51
|
Yuan Q, Peng R, Yu H, Wang S, Chen Z, Dong S, Li W, Cheng B, Jiang Q, Cong Y, Li F, Li C. Disulfiram Protects Against Radiation-Induced Intestinal Injury in Mice. Front Pharmacol 2022; 13:852669. [PMID: 35517788 PMCID: PMC9061966 DOI: 10.3389/fphar.2022.852669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
Radiation-induced intestinal injury (RIII) occurs after high doses of radiation exposure. RIII restricts the therapeutic efficacy of radiotherapy in cancer and increases morbidity and mortality in nuclear disasters. Currently, there is no approved agent for the prevention or treatment of RIII. Here, we reported that the disulfiram, an FDA-approved alcohol deterrent, prolonged the survival in mice after lethal irradiation. Pretreatment with disulfiram inhibited proliferation within 24 h after irradiation, but improved crypt regeneration at 3.5 days post-irradiation. Mechanistically, disulfiram promoted Lgr5+ intestinal stem cells (ISCs) survival and maintained their ability to regenerate intestinal epithelium after radiation. Moreover, disulfiram suppresses DNA damage accumulation, thus inhibits aberrant mitosis after radiation. Unexpectedly, disulfiram treatment did not inhibit crypt cell apoptosis 4 h after radiation and the regeneration of crypts from PUMA-deficient mice after irradiation was also promoted by disulfiram. In conclusion, our findings demonstrate that disulfiram regulates the DNA damage response and survival of ISCs through affecting the cell cycle. Given its radioprotective efficacy and decades of application in humans, disulfiram is a promising candidate to prevent RIII in cancer therapy and nuclear accident.
Collapse
Affiliation(s)
- Qingwen Yuan
- The Postgraduate Training Base of Jinzhou Medical University, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Renjun Peng
- Department of Nuclear Radiation Injury and Monitoring, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Huijie Yu
- Department of Nuclear Radiation Injury and Monitoring, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Sinian Wang
- Department of Nuclear Radiation Injury and Monitoring, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Zhongmin Chen
- Department of Nuclear Radiation Injury and Monitoring, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Suhe Dong
- Department of Nuclear Radiation Injury and Monitoring, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Wei Li
- Department of Nuclear Radiation Injury and Monitoring, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Bo Cheng
- Department of Pathology, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Qisheng Jiang
- Department of Nuclear Radiation Injury and Monitoring, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Yuwen Cong
- Department of Pathophysiology, Beijing Institute of Radiation Medicine, Beijing Key Laboratory for Radiobiology (BKLRB), Beijing, China
| | - Fengsheng Li
- Department of Nuclear Radiation Injury and Monitoring, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Changzheng Li
- Department of Gastroenterology, The PLA Rocket Force Characteristic Medical Center, Beijing, China
| |
Collapse
|
52
|
Development and Evaluation of a Machine Learning Prediction Model for Small-for-Gestational-Age Births in Women Exposed to Radiation before Pregnancy. J Pers Med 2022; 12:jpm12040550. [PMID: 35455666 PMCID: PMC9031835 DOI: 10.3390/jpm12040550] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 12/22/2022] Open
Abstract
Exposure to radiation has been associated with increased risk of delivering small-for-gestational-age (SGA) newborns. There are no tools to predict SGA newborns in pregnant women exposed to radiation before pregnancy. Here, we aimed to develop an array of machine learning (ML) models to predict SGA newborns in women exposed to radiation before pregnancy. Patients’ data was obtained from the National Free Preconception Health Examination Project from 2010 to 2012. The data were randomly divided into a training dataset (n = 364) and a testing dataset (n = 91). Eight various ML models were compared for solving the binary classification of SGA prediction, followed by a post hoc explainability based on the SHAP model to identify and interpret the most important features that contribute to the prediction outcome. A total of 455 newborns were included, with the occurrence of 60 SGA births (13.2%). Overall, the model obtained by extreme gradient boosting (XGBoost) achieved the highest area under the receiver-operating-characteristic curve (AUC) in the testing set (0.844, 95% confidence interval (CI): 0.713–0.974). All models showed satisfied AUCs, except for the logistic regression model (AUC: 0.561, 95% CI: 0.355–0.768). After feature selection by recursive feature elimination (RFE), 15 features were included in the final prediction model using the XGBoost algorithm, with an AUC of 0.821 (95% CI: 0.650–0.993). ML algorithms can generate robust models to predict SGA newborns in pregnant women exposed to radiation before pregnancy, which may thus be used as a prediction tool for SGA newborns in high-risk pregnant women.
Collapse
|
53
|
Wang A, Wang L, Fu Q, Shi Z, Chen X, Zhang X, Xu W, Wang T, Yu X, Zhang S, Gao Y, Li W, Hu S. Yiqi Jiedu herbal decoction attenuates the 2 Gy 60Co γ ray induced spleen injury by inhibiting apoptosis and modulating the immune balance. JOURNAL OF ETHNOPHARMACOLOGY 2022; 286:114925. [PMID: 34933086 DOI: 10.1016/j.jep.2021.114925] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/08/2021] [Accepted: 12/14/2021] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Irradiation-induced immunosuppression often occurs during radiotherapy in patients, which would increase the risk of opportunistic infections. Many Chinese herbal prescriptions or natural extracts have recently attracted increased radiation protection and therapy attention due to their low toxicity. AIM OF THE STUDY The present study aimed to investigate the protective effects of Yiqi Jiedu (YQJD) decoction on spleen injury induced by 2 Gy 60Co γ ray in mice. MATERIALS AND METHODS A total of 180 Balb/c mice were randomly divided into five groups: blank control (Ctrl), model (IR), positive drug (IRA), low-dose YQJD decoction (IRL), and high-dose YQJD decoction (IRH). After a ten-day intervention, mice were exposed to a single dose of total body irradiation (2 Gy) and sacrificed on the 1st, 3rd, and 7th day after irradiation. The indicators include general observations and body weight, changes in peripheral hemogram, index and histopathology examination of the spleen, distribution of lymphocyte subsets, cytokine levels, and apoptosis in the spleen. RESULTS In comparison to the Ctrl group, the body weight, spleen index, peripheral blood cell, and splenocyte quantities decreased significantly after exposure, accompanied by a notable increase of apoptosis in spleen cells. Moreover, ionizing radiation also broke the balance of CD4+/CD8+, Th1/Th2, and Th17/Treg, triggering immune imbalance and immunosuppression. The above injuries occurred on the 1st day after exposure, worsened on the 3rd, and were relieved on the 7th day. However, the pretreatment of YQJD decoction increased the spleen index, improved the spleen structure, and inhibited radiation-induced apoptosis after exposure. Additionally, YQJD decoction has shown its ability to promote immunological balance recovery following exposure by regulating CD4+/CD8+, Th1/Th2, and Th17/Treg ratios, which may minimize the risk of infection. In addition, the high-dose of YQJD decoction showed a better protective effect than the low-dose group. CONCLUSION The protective effects of YQJD decoction on 2 Gy 60Coγray induced spleen injury were confirmed in this study. This mechanism may be related to inhibiting apoptosis and modulating immune balance. This exploration might provide new insights into the use of Chinese herbs on radioprotection of the immune system.
