Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Int J Radiat Biol ; 100(3): 317-334, 2024.
Article in English | MEDLINE | ID: mdl-37967239

ABSTRACT

PURPOSE: The growing concern over potential unintended nuclear accidents or malicious activities involving nuclear/radiological devices cannot be overstated. Exposure to whole-body doses of radiation can result in acute radiation syndrome (ARS), colloquially known as "radiation sickness," which can severely damage various organ systems. Long-term health consequences, such as cancer and cardiovascular disease, can develop many years post-exposure. Identifying effective medical countermeasures and devising a strategic medical plan represents an urgent, unmet need. Various clinical studies have investigated the therapeutic use of umbilical cord blood (UCB) for a range of illnesses, including ARS. The objective of this review is to thoroughly discuss ARS and its sub-syndromes, and to highlight recent findings regarding the use of UCB for radiation injury. UCB, a rich source of stem cells, boasts numerous advantages over other stem cell sources, like bone marrow, owing to its ease of collection and relatively low risk of severe graft-versus-host disease. Preclinical studies suggest that treatment with UCB, and often UCB-derived mesenchymal stromal cells (MSCs), results in improved survival, accelerated hematopoietic recovery, reduced gastrointestinal tract damage, and mitigation of radiation-induced pneumonitis and pulmonary fibrosis. Interestingly, recent evidence suggests that UCB-derived exosomes and their microRNAs (miRNAs) might assist in treating radiation-induced damage, largely by inhibiting fibrotic pathways. CONCLUSION: UCB holds substantial potential as a radiation countermeasure, and future research should focus on establishing treatment parameters for ARS victims.


Subject(s)
Acute Radiation Syndrome , Mesenchymal Stem Cells , MicroRNAs , Humans , Acute Radiation Syndrome/prevention & control , Acute Radiation Syndrome/metabolism , Fetal Blood , Stem Cells , MicroRNAs/metabolism , Mesenchymal Stem Cells/metabolism
2.
Biomolecules ; 13(5)2023 05 15.
Article in English | MEDLINE | ID: mdl-37238707

ABSTRACT

In cases of accidental high-dose total-body irradiation (TBI), acute radiation syndrome (ARS) can cause death. We reported that the thrombopoietin receptor agonist romiplostim (RP) has the potential to completely rescue mice exposed to lethal TBI. Extracellular vesicles (EVs) are involved in cell-to-cell communication, and the mechanism of RP action may be related to EVs that reflect the radio-mitigative information. We investigated the radio-mitigative effects of EVs on mice with severe ARS. C57BL/6 mice exposed to lethal TBI were treated with RP, and the EVs were isolated from the serum and intraperitoneally injected into other mice with severe ARS. The 30-day survival rate of lethal TBI mice drastically improved by 50-100% with the administration of EVs in the sera collected weekly from the mice in which radiation damage was alleviated and mortality was avoided by the administration of RP. Four responsive miRNAs, namely, miR-144-5p, miR-3620-5p, miR-6354, and miR-7686-5p showed significant expression changes in an array analysis. In particular, miR-144-5p was expressed only in the EVs of RP-treated TBI mice. Specific EVs may exist in the circulating blood of mice that escaped mortality with an ARS mitigator, and their membrane surface and endogenous molecules may be the key to the survival of mice with severe ARS.


Subject(s)
Acute Radiation Syndrome , Extracellular Vesicles , MicroRNAs , Mice , Animals , Acute Radiation Syndrome/drug therapy , Acute Radiation Syndrome/metabolism , Mice, Inbred C57BL , Radiation, Ionizing , MicroRNAs/genetics , MicroRNAs/metabolism , Extracellular Vesicles/metabolism
3.
Int J Radiat Oncol Biol Phys ; 116(5): 1163-1174, 2023 Aug 01.
Article in English | MEDLINE | ID: mdl-36792018

ABSTRACT

PURPOSE: Victims of acute radiation exposure are susceptible to hematopoietic toxicity due to bone marrow damage and loss of mature blood elements. Here, we evaluated cord blood-derived endothelial progenitor cells (CB-EPCs) as a potential cellular therapy for mitigation of hematologic acute radiation syndrome. CB-EPCs express endothelial cell markers and maintain their growth characteristics beyond 10+ passages without diminishing their doubling capacity. Further, CB-EPCs can be cryopreserved in vapor-phase liquid nitrogen and easily recovered for propagation, making them an attractive nonimmunogenic cellular therapy for off-the-shelf use. Importantly, we show CB-EPCs have the capacity to potently expand adult human bone marrow hematopoietic progenitor cells both in vitro and in vivo. METHODS AND MATERIALS: To demonstrate the role of CB-EPCs in promoting in vivo human immune reconstitution after irradiation, we employed a novel humanized mouse model established by transplant of CD34+ bone marrow cells from 9 unique adult organ donors into immunocompromised NSG-SGM3 mice. The response of the humanized immune system to ionizing irradiation was then tested by exposure to 1 Gy followed by subcutaneous treatment of CB-EPCs, Food and Drug Administration-approved growth factor pegfilgrastim (0.3 mg/kg), or saline. RESULTS: At day 7, total human bone marrow was decreased by 80% in irradiated controls. However, treatment with either growth factor pegfilgrastim or CB-EPCs increased recovery of total human bone marrow by 2.5-fold compared with saline. Notably, CB-EPCs also increased recovery of both human CD34+ progenitors by 5-fold and colony-forming capacity by 3-fold versus saline. Additionally, CB-EPCs promoted recovery of endogenous bone marrow endothelial cells as observed by both increased vessel area and length compared with saline. CONCLUSIONS: These findings indicate the feasibility of using humanized mice engrafted with adult bone marrow for radiation research and the development of CB-EPCs as an off-the-shelf cellular therapy for mitigation of hematologic acute radiation syndrome.


