51
|
Bhanja P, Norris A, Gupta-Saraf P, Hoover A, Saha S. BCN057 induces intestinal stem cell repair and mitigates radiation-induced intestinal injury. Stem Cell Res Ther 2018; 9:26. [PMID: 29394953 PMCID: PMC5797353 DOI: 10.1186/s13287-017-0763-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 12/20/2017] [Accepted: 12/26/2017] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Radiation-induced gastrointestinal syndrome (RIGS) results from the acute loss of intestinal stem cells (ISC), impaired epithelial regeneration, and subsequent loss of the mucosal barrier, resulting in electrolyte imbalance, diarrhea, weight loss, sepsis, and mortality. The high radiosensitivity of the intestinal epithelium limits effective radiotherapy against abdominal malignancies and limits the survival of victims of nuclear accidents or terrorism. Currently, there is no approved therapy to mitigate radiation toxicity in the intestine. Here we demonstrate that BCN057, an anti-neoplastic small molecular agent, induces ISC proliferation and promotes intestinal epithelial repair against radiation injury. METHODS BCN057 (90 mg/kg body weight, subcutaneously) was injected into C57Bl6 male mice (JAX) at 24 h following abdominal irradiation (AIR) and was continued for 8 days post-irradiation. BCN057-mediated rescue of Lgr5-positive ISC was validated in Lgr5-EGFP-Cre-ERT2 mice exposed to AIR. The regenerative response of Lgr5-positive ISC was examined by lineage tracing assay using Lgr5-EGFP-ires-CreERT2-TdT mice with tamoxifen administration to activate Cre recombinase and thereby marking the ISC and their respective progeny. Ex vivo three-dimensional organoid cultures were developed from surgical specimens of human colon or from mice jejunum and were used to examine the radio-mitigating role of BCN057 on ISC ex vivo. Organoid growth was determined by quantifying the budding crypt/total crypt ratio. Statistical analysis was performed using Log-rank (Mantel-Cox) test and paired two-tail t test. RESULTS Treatment with BCN057 24 h after a lethal dose of AIR rescues ISC, promotes regeneration of the intestinal epithelium, and thereby mitigates RIGS. Irradiated mice without BCN057 treatment suffered from RIGS, resulting in 100% mortality within 15 days post-radiation. Intestinal organoids developed from mice jejunum or human colon demonstrated a regenerative response with BCN057 treatment and mitigated radiation toxicity. However, BCN057 did not deliver radio-protection to mouse or human colon tumor tissue. CONCLUSION BCN057 is a potential mitigator against RIGS and may be useful for improving the therapeutic ratio of abdominal radiotherapy. This is the first report demonstrating that a small molecular agent mitigates radiation-induced intestinal injury by inducing ISC self-renewal and proliferation.
Collapse
Affiliation(s)
- Payel Bhanja
- Department of Radiation Oncology, The University of Kansas Medical Center, MS 4033, 3901 Rainbow Boulevard, Kansas City, Kansas, 66160, USA
| | | | - Pooja Gupta-Saraf
- Department of Radiation Oncology, The University of Kansas Medical Center, MS 4033, 3901 Rainbow Boulevard, Kansas City, Kansas, 66160, USA
| | - Andrew Hoover
- Department of Radiation Oncology, The University of Kansas Medical Center, MS 4033, 3901 Rainbow Boulevard, Kansas City, Kansas, 66160, USA
| | - Subhrajit Saha
- Department of Radiation Oncology, The University of Kansas Medical Center, MS 4033, 3901 Rainbow Boulevard, Kansas City, Kansas, 66160, USA. .,Department of Cancer Biology, The University of Kansas Medical Center, MS 4033, 3901 Rainbow Boulevard, Kansas City, Kansas, 66160, USA.
| |
Collapse
|
52
|
Whole thorax irradiation of non-human primates induces persistent nuclear damage and gene expression changes in peripheral blood cells. PLoS One 2018; 13:e0191402. [PMID: 29351567 PMCID: PMC5774773 DOI: 10.1371/journal.pone.0191402] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 01/04/2018] [Indexed: 12/16/2022] Open
Abstract
We investigated the cytogenetic and gene expression responses of peripheral blood cells of non-human primates (NHP, Macaca mulatta) that were whole-thorax irradiated with a single dose of 10 Gy. In this model, partial irradiation of NHPs in the thoracic region (Whole Thorax Lung Irradiation, WTLI) allows the study of late radiation-induced lung injury, while avoiding acute radiation syndromes related to hematopoietic and gastrointestinal injury. A transient drop in circulating lymphocytes and platelets was seen by 9 days, followed by elevations in respiratory rate, circulating neutrophils, lymphocytes, and monocytes at 60-100 days, corresponding to computed tomography (CT) and histologic evidence of pneumonitis, and elective euthanasia of four animals. To evaluate long-term DNA damage in NHP peripheral blood lymphocytes after 10 Gy WTLI, we used the cytokinesis-block micronucleus (CBMN) assay to measure chromosomal aberrations as post-mitotic micronuclei in blood samples collected up to 8 months after irradiation. Regression analysis showed significant induction of micronuclei in NHP blood cells that persisted with a gradual decline over the 8-month study period, suggesting long-term DNA damage in blood lymphocytes after WTLI. We also report transcriptomic changes in blood up to 30 days after WTLI. We isolated total RNA from peripheral blood at 3 days before and then at 2, 5 and 30 days after irradiation. We identified 1187 transcripts that were significantly changed across the 30-day time course. From changes in gene expression, we identified biological processes related to immune responses, which persisted across the 30-day study. Response to oxygen-containing compounds and bacteria were implicated by gene-expression changes at the earliest day 2 and latest, day 30 time-points. Gene expression changes suggest a persistent altered state of the immune system, specifically response to infection, for at least a month after WTLI.
Collapse
|
53
|
Measey TJ, Pouliot M, Wierzbicki W, Swanson C, Brown D, Authier S, Donini O. Pilot Study of Radiation-induced Gastrointestinal Injury in a Hemi-body Shielded Göttingen Minipig Model. HEALTH PHYSICS 2018; 114:43-57. [PMID: 30085969 DOI: 10.1097/hp.0000000000000751] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Development of medical countermeasures (MCMs) for gastrointestinal (GI) injury following acute radiation exposure requires well-characterized models that can assess not only survival but also secondary endpoints, including structural and functional characteristics of GI damage and recovery that ultimately contribute to long-term survival. The authors conducted a pilot study in a hemi-body shielded Göttingen minipig model of radiation-induced GI injury that enables radiation damage to the GI tract to be evaluated and reduces the potential for hemorrhage and/or damage in other more sensitive organ systems. With shielding of the head, chest, and front legs, radiation dose levels of 14 Gy were required to see significant GI-related morbidity, while dose levels of 16 Gy resulted in significant mortality by day 45 post-irradiation. Periodic scheduled necropsies showed significant reduction in and slow recovery of intestinal crypt count at 14 and 16 Gy. Intestinal proliferative activity was initially increased and then gradually decreased over the course of the study. Histological evidence of marked inflammatory infiltrates was noted in the GI tract at day 5, while collagen deposition, indicative of fibrosis, was observed as early as day 15, peaking at day 30. The radiation dose-responsive indicators of GI damage identified in this model (i.e., intestinal crypt count and proliferative activity) may serve as useful endpoints for evaluation of the efficacy of potential MCMs.
Collapse
|
54
|
Measey TJ, Pouliot M, Wierzbicki W, Swanson C, Brown D, Stamatopoulos J, Proulx D, Authier S, Donini O. Expanded Characterization of a Hemi-Body Shielded Göttingen Minipig Model of Radiation-induced Gastrointestinal Injury Incorporating Oral Dosing Procedures. HEALTH PHYSICS 2018; 114:32-42. [PMID: 30085968 DOI: 10.1097/hp.0000000000000750] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
In collaboration with the Biomedical Advanced Research and Development Authority (BARDA), the authors recently conducted a pilot study in a hemi-body shielded model of radiation-induced gastrointestinal (GI) injury in Göttingen minipigs following exposure to radiation dose levels between 8-16 Gy. Herein, the impact of oral dosing procedures is assessed, as well as the specific causes of death in animals exposed to radiation doses of 14 and 16 Gy (n = 64; 32 male, 32 female, between 6 and 8 mo of age). Oral dosing using a 2-tablet placebo system comprised of both immediate release and enteric-coated tablets starting 24 h post-irradiation resulted in inhibited gastric emptying of the enteric-coated tablets, which were found to be retained in the stomach and/or regurgitated. This finding appears to be species-specific, as similar findings have not been reported for other large animal species (e.g., non-human primates). Mortality was primarily dictated by decreased activity, body weight loss (>35%), and/or respiratory distress, despite shielding of the lung. The cause of respiratory distress in animals that were pre-terminally euthanized varied according to the timing of death, with interstitial inflammation and extensive fibrosis observed >20 days post-irradiation. Kidney damage was also identified in most animals after day 10. Changes in the GI tract were consistent with previous studies and included collagen deposition/fibrosis. Observations of inflammatory infiltrates and interstitial inflammation/fibrosis in both shielded and unshielded organs support a strong secondary inflammatory syndrome post-irradiation.
Collapse
|
55
|
Cohen EP, Hankey KG, Bennett AW, Farese AM, Parker GA, MacVittie TJ. Acute and Chronic Kidney Injury in a Non-Human Primate Model of Partial-Body Irradiation with Bone Marrow Sparing. Radiat Res 2017; 188:661-671. [PMID: 29035153 DOI: 10.1667/rr24857.1] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The development of medical countermeasures against acute and delayed multi-organ injury requires animal models predictive of the human response to radiation and its treatment. Late chronic injury is a well-known feature of radiation nephropathy, but acute kidney injury has not been reported in an appropriate animal model. We have established a single-fraction partial-body irradiation model with minimal marrow sparing in non-human primates. Subject-based medical management was used including parenteral fluids according to prospective morbidity criteria. We show herein that 10 or 11 Gy exposures caused both acute and chronic kidney injury. Acute and chronic kidney injury appear to be dose-independent between 10 and 11 Gy. Acute kidney injury was identified during the first 50 days postirradiation and appeared to resolve before the occurrence of chronic kidney injury, which was progressively more severe up to 180 days postirradiation, which was the end of the study. These findings show that mitigation of the acute radiation syndrome by medical management will unmask delayed late effects that occur months after partial-body irradiation. They further emphasize that both acute and chronic changes in kidney function must be taken into account in the use and timing of mitigators and medical management for acute radiation syndrome and delayed effects of acute radiation exposure (DEARE).
