2014] [Accepted: 08/26/2014] [Indexed: 01/16/2023]
Affiliation(s)
- Qiu Chen
- School of Radiation Medicine and Protection; Soochow University; Suzhou China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions; Soochow University; Suzhou China
| | - Xiaochun Xia
- School of Radiation Medicine and Protection; Soochow University; Suzhou China
- Nantong Tumor Hospital; Nantong China
| | - Shu Wu
- School of Radiation Medicine and Protection; Soochow University; Suzhou China
| | - Anqing Wu
- School of Radiation Medicine and Protection; Soochow University; Suzhou China
| | - Dandan Qi
- Stem Cell Research Laboratory of Jiangsu Province; Suzhou China
| | - Wei Liu
- School of Radiation Medicine and Protection; Soochow University; Suzhou China
| | - Fengmei Cui
- School of Radiation Medicine and Protection; Soochow University; Suzhou China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions; Soochow University; Suzhou China
| | - Yang Jiao
- School of Radiation Medicine and Protection; Soochow University; Suzhou China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions; Soochow University; Suzhou China
| | - Wei Zhu
- School of Radiation Medicine and Protection; Soochow University; Suzhou China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions; Soochow University; Suzhou China
| | - Yongping Gu
- Experimental Centre of Medical College; Soochow University; Suzhou China
| | - Hongjian Gao
- Electron Microscopy Core Labratory, Shanghai Medical College; Fudan University; Shanghai China
| | - Xueguang Zhang
- Stem Cell Research Laboratory of Jiangsu Province; Suzhou China
- Jiangsu Institute of Clinical Immunology; Suzhou China
- Institute of Medical Biotechnology; Soochow University; Suzhou Jiangsu Province China
| | - Jianping Cao
- School of Radiation Medicine and Protection; Soochow University; Suzhou China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions; Soochow University; Suzhou China
| |
Collapse
|
76
|
Gao F, Fish BL, Szabo A, Schock A, Narayanan J, Jacobs ER, Moulder JE, Lazarova Z, Medhora M. Enhanced survival from radiation pneumonitis by combined irradiation to the skin. Int J Radiat Biol 2014; 90:753-61. [PMID: 24827855 DOI: 10.3109/09553002.2014.922722] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE To develop mitigators for combined irradiation to the lung and skin. METHODS Rats were treated with X-rays as follows: (1) 12.5 or 13 Gy whole thorax irradiation (WTI); (2) 30 Gy soft X-rays to 10% area of the skin only; (3) 12.5 or 13 Gy WTI + 30 Gy skin irradiation after 3 hours; (4) 12.5 Gy WTI + skin irradiation and treated with captopril (160 mg/m(2)/day) started after 7 days. Our end points were survival (primary) based on IACUC euthanization criteria and secondary measurements of breathing intervals and skin injury. Lung collagen at 210 days was measured in rats surviving 13 Gy WTI. RESULTS After 12.5 Gy WTI with or without skin irradiation, one rat (12.5 Gy WTI) was euthanized. Survival was less than 10% in rats receiving 13 Gy WTI, but was enhanced when combined with skin irradiation (p < 0.0001). Collagen content was increased at 210 days after 13 Gy WTI vs. 13 Gy WTI + 30 Gy skin irradiation (p < 0.05). Captopril improved radiation-dermatitis after 12.5 Gy WTI + 30 Gy skin irradiation (p = 0.008). CONCLUSIONS Radiation to the skin given 3 h after WTI mitigated morbidity during pneumonitis in rats. Captopril enhanced the rate of healing of radiation-dermatitis after combined irradiations to the thorax and skin.
Collapse
Affiliation(s)
- Feng Gao
- Department of Radiation Oncology
| | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Kiang JG, Fukumoto R. Ciprofloxacin increases survival after ionizing irradiation combined injury: γ-H2AX formation, cytokine/chemokine, and red blood cells. HEALTH PHYSICS 2014; 106:720-6. [PMID: 24776905 PMCID: PMC4007686 DOI: 10.1097/hp.0000000000000108] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Exposure to ionizing radiation alone (radiation injury, RI) or combined with traumatic tissue injury (radiation combined injury, CI) is a crucial life-threatening factor in nuclear and radiological accidents. It is well documented that RI and CI occur at the molecular, cellular, tissue, and system levels. However, their mechanisms remain largely unclear. It has been observed in dogs, pigs, rats, guinea pigs, and mice that radiation exposure combined with burns, wounds, or bacterial infection results in greater mortality than radiation exposure alone. In this laboratory, the authors found that B6D2F1/J female mice exposed to 9.75 Gy ⁶⁰Co-γ photon radiation followed by 15% total body surface area wounds experienced 50% higher mortality (over a 30-d observation period) compared to irradiation alone. CI enhanced DNA damages, amplified iNOS activation, induced massive release of pro-inflammatory cytokines, overexpressed MMPs and TLRs, and aggravated sepsis that led to cell death. In the present study, B6D2F1/J mice that received CI were treated with ciprofloxacin (CIP, 90 mg/kg p.o., q.d. within 2 h after CI through day 21). At day 1, CIP treatment reduced CI-induced γ-H2AX formation significantly. At day 10, CIP treatment not only reduced cytokine/chemokine concentrations significantly, including IL-6 and KC (i.e., IL-8 in humans), but also enhanced IL-3 production compared to vehicle-treated controls. CIP also elevated red blood cell counts, hemoglobin levels, and hematocrits. At day 30, CIP treatment increased 45% survival after CI (i.e., 2.3-fold increase over vehicle treatment). The results suggest that CIP may prove to be an effective therapeutic drug for CI.
