Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
JCI Insight ; 6(8)2021 04 22.
Article in English | MEDLINE | ID: mdl-33724956

ABSTRACT

After 9/11, threat of nuclear attack on American urban centers prompted government agencies to develop medical radiation countermeasures to mitigate hematopoietic acute radiation syndrome (H-ARS) and higher-dose gastrointestinal acute radiation syndrome (GI-ARS) lethality. While repurposing leukemia drugs that enhance bone marrow repopulation successfully treats H-ARS in preclinical models, no mitigator potentially deliverable under mass casualty conditions preserves GI tract. Here, we report generation of an anti-ceramide 6B5 single-chain variable fragment (scFv) and show that s.c. 6B5 scFv delivery at 24 hours after a 90% lethal GI-ARS dose of 15 Gy mitigated mouse lethality, despite administration after DNA repair was complete. We defined an alternate target to DNA repair, an evolving pattern of ceramide-mediated endothelial apoptosis after radiation, which when disrupted by 6B5 scFv, initiates a durable program of tissue repair, permitting crypt, organ, and mouse survival. We posit that successful preclinical development will render anti-ceramide 6B5 scFv a candidate for inclusion in the Strategic National Stockpile for distribution after a radiation catastrophe.


Subject(s)
Acute Radiation Syndrome/drug therapy , Ceramides/immunology , Gastrointestinal Diseases/drug therapy , Intestine, Small/drug effects , Intestine, Small/radiation effects , Single-Chain Antibodies/pharmacology , Acute Radiation Syndrome/mortality , Animals , DNA Repair , Gastrointestinal Diseases/mortality , Humans , Injections, Subcutaneous , Intestine, Small/pathology , Jurkat Cells/drug effects , Jurkat Cells/radiation effects , Mice , Single-Chain Antibodies/therapeutic use
2.
Clin Transl Sci ; 13(4): 807-817, 2020 07.
Article in English | MEDLINE | ID: mdl-32112517

ABSTRACT

Acute exposure to high doses of radiation leads to severe myelosuppression, but few treatments are currently available to treat hematopoietic syndrome of acute radiation syndrome. Granulocyte colony stimulating factors (e.g., filgrastim) stimulate proliferation of neutrophil precursors and enhance mature neutrophil function. Owing to ethical constraints on conducting clinical research in lethally irradiated humans, we developed a model-based strategy to integrate preclinical experience in irradiated nonhuman primates (NHPs) and other clinical myelosuppressive conditions to inform filgrastim dosing to treat hematopoietic syndrome of acute radiation syndrome. Models predicting neutrophil counts and overall survival based on drug exposures were calibrated and scaled from NHPs to adult and pediatric human subjects. Several scenarios were examined investigating variations in filgrastim doses, dose frequency, treatment initiation, and duration, as well as the effect of age and radiation dose rate. Model-based simulations and established safety profiles supported that a subcutaneous filgrastim dose of 10 µg/kg once daily provides a significant survival benefit (50%) over placebo in both adults and children, provided that the treatment is initiated within 1-14 days after radiation exposure and lasts 2-3 weeks. For treatment durations of longer than 3 weeks, filgrastim treatment is not expected to provide significantly greater benefit. This survival benefit is expected to hold for the wide range of radiation doses and dose rates (0.01-1,000 Gy/hours) examined.


Subject(s)
Acute Radiation Syndrome/drug therapy , Filgrastim/administration & dosage , Hematologic Agents/administration & dosage , Acute Radiation Syndrome/mortality , Adult , Age Factors , Animals , Child , Computer Simulation , Disease Models, Animal , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Drug Evaluation, Preclinical , Female , Granulocyte Precursor Cells/drug effects , Humans , Injections, Subcutaneous , Macaca mulatta , Male , Myelopoiesis/drug effects , Risk Assessment/methods , Treatment Outcome
3.
Health Phys ; 119(3): 351-357, 2020 09.
Article in English | MEDLINE | ID: mdl-31934930

ABSTRACT

Studies performed decades ago in the canine and nonhuman primate established the dose response relationships for the hematopoietic acute radiation syndrome in response to mixed neutron/gamma, x-radiation, and Co gamma radiation. There were no published studies that determined the dose response relationships for the gastrointestinal acute radiation syndrome in response to either noted radiation quality. This analysis of a retrospective, unpublished study provided the dose response relationships in a canine model for the acute gastrointestinal syndrome relative to the acute hematopoietic syndrome due to mixed neutron/gamma radiation. Canines were exposed to total-body, steady state, bilateral, 0.40 Gy min, mixed neutron/gamma (5.4:1) radiation from a TRIGA reactor. The average neutron/gamma energy (MeV) was 0.85/0.9, and exposure was reported as midline tissue dose. Medical management was not administered. The mixed neutron/gamma exposure resulted in an estimated LD50/6 of 2.83 Gy [2.76, 2.94] and LD50/30 of 2.16 Gy [2.01, 2.24] for the GI- and H-ARS respectively. The mean survival times for decedents after mixed neutron/gamma exposure approximate to the LD50/6 were 8.5 d, 10.5 d, and 4 d for 2.75 Gy, 2.80 Gy, 3.00, and 3.12 Gy exposures, respectively. The mean survival times for decedents for mixed neutron/gamma exposure approximate to the LD50/30 were 21.3 d and 15.6 d for 2.00 Gy and 2.25 Gy, respectively. Furthermore, the dose response relationships for the acute hematopoietic syndrome due to mixed neutron/gamma exposure (0.85/0.9 MeV; 5.4:1) resulted in an estimated relative biological effectiveness of 1.2 as compared with reference Co gamma radiation.