Collapse
Affiliation(s)
| | - Kim G Hankey
- b Radiation Oncology, University of Maryland, School of Medicine, Baltimore, Maryland; and
| | - Alexander W Bennett
- b Radiation Oncology, University of Maryland, School of Medicine, Baltimore, Maryland; and
| | - Ann M Farese
- b Radiation Oncology, University of Maryland, School of Medicine, Baltimore, Maryland; and
| | | | - Thomas J MacVittie
- b Radiation Oncology, University of Maryland, School of Medicine, Baltimore, Maryland; and
| |
Collapse
|
56
|
Prado C, MacVittie TJ, Bennett AW, Kazi A, Farese AM, Prado K. Organ Doses Associated with Partial-Body Irradiation with 2.5% Bone Marrow Sparing of the Non-Human Primate: A Retrospective Study. Radiat Res 2017; 188:615-625. [PMID: 28985133 DOI: 10.1667/rr14804.1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
A partial-body irradiation model with approximately 2.5% bone marrow sparing (PBI/BM2.5) was established to determine the radiation dose-response relationships for the prolonged and delayed multi-organ effects of acute radiation exposure. Historically, doses reported to the entire body were assumed to be equal to the prescribed dose at some defined calculation point, and the dose-response relationship for multi-organ injury has been defined relative to the prescribed dose being delivered at this point, e.g., to a point at mid-depth at the level of the xiphoid of the non-human primate (NHP). In this retrospective-dose study, the true distribution of dose within the major organs of the NHP was evaluated, and these doses were related to that at the traditional dose-prescription point. Male rhesus macaques were exposed using the PBI/BM2.5 protocol to a prescribed dose of 10 Gy using 6-MV linear accelerator photons at a rate of 0.80 Gy/min. Point and organ doses were calculated for each NHP from computed tomography (CT) scans using heterogeneous density data. The prescribed dose of 10.0 Gy to a point at midline tissue assuming homogeneous media resulted in 10.28 Gy delivered to the prescription point when calculated using the heterogeneous CT volume of the NHP. Respective mean organ doses to the volumes of nine organs, including the heart, lung, bowel and kidney, were computed. With modern treatment planning systems, utilizing a three-dimensional reconstruction of the NHP's CT images to account for the variations in body shape and size, and using density corrections for each of the tissue types, bone, water, muscle and air, accurate determination of the differences in dose to the NHP can be achieved. Dose and volume statistics can be ascertained for any body structure or organ that has been defined using contouring tools in the planning system. Analysis of the dose delivered to critical organs relative to the total-body target dose will permit a more definitive analysis of organ-specific effects and their respective influence in multiple organ injury.
Collapse
Affiliation(s)
- C Prado
- a Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland; and
| | - T J MacVittie
- a Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland; and
| | - A W Bennett
- a Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland; and
| | - A Kazi
- b Veterans Administration, Maryland Health Care System, Baltimore, Maryland
| | - A M Farese
- a Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland; and
| | - K Prado
- a Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland; and
| |
Collapse
|
57
|
Carter CL, Jones JW, Farese AM, MacVittie TJ, Kane MA. Lipidomic dysregulation within the lung parenchyma following whole-thorax lung irradiation: Markers of injury, inflammation and fibrosis detected by MALDI-MSI. Sci Rep 2017; 7:10343. [PMID: 28871103 PMCID: PMC5583385 DOI: 10.1038/s41598-017-10396-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 08/08/2017] [Indexed: 12/23/2022] Open
Abstract
Radiation-induced lung injury (RILI) is a delayed effect of acute radiation exposure that can limit curative cancer treatment therapies and cause lethality following high-dose whole-thorax lung irradiation (WTLI). To date, the exact mechanisms of injury development following insult remain ill-defined and there are no FDA approved pharmaceutical agents or medical countermeasures. Traditionally, RILI development is considered as three phases, the clinically latent period, the intermediate acute pneumonitis phase and the later fibrotic stage. Utilizing matrix-assisted laser desorption ionization mass spectrometry imaging, we identified a number of lipids that were reflective of disease state or injury. Lipids play central roles in metabolism and cell signaling, and thus reflect the phenotype of the tissue environment, making these molecules pivotal biomarkers in many disease processes. We detected decreases in specific surfactant lipids irrespective of the different pathologies that presented within each sample at 180 days post whole-thorax lung irradiation. We also detected regional increases in ether-linked phospholipids that are the precursors of PAF, and global decreases in lipids that were reflective of severe fibrosis. Taken together our results provide panels of lipids that can differentiate between naïve and irradiated samples, as well as providing potential markers of inflammation and fibrosis.
Collapse
Affiliation(s)
- Claire L Carter
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, 21201, Baltimore, MD, USA
| | - Jace W Jones
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, 21201, Baltimore, MD, USA
| | - Ann M Farese
- University of Maryland, School of Medicine, Department of Radiation Oncology, 21201, Baltimore, MD, USA
| | - Thomas J MacVittie
- University of Maryland, School of Medicine, Department of Radiation Oncology, 21201, Baltimore, MD, USA
| | - Maureen A Kane
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, 21201, Baltimore, MD, USA.
| |
Collapse
|
58
|
Singh VK, Olabisi AO. Nonhuman primates as models for the discovery and development of radiation countermeasures. Expert Opin Drug Discov 2017; 12:695-709. [PMID: 28441902 DOI: 10.1080/17460441.2017.1323863] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Despite significant scientific advances over the past six decades toward the development of safe and effective radiation countermeasures for humans using animal models, only two pharmaceutical agents have been approved by United States Food and Drug Administration (US FDA) for hematopoietic acute radiation syndrome (H-ARS). Additional research efforts are needed to further develop large animal models for improving the prediction of clinical safety and effectiveness of radiation countermeasures for ARS and delayed effects of acute radiation exposure (DEARE) in humans. Area covered: The authors review the suitability of animal models for the development of radiation countermeasures for ARS following the FDA Animal Rule with a special focus on nonhuman primate (NHP) models of ARS. There are seven centers in the United States currently conducting studies with irradiated NHPs, with the majority of studies being conducted with rhesus monkeys. Expert opinion: The NHP model is considered the gold standard animal model for drug development and approval by the FDA. The lack of suitable substitutes for NHP models for predicting response in humans serves as a bottleneck for the development of radiation countermeasures. Additional large animal models need to be characterized to support the development and FDA-approval of new radiation countermeasures.
Collapse
Affiliation(s)
- Vijay K Singh
- a Department of Pharmacology and Molecular Therapeutics , F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences , Bethesda , MD , USA.,b Armed Forces Radiobiology Research Institute , Uniformed Services University of the Health Sciences , Bethesda , MD , USA
| | - Ayodele O Olabisi
- b Armed Forces Radiobiology Research Institute , Uniformed Services University of the Health Sciences , Bethesda , MD , USA
| |
Collapse
|
59
|
Pannkuk EL, Laiakis EC, Authier S, Wong K, Fornace AJ. Gas Chromatography/Mass Spectrometry Metabolomics of Urine and Serum from Nonhuman Primates Exposed to Ionizing Radiation: Impacts on the Tricarboxylic Acid Cycle and Protein Metabolism. J Proteome Res 2017; 16:2091-2100. [PMID: 28351153 PMCID: PMC5720681 DOI: 10.1021/acs.jproteome.7b00064] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Ionizing radiation (IR) directly damages cells and tissues or indirectly damages them through reactive free radicals that may lead to longer term adverse sequelae such as cancers, persistent inflammation, or possible death. Potential exposures include nuclear reactor accidents, improper disposal of equipment containing radioactive materials or medical errors, and terrorist attacks. Metabolomics (comprehensive analysis of compounds <1 kDa) by mass spectrometry (MS) has been proposed as a tool for high-throughput biodosimetry and rapid assessment of exposed dose and triage needed. While multiple studies have been dedicated to radiation biomarker discovery, many have utilized liquid chromatography (LC) MS platforms that may not detect particular compounds (e.g., small carboxylic acids or isomers) that complementary analytical tools, such as gas chromatography (GC) time-of-flight (TOF) MS, are ideal for. The current study uses global GC-TOF-MS metabolomics to complement previous LC-MS analyses on nonhuman primate biofluids (urine and serum) 7 days after exposure to 2, 4, 6, 7, and 10 Gy IR. Multivariate data analysis was used to visualize differences between control and IR exposed groups. Univariate analysis was used to determine a combined 26 biomarkers in urine and serum that significantly changed after exposure to IR. We found several metabolites involved in tricarboxylic acid cycle function, amino acid metabolism, and host microbiota that were not previously detected by global and targeted LC-MS studies.
Collapse
Affiliation(s)
- Evan L. Pannkuk
- Tumor Biology Program, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, D.C. 20057, United States
| | - Evagelia C. Laiakis
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, D.C. 20057, United States
| | - Simon Authier
- CiToxLAB North America, Laval, Quebec H7V 4B3, Canada
| | - Karen Wong
- CiToxLAB North America, Laval, Quebec H7V 4B3, Canada
| | - Albert J. Fornace
- Tumor Biology Program, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, D.C. 20057, United States
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, D.C. 20057, United States
| |
Collapse
|
60
|
Brodin NP, Velcich A, Guha C, Tomé WA. A Model for Precise and Uniform Pelvic- and Limb-Sparing Abdominal Irradiation to Study the Radiation-Induced Gastrointestinal Syndrome in Mice Using Small Animal Irradiation Systems. Dose Response 2017; 15:1559325816685798. [PMID: 28203121 PMCID: PMC5298432 DOI: 10.1177/1559325816685798] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Background and Purpose: Currently, no readily available mitigators exist for acute abdominal radiation injury. Here, we present an animal model for precise and homogenous limb-sparing abdominal irradiation (LSAIR) to study the radiation-induced gastrointestinal syndrome (RIGS). Materials and Methods: The LSAIR technique was developed using the small animal radiation research platform (SARRP) with image guidance capabilities. We delivered LSAIR at doses between 14 and 18 Gy on 8- to 10-week-old male C57BL/6 mice. Histological analysis was performed to confirm that the observed mortality was due to acute abdominal radiation injury. Results: A steep dose–response relationship was found for survival, with no deaths seen at doses below 16 Gy and 100% mortality at above 17 Gy. All deaths occurred between 6 and 10 days after irradiation, consistent with the onset of RIGS. This was further confirmed by histological analysis showing clear differences in the number of regenerative intestinal crypts between animals receiving sublethal (14 Gy) and 100% lethal (18 Gy) radiation. Conclusion: The developed LSAIR technique provides uniform dose delivery with a clear dose response, consistent with acute abdominal radiation injury on histological examination. This model can provide a useful tool for researchers investigating the development of mitigators for accidental or clinical high-dose abdominal irradiation.