Collapse
Affiliation(s)
- Juliann G. Kiang
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD, U.S.A
- Department of Radiation Biology, Uniformed Services University of the Health Sciences, Bethesda, MD, U.S.A
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, U.S.A
| | - Risaku Fukumoto
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD, U.S.A
| |
Collapse
|
78
|
Kiang JG, Zhai M, Liao PJ, Bolduc DL, Elliott TB, Gorbunov NV. Pegylated G-CSF inhibits blood cell depletion, increases platelets, blocks splenomegaly, and improves survival after whole-body ionizing irradiation but not after irradiation combined with burn. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:481392. [PMID: 24738019 PMCID: PMC3964894 DOI: 10.1155/2014/481392] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 01/22/2014] [Indexed: 01/12/2023]
Abstract
Exposure to ionizing radiation alone (radiation injury, RI) or combined with traumatic tissue injury (radiation combined injury, CI) is a crucial life-threatening factor in nuclear and radiological accidents. As demonstrated in animal models, CI results in greater mortality than RI. In our laboratory, we found that B6D2F1/J female mice exposed to (60)Co-γ-photon radiation followed by 15% total-body-surface-area skin burns experienced an increment of 18% higher mortality over a 30-day observation period compared to irradiation alone; that was accompanied by severe cytopenia, thrombopenia, erythropenia, and anemia. At the 30th day after injury, neutrophils, lymphocytes, and platelets still remained very low in surviving RI and CI mice. In contrast, their RBC, hemoglobin, and hematocrit were similar to basal levels. Comparing CI and RI mice, only RI induced splenomegaly. Both RI and CI resulted in bone marrow cell depletion. It was observed that only the RI mice treated with pegylated G-CSF after RI resulted in 100% survival over the 30-day period, and pegylated G-CSF mitigated RI-induced body-weight loss and depletion of WBC and platelets. Peg-G-CSF treatment sustained RBC balance, hemoglobin levels, and hematocrits and inhibited splenomegaly after RI. The results suggest that pegylated G-CSF effectively sustained animal survival by mitigating radiation-induced cytopenia, thrombopenia, erythropenia, and anemia.
Collapse
Affiliation(s)
- Juliann G. Kiang
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889, USA
- Department of Radiation Biology, 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
| | - Min Zhai
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889, USA
| | - Pei-Jyun Liao
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889, USA
| | - David L. Bolduc
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889, USA
| | - Thomas B. Elliott
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889, USA
| | - Nikolai V. Gorbunov
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889, USA
| |
Collapse
|
79
|
Fukumoto R, Burns TM, Kiang JG. Ciprofloxacin enhances stress erythropoiesis in spleen and increases survival after whole-body irradiation combined with skin-wound trauma. PLoS One 2014; 9:e90448. [PMID: 24587369 PMCID: PMC3938753 DOI: 10.1371/journal.pone.0090448] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 02/02/2014] [Indexed: 02/04/2023] Open
Abstract
Severe hematopoietic loss is one of the major therapeutic targets after radiation-combined injury (CI), a kind of injury resulting from radiation exposure combined with other traumas. In this study, we tested the use of ciprofloxacin (CIP) as a treatment, because of recently reported immunomodulatory effects against CI that may improve hematopoiesis. The CIP regimen was a daily, oral dose for 3 weeks, with the first dose 2 h after CI. CIP treatment improved 30-day survival in mice at 80% compared to 35% for untreated controls. Study of early changes in hematological parameters identified CI-induced progressive anemia by 10 days that CIP significantly ameliorated. CI induced erythropoietin (EPO) mRNA in kidney and protein in kidney and serum; CIP stimulated EPO mRNA expression. In spleens of CI mice, CIP induced bone morphogenetic protein 4 (BMP4) in macrophages with EPO receptors. Splenocytes from CIP-treated CI mice formed CD71+ colony-forming unit-erythroid significantly better than those from controls. Thus, CIP-mediated BMP4-dependent stress erythropoiesis may play a role in improving survival after CI.
Collapse
Affiliation(s)
- Risaku Fukumoto
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, Maryland, United States of America
| | - True M. Burns
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, Maryland, United States of America
| | - Juliann G. Kiang
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, Maryland, United States of America
- Department of Radiation Biology, 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
- * E-mail:
| |
Collapse
|
80
|
Chen X, Liao C, Chu Q, Zhou G, Lin X, Li X, Lu H, Xu B, Yue Z. Dissecting the molecular mechanism of ionizing radiation-induced tissue damage in the feather follicle. PLoS One 2014; 9:e89234. [PMID: 24586618 PMCID: PMC3930710 DOI: 10.1371/journal.pone.0089234] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Accepted: 01/16/2014] [Indexed: 11/18/2022] Open
Abstract
Ionizing radiation (IR) is a common therapeutic agent in cancer therapy. It damages normal tissue and causes side effects including dermatitis and mucositis. Here we use the feather follicle as a model to investigate the mechanism of IR-induced tissue damage, because any perturbation of feather growth will be clearly recorded in its regular yet complex morphology. We find that IR induces defects in feather formation in a dose-dependent manner. No abnormality was observed at 5 Gy. A transient, reversible perturbation of feather growth was induced at 10 Gy, leading to defects in the feather structure. This perturbation became irreversible at 20 Gy. Molecular and cellular analysis revealed P53 activation, DNA damage and repair, cell cycle arrest and apoptosis in the pathobiology. IR also induces patterning defects in feather formation, with disrupted branching morphogenesis. This perturbation is mediated by cytokine production and Stat1 activation, as manipulation of cytokine levels or ectopic Stat1 over-expression also led to irregular feather branching. Furthermore, AG-490, a chemical inhibitor of Stat1 signaling, can partially rescue IR-induced tissue damage. Our results suggest that the feather follicle could serve as a useful model to address the in vivo impact of the many mechanisms of IR-induced tissue damage.
Collapse
Affiliation(s)
- Xi Chen
- Institute of Life Sciences, Fuzhou University, Fuzhou, Fujian, China
| | - Chunyan Liao
- Institute of Life Sciences, Fuzhou University, Fuzhou, Fujian, China
| | - Qiqi Chu
- Institute of Life Sciences, Fuzhou University, Fuzhou, Fujian, China
| | - Guixuan Zhou
- Institute of Life Sciences, Fuzhou University, Fuzhou, Fujian, China
| | - Xiang Lin
- Institute of Life Sciences, Fuzhou University, Fuzhou, Fujian, China
| | - Xiaobo Li
- Department of Radiation Oncology, Union Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Haijie Lu
- Department of Radiation Oncology, Union Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Benhua Xu
- Department of Radiation Oncology, Union Hospital of Fujian Medical University, Fuzhou, Fujian, China
- * E-mail: (BX); (ZY)
| | - Zhicao Yue
- Institute of Life Sciences, Fuzhou University, Fuzhou, Fujian, China
- * E-mail: (BX); (ZY)
| |
Collapse
|
81
|
Kiang JG, Gorbunov NV. Bone Marrow Mesenchymal Stem Cells Increase Survival after Ionizing Irradiation Combined with Wound Trauma: Characterization and Therapy. JOURNAL OF CELL SCIENCE & THERAPY 2014; 5:190. [PMID: 34457993 PMCID: PMC8396709 DOI: 10.4172/2157-7013.1000190] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The aim of this study was to investigate whether treatment with mesenchymal stem cells (MSCs) could improve survival after radiation combined injury. Bone marrow MSCs (BMSCs) were isolated from femurs of B6D2F1/J female mice and were expanded and cultivated in hypoxic conditions (5% O2, 10% CO2, 85% N2) over 30 days. BMSCs were transfused to mice 24 hr after combined injury due to 60Co-γ-photon irradiation (9.25 and 9.75 Gy, 0.4 Gy/min, bilateral) followed by skin wounding (CI). Water consumption, body weight, wound healing, and survival tallies were monitored during observation period. Mice subjected to CI experienced a dramatic moribundity over a 30-day observation period. Thus, CI (9.25 Gy)-animal group was characterized by 40% mortality rate while CI (9.75 Gy)-animal group had 100% mortality rate. CI-induced sickness was accompanied by body weight loss, increased water intake, and delayed wound healing. At the 30th day post-injury, bone marrow cell depletion still remained in surviving CI mice. Treatment of CI (9.25 Gy)-animal group with BMSCs led to an increase in 30-day survival rate by 30%, attenuated body weight loss, accelerated wound healing rate, and ameliorated bone-marrow cell depletion. Our novel results are the first to suggest that BMSC therapy is efficacious to sustain animal survival after CI.