Subject(s)
Acute Radiation Syndrome/etiology , Cobalt Radioisotopes/adverse effects , Gamma Rays/adverse effects , Acute Radiation Syndrome/mortality , Animals , Cobalt Radioisotopes/administration & dosage , Disease Models, Animal , Dogs , Dose-Response Relationship, Radiation , Female , Male , Radiation Injuries, Experimental/etiology , Retrospective Studies , Whole-Body Irradiation/adverse effects , Whole-Body Irradiation/mortality
4.
Health Phys ; 116(4): 546-557, 2019 04.
Article in English | MEDLINE | ID: mdl-30789496

ABSTRACT

Accurate analyses of the delayed effects of acute radiation exposure in survivors of the hematopoietic acute radiation syndrome are hampered by low numbers of mice for examination due to high lethality from the acute syndrome, increased morbidity and mortality in survivors, high cost of husbandry for long-term studies, biological variability, and inconsistencies of models from different laboratories complicating meta-analyses. To address this, a compilation of 38 similar hematopoietic acute radiation syndrome studies conducted over a 7-y period in the authors' laboratory, comprising more than 1,500 irradiated young adult C57BL/6 mice and almost 600 day-30 survivors, was assessed for hematopoietic delayed effects of acute radiation exposure at various times up to 30 mo of age. Significant loss of long-term repopulating potential of phenotypically defined primitive hematopoietic stem cells was documented in hematopoietic acute radiation syndrome survivors, as well as significant decreases in all hematopoietic lineages in peripheral blood, prominent myeloid skew, significantly decreased bone marrow cellularity, and numbers of lineage-negative Sca-1+ cKit+ CD150+ cells (KSL CD150+; the phenotype known to be enriched for hematopoietic stem cells), and increased cycling of KSL CD150+ cells. Studies interrogating the phenotype of bone marrow cells capable of initiation of suspension cultures and engraftment in competitive transplantation assays documented the phenotype of hematopoietic stem cells in hematopoietic acute radiation syndrome survivors to be the same as that in nonirradiated age-matched controls. This compilation study adds rigor and validity to our initial findings of persistent hematopoietic dysfunction in hematopoietic acute radiation syndrome survivors that arises at the level of the hematopoietic stem cell and which affects all classes of hematopoietic cells for the life of the survivor.


Subject(s)
Acute Radiation Syndrome/mortality , Bone Marrow/radiation effects , Hematopoiesis/radiation effects , Radiation Injuries, Experimental/mortality , Acute Radiation Syndrome/pathology , Animals , Bone Marrow/pathology , Cell Cycle/radiation effects , Female , Male , Mice , Mice, Inbred C57BL , Radiation Injuries, Experimental/pathology
5.
Radiat Res ; 191(4): 323-334, 2019 04.
Article in English | MEDLINE | ID: mdl-30730284

ABSTRACT

Intensive research is underway to find new agents that can successfully mitigate the acute effects of radiation exposure. This is primarily in response to potential counterthreats of radiological terrorism and nuclear accidents but there is some hope that they might also be of value for cancer patients treated with radiation therapy. Research into mitigation countermeasures typically employs classic animal models of acute radiation syndromes (ARS) that develop after whole-body irradiation (WBI). While agents are available that successfully mitigate ARS when given after radiation exposure, their success raises questions as to whether they simply delay lethality or unmask potentially lethal radiation pathologies that may appear later in time. Life shortening is a well-known consequence of WBI in humans and experimental animals, but it is not often examined in a mitigation setting and its causes, other than cancer, are not well-defined. This is in large part because delayed effects of acute radiation exposure (DEARE) do not follow the strict time-dose phenomena associated with ARS and present as a diverse range of symptoms and pathologies with low mortality rates that can be evaluated only with the use of large cohorts of subjects, as in this study. Here, we describe chronically increased mortality rates up to 660 days in large numbers of mice given LD70/30 doses of WBI. Systemic myeloid cell activation after WBI persists in some mice and is associated with late immunophenotypic changes and hematopoietic imbalance. Histopathological changes are largely of a chronic inflammatory nature and variable incidence, as are the clinical symptoms, including late diarrhea that correlates temporally with changes in the content of the microbiome. We also describe the acute and long-term consequences of mitigating hematopoietic ARS (H-ARS) lethality after LD70/30 doses of WBI in multiple cohorts of mice treated uniformly with radiation mitigators that have a common 4-nitro-phenylsulfonamide (NPS) pharmacophore. Effective NPS mitigators dramatically decrease ARS mortality. There is slightly increased subacute mortality, but the rate of late mortalities is slowed, allowing some mice to live a normal life span, which is not the case for WBI controls. The study has broad relevance to radiation late effects and their potential mitigation and epitomizes the complex interaction between radiation-damaged tissues and immune homeostasis.