Collapse
Affiliation(s)
- N Patrik Brodin
- Department of Radiation Oncology, Institute for Onco-Physics, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Radiation Oncology, Montefiore Medical Center, Bronx, NY, USA
| | - Anna Velcich
- Department of Radiation Oncology, Montefiore Medical Center, Bronx, NY, USA
| | - Chandan Guha
- Department of Radiation Oncology, Institute for Onco-Physics, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Radiation Oncology, Montefiore Medical Center, Bronx, NY, USA
| | - Wolfgang A Tomé
- Department of Radiation Oncology, Institute for Onco-Physics, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Radiation Oncology, Montefiore Medical Center, Bronx, NY, USA
| |
Collapse
|
61
|
Homer MJ, Raulli R, DiCarlo-Cohen AL, Esker J, Hrdina C, Maidment BW, Moyer B, Rios C, Macchiarini F, Prasanna PG, Wathen L. UNITED STATES DEPARTMENT OF HEALTH AND HUMAN SERVICES BIODOSIMETRY AND RADIOLOGICAL/NUCLEAR MEDICAL COUNTERMEASURE PROGRAMS. RADIATION PROTECTION DOSIMETRY 2016; 171:85-98. [PMID: 27590469 PMCID: PMC6280724 DOI: 10.1093/rpd/ncw226] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The United States Department of Health and Human Services (HHS) is fully committed to the development of medical countermeasures to address national security threats from chemical, biological, radiological, and nuclear agents. Through the Public Health Emergency Medical Countermeasures Enterprise, HHS has launched and managed a multi-agency, comprehensive effort to develop and operationalize medical countermeasures. Within HHS, development of medical countermeasures includes the National Institutes of Health (NIH), (led by the National Institute of Allergy and Infectious Diseases), the Office of the Assistant Secretary of Preparedness and Response/Biomedical Advanced Research and Development Authority (BARDA); with the Division of Medical Countermeasure Strategy and Requirements, the Centers for Disease Control and Prevention, and the Food and Drug Administration as primary partners in this endeavor. This paper describes various programs and coordinating efforts of BARDA and NIH for the development of medical countermeasures for radiological and nuclear threats.
Collapse
Affiliation(s)
- Mary J Homer
- Biomedical Advanced Research and Development Authority, Office of the Assistant Secretary for Preparedness and Response, US Department of Health and Human Services , 330 Independence Ave., SW, Room G644, Washington, DC 20201, USA
| | - Robert Raulli
- Biomedical Advanced Research and Development Authority, Office of the Assistant Secretary for Preparedness and Response, US Department of Health and Human Services , 330 Independence Ave., SW, Room G644, Washington, DC 20201, USA
| | - Andrea L DiCarlo-Cohen
- Radiation Nuclear Countermeasures Program, Division of Allergy, Immunology, and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, US Department of Health and Human Services, 5601 Fishers Lane, Rockville, MD 20892-9828, USA
| | - John Esker
- Biomedical Advanced Research and Development Authority, Office of the Assistant Secretary for Preparedness and Response, US Department of Health and Human Services , 330 Independence Ave., SW, Room G644, Washington, DC 20201, USA
| | - Chad Hrdina
- Medical Utilization and Response Integration, Division of Medical Countermeasure Strategy and Requirements, Office of Policy and Planning, Office of the Assistant Secretary for Preparedness and Response, US Department of Health and Human Services , 330 Independence Ave., SW, Room G644, Washington, DC 20201, USA
| | - Bert W Maidment
- Radiation Nuclear Countermeasures Program, Division of Allergy, Immunology, and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, US Department of Health and Human Services, 5601 Fishers Lane, Rockville, MD 20892-9828, USA
| | - Brian Moyer
- Biomedical Advanced Research and Development Authority, Office of the Assistant Secretary for Preparedness and Response, US Department of Health and Human Services , 330 Independence Ave., SW, Room G644, Washington, DC 20201, USA
| | - Carmen Rios
- Radiation Nuclear Countermeasures Program, Division of Allergy, Immunology, and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, US Department of Health and Human Services, 5601 Fishers Lane, Rockville, MD 20892-9828, USA
| | - Francesca Macchiarini
- Radiation Nuclear Countermeasures Program, Division of Allergy, Immunology, and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, US Department of Health and Human Services, 5601 Fishers Lane, Rockville, MD 20892-9828, USA
| | - Pataje G Prasanna
- Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute; National Institutes of Health, US Department of Health and Human Services , 9608 Medical Center Drive, Room 3W230, MSC9727 , Bethesda, MD 20892-9727, USA
| | - Lynne Wathen
- Biomedical Advanced Research and Development Authority, Office of the Assistant Secretary for Preparedness and Response, US Department of Health and Human Services , 330 Independence Ave., SW, Room G644, Washington, DC 20201, USA
| |
Collapse
|
62
|
DeBo RJ, Lees CJ, Dugan GO, Caudell DL, Michalson KT, Hanbury DB, Kavanagh K, Cline JM, Register TC. Late Effects of Total-Body Gamma Irradiation on Cardiac Structure and Function in Male Rhesus Macaques. Radiat Res 2016; 186:55-64. [PMID: 27333082 PMCID: PMC5068576 DOI: 10.1667/rr14357.1] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Heart disease is an increasingly recognized, serious late effect of radiation exposure, most notably among breast cancer and Hodgkin's disease survivors, as well as the Hiroshima and Nagasaki atomic bomb survivors. The purpose of this study was to evaluate the late effects of total-body irradiation (TBI) on cardiac morphology, function and selected circulating biomarkers in a well-established nonhuman primate model. For this study we used male rhesus macaques that were exposed to a single total-body dose of ionizing gamma radiation (6.5-8.4 Gy) 5.6-9.7 years earlier at ages ranging from ∼3-10 years old and a cohort of nonirradiated controls. Transthoracic echocardiography was performed annually for 3 years on 20 irradiated and 11 control animals. Myocardium was examined grossly and histologically, and myocardial fibrosis/collagen was assessed microscopically and by morphometric analysis of Masson's trichrome-stained sections. Serum/plasma from 27 irradiated and 13 control animals was evaluated for circulating biomarkers of cardiac damage [N-terminal pro B-type natriuretic protein (nt-proBNP) and troponin-I], inflammation (CRP, IL-6, MCP-1, sICAM) and microbial translocation [LPS-binding protein (LBP) and sCD14]. A higher prevalence of histological myocardial fibrosis was observed in the hearts obtained from the irradiated animals (9/14) relative to controls (0/3) (P = 0.04, χ(2)). Echocardiographically determined left ventricular end diastolic and systolic diameters were significantly smaller in irradiated animals (repeated measures ANOVA, P < 0.001 and P < 0.008, respectively). Histomorphometric analysis of trichrome-stained sections of heart tissue demonstrated ∼14.9 ± 1.4% (mean ± SEM) of myocardial area staining for collagen in irradiated animals compared to 9.1 ± 0.9 % in control animals. Circulating levels of MCP-1 and LBP were significantly higher in irradiated animals (P < 0.05). A high incidence of diabetes in the irradiated animals was associated with higher plasma triglyceride and lower HDLc but did not appear to be associated with cardiovascular phenotypes. These results demonstrate that single total-body doses of 6.5-8.4 Gy produced long-term effects including a high incidence of myocardial fibrosis, reduced left ventricular diameter and elevated systemic inflammation. Additional prospective studies are required to define the time course and mechanisms underlying radiation-induced heart disease in this model.
Collapse
Affiliation(s)
| | - Cynthia J. Lees
- Department of Pathology/Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Greg O. Dugan
- Department of Pathology/Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - David L. Caudell
- Department of Pathology/Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Kris T. Michalson
- Department of Pathology/Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - David B. Hanbury
- Department of Pathology/Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Kylie Kavanagh
- Department of Pathology/Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - J. Mark Cline
- Department of Pathology/Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Thomas C. Register
- Department of Pathology/Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
63
|
|
64
|
Shea-Donohue T, Fasano A, Zhao A, Notari L, Yan S, Sun R, Bohl JA, Desai N, Tudor G, Morimoto M, Booth C, Bennett A, Farese AM, MacVittie TJ. Mechanisms Involved in the Development of the Chronic Gastrointestinal Syndrome in Nonhuman Primates after Total-Body Irradiation with Bone Marrow Shielding. Radiat Res 2016; 185:591-603. [PMID: 27223826 DOI: 10.1667/rr14024.1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In this study, nonhuman primates (NHPs) exposed to lethal doses of total body irradiation (TBI) within the gastrointestinal (GI) acute radiation syndrome range, sparing ∼5% of bone marrow (TBI-BM5), were used to evaluate the mechanisms involved in development of the chronic GI syndrome. TBI increased mucosal permeability in the jejunum (12-14 Gy) and proximal colon (13-14 Gy). TBI-BM5 also impaired mucosal barrier function at doses ranging from 10-12.5 Gy in both small intestine and colon. Timed necropsies of NHPs at 6-180 days after 10 Gy TBI-BM5 showed that changes in small intestine preceded those in the colon. Chronic GI syndrome in NHPs is characterized by continued weight loss and intermittent GI syndrome symptoms. There was a long-lasting decrease in jejunal glucose absorption coincident with reduced expression of the sodium-linked glucose transporter. The small intestine and colon showed a modest upregulation of several different pro-inflammatory mediators such as NOS-2. The persistent inflammation in the post-TBI-BM5 period was associated with a long-lasting impairment of mucosal restitution and a reduced expression of intestinal and serum levels of alkaline phosphatase (ALP). Mucosal healing in the postirradiation period is dependent on sparing of stem cell crypts and maturation of crypt cells into appropriate phenotypes. At 30 days after 10 Gy TBI-BM5, there was a significant downregulation in the gene and protein expression of the stem cell marker Lgr5 but no change in the gene expression of enterocyte or enteroendocrine lineage markers. These data indicate that even a threshold dose of 10 Gy TBI-BM5 induces a persistent impairment of both mucosal barrier function and restitution in the GI tract and that ALP may serve as a biomarker for these events. These findings have important therapeutic implications for the design of medical countermeasures.