Collapse
Affiliation(s)
- An Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China; Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China.
| | - Lei Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Qian Fu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Zhongyu Shi
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Xiaoying Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Xiaomeng Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Wenhui Xu
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Tieshan Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Xue Yu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Shujing Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Yushan Gao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Wei Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Sumin Hu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
54
|
Mitigation of total body irradiation-induced mortality and hematopoietic injury of mice by a thrombopoietin mimetic (JNJ-26366821). Sci Rep 2022; 12:3485. [PMID: 35241733 PMCID: PMC8894488 DOI: 10.1038/s41598-022-07426-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/11/2022] [Indexed: 11/08/2022] Open
Abstract
The threat of a nuclear attack has increased in recent years highlighting the benefit of developing additional therapies for the treatment of victims suffering from Acute Radiation Syndrome (ARS). In this work, we evaluated the impact of a PEGylated thrombopoietin mimetic peptide, JNJ-26366821, on the mortality and hematopoietic effects associated with ARS in mice exposed to lethal doses of total body irradiation (TBI). JNJ-26366821 was efficacious as a mitigator of mortality and thrombocytopenia associated with ARS in both CD2F1 and C57BL/6 mice exposed to TBI from a cobalt-60 gamma-ray source. Single administration of doses ranging from 0.3 to 1 mg/kg, given 4, 8, 12 or 24 h post-TBI (LD70 dose) increased survival by 30-90% as compared to saline control treatment. At the conclusion of the 30-day study, significant increases in bone marrow colony forming units and megakaryocytes were observed in animals administered JNJ-26366821 compared to those administered saline. In addition, enhanced recovery of FLT3-L levels was observed in JNJ-26366821-treated animals. Probit analysis of survival in the JNJ-26366821- and saline-treated cohorts revealed a dose reduction factor of 1.113 and significant increases in survival for up to 6 months following irradiation. These results support the potential use of JNJ-26366821 as a medical countermeasure for treatment of acute TBI exposure in case of a radiological/nuclear event when administered from 4 to 24 h post-TBI.
Collapse
|
55
|
Gamma-tocotrienol, a radiation countermeasure, reverses proteomic changes in serum following total-body gamma irradiation in mice. Sci Rep 2022; 12:3387. [PMID: 35233005 PMCID: PMC8888544 DOI: 10.1038/s41598-022-07266-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/15/2022] [Indexed: 12/14/2022] Open
Abstract
Radiological incidents or terrorist attacks would likely expose civilians and military personnel to high doses of ionizing radiation, leading to the development of acute radiation syndrome. We examined the effectiveness of prophylactic administration of a developmental radiation countermeasure, γ-tocotrienol (GT3), in a total-body irradiation (TBI) mouse model. CD2F1 mice received GT3 24 h prior to 11 Gy cobalt-60 gamma-irradiation. This dose of radiation induces severe hematopoietic acute radiation syndrome and moderate gastrointestinal injury. GT3 provided 100% protection, while the vehicle control group had 100% mortality. Two-dimensional differential in-gel electrophoresis was followed by mass spectrometry and Ingenuity Pathway Analysis (IPA). Analysis revealed a change in expression of 18 proteins in response to TBI, and these changes were reversed with prophylactic treatment of GT3. IPA revealed a network of associated proteins involved in cellular movement, immune cell trafficking, and inflammatory response. Of particular interest, significant expression changes in beta-2-glycoprotein 1, alpha-1-acid glycoprotein 1, alpha-2-macroglobulin, complement C3, mannose-binding protein C, and major urinary protein 6 were noted after TBI and reversed with GT3 treatment. This study reports the untargeted approach, the network, and specific serum proteins which could be translated as biomarkers of both radiation injury and protection by countermeasures.
Collapse
|
56
|
Vasin MV. B-190 (Indralin) in Light of the History of the Formation of Ideas about the Mechanism of Action of Radioprotectors. BIOL BULL+ 2022. [DOI: 10.1134/s1062359021110091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
57
|
Kumar A, Choudhary S, Kumar S, Adhikari JS, Kapoor S, Chaudhury NK. Role of melatonin mediated G-CSF induction in hematopoietic system of gamma-irradiated mice. Life Sci 2022; 289:120190. [PMID: 34883100 DOI: 10.1016/j.lfs.2021.120190] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 11/24/2021] [Accepted: 11/25/2021] [Indexed: 11/16/2022]
Abstract
AIMS Hematopoietic acute radiation syndrome (H-ARS) can cause lethality, and therefore, the necessity of a safe radioprotector. The present study was focused on investigating the role of melatonin in granulocytes colony-stimulating factor (G-CSF) and related mechanisms underlying the reduction of DNA damage in hematopoietic system of irradiated mice. MAIN METHODS C57BL/6 male mice were exposed to 2, 5, and 7.5Gy of whole-body irradiation (WBI), 30 min after intra-peritoneal administration of melatonin with different doses. Mice were sacrificed at different time intervals after WBI, and bone marrow, splenocytes, and peripheral blood lymphocytes were isolated for studying various parameters including micronuclei (MN), cell cycle, comet, γ-H2AX, gene expression, amino acid profiling, and hematology. KEY FINDINGS Melatonin100mg/kg ameliorated radiation (7.5Gy and 5Gy) induced MN frequency and cell death in bone marrow without mortality. At 24 h of post-WBI (2Gy), the frequency of micronucleated polychromatic erythrocytes (mnPCE) with different melatonin doses revealed 20 mg/kg as optimal i.p. dose for protecting the hematopoietic system against radiation injury. In comet assay, a significant reduction in radiation-induced % DNA tail (p ≤ 0.05) was observed at this dose. Melatonin reduced γ-H2AX foci/cell and eventually reached to the control level. Melatonin also decreased blood arginine levels in mice after 24 h of WBI. The gene expression of G-CSF, Bcl-2-associated X protein (BAX), and Bcl2 indicated the role of melatonin in G-CSF regulation and downstream pro-survival pathways along with anti-apoptotic activity. SIGNIFICANCE The results revealed that melatonin recovers the hematopoietic system of irradiated mice by inducing G-CSF mediated radioprotection.
Collapse
Affiliation(s)
- Arun Kumar
- Division of Radiation Biodosimetry, Institute of Nuclear Medicine and Allied Sciences (INMAS)-Defence Research and Development Organisation (DRDO), Brig. SK Mazumdar Marg, Timarpur, Delhi 110054, India
| | - Sandeep Choudhary
- Division of Radiation Biodosimetry, Institute of Nuclear Medicine and Allied Sciences (INMAS)-Defence Research and Development Organisation (DRDO), Brig. SK Mazumdar Marg, Timarpur, Delhi 110054, India; Department of Pharmacology, School of Pharmaceutical Education and Research, Hamdard University, Hamdard nagar, New Delhi 110062, India
| | - Somesh Kumar
- Pediatrics Genetics & Research Laboratory, Department of Pediatrics, Maulana Azad Medical College & Associated Lok Nayak Hospital, Delhi 110002, India
| | - Jawahar S Adhikari
- Division of Radiation Biodosimetry, Institute of Nuclear Medicine and Allied Sciences (INMAS)-Defence Research and Development Organisation (DRDO), Brig. SK Mazumdar Marg, Timarpur, Delhi 110054, India
| | - Seema Kapoor
- Pediatrics Genetics & Research Laboratory, Department of Pediatrics, Maulana Azad Medical College & Associated Lok Nayak Hospital, Delhi 110002, India
| | - Nabo K Chaudhury
- Division of Radiation Biodosimetry, Institute of Nuclear Medicine and Allied Sciences (INMAS)-Defence Research and Development Organisation (DRDO), Brig. SK Mazumdar Marg, Timarpur, Delhi 110054, India.