Collapse
Affiliation(s)
- Juliann G Kiang
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, Maryland, 20889, USA
- Department of Radiation Biology, 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
- Corresponding author: Juliann G Kiang, Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, Maryland, 20889, USA, Tel: 301-295-0530;
| | - Nikolai V Gorbunov
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, Maryland, 20889, USA
| |
Collapse
|
82
|
Singh VK, Wise SY, Fatanmi OO, Beattie LA, Ducey EJ, Seed TM. Alpha-tocopherol succinate- and AMD3100-mobilized progenitors mitigate radiation combined injury in mice. JOURNAL OF RADIATION RESEARCH 2014; 55:41-53. [PMID: 23814114 PMCID: PMC3885121 DOI: 10.1093/jrr/rrt088] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 05/23/2013] [Accepted: 05/27/2013] [Indexed: 05/28/2023]
Abstract
The purpose of this study was to elucidate the role of alpha-tocopherol succinate (TS)- and AMD3100-mobilized progenitors in mitigating combined injury associated with acute radiation exposure in combination with secondary physical wounding. CD2F1 mice were exposed to high doses of cobalt-60 gamma-radiation and then transfused intravenously with 5 million peripheral blood mononuclear cells (PBMCs) from TS- and AMD3100-injected mice after irradiation. Within 1 h after irradiation, mice were exposed to secondary wounding. Mice were observed for 30 d after irradiation and cytokine analysis was conducted by multiplex Luminex assay at various time-points after irradiation and wounding. Our results initially demonstrated that transfusion of TS-mobilized progenitors from normal mice enhanced survival of acutely irradiated mice exposed 24 h prior to transfusion to supralethal doses (11.5-12.5 Gy) of (60)Co gamma-radiation. Subsequently, comparable transfusions of TS-mobilized progenitors were shown to significantly mitigate severe combined injuries in acutely irradiated mice. TS administered 24 h before irradiation was able to protect mice against combined injury as well. Cytokine results demonstrated that wounding modulates irradiation-induced cytokines. This study further supports the conclusion that the infusion of TS-mobilized progenitor-containing PBMCs acts as a bridging therapy in radiation-combined-injury mice. We suggest that this novel bridging therapeutic approach involving the infusion of TS-mobilized hematopoietic progenitors following acute radiation exposure or combined injury might be applicable to humans.
Collapse
Affiliation(s)
- Vijay K. Singh
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, 8901 Wisconsin Ave, Bethesda, MD 20889-5603, USA
- Department of Radiation Biology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4417 Maple Avenue, Bethesda, MD, USA
| | - Stephen Y. Wise
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, 8901 Wisconsin Ave, Bethesda, MD 20889-5603, USA
| | - Oluseyi O. Fatanmi
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, 8901 Wisconsin Ave, Bethesda, MD 20889-5603, USA
| | - Lindsay A. Beattie
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, 8901 Wisconsin Ave, Bethesda, MD 20889-5603, USA
| | - Elizabeth J. Ducey
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, 8901 Wisconsin Ave, Bethesda, MD 20889-5603, USA
| | | |
Collapse
|
83
|
Kiang JG, Ledney GD. Skin injuries reduce survival and modulate corticosterone, C-reactive protein, complement component 3, IgM, and prostaglandin E 2 after whole-body reactor-produced mixed field (n + γ-photons) irradiation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:821541. [PMID: 24175013 PMCID: PMC3791621 DOI: 10.1155/2013/821541] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 08/05/2013] [Accepted: 08/13/2013] [Indexed: 01/19/2023]
Abstract
Skin injuries such as wounds or burns following whole-body γ-irradiation (radiation combined injury (RCI)) increase mortality more than whole-body γ-irradiation alone. Wound-induced decreases in survival after irradiation are triggered by sustained activation of inducible nitric oxide synthase pathways, persistent alteration of cytokine homeostasis, and increased susceptibility to systemic bacterial infection. Among these factors, radiation-induced increases in interleukin-6 (IL-6) concentrations in serum were amplified by skin wound trauma. Herein, the IL-6-induced stress proteins including C-reactive protein (CRP), complement 3 (C3), immunoglobulin M (IgM), and prostaglandin E2 (PGE2) were evaluated after skin injuries given following a mixed radiation environment that might be found after a nuclear incident. In this report, mice received 3 Gy of reactor-produced mixed field (n + γ-photons) radiations at 0.38 Gy/min followed by nonlethal skin wounding or burning. Both wounds and burns reduced survival and increased CRP, C3, and PGE2 in serum after radiation. Decreased IgM production along with an early rise in corticosterone followed by a subsequent decrease was noted for each RCI situation. These results suggest that RCI-induced alterations of corticosterone, CRP, C3, IgM, and PGE2 cause homeostatic imbalance and may contribute to reduced survival. Agents inhibiting these responses may prove to be therapeutic for RCI and improve related survival.