Subject(s)
Acute Radiation Syndrome/immunology , Acute Radiation Syndrome/prevention & control , Hematopoietic System/drug effects , Hematopoietic System/radiation effects , Radiation-Protective Agents/pharmacology , Acute Radiation Syndrome/microbiology , Acute Radiation Syndrome/mortality , Animals , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/radiation effects , Heart/drug effects , Heart/radiation effects , Male , Mice , Neoplasms, Radiation-Induced/immunology , Neoplasms, Radiation-Induced/microbiology , Neoplasms, Radiation-Induced/mortality , Neoplasms, Radiation-Induced/prevention & control , Sulfonamides/pharmacology , Survival Analysis
6.
Sci Rep ; 9(1): 2198, 2019 02 18.
Article in English | MEDLINE | ID: mdl-30778109

ABSTRACT

The increasing potential for accidental radiation exposure from either nuclear accidents or terrorist activities has escalated the need for radiation countermeasure development. We previously showed that a 30-day course of high-dose captopril, an ACE inhibitor, initiated 1-4 h after total body irradiation (TBI), improved Hematopoietic Acute Radiation Syndrome (H-ARS) and increased survival in mice. However, because of the time likely required for the deployment of a stockpiled radiation countermeasure to a radiation mass casualty site, there is a need for therapies that can be administered 24-48 hours after initial exposure. Using C57BL/6 mice exposed to an LD50-80/30 of 60Co TBI (7.75-7.9 Gy, 0.615 Gy/min), we show that low-dose captopril administration, initiated as late as 48 h post-TBI and continued for 14 days, significantly enhanced overall survival similarly to high-dose, rapid administration. Captopril treatment did not affect radiation-induced cell cycle arrest genes or the immediate loss of hematopoietic precursors. Reduced mortality was associated with the recovery of bone marrow cellularity and mature blood cell recovery at 21-30 days post-irradiation. Captopril reduced radiation-induced cytokines EPO, G-CSF, and SAA in the plasma. Finally, delayed captopril administration mitigated brain micro-hemorrhage at 21 days post-irradiation. These data indicate that low dose captopril administered as late as 48 h post-TBI for only two weeks improves survival that is associated with hematopoietic recovery and reduced inflammatory response. These data suggest that captopril may be an ideal countermeasure to mitigate H-ARS following accidental radiation exposure.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Captopril/administration & dosage , Hematopoiesis/drug effects , Hematopoiesis/radiation effects , Radiation-Protective Agents/administration & dosage , Whole-Body Irradiation , Acute Radiation Syndrome/blood , Acute Radiation Syndrome/etiology , Acute Radiation Syndrome/mortality , Acute Radiation Syndrome/prevention & control , Animals , Blood Cell Count , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cytokines/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/radiation effects , Inflammation Mediators/metabolism , Mice , Radiation Dosage , Radiation Exposure , Time-to-Treatment , Whole-Body Irradiation/adverse effects
7.
Health Phys ; 116(4): 529-545, 2019 04.
Article in English | MEDLINE | ID: mdl-30624354

ABSTRACT

Our goal is to develop lisinopril as a mitigator of delayed effects of acute radiation exposure in the National Institute of Allergy and Infectious Diseases program for radiation countermeasures. Published studies demonstrated mitigation of delayed effects of acute radiation exposure by lisinopril in adult rats. However, juvenile or old rats beyond their reproductive lifespans have never been tested. Since no preclinical models of delayed effects of acute radiation exposure were available in these special populations, appropriate rat models were developed to test lisinopril after irradiation. Juvenile (42-d-old, prepubertal) female and male WAG/RijCmcr (Wistar) rats were given 13-Gy partial-body irradiation with only part of one hind limb shielded. Lethality from lung injury between 39-58 d and radiation nephropathy between 106-114 d were recorded. All irradiated-only juvenile rats were morbid from delayed effects of acute radiation exposure by 114 d, while lisinopril (24 mg m d) started 7 d after irradiation and continued improved survival to 88% (p = 0.0015, n ≥ 8/group). Old rats (>483-d-old, reproductively senescent) were irradiated with 13-Gy partial-body irradiation keeping part of one leg shielded and additionally shielding the head in some animals. Irradiated old females developed lethal nephropathy, and all became morbid by 170 d after irradiation, though no rats displayed lethal radiation pneumonitis. Similar results were observed for irradiated geriatric males, though 33% of rats remained alive at 180 d after irradiation. Lisinopril mitigated radiation nephropathy in old rats of both sexes. Finally, comparison of delayed effects of acute radiation exposure between irradiated juvenile, adult, and old rats showed younger rats were more sensitive to delayed effects of acute radiation exposure with earlier manifestation of injuries to some organs.


