1
|
Mitra D, Armijo GK, Ober EH, Baker SM, Turner HC, Broustas CG. MIIST305 mitigates gastrointestinal acute radiation syndrome injury and ameliorates radiation-induced gut microbiome dysbiosis. Gut Microbes 2025; 17:2458189. [PMID: 39930324 PMCID: PMC11817531 DOI: 10.1080/19490976.2025.2458189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/07/2025] [Accepted: 01/21/2025] [Indexed: 02/14/2025] Open
Abstract
High-dose radiation exposure results in gastrointestinal (GI) acute radiation syndrome identified by the destruction of mucosal layer, intestinal growth barrier dysfunction, and aberrant inflammatory responses. Further, radiation causes gut microbiome dysbiosis characterized by diminished microbial diversity, mostly commensal bacteria, and the spread of bacterial pathogens that trigger the recruitment of immune cells and the production of pro-inflammatory factors that lead to further GI tissue damage. Currently, there are no U.S. Food and Drug Administration (FDA) approved countermeasures that can treat radiation-induced GI injuries. To meet this critical need, Synedgen Inc. has developed a glycopolymer radiomitigator (MIIST305) that is specifically targeted to the GI tract, which acts by intercalating into the mucus layer and the glycocalyx of intestinal epithelial cells that could potentially ameliorate the deleterious effects of radiation. Male C57BL/6J adult mice were exposed to 13 Gy partial body X-irradiation with 5% bone marrow shielding and MIIST305 was administered on days 1, 3, and 5 post-irradiation. Approximately 85% of the animals survived the irradiation exposure and were apparently healthy until the end of the 30-day study period. In contrast, no control, Vehicle-treated animals survived past day 10 at this radiation dose. We show that MIIST305 improved intestinal epithelial barrier function and suppressed systemic inflammatory responses mediated by radiation-induced pro-inflammatory cytokines. Taxonomic profiling and community structure of the fecal and colonic mucosa microbiota demonstrated that MIIST305 treatment increased microbial diversity and restored abundance of beneficial commensal bacteria, including Lactobacillus and Bifidobacterium genera while suppressing potentially pathogenic bacteria Enterococcus and Staphylococcus compared with Vehicle-treated animals. In summary, MIIST305 is a novel GI-targeted therapeutic that greatly enhances survival in mice exposed to lethal radiation and protects the GI tract from injury by restoring a balanced gut microbiota and reducing pro-inflammatory responses. Further development of this drug as an FDA-approved medical countermeasure is of critical importance.
Collapse
Affiliation(s)
- Debmalya Mitra
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Gabriel K. Armijo
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Elizabeth H. Ober
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Helen C. Turner
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Constantinos G. Broustas
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
2
|
Sproull M, Camphausen K. Partial-body Models of Radiation Exposure. Radiat Res 2025; 203:129-141. [PMID: 39923796 PMCID: PMC11973700 DOI: 10.1667/rade-24-00189.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 01/29/2025] [Indexed: 02/11/2025]
Abstract
The events of 9/11 sparked a revitalization of civil defense in the U.S. for emergency planning and preparedness for future radiological or nuclear event scenarios and specifically for mass casualty medical management of radiation exposure and injury. Research in medical countermeasure development in the form of novel pharmaceuticals to treat radiation injury and new radiation biodosimetry diagnostics, primarily focused on development of research models of uniform total-body irradiation (TBI). With the success of those models, it was recognized that most radiation exposures in the field will involve non-uniform heterogeneous irradiations and many partial-body or organ-specific irradiation models have been utilized. This review examines partial-body models of irradiations developed in the last decade for heterogeneous radiation exposures and organ-specific radiation exposure patterns. These research models have been used to further our understanding of radiation injury, novel medical countermeasures and biodosimetry diagnostics in development for future radiological and nuclear event scenarios.