Collapse
Affiliation(s)
- Juliann G. Kiang
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889, USA
- Department of Radiation Biology, 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
| | - G. David Ledney
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889, USA
| |
Collapse
|
84
|
Lu X, Nurmemet D, Bolduc DL, Elliott TB, Kiang JG. Radioprotective effects of oral 17-dimethylaminoethylamino-17-demethoxygeldanamycin in mice: bone marrow and small intestine. Cell Biosci 2013; 3:36. [PMID: 24499553 PMCID: PMC3852109 DOI: 10.1186/2045-3701-3-36] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 08/01/2013] [Indexed: 01/05/2023] Open
Abstract
Background Our previous research demonstrated that one subcutaneous injection of 17-Dimethylaminoethylamino-17-demethoxygeldanamycin (17-DMAG) 24 hours (h) before irradiation (8.75 Gy) increased mouse survival by 75%. However, the protective mechanism of 17-DMAG is currently unknown. The present study aimed to investigate whether oral administration of 17-DMAG was also radioprotective and the potential role it may play in radioprotection. Results A single dose of orally pre-administered (24, 48, or 72 h) 17-DMAG (10 mg/kg) increased irradiated mouse survival, reduced body weight loss, improved water consumption, and decreased facial dropsy, whereas orally post-administered 17-DMAG failed. Additional oral doses of pre-treatment did not improve 30-day survival. The protective effect of multiple pre-administrations (2−3 times) of 17-DMAG at 10 mg/kg was equal to the outcome of a single pre-treatment. In 17-DMAG-pretreated mice, attenuation of bone marrow aplasia in femurs 30 days after irradiation with recovered expressions of cluster of differentiation 34, 44 (CD34, CD44), and survivin in bone marrow cells were observed. 17-DMAG also elevated serum granulocyte-colony stimulating factor (G-CSF), decreased serum fms-related tyrosine kinase 3 ligand, and reduced white blood cell depletion. 17-DMAG ameliorated small intestinal histological damage, promoted recovery of villus heights and intestinal crypts including stem cells, where increased leucine-rich repeat-containing G-protein coupled receptor 5 (Lgr5) was found 30 days after irradiation. Conclusions 17-DMAG is a potential radioprotectant for bone marrow and small intestine that results in survival improvement.
Collapse
Affiliation(s)
- Xinyue Lu
- Radiation Combined Injury Program, Scientific Research Department, Armed Forces Radiobiology Research Institute, 8901 Wisconsin Avenue, Bethesda, MD 20889-5603, USA.
| | | | | | | | | |
Collapse
|
85
|
Palmer JL, Deburghgraeve CR, Bird MD, Hauer-Jensen M, Chen MM, Yong S, Kovacs EJ. Combined radiation and burn injury results in exaggerated early pulmonary inflammation. Radiat Res 2013; 180:276-83. [PMID: 23899376 DOI: 10.1667/rr3104.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Events such as a nuclear meltdown accident or nuclear attack have potential for severe radiation injuries. Radiation injury frequently occurs in combination with other forms of trauma, most often burns. Thus far, combined injury studies have focused mainly on skin wound healing and damage to the gut. Since both radiation exposure and remote burn have pulmonary consequences, we examined the early effects of combined injury on the lung. C57BL/6 male mice were irradiated with 5 Gy of total body irradiation followed by a 15% total body surface area scald burn. Lungs from surviving animals were examined for evidence of inflammation and pneumonitis. At 48 h post-injury, pathology of the lungs from combined injury mice showed greater inflammation compared to all other treatment groups, with marked red blood cell and leukocyte congestion of the pulmonary vasculature. There was excessive leukocyte accumulation, primarily neutrophils, in the vasculature and interstitium, with occasional cells in the alveolar space. At 24 and 48 h post-injury, myeloperoxidase levels in lungs of combined injury mice were elevated compared to all other treatment groups (P < 0.01), confirming histological evidence of neutrophil accumulation. Pulmonary levels of the neutrophil chemoattractant KC (CXCL1) were 3 times above that of either injury alone (P < 0.05). Further, monocyte chemotactic protein-1 (MCP-1, CCL2) was increased two- and threefold compared to burn injury or radiation injury, respectively (P < 0.05). Together, these data suggest that combined radiation and burn injury augments early pulmonary congestion and inflammation. Currently, countermeasures for this unique type of injury are extremely limited. Further research is needed to elucidate the mechanisms behind the synergistic effects of combined injury in order to develop appropriate treatments.
Collapse
Affiliation(s)
- Jessica L Palmer
- a Burn and Shock Trauma Institute, Department of Surgery, and Loyola University Medical Center, Maywood, Illinois 60153
| | | | | | | | | | | | | |
Collapse
|
86
|
Combination of radiation and burn injury alters [¹⁸F] 2-fluoro-2-deoxy-D-glucose uptake in mice. J Burn Care Res 2013; 33:723-30. [PMID: 23143615 DOI: 10.1097/bcr.0b013e31825d678f] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Radiation exposure and burn injury have both been shown to alter glucose utilization in vivo. The present study was designed to study the effect of burn injury combined with radiation exposure on glucose metabolism in mice using [¹⁸F] 2-fluoro-2-deoxy-D-glucose (¹⁸FDG). Groups of male mice weighing approximately 30 g were studied. Group 1 was irradiated with a ¹³⁷Cs source (9 Gy). Group 2 received full thickness burn injury on 25% TBSA followed by resuscitation with saline (2 ml, IP). Group 3 received radiation followed 10 minutes later by burn injury. Group 4 were sham-treated controls. After treatment, the mice were fasted for 23 hours and then injected (IV) with 50 μCi of ¹⁸FDG. One hour postinjection, the mice were sacrificed, and biodistribution was measured. Positive blood cultures were observed in all groups of animals compared to the shams. Increased mortality was observed after 6 days in the burn plus radiated group as compared to the other groups. Radiation and burn treatments separately or in combination produced major changes in ¹⁸FDG uptake by many tissues. In the heart, brown adipose tissue, and spleen, radiation plus burn produced a much greater increase (P < .0001) in ¹⁸FDG accumulation than either treatment separately. All three treatments produced moderate decreases in ¹⁸FDG accumulation (P < .01) in the brain and gonads. Burn injury, but not irradiation, increased ¹⁸FDG accumulation in skeletal muscle; however, the combination of burn plus radiation decreased ¹⁸FDG accumulation in skeletal muscle. This model may be useful for understanding the effects of burns plus irradiation injury on glucose metabolism and in developing treatments for victims of injuries produced by the combination of burn plus irradiation.