| |
Collapse
|
58
|
MacVittie TJ, Farese AM. Recent advances in medical countermeasure development against acute radiation exposure based on the US FDA animal rule. JOURNAL OF RADIOLOGICAL PROTECTION : OFFICIAL JOURNAL OF THE SOCIETY FOR RADIOLOGICAL PROTECTION 2021; 41:S438-S453. [PMID: 34433144 DOI: 10.1088/1361-6498/ac20e0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 08/25/2021] [Indexed: 06/13/2023]
Abstract
Recent advances in medical countermeasures (MCMs) has been dependent on the Food and Drug Administration (FDA) animal rule (AR) and the final guidance document provided for industry on product development. The criteria outlined therein establish the path for approval under the AR. The guidance document, along with the funding and requirements from the federal agencies provided the basic considerations for animal model development in assessing radiation effects and efficacy against the potential lethal effects of acute radiation injury and the delayed effects of acute exposure. Animal models, essential for determining MCM efficacy, were developed and validated to assess organ-specific, potentially lethal, radiation effects against the gastrointestinal (GI) and hematopoietic acute radiation syndrome (H-ARS), and radiation-induced delayed effects to lung and associated comorbidities of prolonged immune suppression, GI, kidney and heart injury. Partial-body irradiation models where marginal bone marrow was spared resulted in the ability to evaluate the concomitant evolution of multiple organ injury in the acute and delayed effects in survivors of acute radiation exposure. There are no MCMs for prophylaxis against the major sequelae of the ARS or the delayed effects of acute exposure. Also lacking are MCMs that will mitigate the GI ARS consequent to potentially lethal exposure from a terrorist event or major radiation accident. Additionally, the gap in countermeasures for prophylaxis may extend to mixed neutron/gamma radiation if current modelling predicts prompt exposure from an improvised nuclear device. However, progress in the field of MCM development has been made due to federal and corporate funding, clarification of the critical criteria for efficacy within the FDA AR and the concomitant development and validation of additional animal models. These models provided for a strategic and tactical approach to determine radiation effects and MCM efficacy.
Collapse
Affiliation(s)
- Thomas J MacVittie
- Department of Radiation Oncology, University of Maryland, School of Medicine, Baltimore, MD, 21201, United States of America
| | - Ann M Farese
- Department of Radiation Oncology, University of Maryland, School of Medicine, Baltimore, MD, 21201, United States of America
| |
Collapse
|
59
|
Lazarus HM, Armitage JO, Gale RP. Role of molecularly-cloned hematopoietic growth factors after acute high-dose radiation exposures. JOURNAL OF RADIOLOGICAL PROTECTION : OFFICIAL JOURNAL OF THE SOCIETY FOR RADIOLOGICAL PROTECTION 2021; 41:S478-S489. [PMID: 34134098 DOI: 10.1088/1361-6498/ac0bff] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 06/16/2021] [Indexed: 06/12/2023]
Abstract
Therapy of acute, high-dose whole-body exposures of humans to ionizing radiations is a complex medical challenge. Since 1944 more than 400 radiologic accidents have been registered with more than 3000 substantial radiation exposures and 127 fatalities. There are several potential interventions including supportive care, transfusions, preventative or therapeutic anti-infection drugs, molecularly-cloned myeloid growth factors and hematopoietic cell transplants. We discuss the use of the granulocyte and granulocyte-macrophage colony-stimulating factor (G-CSF and GM-CSF) to treat acute high-dose ionizing radiation exposures. Considerable data in experimental models including monkeys indicate use of these drugs accelerates bone marrow recovery and in some but not all instances increases survival. In ten accidents since 1996, 30 victims received G-CSF alone or with other growth factors. Twenty-six victims survived. In seven accidents since 1986, 28 victims received GM-CSF alone or with other growth factors; 18 victims survived. However, absent control or data from randomized trials, it is not possible to know with certainty what role, if any, receiving G-CSF or GM-CSF was of benefit. Given the favorablebenefit-to-riskratio of molecularly-cloned myeloid growth factors, their use soon after exposure to acute, high-dose whole-body ionizing radiations is reasonable.
Collapse
Affiliation(s)
- Hillard M Lazarus
- Department of Medicine, Division of Hematology and Oncology, Case Western Reserve University, Cleveland, OH, United States of America
| | - James O Armitage
- Department of Medicine, University of Nebraska, Omaha, NE, United States of America
| | - Robert Peter Gale
- Centre for Haematology Research, Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| |
Collapse
|
60
|
Cheema AK, Li Y, Singh J, Johnson R, Girgis M, Wise SY, Fatanmi OO, Kaytor MD, Singh VK. Microbiome study in irradiated mice treated with BIO 300, a promising radiation countermeasure. Anim Microbiome 2021; 3:71. [PMID: 34627406 PMCID: PMC8501697 DOI: 10.1186/s42523-021-00132-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 09/22/2021] [Indexed: 01/04/2023] Open
Abstract
Background The mammalian gut harbors very complex and diverse microbiota that play an important role in intestinal homeostasis and host health. Exposure to radiation results in dysbiosis of the gut microbiota leading to detrimental pathophysiological changes to the host. To alleviate the effects of irradiation, several candidate countermeasures are under investigation. BIO 300, containing synthetic genistein formulated as an amorphous solid dispersion or as an aqueous suspension of nanoparticles, is a promising candidate under advanced development. The aim of this study was to investigate the effects of BIO 300 on the gut microbiome and metabolome of mice exposed to 60Co gamma-radiation. The gut microbiota and metabolome of control and drug-treated mice exposed to radiation was characterized by bacterial 16S rRNA amplicon sequencing and untargeted metabolomics. Results We found that irradiation altered the Firmicutes/Bacteroidetes ratio and significantly decreased the relative abundance of Lactobacillus, both in BIO 300-treated and control mice; however, the ratio returned to near normal levels in BIO 300-treated mice by day 14 post-irradiation. Concomitantly, we also observed corrective shifts in metabolic pathways that were perturbed after irradiation. Conclusions Overall, the data presented show that radiation exposure led to a relative depletion of commensals like Lactobacillus leading to an inflammatory metabolic phenotype while the majority of the drug-treated mice showed alleviation of this condition primarily by restoration of normal gut microbiota. These results indicate that the radioprotective effects of BIO 300, at least in part, may involve correction of the host-microbiome metabolic axis. Supplementary Information The online version contains supplementary material available at 10.1186/s42523-021-00132-1.