Collapse
Affiliation(s)
- M. Sproull
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland
| | - K. Camphausen
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
3
|
Mitra D, Armijo GK, Ober EH, Baker SM, Turner HC, Broustas CG. MIIST305 mitigates gastrointestinal acute radiation syndrome injury and ameliorates radiation-induced gut microbiome dysbiosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619652. [PMID: 39484519 PMCID: PMC11526895 DOI: 10.1101/2024.10.22.619652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
High-dose radiation exposure results in gastrointestinal (GI) acute radiation syndrome identified by the destruction of mucosal layer, intestinal epithelial barrier dysfunction, and aberrant inflammatory responses. In addition, radiation causes gut microbiome dysbiosis characterized by diminished microbial diversity, reduction in the abundance of beneficial commensal bacteria, and the spread of bacterial pathogens that trigger the recruitment of immune cells and the production of pro-inflammatory factors that lead to further GI tissue damage. Currently, there are no FDA-approved countermeasures that can treat radiation-induced GI injury. To meet this critical need, Synedgen Inc., has developed a glycopolymer radiomitigator (MIIST305) that is specifically targeted to the GI tract which acts by intercalating into the mucus layer and the glycocalyx of intestinal epithelial cells that could potentially ameliorate the deleterious effects of radiation. Male C57BL/6J adult mice were exposed to 13 Gy total body X-irradiation with 5% bone marrow shielding and MIIST305 was administered on days 1, 3, and 5 post-irradiation. Approximately 85% of the animals survived the irradiation exposure and were apparently healthy until the end of the 30-day study period. In contrast, no control, vehicle-treated animals survived past day 10 at this radiation dose. We show that MIIST305 improved intestinal epithelial barrier function and suppressed systemic inflammatory response mediated by radiation-induced pro-inflammatory cytokines. Taxonomic profiling and community structure of the fecal and colonic mucosa microbiota demonstrated that MIIST305 treatment increased microbial diversity and restored abundance of beneficial commensal bacteria, including Lactobacillus and Bifidobacterium genera, while suppressing potentially pathogenic bacteria compared with vehicle-treated animals. In summary, MIIST305 is a novel GI-targeted therapeutic that greatly enhances survival in mice exposed to lethal radiation and protects the GI tract from injury by restoring a balanced gut microbiota and effectively reducing proinflammatory responses. Further development of this drug as an FDA-approved medical countermeasure will be of critical importance in the event of a radiation public health emergency.
Collapse
Affiliation(s)
- Debmalya Mitra
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Gabriel K. Armijo
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Elizabeth H. Ober
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Helen C. Turner
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Constantinos G. Broustas
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
4
|
Singh VK, Wise SY, Fatanmi OO, Petrus SA, Carpenter AD, Lugo-Roman LA, Lee SH, Hauer-Jensen M, Seed TM. Pathology of acute sub-lethal or near-lethal irradiation of nonhuman primates prophylaxed with the nutraceutical, gamma tocotrienol. Sci Rep 2024; 14:13315. [PMID: 38858439 PMCID: PMC11164941 DOI: 10.1038/s41598-024-64102-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 06/05/2024] [Indexed: 06/12/2024] Open
Abstract
Exposure to high, marginally lethal doses or higher of ionizing radiation, either intentional or accidental, results in injury to various organs. Currently, there is only a limited number of safe and effective radiation countermeasures approved by US Food and Drug Administration for such injuries. These approved agents are effective for only the hematopoietic component of the acute radiation syndrome and must be administered only after the exposure event: currently, there is no FDA-approved agent that can be used prophylactically. The nutraceutical, gamma-tocotrienol (GT3) has been found to be a promising radioprotector of such exposure-related injuries, especially those of a hematopoietic nature, when tested in either rodents or nonhuman primates. We investigated the nature of injuries and the possible protective effects of GT3 within select organ systems/tissues caused by both non-lethal level (4.0 Gy), as well as potentially lethal level (5.8 Gy) of ionizing radiation, delivered as total-body or partial-body exposure. Results indicated that the most severe, dose-dependent injuries occurred within those organ systems with strong self-renewing capacities (e.g., the lymphohematopoietic and gastrointestinal systems), while in other tissues (e.g., liver, kidney, lung) endowed with less self-renewal, the pathologies noted tended to be less pronounced and less dependent on the level of exposure dose or on the applied exposure regimen. The prophylactic use of the test nutraceutical, GT3, appeared to limit the extent of irradiation-associated pathology within blood forming tissues and, to some extent, within the small intestine of the gastrointestinal tract. No distinct, global pattern of bodily protection was noted with the agent's use, although a hint of a possible radioprotective benefit was suggested not only by a lessening of apparent injury within select organ systems, but also by way of noting the lack of early onset of moribundity within select GT3-treated animals.
Collapse
Affiliation(s)
- Vijay K Singh
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814-2712, USA.
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.
| | - Stephen Y Wise
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814-2712, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Oluseyi O Fatanmi
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814-2712, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Sarah A Petrus
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814-2712, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Alana D Carpenter
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814-2712, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Luis A Lugo-Roman
- Department of Laboratory Animal Resources, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Sang-Ho Lee
- Pathology Department, Research Services, Naval Medical Research Center, Silver Spring, MD, 20910, USA
| | - Martin Hauer-Jensen
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Thomas M Seed
- Tech Micro Services, 4417 Maple Avenue, Bethesda, MD, 20814, USA
| |
Collapse
|
5
|
Singh VK, Wise SY, Fatanmi OO, Petrus SA, Carpenter AD, Lee SH, Hauer-Jensen M, Seed TM. Histopathological studies of nonhuman primates exposed to supralethal doses of total- or partial-body radiation: influence of a medical countermeasure, gamma-tocotrienol. Sci Rep 2024; 14:5757. [PMID: 38459144 PMCID: PMC10923821 DOI: 10.1038/s41598-024-56135-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 03/01/2024] [Indexed: 03/10/2024] Open
Abstract
Despite remarkable scientific progress over the past six decades within the medical arts and in radiobiology in general, limited radiation medical countermeasures (MCMs) have been approved by the United States Food and Drug Administration for the acute radiation syndrome (ARS). Additional effort is needed to develop large animal models for improving the prediction of clinical safety and effectiveness of MCMs for acute and delayed effects of radiation in humans. Nonhuman primates (NHPs) are considered the animal models that reproduce the most appropriate representation of human disease and are considered the gold standard for drug development and regulatory approval. The clinical and histopathological effects of supralethal, total- or partial-body irradiations (12 Gy) of NHPs were assessed, along with possible protective actions of a promising radiation MCM, gamma-tocotrienol (GT3). Results show that these supralethal radiation exposures induce severe injuries that manifest both clinically as well as pathologically, as evidenced by the noted functionally crippling lesions within various major organ systems of experimental NHPs. The MCM, GT3, has limited radioprotective efficacy against such supralethal radiation doses.