Collapse
|
87
|
Gorbunov NV, Garrison BR, McDaniel DP, Zhai M, Liao PJ, Nurmemet D, Kiang JG. Adaptive redox response of mesenchymal stromal cells to stimulation with lipopolysaccharide inflammagen: mechanisms of remodeling of tissue barriers in sepsis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:186795. [PMID: 23710283 PMCID: PMC3654342 DOI: 10.1155/2013/186795] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Revised: 03/08/2013] [Accepted: 03/11/2013] [Indexed: 01/07/2023]
Abstract
Acute bacterial inflammation is accompanied by excessive release of bacterial toxins and production of reactive oxygen and nitrogen species (ROS and RNS), which ultimately results in redox stress. These factors can induce damage to components of tissue barriers, including damage to ubiquitous mesenchymal stromal cells (MSCs), and thus can exacerbate the septic multiple organ dysfunctions. The mechanisms employed by MSCs in order to survive these stress conditions are still poorly understood and require clarification. In this report, we demonstrated that in vitro treatment of MSCs with lipopolysaccharide (LPS) induced inflammatory responses, which included, but not limited to, upregulation of iNOS and release of RNS and ROS. These events triggered in MSCs a cascade of responses driving adaptive remodeling and resistance to a "self-inflicted" oxidative stress. Thus, while MSCs displayed high levels of constitutively present adaptogens, for example, HSP70 and mitochondrial Sirt3, treatment with LPS induced a number of adaptive responses that included induction and nuclear translocation of redox response elements such as NFkB, TRX1, Ref1, Nrf2, FoxO3a, HO1, and activation of autophagy and mitochondrial remodeling. We propose that the above prosurvival pathways activated in MSCs in vitro could be a part of adaptive responses employed by stromal cells under septic conditions.
Collapse
Affiliation(s)
- Nikolai V. Gorbunov
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889-1402, USA
| | - Bradley R. Garrison
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889-1402, USA
| | - Dennis P. McDaniel
- Biomedical Instrumentation Center, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Min Zhai
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889-1402, USA
| | - Pei-Jyun Liao
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889-1402, USA
| | - Dilber Nurmemet
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889-1402, USA
| | - Juliann G. Kiang
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889-1402, USA
- Department of Radiation Biology, 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
|
88
|
Abstract
The continued development of nuclear weapons and the potential for thermonuclear injury necessitates the further understanding of the immune consequences after radiation combined with injury (RCI). We hypothesized that sublethal ionization radiation exposure combined with a full-thickness thermal injury would result in the production of immature myeloid cells. Mice underwent either a full-thickness contact burn of 20% total body surface area or sham procedure followed by a single whole-body dose of 5-Gy radiation. Serum, spleen, and peripheral lymph nodes were harvested at 3 and 14 days after injury. Flow cytometry was performed to identify and characterize adaptive and innate cell compartments. Elevated proinflammatory and anti-inflammatory serum cytokines and profound leukopenia were observed after RCI. A population of cells with dual expression of the cell surface markers Gr-1 and CD11b were identified in all experimental groups, but were significantly elevated after burn alone and RCI at 14 days after injury. In contrast to the T-cell-suppressive nature of myeloid-derived suppressor cells found after trauma and sepsis, myeloid cells after RCI augmented T-cell proliferation and were associated with a weak but significant increase in interferon γ and a decrease in interleukin 10. This is consistent with previous work in burn injury indicating that a myeloid-derived suppressor cell-like population increases innate immunity. Radiation combined injury results in the increase in distinct populations of Gr-1CD11b cells within the secondary lymphoid organs, and we propose these immature inflammatory myeloid cells provide innate immunity to the severely injured and immunocompromised host.
Collapse
|
89
|
Fukumoto R, Cary LH, Gorbunov NV, Lombardini ED, Elliott TB, Kiang JG. Ciprofloxacin modulates cytokine/chemokine profile in serum, improves bone marrow repopulation, and limits apoptosis and autophagy in ileum after whole body ionizing irradiation combined with skin-wound trauma. PLoS One 2013; 8:e58389. [PMID: 23520506 PMCID: PMC3592826 DOI: 10.1371/journal.pone.0058389] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2012] [Accepted: 02/04/2013] [Indexed: 01/21/2023] Open
Abstract
Radiation combined injury (CI) is a radiation injury (RI) combined with other types of injury, which generally leads to greater mortality than RI alone. A spectrum of specific, time-dependent pathophysiological changes is associated with CI. Of these changes, the massive release of pro-inflammatory cytokines, severe hematopoietic and gastrointestinal losses and bacterial sepsis are important treatment targets to improve survival. Ciprofloxacin (CIP) is known to have immunomodulatory effect besides the antimicrobial activity. The present study reports that CIP ameliorated pathophysiological changes unique to CI that later led to major mortality. B6D2F1/J mice received CI on day 0, by RI followed by wound trauma, and were treated with CIP (90 mg/kg p.o., q.d. within 2 h after CI through day 10). At day 10, CIP treatment not only significantly reduced pro-inflammatory cytokine and chemokine concentrations, including interleukin-6 (IL-6) and KC (i.e., IL-8 in human), but it also enhanced IL-3 production compared to vehicle-treated controls. Mice treated with CIP displayed a greater repopulation of bone marrow cells. CIP also limited CI-induced apoptosis and autophagy in ileal villi, systemic bacterial infection, and IgA production. CIP treatment led to LD0/10 compared to LD20/10 for vehicle-treated group after CI. Given the multiple beneficial activities of CIP shown in our experiments, CIP may prove to be a useful therapeutic drug for CI.
Collapse
Affiliation(s)
- Risaku Fukumoto
- Radiation Combined Injury Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Lynnette H. Cary
- Radiation Countermeasures Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Nikolai V. Gorbunov
- Radiation Combined Injury Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Eric D. Lombardini
- Veterinary Sciences Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Thomas B. Elliott
- Radiation Combined Injury Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Juliann G. Kiang
- Radiation Combined Injury Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
- Department of Radiation Biology, 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
- * E-mail:
| |
Collapse
|
90
|
Tajima G, Delisle AJ, Hoang K, O'Leary FM, Ikeda K, Hanschen M, Stoecklein VM, Lederer JA. Immune system phenotyping of radiation and radiation combined injury in outbred mice. Radiat Res 2012; 179:101-12. [PMID: 23216446 DOI: 10.1667/rr3120.1] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The complexity of a radionuclear event would be immense due to varying levels of radiation exposures and injuries caused by blast-associated trauma. With this scenario in mind, we developed a mouse model to mimic as closely as possible the potential consequences of radiation injury and radiation combined injury (RCI) on survival, immune system phenotype, and immune function. Using a mouse burn injury model and a (137)CsCl source irradiator to induce injuries, we report that the immunological response to radiation combined injury differs significantly from radiation or burn injury alone. Mice that underwent radiation combined injury showed lower injury survival and cecal ligation and puncture (CLP) induced polymicrobial sepsis survival rates than mice with single injuries. As anticipated, radiation exposure caused dose-dependent losses of immune cell subsets. We found B and T cells to be more radiation sensitive, while macrophages, dendritic cells and NK cells were relatively more resistant. However, radiation and radiation combined injury did induce significant increases in the percentages of CD4(+) regulatory T cells (Tregs) and a subset of macrophages that express cell-surface GR-1 (GR-1(+) macrophages). Immune system phenotyping analysis indicated that spleen cells from radiation combined injury mice produced higher levels of proinflammatory cytokines than cells from mice with radiation or burn injury alone, especially at lower dose radiation exposure levels. Interestingly, this enhanced proinflammatory phenotype induced by radiation combined injury persisted for at least 28 days after injury. In total, our data provide baseline information on differences in immune phenotype and function between radiation injury and radiation combined injury in mice. The establishment of this animal model will aid in future testing for therapeutic strategies to mitigate the immune and pathophysiological consequences of radionuclear events.