Collapse
Affiliation(s)
- Amrita K Cheema
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA.,Department of Biochemistry, Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC, USA
| | - Yaoxiang Li
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Jatinder Singh
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Ryan Johnson
- Department of Preventive Medicine and Biostatistics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Michael Girgis
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, 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, Bethesda, MD, USA.,Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, 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, Bethesda, MD, USA.,Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | | | - 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, Bethesda, MD, USA. .,Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| |
Collapse
|
61
|
Farese AM, Drouet M, Herodin F, Bertho JM, Thrall KD, Authier S, Doyle-Eisele M, MacVittie TJ. Acute Radiation Effects, the H-ARS in the Non-human Primate: A Review and New Data for the Cynomolgus Macaque with Reference to the Rhesus Macaque. HEALTH PHYSICS 2021; 121:304-330. [PMID: 34546214 DOI: 10.1097/hp.0000000000001442] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
ABSTRACT Medical countermeasure development under the US Food and Drug Administration animal rule requires validated animal models of acute radiation effects. The key large animal model is the non-human primate, rhesus macaque. To date, only the rhesus macaque has been used for both critical supportive data and pivotal efficacy trials seeking US Food and Drug Administration approval. The potential for use of the rhesus for other high priority studies such as vaccine development underscores the need to identify another non-human primate model to account for the current lack of rhesus for medical countermeasure development. The cynomolgus macaque, Macaca fascicularis, has an existing database of medical countermeasure development against the hematopoietic acute radiation syndrome, as well as the use of radiation exposure protocols that mimic the likely nonuniform and heterogenous exposure consequent to a nuclear terrorist event. The review herein describes published studies of adult male cynomolgus macaques that used two exposure protocols-unilateral, nonuniform total-body irradiation and partial-body irradiation with bone marrow sparing-with the administration of subject-based medical management to assess mitigation against the hematopoietic acute radiation syndrome. These studies assessed the efficacy of cytokine combinations and cell-based therapy to mitigate acute radiation-induced myelosuppression. Both therapeutics were shown to mitigate the myelosuppression of the hematopoietic acute radiation syndrome. Additional studies being presented herein further defined the dose-dependent hematopoietic acute radiation syndrome of cynomolgus and rhesus macaques and a differential dose-dependent effect with young male and female cynomolgus macaques. The database supports the investigation of the cynomolgus macaque as a comparable non-human primate for efficacy testing under the US Food and Drug Administration animal rule. Critical gaps in knowledge required to validate the models and exposure protocols are also identified.
Collapse
Affiliation(s)
- Ann M Farese
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore
| | - Michel Drouet
- Armed Forces Biomedical Research Institute, France, Department of Radiobiology, Brétigny-sur-Orge, France
| | | | - Jean-Marc Bertho
- Institute of Radiation Protection and Nuclear Safety (IRSN), 31 avenue de la division Leclerc, 92260, Fontenay-aux-Roses, France; Present address: French Nuclear Safety Authority (ASN), 15 rue Louis Lejeune 92540 Montrouge, France
| | | | - Simon Authier
- Charles River, 445 Armand Frappier, Laval, QC, Canada, H7V 4B3
| | - Melanie Doyle-Eisele
- Lovelace Biomedical Research Institute, Laboratory Animal Sciences (Life Sciences), Albuquerque, NM
| | - Thomas J MacVittie
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore
| |
Collapse
|
62
|
Singh VK, Fatanmi OO, Wise SY, Carpenter A, Nakamura-Peek S, Serebrenik AA, Kaytor MD. A novel oral formulation of BIO 300 confers prophylactic radioprotection from acute radiation syndrome in mice. Int J Radiat Biol 2021; 98:958-967. [PMID: 34554032 DOI: 10.1080/09553002.2021.1981556] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
PURPOSE Exposure to high doses of ionizing radiation can result in hematopoietic acute radiation syndrome (H-ARS) and delayed effects of acute radiation exposure (DEARE). There is no radiation medical countermeasure (MCM) approved by the U.S. Food and Drug Administration which can be used prior to radiation exposure to protect exposed individuals. Different formulations containing synthetic genistein (BIO 300) are being developed to counter the harmful effects of radiation exposure. MATERIALS AND METHODS We investigated the efficacy of a BIO 300 oral powder (OP) formulation as a prophylactic radiation MCM against a lethal dose of cobalt-60 gamma-radiation in CD2F1 male mice while comparing to other formulations of BIO 300 and Neulasta (PEGylated filgrastim), a standard of care drug for H-ARS. RESULTS BIO 300 OP provided significant radioprotection against ionizing radiation in mice when administered twice per day for six days prior to total-body radiation exposure. Its radioprotective efficacy in the murine model was comparable to the efficacy of a single subcutaneous (sc) injection of Neulasta administered after total-body radiation exposure. CONCLUSIONS Our results demonstrate that BIO 300 OP, which can be administered orally, is a promising prophylactic radiation countermeasure for H-ARS.
Collapse
Affiliation(s)
- Vijay K Singh
- Department of Pharmacology and Molecular Therapeutics, Division of Radioprotectants, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Oluseyi O Fatanmi
- Department of Pharmacology and Molecular Therapeutics, Division of Radioprotectants, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Stephen Y Wise
- Department of Pharmacology and Molecular Therapeutics, Division of Radioprotectants, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Alana Carpenter
- Department of Pharmacology and Molecular Therapeutics, Division of Radioprotectants, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Sara Nakamura-Peek
- Department of Pharmacology and Molecular Therapeutics, Division of Radioprotectants, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.,Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | | | | |
Collapse
|
63
|
Singh VK, Seed TM. Radiation countermeasures for hematopoietic acute radiation syndrome: growth factors, cytokines and beyond. Int J Radiat Biol 2021; 97:1526-1547. [PMID: 34402734 DOI: 10.1080/09553002.2021.1969054] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE The intent of this article is to report the status of some of the pharmaceuticals currently in late stage development for possible use for individuals unwantedly and acutely injured as a result of radiological/nuclear exposures. The two major questions we attempt to address here are: (a) What medicinals are currently deemed by regulatory authorities (US FDA) to be safe and effective and are being stockpiled? (b) What additional agents might be needed to make the federal/state/local medicinal repositories more robust and useful in effectively managing contingencies involving radiation overexposures? CONCLUSIONS A limited number (precisely four) of medicinals have been deemed safe and effective, and are approved by the US FDA for the 'hematopoietic acute radiation syndrome (H-ARS).' These agents are largely recombinant growth factors (e.g. rhuG-CSF/filgrastim, rhuGM-CSF/sargramostim) that target and stimulate myeloid progenitors within bone marrow. Romiplostim, a small molecular agonist that enhances platelet production via stimulation of bone marrow megakaryocytes, has been recently approved and indicated for H-ARS. It is critical that additional agents for other major sub-syndromes of ARS (gastrointestinal-ARS) be approved. Future success in developing such medicinals will undoubtedly entail some form of a polypharmaceutical strategy, or perhaps novel, bioengineered chimeric agents with multiple, radioprotective/radiomitigative functionalities.