Collapse
Affiliation(s)
- Terez Shea-Donohue
- a Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland;,b Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Alessio Fasano
- c Massachusetts General Hospital, Pediatric Gastroenterology and Nutrition, Boston, Massachusetts
| | - Aiping Zhao
- a Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland;,b Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Luigi Notari
- b Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Shu Yan
- b Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Rex Sun
- b Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jennifer A Bohl
- b Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Neemesh Desai
- a Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Greg Tudor
- d Epistem Ltd., Manchester, United Kingdom
| | - Motoko Morimoto
- e School of Food, Agricultural and Environmental Studies, Miyagi University, Japan
| | | | - Alexander Bennett
- a Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Ann M Farese
- a Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Thomas J MacVittie
- a Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
65
|
Pannkuk EL, Laiakis EC, Mak TD, Astarita G, Authier S, Wong K, Fornace AJ. A Lipidomic and Metabolomic Serum Signature from Nonhuman Primates Exposed to Ionizing Radiation. Metabolomics 2016; 12:80. [PMID: 28220056 PMCID: PMC5314995 DOI: 10.1007/s11306-016-1010-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
INTRODUCTION Due to dangers associated with potential accidents from nuclear energy and terrorist threats, there is a need for high-throughput biodosimetry to rapidly assess individual doses of radiation exposure. Lipidomics and metabolomics are becoming common tools for determining global signatures after disease or other physical insult and provide a "snapshot" of potential cellular damage. OBJECTIVES The current study assesses changes in the nonhuman primate (NHP) serum lipidome and metabolome 7 days following exposure to ionizing radiation (IR). METHODS Serum sample lipids and metabolites were extracted using a biphasic liquid-liquid extraction and analyzed by ultra performance liquid chromatography quadrupole time-of-flight mass spectrometry. Global radiation signatures were acquired in data-independent mode. RESULTS Radiation exposure caused significant perturbations in lipid metabolism, affecting all major lipid species, including free fatty acids, glycerolipids, glycerophospholipids and esterified sterols. In particular, we observed a significant increase in the levels of polyunsaturated fatty acids (PUFA)-containing lipids in the serum of NHPs exposed to 10 Gy radiation, suggesting a primary role played by PUFAs in the physiological response to IR. Metabolomics profiling indicated an increase in the levels of amino acids, carnitine, and purine metabolites in the serum of NHPs exposed to 10 Gy radiation, suggesting perturbations to protein digestion/absorption, biological oxidations, and fatty acid β-oxidation. CONCLUSIONS This is the first report to determine changes in the global NHP serum lipidome and metabolome following radiation exposure and provides information for developing metabolomic biomarker panels in human-based biodosimetry.
Collapse
Affiliation(s)
- Evan L. Pannkuk
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC
| | - Evagelia C. Laiakis
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC
| | - Tytus D. Mak
- Mass Spectrometry Data Center, National Institute of Standards and Technology, Gaithersburg, MD
| | - Giuseppe Astarita
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC
- Health Sciences, Waters Corporation, Milford, MA
| | | | | | - Albert J. Fornace
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
- Address for correspondence: Georgetown University, 3970 Reservoir Road, NW, New, Research Building, Room E504, Washington, DC 20057, , Phone: 202-687-7843, Fax: 202-687-3140
| |
Collapse
|
66
|
Cui W, Bennett AW, Zhang P, Barrow KR, Kearney SR, Hankey KG, Taylor-Howell C, Gibbs AM, Smith CP, MacVittie TJ. A non-human primate model of radiation-induced cachexia. Sci Rep 2016; 6:23612. [PMID: 27029502 PMCID: PMC4814846 DOI: 10.1038/srep23612] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 03/10/2016] [Indexed: 12/25/2022] Open
Abstract
Cachexia, or muscle wasting, is a serious health threat to victims of radiological accidents or patients receiving radiotherapy. Here, we propose a non-human primate (NHP) radiation-induced cachexia model based on clinical and molecular pathology findings. NHP exposed to potentially lethal partial-body irradiation developed symptoms of cachexia such as body weight loss in a time- and dose-dependent manner. Severe body weight loss as high as 20–25% was observed which was refractory to nutritional intervention. Radiographic imaging indicated that cachectic NHP lost as much as 50% of skeletal muscle. Histological analysis of muscle tissues showed abnormalities such as presence of central nuclei, inflammation, fatty replacement of skeletal muscle, and muscle fiber degeneration. Biochemical parameters such as hemoglobin and albumin levels decreased after radiation exposure. Levels of FBXO32 (Atrogin-1), ActRIIB and myostatin were significantly changed in the irradiated cachectic NHP compared to the non-irradiated NHP. Our data suggest NHP that have been exposed to high dose radiation manifest cachexia-like symptoms in a time- and dose-dependent manner. This model provides a unique opportunity to study the mechanism of radiation-induced cachexia and will aid in efficacy studies of mitigators of this disease.
Collapse
Affiliation(s)
- Wanchang Cui
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland, 21201 USA 10 South Pine Street, MSTF Room 604, Baltimore, MD 21201 USA
| | - Alexander W Bennett
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland, 21201 USA 10 South Pine Street, MSTF Room 604, Baltimore, MD 21201 USA
| | - Pei Zhang
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland, 21201 USA 10 South Pine Street, MSTF Room 604, Baltimore, MD 21201 USA
| | - Kory R Barrow
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland, 21201 USA 10 South Pine Street, MSTF Room 604, Baltimore, MD 21201 USA
| | - Sean R Kearney
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland, 21201 USA 10 South Pine Street, MSTF Room 604, Baltimore, MD 21201 USA
| | - Kim G Hankey
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland, 21201 USA 10 South Pine Street, MSTF Room 604, Baltimore, MD 21201 USA
| | - Cheryl Taylor-Howell
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland, 21201 USA 10 South Pine Street, MSTF Room 604, Baltimore, MD 21201 USA
| | - Allison M Gibbs
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland, 21201 USA 10 South Pine Street, MSTF Room 604, Baltimore, MD 21201 USA
| | - Cassandra P Smith
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland, 21201 USA 10 South Pine Street, MSTF Room 604, Baltimore, MD 21201 USA
| | - Thomas J MacVittie
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland, 21201 USA 10 South Pine Street, MSTF Room 604, Baltimore, MD 21201 USA
| |
Collapse
|
67
|
MacVittie TJ, Bennett AW, Farese AM, Taylor-Howell C, Smith CP, Gibbs AM, Prado K, Jackson W. The Effect of Radiation Dose and Variation in Neupogen® Initiation Schedule on the Mitigation of Myelosuppression during the Concomitant GI-ARS and H-ARS in a Nonhuman Primate Model of High-dose Exposure with Marrow Sparing. HEALTH PHYSICS 2015; 109:427-39. [PMID: 26425903 PMCID: PMC9442798 DOI: 10.1097/hp.0000000000000350] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
A nonhuman primate (NHP) model of acute high-dose, partial-body irradiation with 5% bone marrow (PBI/BM5) sparing was used to assess the effect of Neupogen® [granulocyte colony stimulating factor (G-CSF)] to mitigate the associated myelosuppression when administered at an increasing interval between exposure and initiation of treatment. A secondary objective was to assess the effect of Neupogen® on the mortality or morbidity of the hematopoietic (H)- acute radiation syndrome (ARS) and concurrent acute gastrointestinal radiation syndrome (GI-ARS). NHP were exposed to 10.0 or 11.0 Gy with 6 MV LINAC-derived photons at approximately 0.80 Gy min. All NHP received medical management. NHP were dosed daily with control article (5% dextrose in water) initiated on day 1 post-exposure or Neupogen® (10 μg kg) initiated on day 1, day 3, or day 5 until recovery [absolute neutrophil count (ANC) ≥ 1,000 cells μL for three consecutive days]. Mortality in both the 10.0 Gy and 11.0 Gy cohorts suggested that early administration of Neupogen® at day 1 post exposure may affect acute GI-ARS mortality, while Neupogen® appeared to mitigate mortality due to the H-ARS. However, the study was not powered to detect statistically significant differences in survival. The ability of Neupogen® to stimulate granulopoiesis was assessed by evaluating key parameters for ANC recovery: the depth of nadir, duration of neutropenia (ANC < 500 cells μL) and recovery time to ANC ≥ 1,000 cells μL. Following 10.0 Gy PBI/BM5, the mean duration of neutropenia was 11.6 d in the control cohort vs. 3.5 d and 4.6 d in the day 1 and day 3 Neupogen® cohorts, respectively. The respective ANC nadirs were 94 cells μL, 220 cells μL, and 243 cells μL for the control and day 1 and day 3 Neupogen® cohorts. Following 11.0 Gy PBI/BM5, the duration of neutropenia was 10.9 d in the control cohort vs. 2.8 d, 3.8 d, and 4.5 d in the day 1, day 3, and day 5 Neupogen® cohorts, respectively. The respective ANC nadirs for the control and day 1, day 3, and day 5 Neupogen® cohorts were 131 cells μL, 292 cells μL, 236 cells μL, and 217 cells μL, respectively. Therefore, the acceleration of granulopoiesis by Neupogen® in this model is independent of the time interval between radiation exposure and treatment initiation up to 5 d post-exposure. The PBI/BM5 model can be used to assess medical countermeasure efficacy in the context of the concurrent GI- and H-ARS.
Collapse
Affiliation(s)
- Thomas J MacVittie
- *University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD; †University of Maryland Medical Center, Department of Radiation Oncology, Baltimore, MD; ‡Statistician, Rockville, MD
| | | | | | | | | | | | | | | |
Collapse
|
68
|
Zhang P, Cui W, Hankey KG, Gibbs AM, Smith CP, Taylor-Howell C, Kearney SR, MacVittie TJ. Increased Expression of Connective Tissue Growth Factor (CTGF) in Multiple Organs After Exposure of Non-Human Primates (NHP) to Lethal Doses of Radiation. HEALTH PHYSICS 2015; 109:374-90. [PMID: 26425899 PMCID: PMC4593333 DOI: 10.1097/hp.0000000000000343] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Exposure to sufficiently high doses of ionizing radiation is known to cause fibrosis in many different organs and tissues. Connective tissue growth factor (CTGF/CCN2), a member of the CCN family of matricellular proteins, plays an important role in the development of fibrosis in multiple organs. The aim of the present study was to quantify the gene and protein expression of CTGF in a variety of organs from non-human primates (NHP) that were previously exposed to potentially lethal doses of radiation. Tissues from non-irradiated NHP and NHP exposed to whole thoracic lung irradiation (WTLI) or partial-body irradiation with 5% bone marrow sparing (PBI/BM5) were examined by real-time quantitative reverse transcription PCR, western blot, and immunohistochemistry. Expression of CTGF was elevated in the lung tissues of NHP exposed to WTLI relative to the lung tissues of the non-irradiated NHP. Increased expression of CTGF was also observed in multiple organs from NHP exposed to PBI/BM5 compared to non-irradiated NHP; these included the lung, kidney, spleen, thymus, and liver. These irradiated organs also exhibited histological evidence of increased collagen deposition compared to the control tissues. There was significant correlation of CTGF expression with collagen deposition in the lung and spleen of NHP exposed to PBI/BM5. Significant correlations were observed between spleen and multiple organs on CTGF expression and collagen deposition, respectively, suggesting possible crosstalk between spleen and other organs. These data suggest that CTGF levels are increased in multiple organs after radiation exposure and that inflammatory cell infiltration may contribute to the elevated levels of CTGF in multiple organs.