Subject(s)
Lisinopril/therapeutic use , Radiation Injuries, Experimental/drug therapy , Radiation-Protective Agents/therapeutic use , Acute Radiation Syndrome/drug therapy , Acute Radiation Syndrome/mortality , Acute Radiation Syndrome/pathology , Acute Radiation Syndrome/prevention & control , Age Factors , Animals , Female , Male , Medical Countermeasures , Radiation Injuries, Experimental/mortality , Radiation Injuries, Experimental/pathology , Radiation Injuries, Experimental/prevention & control , Rats , Rats, Wistar , Time Factors
8.
Health Phys ; 116(3): 339-353, 2019 03.
Article in English | MEDLINE | ID: mdl-30281533

ABSTRACT

A nonhuman primate model of acute, partial-body, high-dose irradiation with minimal (2.5%) bone marrow sparing was used to assess endogenous gastrointestinal and hematopoietic recovery and the ability of Neulasta (pegylated granulocyte colony-stimulating factor) or Neupogen (granulocyte colony-stimulating factor) to enhance recovery from myelosuppression when administered at an increased interval between exposure and initiation of treatment. A secondary objective was to assess the effect of Neulasta or Neupogen on mortality and morbidity due to the hematopoietic acute radiation syndrome and concomitant gastrointestinal acute radiation syndrome. Nonhuman primates were exposed to 10.0 Gy, 6 MV, linear accelerator-derived photons delivered at 0.80 Gy min. All nonhuman primates received subject-based medical management. Nonhuman primates were dosed daily with control article (5% dextrose in water), initiated on day 1 postexposure; Neulasta (300 µg kg), administered on days 1, 8, and 15 or days 3, 10, and 17 postexposure; or Neupogen (10 µg kg), administered daily postexposure following its initiation on day 1 or day 3 until neutrophil recovery (absolute neutrophil count ≥1,000 cells µL for 3 consecutive days). Mortality in the irradiated cohorts suggested that administration of Neulasta or Neupogen on either schedule did not affect mortality due to gastrointestinal acute radiation syndrome or mitigate mortality due to hematopoietic acute radiation syndrome (plus gastrointestinal damage). Following 10.0 Gy partial-body irradiation with 2.5% bone marrow sparing, the mean duration of neutropenia (absolute neutrophil count <500 cells µL) was 22.4 d in the control cohort vs. 13.0 and 15.3 d in the Neulasta day 1, 8, 15 and day 3, 10, 17 cohorts, relative to 16.2 and 17.4 d in the Neupogen cohorts initiated on day 1 and day 3, respectively. The absolute neutrophil count nadirs were 48 cells µL in the controls; 117 cells µL and 40 cells µL in the Neulasta days 1, 8, and 15 or days 3, 10, and 17 cohorts, respectively; and 75 cells µL and 37 cells µL in the Neupogen day 1 and day 3 cohorts, respectively. Therefore, the earlier administration of Neulasta or Neupogen was more effective in this model of marginal 2.5% bone marrow sparing. The approximate 2.5% bone marrow sparing may approach the threshold for efficacy of the lineage-specific medical countermeasure. The partial-body irradiation with 2.5% bone marrow sparing model can be used to assess medical countermeasure efficacy in the context of the concomitant gastrointestinal and hematopoietic acute radiation syndrome sequelae.


Subject(s)
Acute Radiation Syndrome/drug therapy , Bone Marrow/radiation effects , Filgrastim/therapeutic use , Gastrointestinal Tract/radiation effects , Hematinics/therapeutic use , Hematopoiesis/radiation effects , Polyethylene Glycols/therapeutic use , Acute Radiation Syndrome/mortality , Animals , Bone Marrow/drug effects , Gastrointestinal Tract/drug effects , Hematopoiesis/drug effects , Macaca mulatta , Male
9.
Radiat Res ; 191(1): 107-121, 2019 01.
Article in English | MEDLINE | ID: mdl-30430918