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, Bethesda, MD, USA.,Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | | |
Collapse
|
64
|
Forsberg MH, Kink JA, Thickens AS, Lewis BM, Childs CJ, Hematti P, Capitini CM. Exosomes from primed MSCs can educate monocytes as a cellular therapy for hematopoietic acute radiation syndrome. Stem Cell Res Ther 2021; 12:459. [PMID: 34407878 PMCID: PMC8371870 DOI: 10.1186/s13287-021-02491-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 07/04/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Acute radiation syndrome (ARS) is caused by acute exposure to ionizing radiation that damages multiple organ systems but especially the bone marrow (BM). We have previously shown that human macrophages educated with exosomes from human BM-derived mesenchymal stromal cells (MSCs) primed with lipopolysaccharide (LPS) prolonged survival in a xenogeneic lethal ARS model. The purpose of this study was to determine if exosomes from LPS-primed MSCs could directly educate human monocytes (LPS-EEMos) for the treatment of ARS. METHODS Human monocytes were educated by exosomes from LPS-primed MSCs and compared to monocytes educated by unprimed MSCs (EEMos) and uneducated monocytes to assess survival and clinical improvement in a xenogeneic mouse model of ARS. Changes in surface molecule expression of exosomes and monocytes after education were determined by flow cytometry, while gene expression was determined by qPCR. Irradiated human CD34+ hematopoietic stem cells (HSCs) were co-cultured with LPS-EEMos, EEMos, or uneducated monocytes to assess effects on HSC survival and proliferation. RESULTS LPS priming of MSCs led to the production of exosomes with increased expression of CD9, CD29, CD44, CD146, and MCSP. LPS-EEMos showed increases in gene expression of IL-6, IL-10, IL-15, IDO, and FGF-2 as compared to EEMos generated from unprimed MSCs. Generation of LPS-EEMos induced a lower percentage of CD14+ monocyte subsets that were CD16+, CD73+, CD86+, or CD206+ but a higher percentage of PD-L1+ cells. LPS-EEMos infused 4 h after lethal irradiation significantly prolonged survival, reducing clinical scores and weight loss as compared to controls. Complete blood counts from LPS-EEMo-treated mice showed enhanced hematopoietic recovery post-nadir. IL-6 receptor blockade completely abrogated the radioprotective survival benefit of LPS-EEMos in vivo in female NSG mice, but only loss of hematopoietic recovery was noted in male NSG mice. PD-1 blockade had no effect on survival. Furthermore, LPS-EEMos also showed benefits in vivo when administered 24 h, but not 48 h, after lethal irradiation. Co-culture of unprimed EEMos or LPS-EEMos with irradiated human CD34+ HSCs led to increased CD34+ proliferation and survival, suggesting hematopoietic recovery may be seen clinically. CONCLUSION LPS-EEMos are a potential counter-measure for hematopoietic ARS, with a reduced biomanufacturing time that facilitates hematopoiesis.
Collapse
Affiliation(s)
- Matthew H Forsberg
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, 1111 Highland Ave, WIMR 4137, Madison, WI, 53705, USA
| | - John A Kink
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, 1111 Highland Ave, WIMR 4033, Madison, WI, 53705, USA
| | - Anna S Thickens
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, 1111 Highland Ave, WIMR 4033, Madison, WI, 53705, USA
| | - Bryson M Lewis
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, 1111 Highland Ave, WIMR 4033, Madison, WI, 53705, USA
| | - Charlie J Childs
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, 1111 Highland Ave, WIMR 4033, Madison, WI, 53705, USA
| | - Peiman Hematti
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA.
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, 1111 Highland Ave, WIMR 4033, Madison, WI, 53705, USA.
| | - Christian M Capitini
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, 1111 Highland Ave, WIMR 4137, Madison, WI, 53705, USA.
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA.
| |
Collapse
|
65
|
Yamamoto Y, Minami M, Yoshida K, Nagata M, Miyata T, Yang T, Takayama N, Suzuki K, Okawa M, Yamada K, Miyamoto S. Irradiation Accelerates Plaque Formation and Cellular Senescence in Flow-Altered Carotid Arteries of Apolipoprotein E Knock-Out Mice. J Am Heart Assoc 2021; 10:e020712. [PMID: 34227406 PMCID: PMC8483483 DOI: 10.1161/jaha.120.020712] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Background Chronic inflammation through cellular senescence, known as the senescence‐associated secretory phenotype, is a mechanism of various organ diseases, including atherosclerosis. Particularly, ionizing radiation (IR) contributes to cellular senescence by causing DNA damage. Although previous clinical studies have demonstrated that radiotherapy causes atherosclerosis as a long‐term side effect, the detailed mechanism is unclear. This study was conducted to investigate the relationship between radiation‐induced atherosclerosis and senescence‐associated secretory phenotype in murine carotid arteries. Methods and Results Partial ligation of the left carotid artery branches in 9‐week‐old male apolipoprotein E‐deficient mice was performed to induce atherosclerosis. The mice received total body irradiation at a dose of 6 Gy using gamma rays at 2 weeks post operation. We compared the samples collected 4 weeks after IR with unirradiated control samples. The IR and control groups presented pathologically progressive lesions in 90.9% and 72.3% of mice, respectively. Plaque volume, macrophage accumulation, and phenotype switching of vascular smooth muscle cells were advanced in the IR group. Irradiated samples showed increased persistent DNA damage response (53BP1 [p53 binding protein 1]), upregulated cyclin‐dependent kinase inhibitors (p16INK4a and p21), and elevated inflammatory chemokines expression (monocyte chemotactic protein‐1, keratinocyte‐derived chemokine, and macrophage inflammatory protein 2). Conclusions IR promoted plaque growth in murine carotid arteries. Our findings support the possibility that senescence‐associated secretory phenotype aggravates atherogenesis in irradiated artery. This mice model might contribute to mechanism elucidation of radiation‐induced atherosclerosis.
Collapse
Affiliation(s)
- Yu Yamamoto
- Department of Neurosurgery Kyoto University Graduate School of Medicine Kyoto Japan.,Department of Clinical Innovative Medicine Kyoto University Graduate School of Medicine Kyoto Japan
| | - Manabu Minami
- Department of Clinical Innovative Medicine Kyoto University Graduate School of Medicine Kyoto Japan.,Department of Data Science National Cerebral and Cardiovascular Center Suita Japan
| | - Kazumichi Yoshida
- Department of Neurosurgery Kyoto University Graduate School of Medicine Kyoto Japan
| | - Manabu Nagata
- Department of Neurosurgery Kyoto University Graduate School of Medicine Kyoto Japan.,Department of Clinical Innovative Medicine Kyoto University Graduate School of Medicine Kyoto Japan
| | - Takeshi Miyata
- Department of Neurosurgery Kyoto University Graduate School of Medicine Kyoto Japan.,Department of Clinical Innovative Medicine Kyoto University Graduate School of Medicine Kyoto Japan
| | - Tao Yang
- Department of Neurosurgery Kyoto University Graduate School of Medicine Kyoto Japan.,Department of Clinical Innovative Medicine Kyoto University Graduate School of Medicine Kyoto Japan
| | - Naoki Takayama
- Department of Neurosurgery Kyoto University Graduate School of Medicine Kyoto Japan
| | - Keita Suzuki
- Department of Neurosurgery Kyoto University Graduate School of Medicine Kyoto Japan
| | - Masakazu Okawa
- Department of Neurosurgery Kyoto University Graduate School of Medicine Kyoto Japan
| | - Kiyofumi Yamada
- Department of Neurosurgery Kyoto University Graduate School of Medicine Kyoto Japan
| | - Susumu Miyamoto
- Department of Neurosurgery Kyoto University Graduate School of Medicine Kyoto Japan
| |
Collapse
|
66
|
Sanguri S, Gupta D. Prebiotic Mannan Oligosaccharide Pretreatment Improves Mice Survival Against Lethal Effects of Gamma Radiation by Protecting GI Tract and Hematopoietic Systems. Front Oncol 2021; 11:677781. [PMID: 34249717 PMCID: PMC8266395 DOI: 10.3389/fonc.2021.677781] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/27/2021] [Indexed: 11/13/2022] Open
Abstract
Total body irradiation (TBI) results in critical injuries in a dose dependent manner that primarily damages highly proliferating tissues including hematopoietic stem cells (HSCs) and intestinal crypt stem cells etc. This may result in hematopoietic syndrome leading to bone marrow failure and gastrointestinal syndrome leading to chronic intestinal functional alterations. Death results from the gastrointestinal syndrome due to sepsis, bleeding, dehydration, and multi-system organ failure. We demonstrate that the prebiotic mannan oligosaccharide (MOS) pretreatment substantially prolongs survival in both male and female mice when administered 2 h prior to radiation either through oral or intraperitoneal route. The radioprotective efficacy of MOS was found to be age dependent and improves survival even in aged mice (12–13 months old). MOS pretreatment effectively abrogates radiation-induced hematopoietic injury and accelerates recovery of lymphocytes and WBCs and alleviates depletion of circulatory blood cells. Results also illustrate that MOS pretreatment abolishes crypt cell death and denudation of villi in comparison to the respective irradiated animals and ameliorates the overall radiation-induced damage to the GI system. MOS pretreatment facilitates intestinal recovery leading to enhanced animal survival demonstrating its protection efficacy against TBI induced mortality. Moreover, MOS pretreated animals show signs of accelerated recovery in terms of severity of radiation sickness symptoms including weight loss and completely abolish TBI associated mortality.