Collapse
Affiliation(s)
- Vijay K Singh
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814-2712, USA.
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.
| | - Stephen Y Wise
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814-2712, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Oluseyi O Fatanmi
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814-2712, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Sarah A Petrus
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814-2712, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Alana D Carpenter
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814-2712, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Sang-Ho Lee
- Pathology Department, Research Services, Naval Medical Research Center, Silver Spring, MD, 20910, USA
| | - Martin Hauer-Jensen
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Thomas M Seed
- Tech Micro Services, 4417 Maple Avenue, Bethesda, MD, 20814, USA
| |
Collapse
|
6
|
Molinar-Inglis O, DiCarlo AL, Lapinskas PJ, Rios CI, Satyamitra MM, Silverman TA, Winters TA, Cassatt DR. Radiation-induced multi-organ injury. Int J Radiat Biol 2024; 100:486-504. [PMID: 38166195 PMCID: PMC11874064 DOI: 10.1080/09553002.2023.2295298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/19/2023] [Accepted: 11/15/2023] [Indexed: 01/04/2024]
Abstract
PURPOSE Natural history studies have been informative in dissecting radiation injury, isolating its effects, and compartmentalizing injury based on the extent of exposure and the elapsed time post-irradiation. Although radiation injury models are useful for investigating the mechanism of action in isolated subsyndromes and development of medical countermeasures (MCMs), it is clear that ionizing radiation exposure leads to multi-organ injury (MOI). METHODS The Radiation and Nuclear Countermeasures Program within the National Institute of Allergy and Infectious Diseases partnered with the Biomedical Advanced Research and Development Authority to convene a virtual two-day meeting titled 'Radiation-Induced Multi-Organ Injury' on June 7-8, 2022. Invited subject matter experts presented their research findings in MOI, including study of mechanisms and possible MCMs to address complex radiation-induced injuries. RESULTS This workshop report summarizes key information from each presentation and discussion by the speakers and audience participants. CONCLUSIONS Understanding the mechanisms that lead to radiation-induced MOI is critical to advancing candidate MCMs that could mitigate the injury and reduce associated morbidity and mortality. The observation that some of these mechanisms associated with MOI include systemic injuries, such as inflammation and vascular damage, suggests that MCMs that address systemic pathways could be effective against multiple organ systems.
Collapse
Affiliation(s)
- Olivia Molinar-Inglis
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, MD, USA
| | - Andrea L. DiCarlo
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, MD, USA
| | - Paula J. Lapinskas
- Biomedical Advanced Research and Development Authority (BARDA), Administration for Strategic Preparedness and Response (ASPR), Department of Health and Human Services (HHS), Washington, DC, USA
| | - Carmen I. Rios
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, MD, USA
| | - Merriline M. Satyamitra
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, MD, USA
| | - Toby A. Silverman
- Biomedical Advanced Research and Development Authority (BARDA), Administration for Strategic Preparedness and Response (ASPR), Department of Health and Human Services (HHS), Washington, DC, USA
| | - Thomas A. Winters
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, MD, USA
| | - David R. Cassatt
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, MD, USA
| |
Collapse
|
7
|
Martello S, Bylicky MA, Shankavaram U, May JM, Chopra S, Sproull M, Scott KMK, Aryankalayil MJ, Coleman CN. Comparative Analysis of miRNA Expression after Whole-Body Irradiation Across Three Strains of Mice. Radiat Res 2023; 200:266-280. [PMID: 37527359 PMCID: PMC10635637 DOI: 10.1667/rade-23-00007.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 06/19/2023] [Indexed: 08/03/2023]
Abstract
Whole- or partial-body exposure to ionizing radiation damages major organ systems, leading to dysfunction on both acute and chronic timescales. Radiation medical countermeasures can mitigate acute damages and may delay chronic effects when delivered within days after exposure. However, in the event of widespread radiation exposure, there will inevitably be scarce resources with limited countermeasures to distribute among the affected population. Radiation biodosimetry is necessary to separate exposed from unexposed victims and determine those who requires the most urgent care. Blood-based, microRNA signatures have great potential for biodosimetry, but the affected population in such a situation will be genetically heterogeneous and have varying miRNA responses to radiation. Thus, there is a need to understand differences in radiation-induced miRNA expression across different genetic backgrounds to develop a