ABSTRACT

The development of effective biomarkers for detecting the magnitude of radiation exposure and resiliency of host response is crucial to identifying appropriate treatment strategies after radiation exposure. We hypothesized that the gastrointestinal resident bacteria would demonstrate predictable, dose-dependent changes after radiation exposure across two large animal models of acute radiation syndrome. Here, Göttingen minipigs (GMP) (n = 50) and rhesus macaques (n = 48) were exposed to five dose levels (resulting in mortality rates of 33-100% and 25-68.7%, respectively). Fecal samples taken prior to and after irradiation (day 0 for GMP; day 0, 3 and 14 for macaques) were used for 16S rRNA gene sequence amplicon high-throughput sequencing. Baseline gut microbiota profiles were dissimilar between GMP and macaques, however, radiation appeared to have similar effect at the phylum level, resulting in Bacteroidetes decrease and Firmicutes increase in both models. The abundance of the main Bacteroidetes genus ( Bacteroides for GMP, Prevotella for macaques) was profoundly decreased by irradiation. Intracellular symbionts [Elusimicrobia in GMP, Treponema (Spirochaetes) in macaques] consistently increased after irradiation, suggesting their use as potential biomarkers of intestinal injury, and potential negative effect on health. Prevotella, Lactobacillus, Clostridium XIVa, Oscillibacter and Elusimicrobium/ Treponema abundances were found to be very significantly correlated with radiation intensity. Furthermore, Prevotella, Enterorhabdus and Ruminococcus and Enterorhabdus maintenance was strongly associated with survival in GMP, while Prevotella, Oscillibacter and Treponema were strongly associated with survival and Streptococcus with death in macaques. Overall, we found that a wide range of gut bacterial genera known to be abundant in the human gut microbiota are excellent biomarkers of radiation intensity and resilience in animal models, and that detrimental effects can be monitored, and potentially prevented, by targeting selected genera.


Subject(s)
Acute Radiation Syndrome/mortality , Gastrointestinal Microbiome , Models, Animal , Radiation Dosage , Acute Radiation Syndrome/etiology , Animals , Biomarkers/metabolism , High-Throughput Nucleotide Sequencing , Humans , Macaca mulatta , RNA, Ribosomal, 16S/genetics , Reproducibility of Results , Swine , Swine, Miniature
10.
Health Phys ; 115(1): 161-169, 2018 07.
Article in English | MEDLINE | ID: mdl-29787442

ABSTRACT

This article summarizes the results of 30 y of follow-up of cancer and noncancer effects in Ukrainian cleanup workers after the Chornobyl accident. The number of power plant employees and first responders with acute radiation syndrome under follow-up by the National Research Center for Radiation Medicine decreased from 179 in 1986-1991 to 105 in 2011-2015. Cancers and leukemia (19) and cardiovascular diseases (21) were the main causes of deaths among acute radiation syndrome survivors (54) during the postaccident period. Increased radiation risks of leukemia in the Ukrainian cohort of 110,645 cleanup workers exposed to low doses are comparable to those among survivors of the atomic bomb explosions in Japan in 1945. Additionally, an excess of chronic lymphocytic leukemia was demonstrated in the cleanup workers cohort for 26 y after the exposure. A significant excess of multiple myeloma incidence [standardized incidence rate (SIR) 1.61 %, 95% confidence interval (CI) 1.01-2.21], thyroid cancer (SIR 4.18, 95% CI 3.76-4.59), female breast cancer (SIR 1.57 CI 1.40-1.73), and all cancers combined (SIR 1.07; 95% CI 1.05-1.09) was registered. High prevalence was demonstrated for cardio- and cerebrovascular diseases and mental health changes. However, the reasons for the increases require further investigation. To monitor other possible late effects of radiation exposure in Chornobyl cleanup workers, analytical cohort and case-control studies need to include cardiovascular pathology, specifically types of potentially radiogenic cancers using a molecular epidemiology approach. Possible effects for further study include increased rates of thyroid, breast, and lung cancers and multiple myeloma; reduction of radiation risks of leukemia to population levels; and increased morbidity and mortality of cleanup workers from cardio- and cerebrovascular pathology.


Subject(s)
Acute Radiation Syndrome/epidemiology , Chernobyl Nuclear Accident , Neoplasms, Radiation-Induced/epidemiology , Occupational Diseases/epidemiology , Occupational Exposure/adverse effects , Population Surveillance , Radiation Exposure/adverse effects , Acute Radiation Syndrome/diagnosis , Acute Radiation Syndrome/etiology , Acute Radiation Syndrome/mortality , Adolescent , Adult , Aged , Case-Control Studies , Cohort Studies , Female , Humans , Incidence , Male , Middle Aged , Neoplasms, Radiation-Induced/diagnosis , Neoplasms, Radiation-Induced/etiology , Neoplasms, Radiation-Induced/mortality , Occupational Diseases/diagnosis , Occupational Diseases/etiology , Occupational Diseases/mortality , Radiation Dosage , Risk Assessment , Risk Factors , Ukraine/epidemiology , Young Adult
11.
Sci Rep ; 8(1): 1302, 2018 01 22.
Article in English | MEDLINE | ID: mdl-29358747