Collapse
Affiliation(s)
- G Tajima
- Department of Surgery, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
91
|
Zhou Y, Ni H, Li M, Sanzari JK, Diffenderfer ES, Lin L, Kennedy AR, Weissman D. Effect of solar particle event radiation and hindlimb suspension on gastrointestinal tract bacterial translocation and immune activation. PLoS One 2012; 7:e44329. [PMID: 23028522 PMCID: PMC3446907 DOI: 10.1371/journal.pone.0044329] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Accepted: 08/01/2012] [Indexed: 01/26/2023] Open
Abstract
The environmental conditions that could lead to an increased risk for the development of an infection during prolonged space flight include: microgravity, stress, radiation, disturbance of circadian rhythms, and altered nutritional intake. A large body of literature exists on the impairment of the immune system by space flight. With the advent of missions outside the Earth's magnetic field, the increased risk of adverse effects due to exposure to radiation from a solar particle event (SPE) needs to be considered. Using models of reduced gravity and SPE radiation, we identify that either 2 Gy of radiation or hindlimb suspension alone leads to activation of the innate immune system and the two together are synergistic. The mechanism for the transient systemic immune activation is a reduced ability of the GI tract to contain bacterial products. The identification of mechanisms responsible for immune dysfunction during extended space missions will allow the development of specific countermeasures.
Collapse
Affiliation(s)
- Yu Zhou
- Division of Infectious Diseases, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Houping Ni
- Division of Infectious Diseases, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Minghong Li
- Division of Infectious Diseases, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jenine K. Sanzari
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Eric S. Diffenderfer
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Liyong Lin
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Ann R. Kennedy
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Drew Weissman
- Division of Infectious Diseases, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
92
|
Haston CK. Mouse genetic approaches applied to the normal tissue radiation response. Front Oncol 2012; 2:94. [PMID: 22891164 PMCID: PMC3413016 DOI: 10.3389/fonc.2012.00094] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Accepted: 07/22/2012] [Indexed: 01/20/2023] Open
Abstract
The varying responses of inbred mouse models to radiation exposure present a unique opportunity to dissect the genetic basis of radiation sensitivity and tissue injury. Such studies are complementary to human association studies as they permit both the analysis of clinical features of disease, and of specific variants associated with its presentation, in a controlled environment. Herein I review how animal models are studied to identify specific genetic variants influencing predisposition to radiation-induced traits. Among these radiation-induced responses are documented strain differences in repair of DNA damage and in extent of tissue injury (in the lung, skin, and intestine) which form the base for genetic investigations. For example, radiation-induced DNA damage is consistently greater in tissues from BALB/cJ mice, than the levels in C57BL/6J mice, suggesting there may be an inherent DNA damage level per strain. Regarding tissue injury, strain specific inflammatory and fibrotic phenotypes have been documented for principally, C57BL/6 C3H and A/J mice but a correlation among responses such that knowledge of the radiation injury in one tissue informs of the response in another is not evident. Strategies to identify genetic differences contributing to a trait based on inbred strain differences, which include linkage analysis and the evaluation of recombinant congenic (RC) strains, are presented, with a focus on the lung response to irradiation which is the only radiation-induced tissue injury mapped to date. Such approaches are needed to reveal genetic differences in susceptibility to radiation injury, and also to provide a context for the effects of specific genetic variation uncovered in anticipated clinical association studies. In summary, mouse models can be studied to uncover heritable variation predisposing to specific radiation responses, and such variations may point to pathways of importance to phenotype development in the clinic.
Collapse
Affiliation(s)
- Christina K Haston
- Meakins-Christie Laboratories and the Department of Medicine, McGill University Montreal, QC, Canada
| |
Collapse
|
93
|
Abstract
Enhanced radiation weapons (ERW) are fission-fusion devices where the massive numbers of neutrons generated during the fusion process are intentionally allowed to escape rather than be confined to increase yield (and fallout products). As a result, the energy partition of the weapon output shifts from blast and thermal energies toward prompt radiation. The neutron/gamma output ratio is also increased. Neutrons emitted from ERW are of higher energy than the Eave of neutrons from fission weapons. These factors affect the patterns of injury distribution; delay wound healing in combined injuries; reduce the therapeutic efficacy of medical countermeasures; and increase the dose to radiation-only casualties, thus potentiating the likelihood of encountering radiation-induced incapacitation. The risk of radiation-induced carcinogenesis is also increased. Radiation exposure to first responders from activation products is increased over that expected from a fission weapon of similar yield. However, the zone of dangerous fallout is significantly reduced in area. At least four nations have developed the potential to produce such weapons. Although the probability of detonation of an ERW in the near future is very small, it is nonzero, and clinicians and medical planners should be aware of the medical effects of ERW.
Collapse
Affiliation(s)
- Glen I Reeves
- Applied Research Associates, Inc., 801 N. Quincy Street Suite 700, Arlington, VA 22315, USA.