Subject(s)
Acute Radiation Syndrome , Endothelial Progenitor Cells , Hematopoietic Stem Cell Transplantation , Adult , Humans , Mice , Animals , Bone Marrow , Hematopoietic Stem Cells/physiology , Fetal Blood/metabolism , Acute Radiation Syndrome/metabolism , Bone Marrow Cells , Intercellular Signaling Peptides and Proteins/metabolism , Hematopoietic Stem Cell Transplantation/methods
4.
Life Sci ; 289: 120190, 2022 Jan 15.
Article in English | MEDLINE | ID: mdl-34883100

ABSTRACT

AIMS: Hematopoietic acute radiation syndrome (H-ARS) can cause lethality, and therefore, the necessity of a safe radioprotector. The present study was focused on investigating the role of melatonin in granulocytes colony-stimulating factor (G-CSF) and related mechanisms underlying the reduction of DNA damage in hematopoietic system of irradiated mice. MAIN METHODS: C57BL/6 male mice were exposed to 2, 5, and 7.5Gy of whole-body irradiation (WBI), 30 min after intra-peritoneal administration of melatonin with different doses. Mice were sacrificed at different time intervals after WBI, and bone marrow, splenocytes, and peripheral blood lymphocytes were isolated for studying various parameters including micronuclei (MN), cell cycle, comet, γ-H2AX, gene expression, amino acid profiling, and hematology. KEY FINDINGS: Melatonin100mg/kg ameliorated radiation (7.5Gy and 5Gy) induced MN frequency and cell death in bone marrow without mortality. At 24 h of post-WBI (2Gy), the frequency of micronucleated polychromatic erythrocytes (mnPCE) with different melatonin doses revealed 20 mg/kg as optimal i.p. dose for protecting the hematopoietic system against radiation injury. In comet assay, a significant reduction in radiation-induced % DNA tail (p ≤ 0.05) was observed at this dose. Melatonin reduced γ-H2AX foci/cell and eventually reached to the control level. Melatonin also decreased blood arginine levels in mice after 24 h of WBI. The gene expression of G-CSF, Bcl-2-associated X protein (BAX), and Bcl2 indicated the role of melatonin in G-CSF regulation and downstream pro-survival pathways along with anti-apoptotic activity. SIGNIFICANCE: The results revealed that melatonin recovers the hematopoietic system of irradiated mice by inducing G-CSF mediated radioprotection.


Subject(s)
Acute Radiation Syndrome/metabolism , Gamma Rays/adverse effects , Granulocyte Colony-Stimulating Factor/metabolism , Hematopoiesis , Melatonin/pharmacology , Radiation Injuries, Experimental/metabolism , Animals , Hematopoiesis/drug effects , Hematopoiesis/radiation effects , Male , Mice , Radiation Injuries, Experimental/drug therapy , Radiation Injuries, Experimental/pathology
5.
Health Phys ; 121(4): 345-351, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34546216

ABSTRACT

ABSTRACT: Near total body exposure to high-dose ionizing radiation results in organ-specific sequelae, including acute radiation syndromes and delayed effects of acute radiation exposure. Among these sequelae are acute kidney injury and chronic kidney injury. Reports that neither oxidative stress nor inflammation are dominant mechanisms defining radiation nephropathy inspired an unbiased, discovery-based proteomic interrogation in order to identify mechanistic pathways of injury. We quantitatively profiled the proteome of kidney from non-human primates following 12 Gy partial body irradiation with 2.5% bone marrow sparing over a time period of 3 wk. Kidney was analyzed by liquid chromatography-tandem mass spectrometry. Out of the 3,432 unique proteins that were identified, we found that 265 proteins showed significant and consistent responses across at least three time points post-irradiation, of which 230 proteins showed strong upregulation while 35 proteins showed downregulation. Bioinformatics analysis revealed significant pathway and upstream regulator perturbations post-high dose irradiation and shed light on underlying mechanisms of radiation damage. These data will be useful for a greater understanding of the molecular mechanisms of injury in well-characterized animal models of partial body irradiation with minimal bone marrow sparing. These data may be potentially useful in the future development of medical countermeasures.