ABSTRACT

Exposure to high-doses of ionizing radiation (IR) leads to development of a strong acute radiation syndrome (ARS) in mammals. ARS manifests after a latency period and it is important to develop fast prognostic biomarkers for its early detection and assessment. Analysis of chromosomal aberrations in peripheral blood lymphocytes is the gold standard of biological dosimetry, but it fails after high doses of IR. Therefore, it is important to establish novel biomarkers of exposure that are fast and reliable also in the high dose range. Here, we investigated the applicability of miRNA levels in mouse serum. We found significantly increased levels of miR-375-3p following whole body exposure to 7 Gy of X-rays. In addition, we analyzed their levels in various organs of control mice and found them to be especially abundant in the pancreas and the intestine. Following a dose of 7 Gy, extensive cell death occurred in these tissues and this correlated negatively with the levels of miR-375-3p in the organs. We conclude that high expressing tissues of miR-375-3p may secrete this miRNA in serum following exposure to 7 Gy. Therefore, elevated miR-375-3p in serum may be a predictor of tissue damage induced by exposure to a high radiation dose.


Subject(s)
Acute Radiation Syndrome/diagnosis , Chromosome Aberrations/radiation effects , MicroRNAs/genetics , X-Rays/adverse effects , Acute Radiation Syndrome/blood , Acute Radiation Syndrome/etiology , Acute Radiation Syndrome/mortality , Animals , Biomarkers/blood , Disease Models, Animal , Dose-Response Relationship, Radiation , Humans , Lymphocytes/metabolism , Lymphocytes/pathology , Lymphocytes/radiation effects , Male , Mice , Mice, Inbred C57BL , MicroRNAs/blood , Survival Analysis , Whole-Body Irradiation
12.
Int J Radiat Oncol Biol Phys ; 96(3): 629-36, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27681759

ABSTRACT

PURPOSE: After radiation therapy (RT), various radiation-induced toxicities can develop in about one-fourth of patients. An international interest in using morbidity and mortality rates to monitor the quality of care and integrate morbidity and mortality review (MMR) meetings into organizations' governance processes has arisen. We report the first results of patients included in our MMR procedure that included biological assays for individual intrinsic radiosensitivity (IIRS). METHODS AND MATERIALS: Twenty-three patients were prospectively included in the MMR database. Twenty-two were evaluable for IIRS. Prostate (n=10) and breast (n=8) cancers were the most frequent disease types. The total dose delivered, determined according to the type of disease, ranged from 30 to 74 Gy. Our MMR procedure requires strict criteria: patients with unresolved toxicity of grade 3 or higher with availability of clinical (photographic) data, IIRS results obtained from skin biopsy assays, treatment modalities, and follow-up data. The RT technique and dosimetry were reviewed. RESULTS: Our prospective registration of toxicities showed mainly rectitis, occurring in 7 cases, and skin toxicities, occurring in 9. Of the 7 patients with rectitis, 5 received 66 Gy of post-prostatectomy RT with V50 (rectum volume receiving 50 Gy) ranging from 45% to 75% and a mean maximal dose of 66.5 Gy. For dermatitis and cystitis, the mean maximal doses were in the range of classical constraints without any overdosage or dose heterogeneity. No errors were found in the review of treatment planning and positioning. Conversely, all the patients were considered biologically as radiosensitive with genomic instability and ATM (ataxia telangiectasia mutated)-dependent DNA double-strand break repair impairments. CONCLUSIONS: The MMR review of files allowed clear answers for patients on the relationship between clinical events and their IIRS. Our procedure has allowed education of all our staff to monitor, identify, and document clinical, physical, and biological aspects of radiation-induced toxicities. Thus we recommend the introduction of the MMR procedure in RT departments.


Subject(s)
Acute Radiation Syndrome/mortality , Dose-Response Relationship, Radiation , Radiation Monitoring/statistics & numerical data , Radiation Tolerance , Radiotherapy/mortality , Registries , Adult , Aged , Aged, 80 and over , Female , France/epidemiology , Humans , Incidence , Male , Middle Aged , Prospective Studies , Radiotherapy/statistics & numerical data , Risk Factors , Survival Analysis
13.
Purinergic Signal ; 11(1): 79-85, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25358454

ABSTRACT

Adenosine A3 receptor knockout (A3AR KO) mice and their wild-type (WT) counterparts were compared from the point of view of their abilities to survive exposures to lethal doses of γ-radiation belonging to the range of radiation doses inducing the bone marrow acute radiation syndrome. Parameters of cumulative 30-day survival (experiment using a midlethal radiation dose) or cumulative 11-day survival (experiment using an absolutely lethal radiation dose), and of mean survival time were evaluated. The values of A3AR KO mice always reflected their higher survival in comparison with WT ones, the P values being above the limit for statistical significance after the midlethal radiation dose and standing for statistical significance after the absolutely lethal radiation dose. This finding was considered surprising, taking into account the previously obtained findings on defects in numbers and functional properties of peripheral blood cells in A3AR KO mice. Therefore, previous hematological analyses of A3AR KO mice were supplemented in the present studies with determination of serum levels of the granulocyte colony-stimulating factor, erythropoietin, and thrombopoietin. Though distinct differences in these parameters were observed between A3AR KO and WT mice, none of them could explain the relatively high postirradiation survival of A3AR KO mice. Further studies on these mice comprising also those on other than hemopoietic tissues and organs can help to clarify their relative radioresistance.