robust signature. We used inbred mouse strains C3H/HeJ and BALB/c mice to determine how accurate miRNA in blood would be in developing markers for radiation vs. no radiation, low dose (1 Gy, 2 Gy) vs. high dose (4 Gy, 8 Gy), and high risk (8 Gy) vs. low risk (1 Gy, 2 Gy, 4 Gy). Mice were exposed to whole-body doses of 0 Gy, 1 Gy, 2 Gy, 4 Gy, or 8 Gy of X rays. MiRNA expression changes were identified using NanoString nCounter panels on blood RNA collected 1, 2, 3 or 7 days postirradiation. Overall, C3H/HeJ mice had more differentially expressed miRNAs across all doses and timepoints than BALB/c mice. The highest amount of differential expression occurred at days 2 and 3 postirradiation for both strains. Comparison of C3H/HeJ and BALB/c expression profiles to those previously identified in C57BL/6 mice revealed 12 miRNAs that were commonly expressed across all three strains, only one of which, miR-340-5p, displayed a consistent regulation pattern in all three miRNA data. Notably multiple Let-7 family members predicted high-dose and high-risk radiation exposure (Let-7a, Let-7f, Let-7e, Let-7g, and Let-7d). KEGG pathway analysis demonstrated involvement of these predicted miRNAs in pathways related to: Fatty acid metabolism, Lysine degradation and FoxO signaling. These findings indicate differences in the miRNA response to radiation across various genetic backgrounds, and highlights key similarities, which we exploited to discover miRNAs that predict radiation exposure. Our study demonstrates the need and the utility of including multiple animal strains in developing and validating biodosimetry diagnostic signatures. From this data, we developed highly accurate miRNA signatures capable of predicting exposed and unexposed subjects within a genetically heterogeneous population as quickly as 24 h of exposure to radiation.
Collapse
Affiliation(s)
- Shannon Martello
- Radiation Oncology Branch, Center for Cancer, National Institutes of Health, Rockville, Maryland 20850
| | - Michelle A. Bylicky
- Radiation Oncology Branch, Center for Cancer, National Institutes of Health, Rockville, Maryland 20850
| | - Uma Shankavaram
- Radiation Oncology Branch, Center for Cancer, National Institutes of Health, Rockville, Maryland 20850
| | - Jared M. May
- Radiation Oncology Branch, Center for Cancer, National Institutes of Health, Rockville, Maryland 20850
| | - Sunita Chopra
- Radiation Oncology Branch, Center for Cancer, National Institutes of Health, Rockville, Maryland 20850
| | - Mary Sproull
- Radiation Oncology Branch, Center for Cancer, National Institutes of Health, Rockville, Maryland 20850
| | - Kevin MK Scott
- Radiation Oncology Branch, Center for Cancer, National Institutes of Health, Rockville, Maryland 20850
| | - Molykutty J. Aryankalayil
- Radiation Oncology Branch, Center for Cancer, National Institutes of Health, Rockville, Maryland 20850
| | - C. Norman Coleman
- Radiation Oncology Branch, Center for Cancer, National Institutes of Health, Rockville, Maryland 20850
- Radiation Research Program, National Cancer Institute, National Institutes of Health, Rockville, Maryland 20850
| |
Collapse
|
8
|
Kumari P, Kumar R, Singh D, Kumar R. N-acetyl-L-tryptophan (NAT) provides protection to intestinal epithelial cells (IEC-6) against radiation-induced apoptosis via modulation of oxidative stress and mitochondrial membrane integrity. Mol Biol Rep 2023; 50:6381-6397. [PMID: 37322322 DOI: 10.1007/s11033-023-08579-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 06/02/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Ionizing radiation generates oxidative stress in biological systems via inducing free radicals. Gastro-intestinal system has been known for its high radiosensitivity. Therefore, to develop an effective radiation countermeasure for gastrointestinal system, N-acetyl L-tryptophan was evaluated for its radioprotective efficacy using intestinal epithelial cells-6 (IEC-6) cells as the experimental model. METHODS AND RESULTS Cellular metabolic and lysosomal activity of L-NAT and L-NAT treated irradiated IEC-6 cells were assessed by MTT and NRU staining, respectively. ROS and mitochondrial superoxide levels along with mitochondrial disruption were detected using specific fluorescent probes. Endogenous antioxidants (CAT, SOD, GST, GPx) activities were determined using calorimetric assay. Apoptosis and DNA damage were assessed using flow cytometery and Comet assay, respectively. Results of the study were demonstrated that L-NAT pre-treatment (- 1 h) to irradiated IEC-6 cells significantly contribute to ensuring 84.36% to 87.68% (p < 0.0001) survival at 0.1 μg/mL concentration against LD50 radiation dose (LD50; 20 Gy). Similar level of radioprotection was observed with a clonogenic assay against γ radiation (LD50; 5 Gy). L-NAT was found to provide radioprotection by neutralizing radiation-induced oxidative stress, enhancing antioxidant enzymes (CAT, SOD, GST, and GPx), and protecting DNA from radiation-induced damage. Further, significant restoration of mitochondrial membrane integrity along with apoptosis inhibition was observed with irradiated IEC-6 cells upon L-NAT pretreatment.