Collapse
Affiliation(s)
- Pei Zhang
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland, USA 21201 10 South Pine Street, MSTF Room 604, Baltimore, MD 21201
| | - Wanchang Cui
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland, USA 21201 10 South Pine Street, MSTF Room 604, Baltimore, MD 21201
- Corresponding authors: Wanchang Cui, ; Phone: 410-706-5282; Fax: 410-706-5270. Thomas J. MacVittie, ; Phone: 410-706-5274; Fax: 410-706-5270
| | - Kim G. Hankey
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland, USA 21201 10 South Pine Street, MSTF Room 604, Baltimore, MD 21201
| | - Allison M. Gibbs
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland, USA 21201 10 South Pine Street, MSTF Room 604, Baltimore, MD 21201
| | - Cassandra P. Smith
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland, USA 21201 10 South Pine Street, MSTF Room 604, Baltimore, MD 21201
| | - Cheryl Taylor-Howell
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland, USA 21201 10 South Pine Street, MSTF Room 604, Baltimore, MD 21201
| | - Sean R. Kearney
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland, USA 21201 10 South Pine Street, MSTF Room 604, Baltimore, MD 21201
| | - Thomas J. MacVittie
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland, USA 21201 10 South Pine Street, MSTF Room 604, Baltimore, MD 21201
- Corresponding authors: Wanchang Cui, ; Phone: 410-706-5282; Fax: 410-706-5270. Thomas J. MacVittie, ; Phone: 410-706-5274; Fax: 410-706-5270
| |
Collapse
|
69
|
Jones JW, Bennett A, Carter CL, Tudor G, Hankey KG, Farese AM, Booth C, MacVittie TJ, Kane MA. Citrulline as a Biomarker in the Non-human Primate Total- and Partial-body Irradiation Models: Correlation of Circulating Citrulline to Acute and Prolonged Gastrointestinal Injury. HEALTH PHYSICS 2015; 109:440-51. [PMID: 26425904 PMCID: PMC4593331 DOI: 10.1097/hp.0000000000000347] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The use of plasma citrulline as a biomarker for acute and prolonged gastrointestinal injury via exposure to total- and partial-body irradiation (6 MV LINAC-derived photons; 0.80 Gy min) in nonhuman primate models was investigated. The irradiation exposure covered gastrointestinal injuries spanning lethal, mid-lethal, and sub-lethal doses. The acute gastrointestinal injury was assessed via measurement of plasma citrulline and small intestinal histopathology over the first 15 d following radiation exposure and included total-body irradiation at 13.0 Gy, 10.5 Gy, and 7.5 Gy and partial-body irradiation at 11.0 Gy with 5% bone marrow sparing. The dosing schemes of 7.5 Gy total-body irradiation and 11.0 Gy partial-body irradiation included time points out to day 60 and day 180, respectively, which allowed for correlation of plasma citrulline to prolonged gastrointestinal injury and survival. Plasma citrulline values were radiation-dependent for all radiation doses under consideration, with nadir values ranging from 63-80% lower than radiation-naïve NHP plasma. The nadir values were observed at day 5 to 7 post irradiation. Longitudinal plasma citrulline profiles demonstrated prolonged gastrointestinal injury resulting from acute high-dose irradiation had long lasting effects on enterocyte function. Moreover, plasma citrulline did not discriminate between total-body or partial-body irradiation over the first 15 d following irradiation and was not predictive of survival based on the radiation models considered herein.
Collapse
Affiliation(s)
- Jace W. Jones
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD
| | - Alexander Bennett
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD
| | - Claire L. Carter
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD
| | | | - Kim G. Hankey
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD
| | - Ann M. Farese
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD
| | | | - Thomas J. MacVittie
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD
| | - Maureen A. Kane
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD
| |
Collapse
|
70
|
Graessle DH, Dörr H, Bennett A, Shapiro A, Farese AM, MacVittie TJ, Meineke V. Comparing the Hematopoetic Syndrome Time Course in the NHP Animal Model to Radiation Accident Cases From the Database Search. HEALTH PHYSICS 2015; 109:493-501. [PMID: 26425908 DOI: 10.1097/hp.0000000000000355] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Since controlled clinical studies on drug administration for the acute radiation syndrome are lacking, clinical data of human radiation accident victims as well as experimental animal models are the main sources of information. This leads to the question of how to compare and link clinical observations collected after human radiation accidents with experimental observations in non-human primate (NHP) models. Using the example of granulocyte counts in the peripheral blood following radiation exposure, approaches for adaptation between NHP and patient databases on data comparison and transformation are introduced. As a substitute for studying the effects of administration of granulocyte-colony stimulating factor (G-CSF) in human clinical trials, the method of mathematical modeling is suggested using the example of G-CSF administration to NHP after total body irradiation.
Collapse
Affiliation(s)
- Dieter H Graessle
- *Bundeswehr Institute of Radiobiology affiliated to the University of Ulm, Munich, Germany; †University of Maryland, School of Medicine, Dept. of Radiation Oncology, Baltimore, MD; ‡U.S. Food and Drug Administration (FDA), Counter-Terrorism and Emergency Coordination Staff, FDA-CDER, 10001 New Hampshire Ave, Mail Stop 2163, Silver Spring, MD
| | | | | | | | | | | | | |
Collapse
|
71
|
Carter CL, Jones JW, Barrow K, Kieta K, Taylor-Howell C, Kearney S, Smith CP, Gibbs A, Farese AM, MacVittie TJ, Kane MA. A MALDI-MSI Approach to the Characterization of Radiation-Induced Lung Injury and Medical Countermeasure Development. HEALTH PHYSICS 2015; 109:466-78. [PMID: 26425906 PMCID: PMC4745118 DOI: 10.1097/hp.0000000000000353] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Radiation-induced lung injury is highly complex and characterized by multiple pathologies, which occur over time and sporadically throughout the lung. This complexity makes biomarker investigations and medical countermeasure screenings challenging. Matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) has the ability to resolve differences spatially in molecular profiles within the lung following radiation exposure and can aid in biomarker identification and pharmaceutical efficacy investigations. MALDI-MSI was applied to the investigation of a whole-thorax lung irradiation model in non-human primates (NHP) for lipidomic analysis and medical countermeasure distribution.
Collapse
Affiliation(s)
- Claire L. Carter
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences
| | - Jace W. Jones
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences
| | - Kory Barrow
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD 21201
| | - Kaitlyn Kieta
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD 21201
| | - Cheryl Taylor-Howell
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD 21201
| | - Sean Kearney
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD 21201
| | - Cassandra P. Smith
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD 21201
| | - Allison Gibbs
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD 21201
| | - Ann M. Farese
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD 21201
| | - Thomas J. MacVittie
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD 21201
| | - Maureen A. Kane
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences
| |
Collapse
|
72
|
Wang J, Shao L, Hendrickson HP, Liu L, Chang J, Luo Y, Seng J, Pouliot M, Authier S, Zhou D, Allaben W, Hauer-Jensen M. Total Body Irradiation in the "Hematopoietic" Dose Range Induces Substantial Intestinal Injury in Non-Human Primates. Radiat Res 2015; 184:545-53. [PMID: 26495870 DOI: 10.1667/rr14191.1] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The non-human primate has been a useful model for studies of human acute radiation syndrome (ARS). However, to date structural changes in various parts of the intestine after total body irradiation (TBI) have not been systematically studied in this model. Here we report on our current study of TBI-induced intestinal structural injury in the non-human primate after doses typically associated with hematopoietic ARS. Twenty-four non-human primates were divided into three groups: sham-irradiated control group; and total body cobalt-60 (60Co) 6.7 Gy gamma-irradiated group; and total body 60Co 7.4 Gy gamma-irradiated group. After animals were euthanized at day 4, 7 and 12 postirradiation, sections of small intestine (duodenum, proximal jejunum, distal jejunum and ileum) were collected and fixed in 10% formalin. The intestinal mucosal surface length, villus height and crypt depths were assessed by computer-assisted image analysis. Plasma citrulline levels were determined using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Total bone marrow cells were counted and hematopoietic stem/progenitor cells in bone marrow were analyzed by flow cytometer. Histopathologically, all segments exhibited conspicuous disappearance of plicae circulares and prominent atrophy of crypts and villi. Intestinal mucosal surface length was significantly decreased in all intestinal segments on day 4, 7 and 12 after irradiation (P < 0.02-P < 0.001). Villus height was significantly reduced in all segments on day 4 and 7 (P = 0.02-0.005), whereas it had recovered by day 12 (P > 0.05). Crypt depth was also significantly reduced in all segments on day 4, 7 and 12 after irradiation (P < 0.04-P < 0.001). Plasma citrulline levels were dramatically reduced after irradiation, consistent with intestinal mucosal injury. Both 6.7 and 7.4 Gy TBI reduced total number of bone marrow cells. And further analysis showed that the number and function of CD45(+)CD34(+) hematopoietic stem/progenitors in bone marrow decreased significantly. In summary, TBI in the hematopoietic ARS dose range induces substantial intestinal injury in all segments of the small bowel. These findings underscore the importance of maintaining the mucosal barrier that separates the gut microbiome from the body's interior after TBI.