Collapse
Affiliation(s)
- Sweta Sanguri
- Division of Metabolic Cell Signaling Research, Institute of Nuclear Medicine & Allied Sciences, Delhi, India
| | - Damodar Gupta
- Division of Metabolic Cell Signaling Research, Institute of Nuclear Medicine & Allied Sciences, Delhi, India
| |
Collapse
|
67
|
Haritwal T, Goyal N, Gupta N, Parvez S, Agrawala PK. Trichostatin A mitigates radiation induced teratogenesis in C57 Bl/6 mice. Mutagenesis 2021; 36:303-309. [PMID: 34086940 DOI: 10.1093/mutage/geab018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 06/03/2021] [Indexed: 11/13/2022] Open
Abstract
Radiation exposure in utero is known to lead to serious concerns to both the mother and children, including developmental anomalies in the children. In the recent past trichostatn A, an HDAC inhibitor and epigenetic modifier, has been shown to mitigate radiation induced anomalies in the male reproductive system of C57BL/6 mice. Therefore, the current study was undertaken to evaluate the mitigating effects of trichostatin A against radiation induced developmental anomalies in mice. Fetuses of in utero whole body gamma irradiated mice during active organogenesis period were examined for developmental anomalies on 8.5 and 18.5 day of gestation. In utero radiation exposure caused developmental anomalies like microcephaly, microphthalamia, gastroschisis and kinky tail besides prenatal mortality. Trichostatin A administration post irradiation was observed to reduce 50% of prenatal mortality at E18.5 by reducing congenital and developmental anomalies. Observation of such results could be corroborated with the HDAC inhibitory potential of trichostatin A knowing that developmental anomalies may have epigenetic origin. Trichostatin A therefore can be considered as a potential radiomitigator.
Collapse
Affiliation(s)
- Teena Haritwal
- Department of Radiation Genetics and Epigenetics, Institute of Nuclear Medicine and Allied Sciences, DRDO, Brig SK Mazumdar Road, Timarpur, Delhi 110054, INDIA
| | - Nikita Goyal
- Department of Radiation Genetics and Epigenetics, Institute of Nuclear Medicine and Allied Sciences, DRDO, Brig SK Mazumdar Road, Timarpur, Delhi 110054, INDIA
| | - Noopur Gupta
- Department of Radiation Genetics and Epigenetics, Institute of Nuclear Medicine and Allied Sciences, DRDO, Brig SK Mazumdar Road, Timarpur, Delhi 110054, INDIA
| | - Suhel Parvez
- Department of Medical Entomology and Toxicology, School of Life Sciences, Jamia Hamdard University, Hamdard Nagar, New Delhi 110062, INDIA
| | - Paban K Agrawala
- Department of Radiation Genetics and Epigenetics, Institute of Nuclear Medicine and Allied Sciences, DRDO, Brig SK Mazumdar Road, Timarpur, Delhi 110054, INDIA
| |
Collapse
|
68
|
Yang E, Choi H, Park JS, Noh YW, Choi CM, Lee WJ, Ko JW, Kim J. A first-in-human study of KMRC011, a potential treatment for acute radiation syndrome, to explore tolerability, pharmacokinetics, and pharmacodynamics. Clin Transl Sci 2021; 14:2161-2170. [PMID: 34080313 PMCID: PMC8604209 DOI: 10.1111/cts.13073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/24/2021] [Accepted: 04/02/2021] [Indexed: 11/27/2022] Open
Abstract
KMRC011 is a novel Toll-like receptor 5 agonist under development as a treatment for acute radiation syndrome (ARS). The aim of this first-in-human study was to investigate the tolerability, pharmacokinetics, and pharmacodynamics of a single intramuscular dose of KMRC011 in healthy subjects. A randomized, single-blind, placebo-controlled, single dose-escalation study was conducted with the starting dose of 5 μg. Eight (4 only for 5 μg cohort) subjects per cohort were randomly assigned to KMRC011 or placebo in a 3:1 ratio. Dose-limiting toxicity (DLT) was assessed throughout the study. Serum concentrations of KMRC011, granulocyte colony-stimulating factor (G-CSF), and interleukin-6 (IL-6) were measured up to 48 h postdose. Based on safety review, the dose of KMRC011 escalated up to 20 μg, and consequently, a total of 4 dose levels (5, 10, 15, and 20 μg) were explored. The most common adverse event was injection site reaction, showing no dose-related trend. Three DLTs (2 cases of hepatic enzyme increased and 1 of pyrexia) were observed; 1 in the 15 μg cohort and 2 in the 20 μg cohort. A developed method could not detect any KMRC011 in serum. KMRC011 15 μg and 20 μg showed significant increases of G-CSF, IL-6, and absolute neutrophil counts, compared with the placebo. A single intramuscular administration of KMRC011 ranging from 5 to 15 μg was tolerated in healthy subjects. Doses of KMRC011 equal to or greater than 15 μg exerted TLR5 agonist-like activities by increasing serum G-CSF and IL-6. It suggests that KMRC011 has the potential for a treatment for ARS.