Subject(s)
Acute Radiation Syndrome , Radiation Injuries, Experimental , Acute Radiation Syndrome/diagnosis , Acute Radiation Syndrome/etiology , Acute Radiation Syndrome/metabolism , Animals , Bone Marrow/radiation effects , Kidney/radiation effects , Macaca mulatta , Proteomics , Radiation Injuries, Experimental/etiology , Radiation Injuries, Experimental/metabolism
6.
Sci Rep ; 11(1): 5585, 2021 03 10.
Article in English | MEDLINE | ID: mdl-33692493

ABSTRACT

Recent political unrest has highlighted the importance of understanding the short- and long-term effects of gamma-radiation exposure on human health and survivability. In this regard, effective treatment for acute radiation syndrome (ARS) is a necessity in cases of nuclear disasters. Here, we propose 20 therapeutic targets for ARS identified using a systematic approach that integrates gene coexpression networks obtained under radiation treatment in humans and mice, drug databases, disease-gene association, radiation-induced differential gene expression, and literature mining. By selecting gene targets with existing drugs, we identified potential candidates for drug repurposing. Eight of these genes (BRD4, NFKBIA, CDKN1A, TFPI, MMP9, CBR1, ZAP70, IDH3B) were confirmed through literature to have shown radioprotective effect upon perturbation. This study provided a new perspective for the treatment of ARS using systems-level gene associations integrated with multiple biological information. The identified genes might provide high confidence drug target candidates for potential drug repurposing for ARS.


Subject(s)
Acute Radiation Syndrome , Databases, Nucleic Acid , Drug Delivery Systems , Gene Regulatory Networks , Transcription Factors , Transcriptome , Acute Radiation Syndrome/drug therapy , Acute Radiation Syndrome/genetics , Acute Radiation Syndrome/metabolism , Acute Radiation Syndrome/pathology , Animals , Drug Repositioning , Humans , Mice , Transcription Factors/genetics , Transcription Factors/metabolism
7.
Sci Rep ; 11(1): 89, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33420217

ABSTRACT

Current models to study the hematopoietic syndrome largely rely on the uniform whole-body exposures. However, in the radio-nuclear accidents or terrorist events, exposure can be non-uniform. The data available on the non-uniform exposures is limited. Thus, we have developed a mice model for studying the hematopoietic syndrome in the non-uniform or partial body exposure scenarios using the localized cobalt60 gamma radiation exposure. Femur region of Strain 'A' male mice was exposed to doses ranging from 7 to 20 Gy. The 30 day survival assay showed 19 Gy as LD100 and 17 Gy as LD50. We measured an array of cytokines and important stem cell markers such as IFN-γ, IL-3, IL-6, GM-CSF, TNF-α, G-CSF, IL-1α, IL-1ß, CD 34 and Sca 1. We found significant changes in IL-6, GM-CSF, TNF-α, G-CSF, and IL-1ß levels compared to untreated groups and amplified levels of CD 34 and Sca 1 positive population in the irradiated mice compared to the untreated controls. Overall, we have developed a mouse model of the hematopoietic acute radiation syndrome that might be useful for understanding of the non-uniform body exposure scenarios. This may also be helpful in the screening of drugs intended for individuals suffering from radiation induced hematopoietic syndrome.


Subject(s)
Acute Radiation Syndrome/etiology , Disease Models, Animal , Hematologic Diseases/etiology , Radiation Exposure/adverse effects , Acute Radiation Syndrome/genetics , Acute Radiation Syndrome/metabolism , Animals , Cobalt Radioisotopes/adverse effects , Cobalt Radioisotopes/chemistry , Cytokines/genetics , Cytokines/metabolism , Femur/metabolism , Femur/radiation effects , Gamma Rays/adverse effects , Hematologic Diseases/genetics , Hematologic Diseases/metabolism , Humans , Male , Mice
8.
Sci Rep ; 10(1): 19343, 2020 11 09.
Article in English | MEDLINE | ID: mdl-33168863

ABSTRACT

Hematopoietic acute radiation syndrome (H-ARS) and delayed effects of acute radiation exposure (DEARE) are detrimental health effects that occur after exposure to high doses of ionizing radiation. BIO 300, a synthetic genistein nanosuspension, was previously proven safe and effective against H-ARS when administered (via the oral (po) or intramuscular (im) route) prior to exposure to lethal doses of total-body radiation. In this study, we evaluated the proteomic changes in serum of nonhuman primates (NHP) after administering BIO 300 by different routes (po and im). We utilized nanoflow-ultra-performance liquid chromatography quadrupole time-of-flight mass spectrometry (NanoUPLC-MS/MS) methods for comprehensive global profiling and quantification of serum proteins. The results corroborate previous findings that suggest a very similar metabolic profile following both routes of drug administration. Furthermore, we observed minor alterations in protein levels, 2 hours after drug administration, which relates to the Cmax of BIO 300 for both routes of administration. Taken together, this assessment may provide an insight into the mechanism of radioprotection of BIO 300 and a reasonable illustration of the pharmacodynamics of this radiation countermeasure.


Subject(s)
Acute Radiation Syndrome/metabolism , Acute Radiation Syndrome/prevention & control , Proteomics/methods , Radiation, Ionizing , Animals , Chromatography, Liquid , Female , Genistein/pharmacology , Macaca mulatta , Male , Metabolome/drug effects , Metabolomics/methods , Principal Component Analysis , Radiation-Protective Agents/pharmacology , Tandem Mass Spectrometry , Time Factors , Whole-Body Irradiation
9.
Health Phys ; 119(5): 604-620, 2020 11.
Article in English | MEDLINE | ID: mdl-32947489