Collapse
Affiliation(s)
- Pratibha Kumari
- Radiation Biotechnology Group, Division of Chemical, Biological, Radiological & Nuclear Defence (CBRN), Defence Research and Development Organization (DRDO), Institute of Nuclear Medicine and Allied Sciences, Brig. S.K. Mazumdar Road, Timarpur, Delhi, 110054, India
| | - Ravi Kumar
- Radiation Biotechnology Group, Division of Chemical, Biological, Radiological & Nuclear Defence (CBRN), Defence Research and Development Organization (DRDO), Institute of Nuclear Medicine and Allied Sciences, Brig. S.K. Mazumdar Road, Timarpur, Delhi, 110054, India
| | - Darshana Singh
- Radiation Biotechnology Group, Division of Chemical, Biological, Radiological & Nuclear Defence (CBRN), Defence Research and Development Organization (DRDO), Institute of Nuclear Medicine and Allied Sciences, Brig. S.K. Mazumdar Road, Timarpur, Delhi, 110054, India
| | - Raj Kumar
- Radiation Biotechnology Group, Division of Chemical, Biological, Radiological & Nuclear Defence (CBRN), Defence Research and Development Organization (DRDO), Institute of Nuclear Medicine and Allied Sciences, Brig. S.K. Mazumdar Road, Timarpur, Delhi, 110054, India.
| |
Collapse
|
9
|
Guan B, Li D, Meng A. Development of radiation countermeasure agents for acute radiation syndromes. Animal Model Exp Med 2023; 6:329-336. [PMID: 37642199 PMCID: PMC10486342 DOI: 10.1002/ame2.12339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 07/18/2023] [Indexed: 08/31/2023] Open
Abstract
The risk of internal and external exposure to ionizing radiation (IR) has increased alongside the development and implementation of nuclear technology. Therefore, serious security issues have emerged globally, and there has been an increase in the number of studies focusing on radiological prevention and medical countermeasures. Radioprotective drugs are particularly important components of emergency medical preparedness strategies for the clinical management of IR-induced injuries. However, a few drugs have been approved to date to treat such injuries, and the related mechanisms are not entirely understood. Thus, the aim of the present review was to provide a brief overview of the World Health Organization's updated list of essential medicines for 2023 for the proper management of national stockpiles and the treatment of radiological emergencies. This review also discusses the types of radiation-induced health injuries and the related mechanisms, as well as the development of various radioprotective agents, including Chinese herbal medicines, for which significant survival benefits have been demonstrated in animal models of acute radiation syndrome.
Collapse
Affiliation(s)
- Bowen Guan
- National Human Diseases Animal Model Resource Center, NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesInstitute of Laboratory Animal Sciences Chinese Academy of Medical Sciences (CAMS), Peking Union Medical College (PUMC), National Center of Technology Innovation for Animal ModelBeijingChina
| | - Deguan Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear MedicineInstitute of Radiation Medicine, Chinese Academy of Medical Science, Peking Union Medical CollegeTianjinChina
| | - Aimin Meng
- National Human Diseases Animal Model Resource Center, NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesInstitute of Laboratory Animal Sciences Chinese Academy of Medical Sciences (CAMS), Peking Union Medical College (PUMC), National Center of Technology Innovation for Animal ModelBeijingChina
| |
Collapse
|
10
|
Wu T, Orschell CM. The delayed effects of acute radiation exposure (DEARE): characteristics, mechanisms, animal models, and promising medical countermeasures. Int J Radiat Biol 2023; 99:1066-1079. [PMID: 36862990 PMCID: PMC10330482 DOI: 10.1080/09553002.2023.2187479] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/25/2023] [Accepted: 02/20/2023] [Indexed: 03/04/2023]
Abstract
PURPOSE Terrorist use of nuclear weapons and radiation accidents put the human population at risk for exposure to life-threatening levels of radiation. Victims of lethal radiation exposure face potentially lethal acute injury, while survivors of the acute phase are plagued with chronic debilitating multi-organ injuries for years after exposure. Developing effective medical countermeasures (MCM) for the treatment of radiation exposure is an urgent need that relies heavily on studies conducted in reliable and well-characterized animal models according to the FDA Animal Rule. Although relevant animal models have been developed in several species and four MCM for treatment of the acute radiation syndrome are now FDA-approved, animal models for the delayed effects of acute radiation exposure (DEARE) have only recently been developed, and there are no licensed MCM for DEARE. Herein, we provide a review of the DEARE including key characteristics of the DEARE gleaned from human data as well as animal, mechanisms common to multi-organ DEARE, small and large animal models used to study the DEARE, and promising new or repurposed MCM under development for alleviation of the DEARE. CONCLUSIONS Intensification of research efforts and support focused on better understanding of mechanisms and natural history of DEARE are urgently needed. Such knowledge provides the necessary first steps toward the design and development of MCM that effectively alleviate the life-debilitating consequences of the DEARE for the benefit of humankind worldwide.