Collapse
Affiliation(s)
- Eunsol Yang
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | - Hyejung Choi
- Department of Clinical Pharmacology and Therapeutics, Samsung Medical Center, Seoul, Korea
| | - Jin-Sol Park
- Department of Clinical Pharmacology and Therapeutics, Samsung Medical Center, Seoul, Korea
| | - Young-Woock Noh
- New Drug Development Center, Osong Medical Innovation Foundation, Cheongju-si, Korea
| | | | - Woo-Jong Lee
- Biomedical Manufacturing Technology Center, Korea Institute of Industrial Technology, Yeongcheon-si, Korea
| | - Jae-Wook Ko
- Department of Clinical Pharmacology and Therapeutics, Samsung Medical Center, Seoul, Korea
| | - Jungryul Kim
- Department of Clinical Pharmacology and Therapeutics, Samsung Medical Center, Seoul, Korea.,Department of Clinical Research Design & Evaluation, SAIHST, Sungkyunkwan University, Seoul, Korea
| |
Collapse
|
69
|
Li Y, Girgis M, Wise SY, Fatanmi OO, Seed TM, Maniar M, Cheema AK, Singh VK. Analysis of the metabolomic profile in serum of irradiated nonhuman primates treated with Ex-Rad, a radiation countermeasure. Sci Rep 2021; 11:11449. [PMID: 34075191 PMCID: PMC8169671 DOI: 10.1038/s41598-021-91067-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 05/21/2021] [Indexed: 01/02/2023] Open
Abstract
To date, the United States Food and Drug Administration (FDA) has approved four drugs to mitigate hematopoietic acute radiation syndrome and all four are repurposed radiomitigators. There are several additional drug candidates currently under evaluation that may also be helpful for use during a widespread emergency. One possible candidate is Ex-Rad, also known as ON01210, a chlorobenzyl sulfone derivative (organosulfur compound), which is a novel, small-molecule kinase inhibitor with demonstrated efficacy in the murine model. In this study, we have evaluated the metabolomic and lipidomic profiles in serum samples of nonhuman primates (NHPs) treated with Ex-Rad after exposure to ionizing radiation. Two different dose administration schedules (Ex-Rad I administered 24 and 36 h post-irradiation, and Ex-Rad II administered 48 and 60 h post-irradiation), were used and evaluated using a global molecular profiling approach. We observed alterations in biochemical pathways relating to inflammation and oxidative stress after radiation exposure that were alleviated in animals that received Ex-Rad I or Ex-Rad II. The results from this study lend credence to the possible radiomitigative effects of this drug possibly via a dampening of metabolism-based tissue injury, thus aiding in recovery of vital, radiation-injured organ systems.
Collapse
Affiliation(s)
- Yaoxiang Li
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Michael Girgis
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, 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, USA.,Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, 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, USA.,Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Thomas M Seed
- Tech Micro Services, 4417 Maple Avenue, Bethesda, MD, 20814, USA
| | - Manoj Maniar
- Onconova Therapeutics, Inc, 375 Pheasant Run, Newtown, PA, USA
| | - Amrita K Cheema
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA.,Department of Biochemistry, Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC, USA
| | - 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, USA. .,Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| |
Collapse
|
70
|
Bene BJ, Blakely WF, Burmeister DM, Cary L, Chhetri SJ, Davis CM, Ghosh SP, Holmes-Hampton GP, Iordanskiy S, Kalinich JF, Kiang JG, Kumar VP, Lowy RJ, Miller A, Naeem M, Schauer DA, Senchak L, Singh VK, Stewart AJ, Velazquez EM, Xiao M. Celebrating 60 Years of Accomplishments of the Armed Forces Radiobiology Research Institute1. Radiat Res 2021; 196:129-146. [PMID: 33979439 DOI: 10.1667/21-00064.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 04/20/2021] [Indexed: 11/03/2022]
Abstract
Chartered by the U.S. Congress in 1961, the Armed Forces Radiobiology Research Institute (AFRRI) is a Joint Department of Defense (DoD) entity with the mission of carrying out the Medical Radiological Defense Research Program in support of our military forces around the globe. In the last 60 years, the investigators at AFRRI have conducted exploratory and developmental research with broad application to the field of radiation sciences. As the only DoD facility dedicated to radiation research, AFRRI's Medical Radiobiology Advisory Team provides deployable medical and radiobiological subject matter expertise, advising commanders in the response to a U.S. nuclear weapon incident and other nuclear or radiological material incidents. AFRRI received the DoD Joint Meritorious Unit Award on February 17, 2004, for its exceptionally meritorious achievements from September 11, 2001 to June 20, 2003, in response to acts of terrorism and nuclear/radiological threats at home and abroad. In August 2009, the American Nuclear Society designated the institute a nuclear historic landmark as the U.S.'s primary source of medical nuclear and radiological research, preparedness and training. Since then, research has continued, and core areas of study include prevention, assessment and treatment of radiological injuries that may occur from exposure to a wide range of doses (low to high). AFRRI collaborates with other government entities, academic institutions, civilian laboratories and other countries to research the biological effects of ionizing radiation. Notable early research contributions were the establishment of dose limits for major acute radiation syndromes in primates, applicable to human exposures, followed by the subsequent evolution of radiobiology concepts, particularly the importance of immune collapse and combined injury. In this century, the program has been essential in the development and validation of prophylactic and therapeutic drugs, such as Amifostine, Neupogen®, Neulasta®, Nplate® and Leukine®, all of which are used to prevent and treat radiation injuries. Moreover, AFRRI has helped develop rapid, high-precision, biodosimetry tools ranging from novel assays to software decision support. New drug candidates and biological dose assessment technologies are currently being developed. Such efforts are supported by unique and unmatched radiation sources and generators that allow for comprehensive analyses across the various types and qualities of radiation. These include but are not limited to both 60Co facilities, a TRIGA® reactor providing variable mixed neutron and γ-ray fields, a clinical linear accelerator, and a small animal radiation research platform with low-energy photons. There are five major research areas at AFRRI that encompass the prevention, assessment and treatment of injuries resulting from the effects of ionizing radiation: 1. biodosimetry; 2. low-level and low-dose-rate radiation; 3. internal contamination and metal toxicity; 4. radiation combined injury; and 5. radiation medical countermeasures. These research areas are bolstered by an educational component to broadcast and increase awareness of the medical effects of ionizing radiation, in the mass-casualty scenario after a nuclear detonation or radiological accidents. This work provides a description of the military medical operations as well as the radiation facilities and capabilities present at AFRRI, followed by a review and discussion of each of the research areas.
Collapse
Affiliation(s)
| | | | | | - Lynnette Cary
- Scientific Research Department.,Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | | | - Catherine M Davis
- Scientific Research Department.,Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Sanchita P Ghosh
- Scientific Research Department.,Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Gregory P Holmes-Hampton
- Scientific Research Department.,Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Sergey Iordanskiy
- Scientific Research Department.,Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | | | - Juliann G Kiang
- Scientific Research Department.,Medicine.,Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | | | | | | | | | - David A Schauer
- Radiation Sciences Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | | | - Vijay K Singh
- Scientific Research Department.,Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | | | | | | |
Collapse
|
71
|
Singh VK, Seed TM. Repurposing Pharmaceuticals Previously Approved by Regulatory Agencies to Medically Counter Injuries Arising Either Early or Late Following Radiation Exposure. Front Pharmacol 2021; 12:624844. [PMID: 34040517 PMCID: PMC8141805 DOI: 10.3389/fphar.2021.624844] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 04/26/2021] [Indexed: 12/12/2022] Open
Abstract
The increasing risks of radiological or nuclear attacks or associated accidents have served to renew interest in developing radiation medical countermeasures. The development of prospective countermeasures and the subsequent gain of Food and Drug Administration (FDA) approval are invariably time consuming and expensive processes, especially in terms of generating essential human data. Due to the limited resources for drug development and the need for expedited drug approval, drug developers have turned, in part, to the strategy of repurposing agents for which safety and clinical data are already available. Approval of drugs that are already in clinical use for one indication and are being repurposed for another indication is inherently faster and more cost effective than for new agents that lack regulatory approval of any sort. There are four known growth factors which have been repurposed in the recent past as radiomitigators following the FDA Animal Rule: Neupogen, Neulasta, Leukine, and Nplate. These four drugs were in clinic for several decades for other indications and were repurposed. A large number of additional agents approved by various regulatory authorities for given indications are currently under investigation for dual use for acute radiation syndrome or for delayed pathological effects of acute radiation exposure. The process of drug repurposing, however, is not without its own set of challenges and limitations.