| |
Collapse
|
94
|
Guinan EC, Barbon CM, Kalish LA, Parmar K, Kutok J, Mancuso CJ, Stoler-Barak L, Suter EE, Russell JD, Palmer CD, Gallington LC, Voskertchian A, Vergilio JA, Cole G, Zhu K, D'Andrea A, Soiffer R, Weiss JP, Levy O. Bactericidal/permeability-increasing protein (rBPI21) and fluoroquinolone mitigate radiation-induced bone marrow aplasia and death. Sci Transl Med 2012; 3:110ra118. [PMID: 22116933 DOI: 10.1126/scitranslmed.3003126] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Identification of safe, effective treatments to mitigate toxicity after extensive radiation exposure has proven challenging. Only a limited number of candidate approaches have emerged, and the U.S. Food and Drug Administration has yet to approve any agent for a mass-casualty radiation disaster. Because patients undergoing hematopoietic stem cell transplantation undergo radiation treatment that produces toxicities similar to radiation-disaster exposure, we studied patients early after such treatment to identify new approaches to this problem. Patients rapidly developed endotoxemia and reduced plasma bactericidal/permeability-increasing protein (BPI), a potent endotoxin-neutralizing protein, in association with neutropenia. We hypothesized that a treatment supplying similar endotoxin-neutralizing activity might replace the BPI deficit and mitigate radiation toxicity and tested this idea in mice. A single 7-Gy radiation dose, which killed 95% of the mice by 30 days, was followed 24 hours later by twice-daily, subcutaneous injections of the recombinant BPI fragment rBPI21 or vehicle alone for 14 or 30 days, with or without an oral fluoroquinolone antibiotic with broad-spectrum antibacterial activity, including that against endotoxin-bearing Gram-negative bacteria. Compared to either fluoroquinolone alone or vehicle plus fluoroquinolone, the combined rBPI21 plus fluoroquinolone treatment improved survival, accelerated hematopoietic recovery, and promoted expansion of stem and progenitor cells. The observed efficacy of rBPI21 plus fluoroquinolone initiated 24 hours after lethal irradiation, combined with their established favorable bioactivity and safety profiles in critically ill humans, suggests the potential clinical use of this radiation mitigation strategy and supports its further evaluation.
Collapse
Affiliation(s)
- Eva C Guinan
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Kiang JG, Garrison BR, Burns TM, Zhai M, Dews IC, Ney PH, Cary LH, Fukumoto R, Elliott TB, Ledney GD. Wound trauma alters ionizing radiation dose assessment. Cell Biosci 2012; 2:20. [PMID: 22686656 PMCID: PMC3469379 DOI: 10.1186/2045-3701-2-20] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Accepted: 06/11/2012] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Wounding following whole-body γ-irradiation (radiation combined injury, RCI) increases mortality. Wounding-induced increases in radiation mortality are triggered by sustained activation of inducible nitric oxide synthase pathways, persistent alteration of cytokine homeostasis, and increased susceptibility to bacterial infection. Among these factors, cytokines along with other biomarkers have been adopted for biodosimetric evaluation and assessment of radiation dose and injury. Therefore, wounding could complicate biodosimetric assessments. RESULTS In this report, such confounding effects were addressed. Mice were given 60Co γ-photon radiation followed by skin wounding. Wound trauma exacerbated radiation-induced mortality, body-weight loss, and wound healing. Analyses of DNA damage in bone-marrow cells and peripheral blood mononuclear cells (PBMCs), changes in hematology and cytokine profiles, and fundamental clinical signs were evaluated. Early biomarkers (1 d after RCI) vs. irradiation alone included significant decreases in survivin expression in bone marrow cells, enhanced increases in γ-H2AX formation in Lin+ bone marrow cells, enhanced increases in IL-1β, IL-6, IL-8, and G-CSF concentrations in blood, and concomitant decreases in γ-H2AX formation in PBMCs and decreases in numbers of splenocytes, lymphocytes, and neutrophils. Intermediate biomarkers (7 - 10 d after RCI) included continuously decreased γ-H2AX formation in PBMC and enhanced increases in IL-1β, IL-6, IL-8, and G-CSF concentrations in blood. The clinical signs evaluated after RCI were increased water consumption, decreased body weight, and decreased wound healing rate and survival rate. Late clinical signs (30 d after RCI) included poor survival and wound healing. CONCLUSION Results suggest that confounding factors such as wounding alters ionizing radiation dose assessment and agents inhibiting these responses may prove therapeutic for radiation combined injury and reduce related mortality.
Collapse
Affiliation(s)
- Juliann G Kiang
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889-5603, USA
- Department of Radiation Biology, 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
| | - Bradley R Garrison
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889-5603, USA
| | - True M Burns
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889-5603, USA
| | - Min Zhai
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889-5603, USA
| | - Ian C Dews
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889-5603, USA
| | - Patrick H Ney
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889-5603, USA
| | - Lynnette H Cary
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889-5603, USA
| | - Risaku Fukumoto
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889-5603, USA
| | - Thomas B Elliott
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889-5603, USA
| | - G David Ledney
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute, Bethesda, MD 20889-5603, USA
| |
Collapse
|
96
|
Cary LH, Ngudiankama BF, Salber RE, Ledney GD, Whitnall MH. Efficacy of Radiation Countermeasures Depends on Radiation Quality. Radiat Res 2012; 177:663-75. [DOI: 10.1667/rr2783.1] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
97
|
Lam V, Moulder JE, Salzman NH, Dubinsky EA, Andersen GL, Baker JE. Intestinal microbiota as novel biomarkers of prior radiation exposure. Radiat Res 2012; 177:573-83. [PMID: 22439602 DOI: 10.1667/rr2691.1] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
There is an urgent need for rapid, accurate, and sensitive diagnostic platforms to confirm exposure to radiation and estimate the dose absorbed by individuals subjected to acts of radiological terrorism, nuclear power plant accidents, or nuclear warfare. Clinical symptoms and physical dosimeters, even when available, do not provide adequate diagnostic information to triage and treat life-threatening radiation injuries. We hypothesized that intestinal microbiota act as novel biomarkers of prior radiation exposure. Adult male Wistar rats (n = 5/group) received single or multiple fraction total-body irradiation of 10.0 Gy and 18.0 Gy, respectively. Fresh fecal pellets were obtained from each rat prior to (day 0) and at days 4, 11, and 21 post-irradiation. Fecal microbiota composition was determined using microarray and quantitative PCR (polymerase chain reaction) analyses. The radiation exposure biomarkers consisted of increased 16S rRNA levels of 12 members of the Bacteroidales, Lactobacillaceae, and Streptococcaceae after radiation exposure, unchanged levels of 98 Clostridiaceae and Peptostreptococcaceae, and decreased levels of 47 separate Clostridiaceae members; these biomarkers are present in human and rat feces. As a result of the ubiquity of these biomarkers, this biomarker technique is non-invasive; microbiota provide a sustained level of reporting signals that are increased several-fold following exposure to radiation, and intestinal microbiota that are unaffected by radiation serve as internal controls. We conclude that intestinal microbiota serve as novel biomarkers of prior radiation exposure, and may be able to complement conventional chromosome aberrational analysis to significantly enhance biological dose assessments.