Collapse
Affiliation(s)
- Tong Wu
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Christie M Orschell
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
11
|
MacVittie TJ. Where are the medical countermeasures against the ARS and DEARE? A current topic relative to an animal model research platform, radiation exposure context, the acute and delayed effects of acute exposure, and the FDA animal rule. Int J Radiat Biol 2023:1-15. [PMID: 36811500 DOI: 10.1080/09553002.2023.2181999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
PURPOSE A question echoed by the National Biodefense Science Board (NBSB) in 2010, remains a reasonable question in 2023; 'Where are the Countermeasures?'. A critical path for development of medical countermeasures (MCM) against acute, radiation-induced organ-specific injury within the acute radiation syndrome (ARS) and the delayed effects of acute radiation exposure (DEARE) requires the recognition of problems and solutions inherent in the path to FDA approval under the Animal Rule. Keep Rule number one in mind, It's not easy. CONSIDERATIONS The current topic herein is focused on defining the nonhuman primate model(s) for efficient MCM development relative to consideration of prompt and delayed exposure in the context of the nuclear scenario. The rhesus macaque is a predictive model for human exposure of partial-body irradiation with marginal bone marrow sparing that allows definition of the multiple organ injury in the acute radiation syndrome (ARS) and the delayed effects of acute radiation exposure (DEARE). The continued definition of natural history is required to delineate an associative or causal interaction within the concurrent multi-organ injury characteristic of the ARS and DEARE. A more efficient development of organ specific MCM for both pre-exposure and post-exposure prophylaxis to include acute radiation-induced combined injury requires closing critical gaps in knowledge and urgent support to rectify the national shortage of nonhuman primates. The rhesus macaque is a validated, predictive model of the human response to prompt and delayed radiation exposure, medical management and MCM treatment. A rational approach to further development of the cynomolgus macaque as a comparable model is urgently required for continued development of MCM for FDA approval. CONCLUSION It is imperative to examine the key variables relative to animal model development and validation, The pharmacokinetics, pharmacodynamics and exposure profiles, of candidate MCM relative to route, administration schedule and optimal efficacy define the fully effective dose. The conduct of adequate and well-controlled pivotal efficacy studies as well as safety and toxicity studies support approval under the FDA Animal Rule and label definition for human use.
Collapse
Affiliation(s)
- Thomas J MacVittie
- Department of Radiation Oncology, University of Maryland, School of Medicine, Baltimore, MD, USA
| |
Collapse
|
12
|
Kumar P, Wang P, Farese AM, MacVittie TJ, Kane MA. Metabolomics of Multiorgan Radiation Injury in Non-human Primate Model Reveals System-wide Metabolic Perturbations. HEALTH PHYSICS 2021; 121:395-405. [PMID: 34546220 DOI: 10.1097/hp.0000000000001472] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
ABSTRACT Exposure to ionizing radiation following a nuclear or radiological incident results in potential acute radiation syndromes causing sequelae of multi-organ injury in a dose- and time-dependent manner. Currently, medical countermeasures against radiation injury are limited, and no biomarkers have been approved by regulatory authorities. Identification of circulating plasma biomarkers indicative of radiation injury can be useful for early triage and injury assessment and in the development of novel therapies (medical countermeasures). Aims of this study are to (1) identify metabolites and lipids with consensus signatures that can inform on mechanism of injury in radiation-induced multi-organ injury and (2) identify plasma biomarkers in non-human primate (NHP) that correlate with tissues (kidney, liver, lung, left and right heart, jejunum) indicative of radiation injury, assessing samples collected over 3 wk post-exposure to 12 Gy partial body irradiation with 2.5% bone marrow sparing. About 180 plasma and tissue metabolites and lipids were quantified through Biocrates AbsoluteIDQ p180 kit using liquid chromatography and mass spectrometry. System-wide perturbations of specific metabolites and lipid levels and pathway alterations were identified. Citrulline, Serotonin, PC ae 38:2, PC ae 36:2, and sum of branched chain amino acids were identified as potential biomarkers of radiation injury. Pathway analysis revealed consistent changes in fatty acid oxidation and metabolism and perturbations in multiple other pathways.