Collapse
Affiliation(s)
- Junru Wang
- a Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Lijian Shao
- a Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Howard P Hendrickson
- a Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Liya Liu
- a Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Jianhui Chang
- a Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Yi Luo
- a Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - John Seng
- a Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | | | | | - Daohong Zhou
- a Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - William Allaben
- a Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Martin Hauer-Jensen
- a Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas.,c Surgical Service, Central Arkansas Veterans Healthcare System, Little Rock, Arkansas
| |
Collapse
|
73
|
Pannkuk EL, Laiakis EC, Authier S, Wong K, Fornace AJ. Global Metabolomic Identification of Long-Term Dose-Dependent Urinary Biomarkers in Nonhuman Primates Exposed to Ionizing Radiation. Radiat Res 2015; 184:121-33. [PMID: 26230079 DOI: 10.1667/rr14091.1] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Due to concerns surrounding potential large-scale radiological events, there is a need to determine robust radiation signatures for the rapid identification of exposed individuals, which can then be used to guide the development of compact field deployable instruments to assess individual dose. Metabolomics provides a technology to process easily accessible biofluids and determine rigorous quantitative radiation biomarkers with mass spectrometry (MS) platforms. While multiple studies have utilized murine models to determine radiation biomarkers, limited studies have profiled nonhuman primate (NHP) metabolic radiation signatures. In addition, these studies have concentrated on short-term biomarkers (i.e., <72 h). The current study addresses the need for biomarkers beyond 72 h using a NHP model. Urine samples were collected at 7 days postirradiation (2, 4, 6, 7 and 10 Gy) and processed with ultra-performance liquid chromatography (UPLC) quadrupole time-of-flight (QTOF) MS, acquiring global metabolomic radiation signatures. Multivariate data analysis revealed clear separation between control and irradiated groups. Thirteen biomarkers exhibiting a dose response were validated with tandem MS. There was significantly higher excretion of l-carnitine, l-acetylcarnitine, xanthine and xanthosine in males versus females. Metabolites validated in this study suggest perturbation of several pathways including fatty acid β oxidation, tryptophan metabolism, purine catabolism, taurine metabolism and steroid hormone biosynthesis. In this novel study we detected long-term biomarkers in a NHP model after exposure to radiation and demonstrate differences between sexes using UPLC-QTOF-MS-based metabolomics technology.
Collapse
Affiliation(s)
- Evan L Pannkuk
- a Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC
| | - Evagelia C Laiakis
- a Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC
| | | | | | - Albert J Fornace
- a Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC;,c Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC; and.,d Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah 22254, Saudi Arabia
| |
Collapse
|
74
|
DeBo RJ, Register TC, Caudell DL, Sempowski GD, Dugan G, Gray S, Owzar K, Jiang C, Bourland JD, Chao NJ, Cline JM. Molecular and cellular profiling of acute responses to total body radiation exposure in ovariectomized female cynomolgus macaques. Int J Radiat Biol 2015; 91:510-8. [PMID: 25786585 DOI: 10.3109/09553002.2015.1028597] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE The threat of radiation exposure requires a mechanistic understanding of radiation-induced immune injury and recovery. The study objective was to evaluate responses to ionizing radiation in ovariectomized (surgically post-menopausal) female cynomolgus macaques. MATERIALS AND METHODS Animals received a single total-body irradiation (TBI) exposure at doses of 0, 2 or 5 Gy with scheduled necropsies at 5 days, 8 weeks and 24 weeks post-exposure. Blood and lymphoid tissues were evaluated for morphologic, cellular, and molecular responses. RESULTS Irradiated animals developed symptoms of acute hematopoietic syndrome, and reductions in thymus weight, thymopoiesis, and bone marrow cellularity. Acute, transient increases in plasma monocyte chemoattractant protein 1 (MCP-1) were observed in 5 Gy animals along with dose-dependent alterations in messenger ribonucleic acid (mRNA) signatures in thymus, spleen, and lymph node. Expression of T cell markers was lower in thymus and spleen, while expression of macrophage marker CD68 (cluster of differentiation 68) was relatively elevated in lymphoid tissues from irradiated animals. CONCLUSIONS Ovariectomized female macaques exposed to moderate doses of radiation experienced increased morbidity, including acute, dose-dependent alterations in systemic and tissue-specific biomarkers, and increased macrophage/T cell ratios. The effects on mortality exceeded expectations based on previous studies in males, warranting further investigation.
Collapse
Affiliation(s)
- Ryne J DeBo
- Department of Pathology, Wake Forest School of Medicine , Winston-Salem, NC , USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Singh VK, Newman VL, Berg AN, MacVittie TJ. Animal models for acute radiation syndrome drug discovery. Expert Opin Drug Discov 2015; 10:497-517. [DOI: 10.1517/17460441.2015.1023290] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
76
|
Kavanagh K, Dendinger MD, Davis AT, Register TC, DeBo R, Dugan G, Cline JM. Type 2 Diabetes is a Delayed Late Effect of Whole-Body Irradiation in Nonhuman Primates. Radiat Res 2015; 183:398-406. [PMID: 25811716 DOI: 10.1667/rr13916.1] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
One newly recognized consequence of radiation exposure may be the delayed development of diabetes and metabolic disease. We document the development of type 2 diabetes in a unique nonhuman primate cohort of monkeys that were whole-body irradiated with high doses (6.5-8.4 Gy) 5-9 years earlier. We report here a higher prevalence of type 2 diabetes in irradiated monkeys compared to age-matched nonirradiated monkeys. These irradiated diabetic primates demonstrate insulin resistance and hypertriglyceridemia, however, they lack the typical obese presentation of primate midlife diabetogenesis. Surprisingly, body composition analyses by computed tomography indicated that prior irradiation led to a specific loss of visceral fat mass. Prior irradiation led to reductions in insulin signaling effectiveness in skeletal muscle and higher monocyte chemoattractant protein 1 levels, indicative of increased inflammation. However, there was an absence of large defects in pancreatic function with radiation exposure, which has been documented previously in animal and human studies. Monkeys that remained healthy and did not become diabetic in the years after irradiation were significantly leaner and smaller, and were generally smaller and younger at the time of exposure. Irradiation also resulted in smaller stature in both diabetic and nondiabetic monkeys, compared to nonirradiated age-matched controls. Our study demonstrates that diabetogenesis postirradiation is not a consequence of disrupted adipose accumulation (generalized or in ectopic depots), nor generalized pancreatic failure, but suggests that peripheral tissues such as the musculature are impaired in their response to insulin exposure. Ongoing inflammation in these animals appears to be a consequence of radiation exposure and can interfere with insulin signaling. The reasons that some animals remain protected from diabetes as a late effect of irradiation are not clear, but may be related to body size. The translational relevance for these results suggest that muscle may be an important and underappreciated target organ for the delayed late effect of whole-body irradiation, leading to increased risk of insulin resistance and diabetes development.
Collapse
Affiliation(s)
- Kylie Kavanagh
- Wake Forest School of Medicine, Department of Pathology, Winston-Salem, North Carolina
| | | | | | | | | | | | | |
Collapse
|
77
|
Eaton EB, Varney TR. Mesenchymal stem cell therapy for acute radiation syndrome: innovative medical approaches in military medicine. Mil Med Res 2015; 2:2. [PMID: 25722881 PMCID: PMC4340678 DOI: 10.1186/s40779-014-0027-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 11/20/2014] [Indexed: 01/03/2023] Open
Abstract
After a radiological or nuclear event, acute radiation syndrome (ARS) will present complex medical challenges that could involve the treatment of hundreds to thousands of patients. Current medical doctrine is based on limited clinical data and remains inadequate. Efforts to develop medical innovations that address ARS complications are unlikely to be generated by industry because of market uncertainties specific to this type of injury. A prospective strategy could be the integration of cellular therapy to meet the medical demands of ARS. The most clinically advanced cellular therapy to date is the administration of mesenchymal stem cells (MSCs). Results of currently published investigations describing MSC safety and efficacy in a variety of injury and disease models demonstrate the unique qualities of this reparative cell population in adapting to the specific requirements of the damaged tissue in which the cells integrate. This report puts forward a rationale for the further evaluation of MSC therapy to address the current unmet medical needs of ARS. We propose that the exploration of this novel therapy for the treatment of the multivariate complications of ARS could be of invaluable benefit to military medicine.
Collapse
Affiliation(s)
- Erik B Eaton
- United States Army Medical Research Institute of Chemical Defense, 3100 Ricketts Point Road, Aberdeen Proving Ground, Maryland, 21010 US
| | - Timothy R Varney
- United States Army Medical Research Institute of Chemical Defense, 3100 Ricketts Point Road, Aberdeen Proving Ground, Maryland, 21010 US
| |
Collapse
|
78
|
Ha CT, Li XH, Fu D, Moroni M, Fisher C, Arnott R, Srinivasan V, Xiao M. Circulating interleukin-18 as a biomarker of total-body radiation exposure in mice, minipigs, and nonhuman primates (NHP). PLoS One 2014; 9:e109249. [PMID: 25290447 PMCID: PMC4188589 DOI: 10.1371/journal.pone.0109249] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 09/04/2014] [Indexed: 12/21/2022] Open
Abstract
We aim to develop a rapid, easy-to-use, inexpensive and accurate radiation dose-assessment assay that tests easily obtained samples (e.g., blood) to triage and track radiological casualties, and to evaluate the radioprotective and therapeutic effects of radiation countermeasures. In the present study, we evaluated the interleukin (IL)-1 family of cytokines, IL-1β, IL-18 and IL-33, as well as their secondary cytokines’ expression and secretion in CD2F1 mouse bone marrow (BM), spleen, thymus and serum in response to γ-radiation from sublethal to lethal doses (5, 7, 8, 9, 10, or 12 Gy) at different time points using the enzyme-linked immune sorbent assay (ELISA), immunoblotting, and cytokine antibody array. Our data identified increases of IL-1β, IL-18, and/or IL-33 in mouse thymus, spleen and BM cells after total-body irradiation (TBI). However, levels of these cytokines varied in different tissues. Interestingly, IL-18 but not IL-1β or IL-33 increased significantly (2.5–24 fold) and stably in mouse serum from day 1 after TBI up to 13 days in a radiation dose-dependent manner. We further confirmed our finding in total-body γ-irradiated nonhuman primates (NHPs) and minipigs, and demonstrated that radiation significantly enhanced IL-18 in serum from NHPs 2–4 days post-irradiation and in minipig plasma 1–3 days post-irradiation. Finally, we compared circulating IL-18 with the well known hematological radiation biomarkers lymphocyte and neutrophil counts in blood of mouse, minipigs and NHPs and demonstrated close correlations between these biomarkers in response to radiation. Our results suggest that the elevated levels of circulating IL-18 after radiation proportionally reflect radiation dose and severity of radiation injury and may be used both as a potential biomarker for triage and also to track casualties after radiological accidents as well as for therapeutic radiation exposure.