ABSTRACT

Exposure to ionizing radiation results in injuries of the hematopoietic, gastrointestinal, and respiratory systems, which are the leading causes responsible for morbidity and mortality. Gastrointestinal injury occurs as an acute radiation syndrome. To help inform on the natural history of the radiation-induced injury of the partial body irradiation model, we quantitatively profiled the proteome of jejunum from non-human primates following 12 Gy partial body irradiation with 2.5% bone marrow sparing over a time period of 3 wk. Jejunum was analyzed by liquid chromatography-tandem mass spectrometry, and pathway and gene ontology analysis were performed. A total of 3,245 unique proteins were quantified out of more than 3,700 proteins identified in this study. Also a total of 289 proteins of the quantified proteins showed significant and consistent responses across at least three time points post-irradiation, of which 263 proteins showed strong upregulations while 26 proteins showed downregulations. Bioinformatic analysis suggests significant pathway and upstream regulator perturbations post-high dose irradiation and shed light on underlying mechanisms of radiation damage. Canonical pathways altered by radiation included GP6 signaling pathway, acute phase response signaling, LXR/RXR activation, and intrinsic prothrombin activation pathway. Additionally, we observed dysregulation of proteins of the retinoid pathway and retinoic acid, an active metabolite of vitamin A, as quantified by liquid chromatography-tandem mass spectrometry. Correlation of changes in protein abundance with a well-characterized histological endpoint, corrected crypt number, was used to evaluate biomarker potential. These data further define the natural history of the gastrointestinal acute radiation syndrome in a non-human primate model of partial body irradiation with minimal bone marrow sparing.


Subject(s)
Acute Radiation Syndrome/diagnosis , Gastrointestinal Tract/metabolism , Organ Sparing Treatments/methods , Proteome/metabolism , Radiation Exposure/adverse effects , Radiation Injuries, Experimental/diagnosis , Retinoids/metabolism , Acute Radiation Syndrome/etiology , Acute Radiation Syndrome/metabolism , Animals , Biomarkers/metabolism , Bone Marrow/radiation effects , Disease Models, Animal , Gastrointestinal Tract/radiation effects , Macaca mulatta , Male , Proteome/analysis , Radiation Dosage , Radiation Injuries, Experimental/etiology , Radiation Injuries, Experimental/metabolism
10.
Int J Mol Sci ; 21(14)2020 Jul 17.
Article in English | MEDLINE | ID: mdl-32708958

ABSTRACT

Acute exposure to ionizing radiation leads to Hematopoietic Acute Radiation Syndrome (H-ARS). To understand the inter-strain cellular and molecular mechanisms of radiation sensitivity, adult males of two strains of minipig, one with higher radiosensitivity, the Gottingen minipig (GMP), and another strain with comparatively lower radiosensitivity, the Sinclair minipig (SMP), were exposed to total body irradiation (TBI). Since Insulin-like Growth Factor-1 (IGF-1) signaling is associated with radiation sensitivity and regulation of cardiovascular homeostasis, we investigated the link between dysregulation of cardiac IGF-1 signaling and radiosensitivity. The adult male GMP; n = 48, and SMP; n = 24, were irradiated using gamma photons at 1.7-2.3 Gy doses. The animals that survived to day 45 after irradiation were euthanized and termed the survivors. Those animals that were euthanized prior to day 45 post-irradiation due to severe illness or health deterioration were termed the decedents. Cardiac tissue analysis of unirradiated and irradiated animals showed that inter-strain radiosensitivity and survival outcomes in H-ARS are associated with activation status of the cardiac IGF-1 signaling and nuclear factor erythroid 2-related factor 2 (Nrf2)-mediated induction of antioxidant gene expression. Our data link H-ARS with dysregulation of cardiac IGF-1 signaling, and highlight the role of oxidative stress and cardiac antioxidant response in radiation sensitivity.


Subject(s)
Acute Radiation Syndrome/metabolism , Heart/radiation effects , Hematopoietic System/radiation effects , Insulin-Like Growth Factor I/metabolism , Signal Transduction/radiation effects , Acute Radiation Syndrome/etiology , Acute Radiation Syndrome/pathology , Animals , Gamma Rays/adverse effects , Hematopoietic System/metabolism , Hematopoietic System/pathology , Male , Myocardium/metabolism , Myocardium/pathology , Oxidative Stress/radiation effects , Radiation Tolerance/radiation effects , Swine , Swine, Miniature
11.
Exp Hematol ; 84: 54-66, 2020 04.
Article in English | MEDLINE | ID: mdl-32240658

ABSTRACT

Exposure to high-dose total body irradiation (TBI) can result in hematopoietic acute radiation syndrome (H-ARS), characterized by leukopenia, anemia, and coagulopathy. Death from H-ARS occurs from hematopoietic insufficiency and opportunistic infections. Following radiation exposure, red blood cells (RBCs) undergo hemolysis from radiation-induced hemoglobin denaturation, causing the release of iron. Free iron can have multiple detrimental biological effects, including suppression of hematopoiesis. We investigated the impact of radiation-induced iron release on the bone marrow following TBI and the potential impact of the ACE inhibitor captopril, which improves survival from H-ARS. C57BL/6J mice were exposed to 7.9 Gy, 60Co irradiation, 0.6 Gy/min (LD70-90/30). RBCs and reticulocytes were significantly reduced within 7 days of TBI, with the RBC nadir at 14-21 days. Iron accumulation in the bone marrow correlated with the time course of RBC hemolysis, with an ∼10-fold increase in bone marrow iron at 14-21 days post-irradiation, primarily within the cytoplasm of macrophages. Iron accumulation in the bone marrow was associated with increased expression of genes for iron binding and transport proteins, including transferrin, transferrin receptor 1, ferroportin, and integrin αMß2. Expression of the gene encoding Nrf2, a transcription factor activated by oxidative stress, also increased at 21 days post-irradiation. Captopril did not alter iron accumulation in the bone marrow or expression of iron storage genes, but did suppress Nrf2 expression. Our study suggests that following TBI, iron is deposited in tissues not normally associated with iron storage, which may be a secondary mechanism of radiation-induced tissue injury.