Subject(s)
Acute Radiation Syndrome/mortality , Receptor, Adenosine A3/genetics , Acute Radiation Syndrome/genetics , Acute Radiation Syndrome/metabolism , Animals , Mice , Mice, Knockout , Receptor, Adenosine A3/metabolism , Survival Rate
14.
Bull Exp Biol Med ; 156(6): 776-7, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24824694

ABSTRACT

We studied the effect of long-term administration of melatonin to male C57Bl/6 mice starting from day 3 after whole-body γ-irradiation (9.5-10.0 Gy, 7.7-17.1 cGy/min). It was found that replacement of drinking water with melatonin solution (5 mg/liter) did not reduce the amount of fluid intake throughout the period of acute radiation injury. The daily dose of melatonin was 0.9-1.2 mg/kg body weight (this parameter was lower at the peak of the disease and increased during the recovery stage). Melatonin by more than 20% (p<0.05) improved survival of mice exposed to γ-irradiation in a dose of LD97/30, reduced leukopenia during the stage of acute manifestations of the disease and maximum mortality, and increased blood leukocyte count by 40% (p<0.05) by day 12 after irradiation.


Subject(s)
Acute Radiation Syndrome/drug therapy , Antioxidants/therapeutic use , Melatonin/therapeutic use , Radiation-Protective Agents/therapeutic use , Acute Radiation Syndrome/mortality , Animals , Gamma Rays/adverse effects , Leukocyte Count , Male , Mice , Mice, Inbred C57BL , Whole-Body Irradiation/adverse effects
15.
PLoS One ; 8(11): e78227, 2013.
Article in English | MEDLINE | ID: mdl-24223778

ABSTRACT

A protective reagent for ARI should have the ability to repair injured tissue caused by radiation and prevent continuous damage from secondary risk factors. Trx-1 was explored as a candidate therapy for ARI, as it scavenges reactive oxygen species, regulates cell growth and differentiation, participates in immune reactions, and inhibits apoptosis by acting inside and/or outside cells. Trx-1 can also decrease excessive inflammation in ARI by regulating the creation of inflamed media, by inhibiting the activation of complement, and by reducing the chemotaxis, adhesion, and migration of inflammatory cells. As effectively and stably expressing exogenous genes in the long term and regulating immune inflammation and tissue repair, MSC are a good choice for Trx-1 gene therapy. In this study, Trx-1-overexpressing hucMSC-Trx-1 were obtained by adenoviral vector-mediated infection. We first measured the redox capacity of hucMSC-Trx-1 with an antioxidant capacity (T-AOC) assay, a hydrogen peroxide (H2O2) content determination assay in vivo, a H2O2-induced oxidation hemolysis assay, and a lipid peroxidation assay in vitro. Then, we measured survival time, the protection of the hematopoietic system, and the regulation of inflammation in important organs in three treatment groups of NOD/SCID mice (treated with hucMSC-Trx-1, with hucMSC, and with saline) that were exposed to 4.5 Gy (60)Co-γ-ray radiation. The hucMSC-Trx-1 group achieved superior antioxidation results, protecting bone marrow hematopoietic stem cells (Lin(-)CD117(+): hucMSC-Trx-1 vs. hucMSC, P<0.05; hucMSC-Trx-1 vs. NS, P<0.01), promoting the formation of red blood cells and hemoglobin (hucMSC-Trx-1 vs. hucMSC or NS, P<0.05), reducing inflammation and damage in important organs (Bone marrow and lung: hucMSC-Trx-1 vs. NS, P<0.01; hucMSC-Trx-1 vs. hucMSC, P<0.05. Liver and intestine: hucMSC-Trx-1 vs. NS, P<0.05; hucMSC-Trx-1 vs. hucMSC, P<0.05), and prolonging survival (hucMSC-Trx-1 vs. hucMSC or NS, P<0.01). Therefore, hucMSC-Trx-1 combines the merits of gene and cell therapy as a multifunctional radioprotector for ARI.