Collapse
Affiliation(s)
- Cam T. Ha
- Radiation Countermeasures Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Xiang-Hong Li
- Radiation Countermeasures Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Dadin Fu
- Radiation Countermeasures Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Maria Moroni
- Radiation Countermeasures Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Carolyn Fisher
- Radiation Countermeasures Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Robert Arnott
- Radiation Countermeasures Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Venkataraman Srinivasan
- Radiation Countermeasures Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Mang Xiao
- Radiation Countermeasures Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
79
|
Moulder JE. 2013 Dade W. Moeller lecture: medical countermeasures against radiological terrorism. HEALTH PHYSICS 2014; 107:164-71. [PMID: 24978287 PMCID: PMC4076685 DOI: 10.1097/hp.0000000000000082] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Soon after the 9-11 attacks, politicians and scientists began to question our ability to cope with a large-scale radiological terrorism incident. The outline of what was needed was fairly obvious: the ability to prevent such an attack, methods to cope with the medical consequences, the ability to clean up afterward, and the tools to figure out who perpetrated the attack and bring them to justice. The medical response needed three components: the technology to determine rapidly the radiation doses received by a large number of people, methods for alleviating acute hematological radiation injuries, and therapies for mitigation and treatment of chronic radiation injuries. Research done to date has shown that a realistic medical response plan is scientifically possible, but the regulatory and financial barriers to achieving this may currently be insurmountable.
Collapse
Affiliation(s)
- John E. Moulder
- Center for Medical Countermeasures Against Radiological Terrorism, Radiation Oncology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, Wisconsin, 53226 U. S. A
| |
Collapse
|
80
|
Development and validation of a LC-MS/MS assay for quantitation of plasma citrulline for application to animal models of the acute radiation syndrome across multiple species. Anal Bioanal Chem 2014; 406:4663-75. [DOI: 10.1007/s00216-014-7870-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 04/24/2014] [Accepted: 04/29/2014] [Indexed: 10/25/2022]
|
81
|
Dörr H, Lamkowski A, Graessle DH, Bennett A, Shapiro A, Farese AM, Garofalo M, MacVittie TJ, Meineke V. Linking the human response to unplanned radiation and treatment to the nonhuman primate response to controlled radiation and treatment. HEALTH PHYSICS 2014; 106:129-34. [PMID: 24276556 PMCID: PMC3843145 DOI: 10.1097/hp.0b013e3182a12de0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
A key difficulty in developing countermeasures against radiation-induced health impairments is the clear lack of controlled clinical studies, due to the relatively low number of radiation victims worldwide. Instead, established and accepted animal models, as well as the recommendations of national and international expert panels and committees, are the main sources of information. Therefore, the development of countermeasures requires comparison of data from many sources and accumulation of information consistent with the U. S. Food and Drug Administration's "Animal Rule." A new approach is the comparative analysis of human data from the SEARCH (System for Evaluation and Archiving of Radiation Accidents based on Case Histories) database and data from nonhuman primate (NHP) animal model studies. The SEARCH database contains 824 clinical cases from 81 radiation accidents in 19 countries. This exceptional collection of clinical data from accidentally radiation-exposed persons is analyzed regarding clinical signs and symptoms of radiation-induced health impairments. To analyze the time course of radiation syndromes, clinical parameters common to the SEARCH and NHP databases have to be assigned into comparable categories of clinical severity for each species. The goal is to establish a method for comparison of human and NHP data, validate the NHP data as a surrogate for human efficacy/clinical studies, and open a way for the extraction of diagnostic and treatment methods for humans after radiation exposure according to relevant regulations.
Collapse
Affiliation(s)
- Harald Dörr
- *Bundeswehr Institute of Radiobiology affiliated to the University of Ulm, Munich, Germany; †University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD; ‡U.S. Food and Drug Administration (FDA), Silver Spring, MD
| | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Garofalo M, Bennett A, Farese AM, Harper J, Ward A, Taylor-Howell C, Cui W, Gibbs A, Lasio G, Jackson W, MacVittie TJ. The delayed pulmonary syndrome following acute high-dose irradiation: a rhesus macaque model. HEALTH PHYSICS 2014; 106:56-72. [PMID: 24276550 DOI: 10.1097/hp.0b013e3182a32b3f] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Several radiation dose- and time-dependent tissue sequelae develop following acute high-dose radiation exposure. One of the recognized delayed effects of such exposures is lung injury, characterized by respiratory failure as a result of pneumonitis that may subsequently develop into lung fibrosis. Since this pulmonary subsyndrome may be associated with high morbidity and mortality, comprehensive treatment following high-dose irradiation will ideally include treatments that mitigate both the acute hematologic and gastrointestinal subsyndromes as well as the delayed pulmonary syndrome. Currently, there are no drugs approved by the Food and Drug Administration to counteract the effects of acute radiation exposure. Moreover, there are no relevant large animal models of radiation-induced lung injury that permit efficacy testing of new generation medical countermeasures in combination with medical management protocols under the FDA animal rule criteria. Herein is described a nonhuman primate model of delayed lung injury resulting from whole thorax lung irradiation. Rhesus macaques were exposed to 6 MV photon radiation over a dose range of 9.0-12.0 Gy and medical management administered according to a standardized treatment protocol. The primary endpoint was all-cause mortality at 180 d. A comparative multiparameter analysis is provided, focusing on the lethal dose response relationship characterized by a lethal dose50/180 of 10.27 Gy [9.88, 10.66] and slope of 1.112 probits per linear dose. Latency, incidence, and severity of lung injury were evaluated through clinical and radiographic parameters including respiratory rate, saturation of peripheral oxygen, corticosteroid requirements, and serial computed tomography. Gross anatomical and histological analyses were performed to assess radiation-induced injury. The model defines the dose response relationship and time course of the delayed pulmonary sequelae and consequent morbidity and mortality. Therefore, it may provide an effective platform for the efficacy testing of candidate medical countermeasures against the delayed pulmonary syndrome.
Collapse
Affiliation(s)
- Michael Garofalo
- *University of Maryland, School of Medicine, Department of Radiation Oncology; †Statistician, Rockville, MD
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Scott AJ, Jones JW, Orschell CM, MacVittie TJ, Kane MA, Ernst RK. Mass spectrometry imaging enriches biomarker discovery approaches with candidate mapping. HEALTH PHYSICS 2014; 106:120-8. [PMID: 24276555 PMCID: PMC4015108 DOI: 10.1097/hp.0b013e3182a4ec2f] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Integral to the characterization of radiation-induced tissue damage is the identification of unique biomarkers. Biomarker discovery is a challenging and complex endeavor requiring both sophisticated experimental design and accessible technology. The resources within the National Institute of Allergy and Infectious Diseases (NIAID)-sponsored Consortium, Medical Countermeasures Against Radiological Threats (MCART), allow for leveraging robust animal models with novel molecular imaging techniques. One such imaging technique, MALDI (matrix-assisted laser desorption ionization) mass spectrometry imaging (MSI), allows for the direct spatial visualization of lipids, proteins, small molecules, and drugs/drug metabolites-or biomarkers-in an unbiased manner. MALDI-MSI acquires mass spectra directly from an intact tissue slice in discrete locations across an x, y grid that are then rendered into a spatial distribution map composed of ion mass and intensity. The unique mass signals can be plotted to generate a spatial map of biomarkers that reflects pathology and molecular events. The crucial unanswered questions that can be addressed with MALDI-MSI include identification of biomarkers for radiation damage that reflect the response to radiation dose over time and the efficacy of therapeutic interventions. Techniques in MALDI-MSI also enable integration of biomarker identification among diverse animal models. Analysis of early, sublethally irradiated tissue injury samples from diverse mouse tissues (lung and ileum) shows membrane phospholipid signatures correlated with histological features of these unique tissues. This paper will discuss the application of MALDI-MSI for use in a larger biomarker discovery pipeline.
Collapse
Affiliation(s)
| | - Jace W. Jones
- University of Maryland, School of Pharmacy, Baltimore, MD
| | | | | | | | | |
Collapse
|
84
|
Garofalo MC, Ward AA, Farese AM, Bennett A, Taylor-Howell C, Cui W, Gibbs A, Prado KL, MacVittie TJ. A pilot study in rhesus macaques to assess the treatment efficacy of a small molecular weight catalytic metalloporphyrin antioxidant (AEOL 10150) in mitigating radiation-induced lung damage. HEALTH PHYSICS 2014; 106:73-83. [PMID: 24276551 DOI: 10.1097/hp.0b013e3182a4d967] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
The objective of this pilot study was to explore whether administration of a catalytic antioxidant, AEOL 10150 (C48H56C15MnN12), could reduce radiation-induced lung injury and improve overall survival when administered after 11.5 Gy of whole thorax lung irradiation in a non-human primate model. Thirteen animals were irradiated with a single exposure of 11.5 Gy, prescribed to midplane, and delivered with 6 MV photons at a dose rate of 0.8 Gy min. Beginning at 24 h post irradiation, the AEOL 10150 cohort (n = 7) received daily subcutaneous injections of the catalytic antioxidant at a concentration of 5 mg kg for a total of 4 wk. All animals received medical management, including dexamethasone, based on clinical signs during the planned 180-d in-life phase of the study. All decedent study animals were euthanized for failure to maintain saturation of peripheral oxygen > 88% on room air. Exposure of the whole thorax to 11.5 Gy resulted in radiation-induced lung injury in all animals. AEOL 10150, as administered in this pilot study, demonstrated potential efficacy as a mitigator against fatal radiation-induced lung injury. Treatment with the drug resulted in 28.6% survival following exposure to a radiation dose that proved to be 100% fatal in the control cohort (n = 6). Computed tomography scans demonstrated less quantitative radiographic injury (pneumonitis, fibrosis, effusions) in the AEOL 10150-treated cohort at day 60 post-exposure, and AEOL 10150-treated animals required less dexamethasone support during the in-life phase of the study. Analysis of serial plasma samples suggested that AEOL 10150 treatment led to lower relative transforming growth factor-Beta-1 levels when compared with the control animals. The results of this pilot study demonstrate that treatment with AEOL 10150 results in reduced clinical, radiographic, anatomic, and molecular evidence of radiation-induced lung injury and merits further study as a medical countermeasure against radiation-induced pulmonary injury.