Collapse
Affiliation(s)
- Praveen Kumar
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD, 21201
| | - Pengcheng Wang
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, 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, Baltimore, MD, 21201
| |
Collapse
|
13
|
Aqeel M, Medhora M, Gore E, Borkenhagen J, Klawikowski S, Eastwood D, Banerjee A, Jacobs ER. Evaluation of Radiation-induced Pleural Effusions after Radiotherapy to Support Development of Animal Models of Radiation Pneumonitis. HEALTH PHYSICS 2021; 121:434-443. [PMID: 34546223 PMCID: PMC8500166 DOI: 10.1097/hp.0000000000001462] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
ABSTRACT Not all animal models develop radiation-induced pleural effusions (RIPEs) as a form of radiation-induced lung injury (RILI). Such effusions are also not well characterized in humans. The purpose of this study is to identify occurrences of RIPE in humans, provide justification for development of relevant animal models, and further characterize its risk factors in cancer patients. We also aim to identify dose thresholds for cardiopulmonary toxicity in humans to shed light on possible pathogenic mechanisms for RIPEs. We carried out a retrospective review of medical records of 96 cancer patients receiving thoracic irradiation (TRT) at our institution. Fifty-three (53%) patients developed a new pleural effusion post TRT; 18 (19%) had RIPE; and 67% developed RIPE ipsilateral to the site irradiated. None developed "contralateral only" effusions. Median time to development was 6 mo (IQR; 4-8 mo). Of 18, 8 patients (44%) had concomitant asymptomatic (radiographic only) or symptomatic radiation pneumonitis and pericardial effusion. Dosimetric factors, including combined and ipsilateral mean lung dose (MLD), were significantly associated with increased risk of RIPE. Angiotensin converting enzyme inhibition, steroids, or concurrent chemotherapy did not modify incidence of RIPE. Our results substantiate the occurrence and incidence of RIPEs in humans. In cancer patients, a median time to development of effusions around 6 mo also supports the onset of RIPEs concurrent with radiation pneumonitis. Future work needs to include large populations of cancer survivors in whom delayed RIPEs can be tracked and correlated with cardiovascular changes in the context of injury to multiple organs.
Collapse
Affiliation(s)
- Masooma Aqeel
- Current Affiliation: Section of Pulmonary & Critical
Care Medicine, Department of Medicine, Aga Khan University, Karachi, Pakistan.
Formerly at Division of Pulmonary Medicine, Department of Medicine, Froedtert
Hospital & Medical College of Wisconsin, Milwaukee, WI, United States
| | - Meetha Medhora
- Division of Pulmonary Medicine, Department of Medicine,
Froedtert Hospital & Medical College of Wisconsin, Milwaukee, WI, United
States
- Department of Radiation Oncology, Froedtert Hospital &
Medical College of Wisconsin, Milwaukee, WI, United States
- Research Service, Department of Veteran’s Affairs,
Clement J. Zablocki VA Medical Center, Milwaukee, WI, United States
| | - Elizabeth Gore
- Department of Radiation Oncology, Froedtert Hospital &
Medical College of Wisconsin, Milwaukee, WI, United States
- Research Service, Department of Veteran’s Affairs,
Clement J. Zablocki VA Medical Center, Milwaukee, WI, United States
| | - Jenna Borkenhagen
- Department of Radiation Oncology, Froedtert Hospital &
Medical College of Wisconsin, Milwaukee, WI, United States
| | - Slade Klawikowski
- Department of Radiation Oncology, Froedtert Hospital &
Medical College of Wisconsin, Milwaukee, WI, United States
| | - Daniel Eastwood
- Department of Biostatistics, Medical College of Wisconsin,
Milwaukee, WI, United States
| | - Anjishnu Banerjee
- Department of Biostatistics, Medical College of Wisconsin,
Milwaukee, WI, United States
| | - Elizabeth R. Jacobs
- Division of Pulmonary Medicine, Department of Medicine,
Froedtert Hospital & Medical College of Wisconsin, Milwaukee, WI, United
States
- Research Service, Department of Veteran’s Affairs,
Clement J. Zablocki VA Medical Center, Milwaukee, WI, United States
| |
Collapse
|
14
|
Port M, Hérodin F, Drouet M, Valente M, Majewski M, Ostheim P, Lamkowski A, Schüle S, Forcheron F, Tichy A, Sirak I, Malkova A, Becker BV, Veit DA, Waldeck S, Badie C, O'Brien G, Christiansen H, Wichmann J, Beutel G, Davidkova M, Doucha-Senf S, Abend M. Gene Expression Changes in Irradiated Baboons: A Summary and Interpretation of a Decade of Findings. Radiat Res 2021; 195:501-521. [PMID: 33788952 DOI: 10.1667/rade-20-00217.1] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 05/05/2021] [Indexed: 11/03/2022]
Affiliation(s)
- M Port
- Bundeswehr Institute of Radiobiology, Munich Germany
| | - F Hérodin
- Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | - M Drouet
- Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | - M Valente
- Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | - M Majewski
- Bundeswehr Institute of Radiobiology, Munich Germany
| | - P Ostheim
- Bundeswehr Institute of Radiobiology, Munich Germany
| | - A Lamkowski
- Bundeswehr Institute of Radiobiology, Munich Germany