Subject(s)
Acute Radiation Syndrome/metabolism , Bone Marrow/metabolism , Gamma Rays/adverse effects , Hematopoiesis/radiation effects , Iron/metabolism , Radiation Injuries, Experimental/metabolism , Acute Radiation Syndrome/genetics , Acute Radiation Syndrome/pathology , Animals , Bone Marrow/pathology , Captopril/pharmacology , Erythrocytes/metabolism , Erythrocytes/pathology , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Hematopoiesis/drug effects , Hematopoiesis/genetics , Mice , Mice, Transgenic , NF-E2-Related Factor 2/biosynthesis , NF-E2-Related Factor 2/genetics , Radiation Injuries, Experimental/genetics , Radiation Injuries, Experimental/pathology
12.
Am J Physiol Gastrointest Liver Physiol ; 318(3): G439-G450, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31961718

ABSTRACT

Methionine is an essential amino acid needed for a variety of processes in living organisms. Ionizing radiation depletes tissue methionine concentrations and leads to the loss of DNA methylation and decreased synthesis of glutathione. In this study, we aimed to investigate the effects of methionine dietary supplementation in CBA/CaJ mice after exposure to doses ranging from 3 to 8.5 Gy of 137Cs of total body irradiation. We report that mice fed a methionine-supplemented diet (MSD; 19.5 vs. 6.5 mg/kg in a methionine-adequate diet, MAD) developed acute radiation toxicity at doses as low as 3 Gy. Partial body irradiation performed with hindlimb shielding resulted in a 50% mortality rate in MSD-fed mice exposed to 8.5 Gy, suggesting prevalence of radiation-induced gastrointestinal syndrome in the development of acute radiation toxicity. Analysis of the intestinal microbiome demonstrated shifts in the gut ecology, observed along with the development of leaky gut syndrome and bacterial translocation into the liver. Normal gut physiology impairment was facilitated by alterations in the one-carbon metabolism pathway and was exhibited as decreases in circulating citrulline levels mirrored by decreased intestinal mucosal surface area and the number of surviving crypts. In conclusion, we demonstrate that a relevant excess of methionine dietary intake exacerbates the detrimental effects of exposure to ionizing radiation in the small intestine.NEW & NOTEWORTHY Methionine supplementation, instead of an anticipated health-promoting effect, sensitizes mice to gastrointestinal radiation syndrome. Mechanistically, excess of methionine negatively affects intestinal ecology, leading to a cascade of physiological, biochemical, and molecular alterations that impair normal gut response to a clinically relevant genotoxic stressor. These findings speak toward increasing the role of registered dietitians during cancer therapy and the necessity of a solid scientific background behind the sales of dietary supplements and claims regarding their benefits.


Subject(s)
Acute Radiation Syndrome/etiology , Dietary Supplements/toxicity , Intestine, Small/drug effects , Methionine/toxicity , Radiation Injuries, Experimental/etiology , Acute Radiation Syndrome/metabolism , Acute Radiation Syndrome/microbiology , Acute Radiation Syndrome/pathology , Animals , DNA Methylation/drug effects , Dysbiosis , Energy Metabolism/drug effects , Gastrointestinal Microbiome/drug effects , Intestine, Small/metabolism , Intestine, Small/microbiology , Intestine, Small/pathology , Male , Mice, Inbred C57BL , Mice, Inbred CBA , Radiation Dosage , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/microbiology , Radiation Injuries, Experimental/pathology , Risk Factors , Whole-Body Irradiation
13.
Int J Radiat Biol ; 96(1): 4-11, 2020 01.
Article in English | MEDLINE | ID: mdl-30403905

ABSTRACT

Purpose: In the wake of a nuclear detonation, individuals with acute radiation syndrome will be a significant source of morbidity and mortality. Mathematical modeling can compare response strategies developed for real-world chaotic conditions after a nuclear blast in order to identify optimal strategies for administering effective treatment to these individuals. To maximize responders' abilities to save lives it is critical to understand how treatment efficacy is impacted by real-world conditions and levels of supportive care. To illustrate the importance of these factors, we developed a mathematical model of cytokine administration 24 h after the blast with varying levels of supportive care described in the primary literature.Conclusion: The results highlight the proportionally higher life-saving benefit of administering cytokines to individuals with a moderate to high dose of radiation exposure, compared to those with a lower dose. However, the fidelity of mathematical models is dependent on the primary data informing them. We describe the data needed to fully explore the impact of timing, dosage, and fractional benefit of cytokines and supportive care treatment in non-optimal situations that could be seen after a nuclear detonation. Studies addressing these types of knowledge gaps are essential to evaluating the relative efficacy of countermeasures to refine existing plans and help develop new strategies and priorities.