Collapse
Affiliation(s)
- Michael C Garofalo
- *University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD 21201
| | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Affiliation(s)
- Thomas J MacVittie
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD
| |
Collapse
|
86
|
Kazi AM, MacVittie TJ, Lasio G, Lu W, Prado KL. The MCART radiation physics core: the quest for radiation dosimetry standardization. HEALTH PHYSICS 2014; 106:97-105. [PMID: 24276553 PMCID: PMC3899940 DOI: 10.1097/hp.0b013e3182a2a987] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Dose-related radiobiological research results can only be compared meaningfully when radiation dosimetry is standardized. To this purpose, the National Institute of Allergy and Infectious Diseases (NIAID)-sponsored Medical Countermeasures Against Radiological Threats (MCART) consortium recently created a Radiation Physics Core (RPC) as an entity to assume responsibility of standardizing radiation dosimetry practices among its member laboratories. The animal research activities in these laboratories use a variety of ionizing photon beams from several irradiators such as 250-320 kVp x-ray generators, Cs irradiators, Co teletherapy machines, and medical linear accelerators (LINACs). In addition to this variety of sources, these centers use a range of irradiation techniques and make use of different dose calculation schemes to conduct their experiments. An extremely important objective in these research activities is to obtain a Dose Response Relationship (DRR) appropriate to their respective organ-specific models of acute and delayed radiation effects. A clear and unambiguous definition of the DRR is essential for the development of medical countermeasures. It is imperative that these DRRs are transparent between centers. The MCART RPC has initiated the establishment of standard dosimetry practices among member centers and is introducing a Remote Dosimetry Monitoring Service (RDMS) to ascertain ongoing quality assurance. This paper will describe the initial activities of the MCART RPC toward implementing these standardization goals. It is appropriate to report a summary of initial activities with the intent of reporting the full implementation at a later date.
Collapse
Affiliation(s)
- Abdul M. Kazi
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD
| | - Thomas J. MacVittie
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD
| | - Giovanni Lasio
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD
| | - Wei Lu
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD
| | - Karl L. Prado
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD
| |
Collapse
|
87
|
MacVittie TJ, Bennett AW, V Cohen M, Farese AM, Higgins A, Hankey KG. Immune cell reconstitution after exposure to potentially lethal doses of radiation in the nonhuman primate. HEALTH PHYSICS 2014; 106:84-96. [PMID: 24276552 DOI: 10.1097/hp.0b013e3182a2a9b2] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Delayed immune reconstitution remains a major cause of morbidity associated with myelosuppression induced by cytotoxic therapy or myeloablative conditioning for stem cell transplant, as well as potentially lethal doses of total- or partial-body irradiation. Restoration of a functional immune cell repertoire requires hematopoietic stem cell reconstitution for all immune cells and effective thymopoiesis for T cell recovery. There are no medical countermeasures available to mitigate damage consequent to high-dose, potentially lethal irradiation, and there are no well characterized large animal models of prolonged immunosuppression to assess efficacy of potential countermeasures. Herein, the authors describe a model of T and B cell reconstitution following lethal doses of partial-body irradiation with 5% bone marrow sparing that includes full exposure of the thymus. Rhesus macaques (n = 31 male, 5.5-11.3 kg body weight) were exposed to midline tissue doses of 9.0-12.0 Gy using 6 MV LINAC-derived photons at a dose rate of 0.80 Gy min, sparing approximately 5% of bone marrow (tibiae, ankles, and feet). All animals received medical management and were monitored for myeloid and lymphoid suppression and recovery through 180 d post-exposure. Myeloid recovery was assessed by neutrophil and platelet-related hematological parameters. Reconstitution of B and T cell subsets was assessed by flow cytometric immunophenotyping, and recent thymic emigrants were identified by RT-PCR of T cell receptor excision circles. Mortality was recorded through 180 d post-exposure. Acute myelo-suppression was characterized by severe neutropenia and thrombocytopenia, followed by recovery 30-60 d post-exposure. Total T (CD3+) and B (CD20+) cells were reduced significantly following exposure and exhibited differential recovery patterns post-exposure. Both CD4+ and CD8+ subsets of naïve T cells and total CD4+ T cell counts remained significantly lower than baseline through 180 d post-exposure. The failure of recent thymic emigrants and naïve T cell subsets to recover to normal baseline values reflects the severe radiation effects on the recovery of marrow-derived stem and early thymic progenitor cells, their mobilization and seeding of receptive thymic niches, and slow endogenous thymic regeneration.
Collapse
Affiliation(s)
- Thomas J MacVittie
- *University of Maryland, School of Medicine, Dept. of Radiation Oncology, Baltimore, MD; †Integrated Research Facility, Frederick, MD; ‡Naval Medical Research Center, Silver Spring, MD
| | | | | | | | | | | |
Collapse
|
88
|
Rios CI, Cassatt DR, Dicarlo AL, Macchiarini F, Ramakrishnan N, Norman MK, Maidment BW. Building the strategic national stockpile through the NIAID Radiation Nuclear Countermeasures Program. Drug Dev Res 2013; 75:23-8. [PMID: 24648046 DOI: 10.1002/ddr.21163] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The possibility of a public health radiological or nuclear emergency in the United States remains a concern. Media attention focused on lost radioactive sources and international nuclear threats, as well as the potential for accidents in nuclear power facilities (e.g., Windscale, Three Mile Island, Chernobyl, and Fukushima) highlight the need to address this critical national security issue. To date, no drugs have been licensed to mitigate/treat the acute and long-term radiation injuries that would result in the event of large-scale, radiation, or nuclear public health emergency. However, recent evaluation of several candidate radiation medical countermeasures (MCMs) has provided initial proof-of-concept of efficacy. The goal of the Radiation Nuclear Countermeasures Program (RNCP) of the National Institute of Allergy and Infectious Diseases (National Institutes of Health) is to help ensure the government stockpiling of safe and efficacious MCMs to treat radiation injuries, including, but not limited to, hematopoietic, gastrointestinal, pulmonary, cutaneous, renal, cardiovascular, and central nervous systems. In addition to supporting research in these areas, the RNCP continues to fund research and development of decorporation agents targeting internal radionuclide contamination, and biodosimetry platforms (e.g., biomarkers and devices) to assess the levels of an individual's radiation exposure, capabilities that would be critical in a mass casualty scenario. New areas of research within the program include a focus on special populations, especially pediatric and geriatric civilians, as well as combination studies, in which drugs are tested within the context of expected medical care management (e.g., antibiotics and growth factors). Moving forward, challenges facing the RNCP, as well as the entire radiation research field, include further advancement and qualification of animal models, dose conversion from animal models to humans, biomarker identification, and formulation development. This paper provides a review of recent work and collaborations supported by the RNCP.
Collapse
Affiliation(s)
- Carmen I Rios
- Radiation Nuclear Countermeasures Program, Division of Allergy, Immunology and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | | | | | | | | | | | | |
Collapse
|
89
|
Li XH, Ghosh SP, Ha CT, Fu D, Elliott TB, Bolduc DL, Villa V, Whitnall MH, Landauer MR, Xiao M. Delta-Tocotrienol Protects Mice from Radiation-Induced Gastrointestinal Injury. Radiat Res 2013; 180:649-57. [DOI: 10.1667/rr13398.1] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
90
|
Evaluation of the gamma-H2AX assay for radiation biodosimetry in a swine model. Int J Mol Sci 2013; 14:14119-35. [PMID: 23880859 PMCID: PMC3742235 DOI: 10.3390/ijms140714119] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Revised: 06/18/2013] [Accepted: 06/25/2013] [Indexed: 02/07/2023] Open
Abstract
There is a paucity of large animal models to study both the extent and the health risk of ionizing radiation exposure in humans. One promising candidate for such a model is the minipig. Here, we evaluate the minipig for its potential in γ-H2AX-based biodosimetry after exposure to ionizing radiation using both Cs137 and Co60 sources. γ-H2AX foci were enumerated in blood lymphocytes and normal fibroblasts of human and porcine origin after ex vivo γ-ray irradiation. DNA double-strand break repair kinetics in minipig blood lymphocytes and fibroblasts, based on the γ-H2AX assay, were similar to those observed in their human counterparts. To substantiate the similarity observed between the human and minipig we show that minipig fibroblast radiosensitivity was similar to that observed with human fibroblasts. Finally, a strong γ-H2AX induction was observed in blood lymphocytes following minipig total body irradiation. Significant responses were detected 3 days after 1.8 Gy and 1 week after 3.8 and 5 Gy with residual γ-H2AX foci proportional to the initial radiation doses. These findings show that the Gottingen minipig provides a useful in vivo model for validation of γ-H2AX biodosimetry for dose assessment in humans.
Collapse
|
91
|
MacVittie TJ, Bennett A, Booth C, Garofalo M, Tudor G, Ward A, Shea-Donohue T, Gelfond D, McFarland E, Jackson W, Lu W, Farese AM. The prolonged gastrointestinal syndrome in rhesus macaques: the relationship between gastrointestinal, hematopoietic, and delayed multi-organ sequelae following acute, potentially lethal, partial-body irradiation. HEALTH PHYSICS 2012; 103:427-53. [PMID: 22929471 PMCID: PMC4140097 DOI: 10.1097/hp.0b013e318266eb4c] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
The dose response relationship for the acute gastrointestinal syndrome following total-body irradiation prevents analysis of the full recovery and damage to the gastrointestinal system, since all animals succumb to the subsequent 100% lethal hematopoietic syndrome. A partial-body irradiation model with 5% bone marrow sparing was established to investigate the prolonged effects of high-dose radiation on the gastrointestinal system, as well as the concomitant hematopoietic syndrome and other multi-organ injury including the lung. Herein, cellular and clinical parameters link acute and delayed coincident sequelae to radiation dose and time course post-exposure. Male rhesus Macaca mulatta were exposed to partial-body irradiation with 5% bone marrow (tibiae, ankles, feet) sparing using 6 MV linear accelerator photons at a dose rate of 0.80 Gy min(-1) to midline tissue (thorax) doses in the exposure range of 9.0 to 12.5 Gy. Following irradiation, all animals were monitored for multiple organ-specific parameters for 180 d. Animals were administered medical management including administration of intravenous fluids, antiemetics, prophylactic antibiotics, blood transfusions, antidiarrheals, supplemental nutrition, and analgesics. The primary endpoint was survival at 15, 60, or 180 d post-exposure. Secondary endpoints included evaluation of dehydration, diarrhea, hematologic parameters, respiratory distress, histology of small and large intestine, lung radiographs, and mean survival time of decedents. Dose- and time-dependent mortality defined several organ-specific sequelae, with LD50/15 of 11.95 Gy, LD50/60 of 11.01 Gy, and LD50/180 of 9.73 Gy for respective acute gastrointestinal, combined hematopoietic and gastrointestinal, and multi-organ delayed injury to include the lung. This model allows analysis of concomitant multi-organ sequelae, thus providing a link between acute and delayed radiation effects. Specific and multi-organ medical countermeasures can be assessed for efficacy and interaction during the concomitant evolution of acute and delayed key organ-specific subsyndromes.
Collapse
Affiliation(s)
- Thomas J MacVittie
- Department of Radiation Oncology, University of Maryland, School of Medicine, Baltimore, MD 21201, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|