Subject(s)
Acute Radiation Syndrome/therapy , Cell- and Tissue-Based Therapy/methods , Mesenchymal Stem Cell Transplantation , Thioredoxins/genetics , Transgenes , Acute Radiation Syndrome/metabolism , Acute Radiation Syndrome/mortality , Acute Radiation Syndrome/pathology , Adenoviridae/genetics , Adenoviridae/metabolism , Animals , Antioxidants/metabolism , Bone Marrow/metabolism , Bone Marrow/pathology , Fetal Blood/cytology , Fetal Blood/metabolism , Genetic Therapy , Genetic Vectors , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Inflammation/therapy , Intestinal Mucosa/metabolism , Intestines/pathology , Liver/metabolism , Liver/pathology , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Oxidation-Reduction , Survival Analysis , Thioredoxins/metabolism , Whole-Body Irradiation
16.
Int J Radiat Oncol Biol Phys ; 86(5): 986-92, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23845847

ABSTRACT

PURPOSE: We are characterizing the Gottingen minipig as an additional large animal model for advanced drug testing for the acute radiation syndrome (ARS) to enhance the discovery and development of novel radiation countermeasures. Among the advantages provided by this model, the similarities to human hematologic parameters and dynamics of cell loss/recovery after irradiation provide a convenient means to compare the efficacy of drugs known to affect bone marrow cellularity and hematopoiesis. METHODS AND MATERIALS: Male Gottingen minipigs, 4 to 5 months old and weighing 9 to 11 kg, were used for this study. We tested the standard off-label treatment for ARS, rhG-CSF (Neupogen, 10 µg/kg/day for 17 days), at the estimated LD70/30 total-body γ-irradiation (TBI) radiation dose for the hematopoietic syndrome, starting 24 hours after irradiation. RESULTS: The results indicated that granulocyte colony stimulating factor (G-CSF) enhanced survival, stimulated recovery from neutropenia, and induced mobilization of hematopoietic progenitor cells. In addition, the administration of G-CSF resulted in maturation of monocytes/macrophages. CONCLUSIONS: These results support continuing efforts toward validation of the minipig as a large animal model for advanced testing of radiation countermeasures and characterization of the pathophysiology of ARS, and they suggest that the efficacy of G-CSF in improving survival after total body irradiation may involve mechanisms other than increasing the numbers of circulating granulocytes.


Subject(s)
Acute Radiation Syndrome/drug therapy , Disease Models, Animal , Granulocyte Colony-Stimulating Factor/therapeutic use , Hematopoiesis/drug effects , Swine, Miniature , Whole-Body Irradiation/adverse effects , Acute Radiation Syndrome/blood , Acute Radiation Syndrome/mortality , Animals , C-Reactive Protein/analysis , Drug Evaluation, Preclinical/methods , Filgrastim , Hematopoiesis/radiation effects , Hematopoietic Stem Cell Mobilization/methods , Humans , Macrophages/cytology , Macrophages/drug effects , Male , Monocytes/cytology , Monocytes/drug effects , Neutropenia/drug therapy , Organs at Risk/radiation effects , Recombinant Proteins/therapeutic use , Reproducibility of Results , Swine
17.
Sud Med Ekspert ; 52(6): 17-9, 2009.
Article in Russian | MEDLINE | ID: mdl-20088132

ABSTRACT

The objective of the present study was to perform retrospective thanatological analysis of reports on acute radiation disease in liquidators of the consequences of the accident at the Chernobyl nuclear power plant. It is shown that hemorrhagic syndrome was the main cause of death among the liquidators exposed to ionizing radiation.


Subject(s)
Acute Radiation Syndrome/diagnosis , Acute Radiation Syndrome/mortality , Chernobyl Nuclear Accident , Forensic Pathology , Occupational Diseases/diagnosis , Occupational Diseases/mortality , Acute Radiation Syndrome/etiology , Adult , Cause of Death , Female , Humans , Male , Occupational Diseases/etiology , Russia , Ukraine
18.
Health Phys ; 93(5): 462-9, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18049222

ABSTRACT

The Chernobyl accident resulted in almost one-third of the reported cases of acute radiation sickness (ARS) reported worldwide. Cases occurred among the plant employees and first responders but not among the evacuated populations or general population. The diagnosis of ARS was initially considered for 237 persons based on symptoms of nausea, vomiting, and diarrhea. Ultimately, the diagnosis of ARS was confirmed in 134 persons. There were 28 short term deaths of which 95% occurred at whole body doses in excess of 6.5 Gy. Underlying bone marrow failure was the main contributor to all deaths during the first 2 mo. Allogenic bone marrow transplantation was performed on 13 patients and an additional six received human fetal liver cells. All of these patients died except one individual who later was discovered to have recovered his own marrow and rejected the transplant. Two or three patients were felt to have died as a result of transplant complications. Skin doses exceeded bone marrow doses by a factor of 10-30, and at least 19 of the deaths were felt to be primarily due to infection from large area beta burns. Internal contamination was of relatively minor importance in treatment. By the end of 2001, an additional 14 ARS survivors died from various causes. Long term treatment has included therapy for beta burn fibrosis and skin atrophy as well as for cataracts.


Subject(s)
Acute Radiation Syndrome/therapy , Chernobyl Nuclear Accident , Acute Radiation Syndrome/diagnosis , Acute Radiation Syndrome/mortality , Bone Marrow Transplantation , Follow-Up Studies , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...