Subject(s)
Medical Countermeasures , Nuclear Weapons , Radiation Exposure/adverse effects , Time-to-Treatment , Acute Radiation Syndrome/etiology , Acute Radiation Syndrome/metabolism , Acute Radiation Syndrome/prevention & control , Acute Radiation Syndrome/therapy , Cytokines/metabolism , Humans
14.
Molecules ; 24(22)2019 Nov 06.
Article in English | MEDLINE | ID: mdl-31698831

ABSTRACT

Prostaglandins and inhibitors of their synthesis (cyclooxygenase (COX) inhibitors, non-steroidal anti-inflammatory drugs) were shown to play a significant role in the regulation of hematopoiesis. Partly due to their hematopoiesis-modulating effects, both prostaglandins and COX inhibitors were reported to act positively in radiation-exposed mammalian organisms at various pre- and post-irradiation therapeutical settings. Experimental efforts were targeted at finding pharmacological procedures leading to optimization of therapeutical outcomes by minimizing undesirable side effects of the treatments. Progress in these efforts was obtained after discovery of selective inhibitors of inducible selective cyclooxygenase-2 (COX-2) inhibitors. Recent studies have been able to suggest the possibility to find combined therapeutical approaches utilizing joint administration of prostaglandins and inhibitors of their synthesis at optimized timing and dosing of the drugs which could be incorporated into the therapy of patients with acute radiation syndrome.


Subject(s)
Acute Radiation Syndrome/metabolism , Hematopoiesis/drug effects , Prostaglandins/biosynthesis , Prostaglandins/pharmacology , Radiation-Protective Agents/pharmacology , Acute Radiation Syndrome/blood , Acute Radiation Syndrome/drug therapy , Acute Radiation Syndrome/etiology , Animals , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2 Inhibitors/therapeutic use , Disease Models, Animal , Humans , Metabolic Networks and Pathways/drug effects , Radiation-Protective Agents/therapeutic use
15.
Int Immunopharmacol ; 76: 105913, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31627170

ABSTRACT

Radiation exposure poses a significant threat to public health, which can lead to acute hematopoietic system and intestinal system injuries due to their higher radiation sensitivity. Hence, antioxidants and thiol-reducing agents could have a potential protective effect against this complication. The dithiol compound 1,4-dithiothreitol (DTT) has been used in biochemistry, peptide/protein chemistry and clinical medicine. However, the effect of DTT on ionizing radiation (IR)-induced hematopoietic injury and intestinal injury are unknown. The current investigation was designed to evaluate the effect of DTT as a safe and clinically applicable thiol-radioprotector in irradiated mice. DTT treatment improved the survival of irradiated mice and ameliorated whole body irradiation (WBI)-induced hematopoietic injury by attenuating myelosuppression and myeloid skewing, increasing self-renewal and differentiation of hematopoietic progenitor cells/hematopoietic stem cells (HPCs/HSCs). In addition, DTT treatment protected mice from abdominal irradiation (ABI)-induced changes in crypt-villus structures and function. Furthermore, treatment with DTT significantly enhanced the ABI-induced reduction in Olfm4 positive cells and offspring cells of Lgr5+ stem cells, including lysozyme+ Paneth cells and Ki67+ cells. Moreover, IR-induced DNA strand break damage, and the expression of proapoptotic-p53, Bax, Bak protein and antiapoptotic-Bcl-2 protein were reversed in DTT treated mice, and DTT also promoted small intestine repair after radiation exposure via the p53 intrinsic apoptotic pathway. In general, these results demonstrated the potential of DTT for protection against hematopoietic injury and intestinal injury after radiation exposure, suggesting DTT as a novel effective agent for radioprotection.


Subject(s)
Acute Radiation Syndrome/drug therapy , Dithiothreitol/therapeutic use , Radiation Injuries, Experimental/drug therapy , Radiation-Protective Agents/therapeutic use , Acute Radiation Syndrome/metabolism , Animals , Apoptosis/drug effects , DNA Damage/drug effects , Dithiothreitol/pharmacology , Gamma Rays/adverse effects , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/radiation effects , Intestine, Small/drug effects , Intestine, Small/metabolism , Intestine, Small/pathology , Intestine, Small/radiation effects , Male , Mice, Inbred C57BL , Proto-Oncogene Proteins c-bcl-2/metabolism , Radiation Injuries, Experimental/metabolism , Radiation-Protective Agents/pharmacology , Spleen/drug effects , Spleen/radiation effects , Thymus Gland/drug effects , Thymus Gland/radiation effects , Tumor Suppressor Protein p53/metabolism , Whole-Body Irradiation
16.
Sci Rep ; 9(1): 14134, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31575959