| | - S Schüle
- Bundeswehr Institute of Radiobiology, Munich Germany
| | - F Forcheron
- Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | - A Tichy
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence, Brno, Czech Republic and Biomedical Research Centre, University Hospital Hradec Králové, Hradec Králové, Czech Republic
| | - I Sirak
- Department of Oncology and Radiotherapy, University Hospital, Hradec Králové, Hradec Králové, Czech Republic
| | - A Malkova
- Department of Hygiene and Preventive Medicine, Faculty of Medicine in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - B V Becker
- Bundeswehr Central Hospital, Department of Radiology and Neuroradiology, Koblenz, Germany
| | - D A Veit
- Bundeswehr Central Hospital, Department of Radiology and Neuroradiology, Koblenz, Germany
| | - S Waldeck
- Bundeswehr Central Hospital, Department of Radiology and Neuroradiology, Koblenz, Germany
| | - C Badie
- Cancer Mechanisms and Biomarkers Group, Radiation Effects Department, Centre for Radiation, Chemical and Environmental Hazards, Public Health of England, Didcot, United Kingdom
| | - G O'Brien
- Cancer Mechanisms and Biomarkers Group, Radiation Effects Department, Centre for Radiation, Chemical and Environmental Hazards, Public Health of England, Didcot, United Kingdom
| | - H Christiansen
- Department of Radiation Oncology, Hannover Medical School, Hannover, Germany
| | - J Wichmann
- Department of Radiation Oncology, Hannover Medical School, Hannover, Germany
| | - G Beutel
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - M Davidkova
- Department of Radiation Dosimetry, Nuclear Physics Institute of the Czech Academy of Sciences, Řež, Czech Republic
| | - S Doucha-Senf
- Bundeswehr Institute of Radiobiology, Munich Germany
| | - M Abend
- Bundeswehr Institute of Radiobiology, Munich Germany
| |
Collapse
|
15
|
Majewski M, Ostheim P, Gluzman-Poltorak Z, Vainstein V, Basile L, Schüle S, Haimerl M, Stroszczynski C, Port M, Abend M. Gene expression changes in male and female rhesus macaque 60 days after irradiation. PLoS One 2021; 16:e0254344. [PMID: 34288924 PMCID: PMC8294544 DOI: 10.1371/journal.pone.0254344] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 06/25/2021] [Indexed: 12/13/2022] Open
Abstract
PURPOSE Transcriptome changes can be expected in survivors after lethal irradiation. We aimed to characterize these in males and females and after different cytokine treatments 60 days after irradiation. MATERIAL AND METHODS Male and female rhesus macaques (n = 142) received a whole-body exposure with 700 cGy, from which 60 animals survived. Peripheral whole blood was drawn pre-exposure and before sacrificing the surviving animals after 60 days. RESULTS We evaluated gene expression in a three-phase study design. Phase I was a whole-genome screening (NGS) for mRNAs using five pre- and post-exposure RNA samples from both sexes (n = 20). Differential gene expression (DGE) was calculated between samples of survivors and pre-exposure samples (reference), separately for males and females. 1,243 up- and down-regulated genes were identified with 30-50% more deregulated genes in females. 37 candidate mRNAs were chosen for qRT-PCR validation in phase II using the remaining samples (n = 117). Altogether 17 genes showed (borderline) significant (t-test) DGE in groups of untreated or treated animals. Nine genes (CD248, EDAR, FAM19A5, GAL3ST4, GCNT4, HBG2/1, LRRN1, NOG, SYT14) remained with significant changes and were detected in at least 50% of samples per group. Panther analysis revealed an overlap between both sexes, related to the WNT signaling pathway, cell adhesion and immunological functions. For phase III, we validated the nine genes with candidate genes (n = 32) from an earlier conducted study on male baboons. Altogether 14 out of 41 genes showed a concordantly DGE across both species in a bilateral comparison. CONCLUSIONS Sixty days after radiation exposure, we identified (1) sex and cytokine treatment independent transcriptional changes, (2) females with almost twice as much deregulated genes appeared more radio-responsive than males, (3) Panther analysis revealed an association with immunological processes and WNT pathway for both sexes.
Collapse
Affiliation(s)
- Matthäus Majewski
- Bundeswehr Institute of Radiobiology, Munich, Germany
- Department of Urology, Bundeswehr Hospital Ulm, Ulm, Germany
| | | | - Zoya Gluzman-Poltorak
- Neumedicines Inc, Pasadena, CA, United States of America
- Applied Stem Cell Therapeutics, Milpitas, CA, United States of America
| | - Vladimir Vainstein
- Neumedicines Inc, Pasadena, CA, United States of America
- Hadassah Medical Center, Jerusalem, Israel
| | - Lena Basile
- Neumedicines Inc, Pasadena, CA, United States of America
| | - Simone Schüle
- Bundeswehr Institute of Radiobiology, Munich, Germany
| | - Michael Haimerl
- Department of Radiology, University Hospital Regensburg, Regensburg, Germany
| | | | - Matthias Port
- Bundeswehr Institute of Radiobiology, Munich, Germany
| | - Michael Abend
- Bundeswehr Institute of Radiobiology, Munich, Germany
- * E-mail:
| |
Collapse
|