Collapse
Affiliation(s)
- Vy Lam
- Division of Cardiothoracic Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA
| | | | | | | | | | | |
Collapse
|
98
|
Jourdan MM, Lopez A, Olasz EB, Duncan NE, Demara M, Kittipongdaja W, Fish BL, Mäder M, Schock A, Morrow NV, Semenenko VA, Baker JE, Moulder JE, Lazarova Z. Laminin 332 deposition is diminished in irradiated skin in an animal model of combined radiation and wound skin injury. Radiat Res 2011; 176:636-48. [PMID: 21854211 PMCID: PMC3227557 DOI: 10.1667/rr2422.1] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
Skin exposure to ionizing radiation affects the normal wound healing process and greatly impacts the prognosis of affected individuals. We investigated the effect of ionizing radiation on wound healing in a rat model of combined radiation and wound skin injury. Using a soft X-ray beam, a single dose of ionizing radiation (10-40 Gy) was delivered to the skin without significant exposure to internal organs. At 1 h postirradiation, two skin wounds were made on the back of each rat. Control and experimental animals were euthanized at 3, 7, 14, 21 and 30 days postirradiation. The wound areas were measured, and tissue samples were evaluated for laminin 332 and matrix metalloproteinase (MMP) 2 expression. Our results clearly demonstrate that radiation exposure significantly delayed wound healing in a dose-related manner. Evaluation of irradiated and wounded skin showed decreased deposition of laminin 332 protein in the epidermal basement membrane together with an elevated expression of all three laminin 332 genes within 3 days postirradiation. The elevated laminin 332 gene expression was paralleled by an elevated gene and protein expression of MMP2, suggesting that the reduced amount of laminin 332 in irradiated skin is due to an imbalance between laminin 332 secretion and its accelerated processing by elevated tissue metalloproteinases. Western blot analysis of cultured rat keratinocytes showed decreased laminin 332 deposition by irradiated cells, and incubation of irradiated keratinocytes with MMP inhibitor significantly increased the amount of deposited laminin 332. Furthermore, irradiated keratinocytes exhibited a longer time to close an artificial wound, and this delay was partially corrected by seeding keratinocytes on laminin 332-coated plates. These data strongly suggest that laminin 332 deposition is inhibited by ionizing radiation and, in combination with slower keratinocyte migration, can contribute to the delayed wound healing of irradiated skin.
Collapse
Affiliation(s)
- M. M. Jourdan
- Department of Dermatology, Center for Medical Countermeasures against Radiological Terrorism, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - A. Lopez
- Department of Dermatology, Center for Medical Countermeasures against Radiological Terrorism, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - E. B. Olasz
- Department of Dermatology, Center for Medical Countermeasures against Radiological Terrorism, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - N. E. Duncan
- Department of Dermatology, Center for Medical Countermeasures against Radiological Terrorism, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - M. Demara
- Department of Dermatology, Center for Medical Countermeasures against Radiological Terrorism, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - W. Kittipongdaja
- Department of Dermatology, Center for Medical Countermeasures against Radiological Terrorism, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - B. L. Fish
- Department of Radiation Oncology, Center for Medical Countermeasures against Radiological Terrorism, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - M. Mäder
- Department of Radiation Oncology, Center for Medical Countermeasures against Radiological Terrorism, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - A. Schock
- Department of Radiation Oncology, Center for Medical Countermeasures against Radiological Terrorism, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - N. V. Morrow
- Department of Radiation Oncology, Center for Medical Countermeasures against Radiological Terrorism, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - V. A. Semenenko
- Department of Radiation Oncology, Center for Medical Countermeasures against Radiological Terrorism, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - J. E. Baker
- Department of Surgery, Center for Medical Countermeasures against Radiological Terrorism, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - J. E. Moulder
- Department of Radiation Oncology, Center for Medical Countermeasures against Radiological Terrorism, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Z. Lazarova
- Department of Dermatology, Center for Medical Countermeasures against Radiological Terrorism, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
99
|
Williams JP, McBride WH. After the bomb drops: a new look at radiation-induced multiple organ dysfunction syndrome (MODS). Int J Radiat Biol 2011; 87:851-68. [PMID: 21417595 PMCID: PMC3314299 DOI: 10.3109/09553002.2011.560996] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE There is increasing concern that, since the Cold War era, there has been little progress regarding the availability of medical countermeasures in the event of either a radiological or nuclear incident. Fortunately, since much is known about the acute consequences that are likely to be experienced by an exposed population, the probability of survival from the immediate hematological crises after total body irradiation (TBI) has improved in recent years. Therefore focus has begun to shift towards later down-stream effects, seen in such organs as the gastrointestinal tract (GI), skin, and lung. However, the mechanisms underlying therapy-related normal tissue late effects, resulting from localised irradiation, have remained somewhat elusive and even less is known about the development of the delayed syndrome seen in the context of whole body exposures, when it is likely that systemic perturbations may alter tissue microenvironments and homeostasis. CONCLUSIONS The sequence of organ failures observed after near-lethal TBI doses are similar in many ways to that of multiple organ dysfunction syndrome (MODS), leading to multiple organ failure (MOF). In this review, we compare the mechanistic pathways that underlie both MODS and delayed normal tissue effects since these may impact on strategies to identify radiation countermeasures.
Collapse
Affiliation(s)
- Jacqueline P Williams
- Department of Radiation Oncology, University of Rochester Medical Center Rochester, NY 14642, USA.
| | | |
Collapse
|
100
|
Abstract
Combined radiation and burn injuries are likely to occur after nuclear events, such as a meltdown accident at a nuclear energy plant or a nuclear attack. Little is known about the mechanisms by which combined injuries result in higher mortality than by either insult alone, and few animal models exist for combined radiation and burn injury. Herein, the authors developed a murine model of radiation and scald burn injury. Mice were given a single dose of 0, 2, 4, 5, 6, or 9 Gray (Gy) alone, followed by a 15% TBSA scald burn. All mice receiving ≤4 Gy of radiation with burn survived combined injury. Higher doses of radiation (5, 6, and 9 Gy) followed by scald injury had a dose-dependent increase in mortality (34, 67, and 100%, respectively). Five Gy was determined to be the ideal dose to use in conjunction with burn injury for this model. There was a decrease in circulating white blood cells in burn, irradiated, and combined injury (5 Gy and burn) mice by 48 hours postinjury compared with sham (49.7, 11.6, and 57.3%, respectively). Circulating interleukin-6 and tumor necrosis factor-α were increased in combined injury at 48 hours postinjury compared with all other treatment groups. Prolonged overproduction of proinflammatory cytokines could contribute to subsequent organ damage. Decreased leukocytes might exacerbate immune impairment and susceptibility to infections. Future studies will determine whether there are long lasting consequences of this early proinflammatory response and extended decrease in leukocytes.
Collapse
|