ABSTRACT

Radiation-induced intestinal injury (RIII) constitutes a crucial clinical element of acute radiation syndrome with life-threatening implications posing challenges in devising effective medical countermeasures. Herein, we report the potential of 7, 8-diacetoxy-4-methylthiocoumarin (DAMTC) to mitigate RIII following total-body irradiation (TBI) in C57BL/6 mice and underlying mechanisms. Administration of DAMTC 24 hours post TBI facilitated structural reconstitution and restoration of functional absorption linked to alleviation of radiation-induced apoptotic death of intestinal crypt progenitor/stem (ICPS) and villus stromal cells through induction of Bcl-2 family-mediated anti-apoptotic signalling. Reduction in TBI-induced DNA damage accumulation coupled with inhibition of cell cycle arrest through stimulation of anti-p53- and anti-p21-dependent synergistic signalling protected ICPS cells from radiation injury. Enhanced proliferation of crypt stem cells, induction of anti-oxidant defence, subjugation of TBI-induced lipid peroxidation and phenotypic polarization of intestinal macrophages to anti-inflammatory M2 class underlie amelioration of RIII. Stimulation of multiple mitigative signalling processes by DAMTC appeared to be associated with enhanced protein acetylation, an important regulator of cellular responses to radiation damage. Our findings establish the mitigative potential of DAMTC against RIII by hyper-acetylation-mediated epigenetic regulation, which triggers axes of anti-apoptotic and pro-survival pathways, enabling proliferation and maintenance of ICPS cells leading to epithelial regeneration.


Subject(s)
Abnormalities, Radiation-Induced/drug therapy , Acute Radiation Syndrome/drug therapy , Coumarins/pharmacology , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/radiation effects , Abnormalities, Radiation-Induced/metabolism , Acute Radiation Syndrome/metabolism , Animals , Cell Proliferation/drug effects , Female , Gastrointestinal Tract/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Lipid Peroxidation/drug effects , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/radiation effects , Whole-Body Irradiation/adverse effects
17.
Biol Blood Marrow Transplant ; 25(11): 2124-2133, 2019 11.
Article in English | MEDLINE | ID: mdl-31394269

ABSTRACT

In the setting of radiation-induced trauma, exposure to high levels of radiation can cause an acute radiation syndrome (ARS) causing bone marrow (BM) failure, leading to life-threatening infections, anemia, and thrombocytopenia. We have previously shown that human macrophages educated with human mesenchymal stem cells (MSCs) by coculture can significantly enhance survival of mice exposed to lethal irradiation. In this study, we investigated whether exosomes isolated from MSCs could replace direct coculture with MSCs to generate exosome educated macrophages (EEMs). Functionally unique phenotypes were observed by educating macrophages with exosomes from MSCs (EEMs) primed with bacterial lipopolysaccharide (LPS) at different concentrations (LPS-low EEMs or LPS-high EEMs). LPS-high EEMs were significantly more effective than uneducated macrophages, MSCs, EEMs, or LPS-low EEMs in extending survival after lethal ARS in vivo. Moreover, LPS-high EEMs significantly reduced clinical signs of radiation injury and restored hematopoietic tissue in the BM and spleen as determined by complete blood counts and histology. LPS-high EEMs showed significant increases in gene expression of STAT3, secretion of cytokines like IL-10 and IL-15, and production of growth factors like FLT-3L. LPS-EEMs also showed increased phagocytic activity, which may aid with tissue remodeling. LPS-high EEMs have the potential to be an effective cellular therapy for the management of ARS.


Subject(s)
Acute Radiation Syndrome/therapy , Exosomes/transplantation , Hematopoiesis , Macrophages/metabolism , Mesenchymal Stem Cells/metabolism , Radiation Injuries, Experimental/therapy , Acute Radiation Syndrome/metabolism , Acute Radiation Syndrome/pathology , Animals , Exosomes/metabolism , Exosomes/pathology , Female , Humans , Lipopolysaccharides/pharmacology , Macrophages/pathology , Male , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred NOD , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/pathology
18.
Naunyn Schmiedebergs Arch Pharmacol ; 392(12): 1537-1550, 2019 12.
Article in English | MEDLINE | ID: mdl-31350581

ABSTRACT

The protective role of α-tocopherol succinate (α-TCS) and the therapeutic efficacy of filgrastim were investigated in gastrointestinal acute radiation syndrome (GI-ARS) induced following 10 Gy whole-body γ-irradiation. Mice were randomly allocated into 5 groups: [1] normal-control, [2] irradiated-control, [3] subcutaneous (s.c.) injection of filgrastim (5 µg/kg/day) for 4 consecutive days given 1 h post-irradiation, [4] s.c. injection with α-TCS (400 mg/kg) 1 day prior to irradiation, [5] s.c. injection with α-TCS (400 mg/kg) 1 day prior to irradiation and filgrastim (5 µg/kg/day) for 4 consecutive days 1 h post-irradiation. Histopathological analysis, serum citrulline level, intestinal interleukin-1ß (IL-1ß), reduced glutathione (GSH), and malondialdehyde (MDA) contents as well as myeloperoxidase (MPO) activity were measured. Intestinal caspase-3, p53, cyclooxygenase-2 (COX-2), and inducible nitric oxide synthase (iNOS) immunopositivity were examined. In irradiated-control, MDA increased (249%) and GSH decreased (25%) compared to normal and were unaffected by filgrastim. α-TCS alone significantly reduced MDA (84.5%) and normalized GSH. The combination significantly reduced MDA (59%) and dramatically increased GSH (1573%), pointing to a possible synergistic action. In irradiated-control, MPO and IL-1ß significantly increased (111% and 613%, respectively) compared to normal-control and both were significantly decreased in all treated groups. Compared to normal-control, citrulline significantly declined (68%) in irradiated-control; a significant elevation was achieved by treatments with α-TCS alone or combined with filgrastim (88% and 94%, respectively). The combination therapy significantly decreased the degree of irradiation-induced injury of the epithelium and cellular infiltration and showed the lowest histopathological scoring compared to the other groups (p ≤ 0.05). In irradiated-control, immune-reactive expressions of iNOS, COX-2, caspase-3, and p53 were remarkable (18.62%, 34.27%, 31.19%, and 27.44%, respectively) and after combination therapy were reduced (1.04%, 22.39%, 8.76%, and 4.91%, respectively). The current findings represent a first-hand strategy in dealing with GI-ARS with a potential preference to using a combined therapy of filgrastim and α-TCS.


Subject(s)
Acute Radiation Syndrome/drug therapy , Antioxidants/therapeutic use , Filgrastim/therapeutic use , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/radiation effects , Hematologic Agents/therapeutic use , alpha-Tocopherol/therapeutic use , Acute Radiation Syndrome/metabolism , Acute Radiation Syndrome/pathology , Animals , Antioxidants/pharmacology , Caspase 3/metabolism , Cyclooxygenase 2/metabolism , Filgrastim/pharmacology , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/pathology , Hematologic Agents/pharmacology , Male , Mice , Nitric Oxide Synthase Type II/metabolism , Tumor Suppressor Protein p53/metabolism , Whole-Body Irradiation/adverse effects , alpha-Tocopherol/pharmacology
19.
Int J Mol Sci ; 20(5)2019 Mar 12.
Article in English | MEDLINE | ID: mdl-30870965

ABSTRACT

Genistein is a naturally occurring phytoestrogen isoflavone and is the active drug ingredient in BIO 300, a radiation countermeasure under advanced development for acute radiation syndrome (H-ARS) and for the delayed effects of acute radiation exposure (DEARE). Here we have assessed the pharmacokinetics (PK) and safety of BIO 300 in the nonhuman primate (NHP). In addition, we analyzed serum samples from animals receiving a single dose of BIO 300 for global metabolomic changes using ultra-performance liquid chromatography (UPLC) quadrupole time-of-flight mass spectrometry (QTOF-MS). We present a comparison of how either intramuscularly (im) or orally (po) administered BIO 300 changed the metabolomic profile. We observed transient alterations in phenylalanine, tyrosine, glycerophosphocholine, and glycerophosphoserine which reverted back to near-normal levels 7 days after drug administration. We found a significant overlap in the metabolite profile changes induced by each route of administration; with the po route showing fewer metabolic alterations. Taken together, our results suggest that the administration of BIO 300 results in metabolic shifts that could provide an overall advantage to combat radiation injury. This initial assessment also highlights the utility of metabolomics and lipidomics to determine the underlying physiological mechanisms involved in the radioprotective efficacy of BIO 300.


Subject(s)
Genistein/administration & dosage , Genistein/pharmacokinetics , Metabolome/drug effects , Nanoparticles/administration & dosage , Suspensions/administration & dosage , Suspensions/pharmacokinetics , Acute Radiation Syndrome/drug therapy , Acute Radiation Syndrome/metabolism , Animals , Chromatography, High Pressure Liquid/methods , Female , Genistein/adverse effects , Macaca mulatta , Male , Metabolomics/methods , Nanoparticles/adverse effects , Nanoparticles/metabolism , Primates , Suspensions/adverse effects
20.
Radiat Res ; 191(5): 383-397, 2019 05.
Article in English | MEDLINE | ID: mdl-30901530

ABSTRACT

We have previously shown significant pathology in the heart and kidney of murine hematopoietic-acute radiation syndrome (H-ARS) survivors of 8.7-9.0 Gy total-body irradiation (TBI). The goal of this study was to determine temporal relationships in the development of vasculopathy and the progression of renal and cardiovascular delayed effects of acute radiation exposure (DEARE) at TBI doses less than 9 Gy and to elucidate the potential roles of senescence, inflammation and oxidative stress. Our results show significant loss of endothelial cells in coronary arteries by 4 months post-TBI (8.53 or 8.72 Gy of gamma radiation). This loss precedes renal dysfunction and interstitial fibrosis and progresses to abnormalities in the arterial media and adventitia and loss of coronary arterioles. Major differences in radiation-induced pathobiology exist between the heart and kidney in terms of vasculopathy progression and also in indices of inflammation, senescence and oxidative imbalance. The results of this work suggest a need for different medical countermeasures for multiple targets in different organs and at various times after acute radiation injury to prevent the progression of DEARE.


Subject(s)
Acute Radiation Syndrome/metabolism , Acute Radiation Syndrome/pathology , Blood Vessels/radiation effects , Cellular Senescence/radiation effects , Oxidative Stress/radiation effects , Acute Radiation Syndrome/physiopathology , Animals , Cell Count , Disease Progression , Dose-Response Relationship, Radiation , Female , Heart/radiation effects , Inflammation/etiology , Kidney/metabolism , Kidney/pathology , Kidney/radiation effects , Male , Mice , Mice, Inbred C57BL , Myocardium/metabolism , Myocardium/pathology , Organ Specificity , Time Factors , Whole-Body Irradiation/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...