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, Bethesda, MD, United States
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | | |
Collapse
|
72
|
Li X, Wang X, Miao L, Liu Y, Lin X, Guo Y, Yuan R, Tian H. Synthesis and radioprotective effects of novel hybrid compounds containing edaravone analogue and 3-n-butylphthalide ring-opening derivatives. J Cell Mol Med 2021; 25:5470-5485. [PMID: 33963805 PMCID: PMC8184683 DOI: 10.1111/jcmm.16557] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 03/22/2021] [Accepted: 04/01/2021] [Indexed: 12/12/2022] Open
Abstract
As the potential risk of radiation exposure is increasing, radioprotectors studies are gaining importance. In this study, novel hybrid compounds containing edaravone analogue and 3-n-butylphthalide ring-opening derivatives were synthesized, and their radioprotective effects were evaluated. Among these, compound 10a displayed the highest radioprotective activity in IEC-6 and HFL-1 cells. Its oral administration increased the survival rates of irradiated mice and alleviated total body irradiation (TBI)-induced hematopoietic damage by mitigating myelosuppression and improving hematopoietic stem/progenitor cell frequencies. Furthermore, 10a treatment prevented abdominal irradiation (ABI)-induced structural damage to the small intestine. Experiment results demonstrated that 10a increased the number of Lgr5+ intestinal stem cells, lysozyme+ Paneth cells and Ki67+ transient amplifying cells, and reduced apoptosis of the intestinal epithelium cells in irradiated mice. Moreover, in vitro and in vivo studies demonstrated that the radioprotective activity of 10a is associated to the reduction of oxidative stress and the inhibition of DNA damage. Furthermore, compound 10a downregulated the expressions of p53, Bax, caspase-9 and caspase-3, and upregulated the expression of Bcl-2, suggesting that it could prevent irradiation-induced intestinal damage through the p53-dependent apoptotic pathway. Collectively, these findings demonstrate that 10a is beneficial for the prevention of radiation damage and has the potential to be a radioprotector.
Collapse
Affiliation(s)
- Xuejiao Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, China
| | - Xinxin Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, China
| | - Longfei Miao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, China
| | - Yahong Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, China
| | - Xiaona Lin
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, China
| | - Yuying Guo
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, China
| | - Renbin Yuan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, China
| | - Hongqi Tian
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, China
| |
Collapse
|
73
|
Bandekar M, Maurya DK, Sharma D, Sandur SK. Preclinical Studies and Clinical Prospects of Wharton's Jelly-Derived MSC for Treatment of Acute Radiation Syndrome. CURRENT STEM CELL REPORTS 2021; 7:85-94. [PMID: 33936933 PMCID: PMC8080090 DOI: 10.1007/s40778-021-00188-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2021] [Indexed: 02/07/2023]
Abstract
Purpose of Review Wharton’s jelly-derived mesenchymal stem cells (WJ-MSCs) have received widespread attention from researchers owing to the remarkable benefits offered by these cells over other stem cells. The primitive nature of WJ-MSCs, ease of isolation, differentiation ability, and immuno-modulatory nature make these cells superior to bone marrow MSCs and ideal to treat various human ailments. This review explores ability of WJ-MSCs to mitigate acute radiation syndrome caused by planned or unplanned radiation exposure. Recent Findings Recent reports suggest that WJ-MSCs home to damaged tissues in irradiated host and mitigate radiation induced damage to radiosensitive tissues such as hematopoietic and gastrointestinal systems. WJ-MSCs and conditioned media were found to protect mice from radiation induced mortality and also prevent radiation dermatitis. Local irradiation-induced lung toxicity in mice was significantly reduced by CXCR4 over-expressing WJ-MSCs. Summary Emerging evidences support safety and effectiveness of WJ-MSCs for treatment of acute radiation syndrome and lung injury after planned or accidental exposure. Additionally, conditioned media collected after culturing WJ-MSCs can also be used for mitigation of radiation dermatitis. Clinical translation of these findings would be possible after careful evaluation of resilience, effectiveness, and molecular mechanism of action of xenogeneic WJ-MSCs in non-human primates.
Collapse
Affiliation(s)
- Mayuri Bandekar
- Radiation Biology & Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Trombay, Mumbai, 400085 India.,University of Mumbai, Kalina, Mumbai, 400098 India
| | - Dharmendra K Maurya
- Radiation Biology & Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Trombay, Mumbai, 400085 India.,Homi Bhabha National Institute, Anushaktinagar, Mumbai, 400094 India
| | - Deepak Sharma
- Radiation Biology & Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Trombay, Mumbai, 400085 India.,Homi Bhabha National Institute, Anushaktinagar, Mumbai, 400094 India
| | - Santosh K Sandur
- Radiation Biology & Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Trombay, Mumbai, 400085 India.,Homi Bhabha National Institute, Anushaktinagar, Mumbai, 400094 India
| |
Collapse
|
74
|
Clayton NP, Khan-Malek RC, Dangler CA, Zhang D, Ascah A, Gains M, Gardner B, Mockbee C, Keutzer JM, McManus J, Authier S. Sargramostim (rhu GM-CSF) Improves Survival of Non-Human Primates with Severe Bone Marrow Suppression after Acute, High-Dose, Whole-Body Irradiation. Radiat Res 2021; 195:191-199. [PMID: 33302291 DOI: 10.1667/rade-20-00131.1] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 11/02/2020] [Indexed: 11/03/2022]
Abstract
Exposure to acute, high-dose, whole-body ionizing radiation results in bone marrow failure (hematopoietic acute radiation syndrome with resultant infection, bleeding, anemia, and increased risk of death). Sargramostim (yeast-derived rhu GM-CSF), a yeast-derived, molecularly cloned, hematopoietic growth factor and pleiotropic cytokine supports proliferation, differentiation, maturation and survival of cells of several myeloid lineages. We evaluated the efficacy of sargramostim in non-human primates (rhesus macaques) exposed to whole-body ionizing radiation at a 50-60% lethal dose. The primary end point was day 60 survival. Non-human primates received daily subcutaneous sargramostim (7 mcg/kg/day) or control. To reflect the anticipated setting of a nuclear or radiologic event, treatment began 48 h postirradiation, and non-human primates received only moderate supportive care (no whole blood transfusions or individualized antibiotics). Sargramostim significantly increased day 60 survival to 78% (95% confidence interval, 61-90%) vs. 42% (26-59%; P = 0.0018) in controls. Neutrophil, platelet and lymphocyte recovery rates were accelerated and infection rates decreased. Improved survival when sargramostim was started 48 h postirradiation, without use of intensive supportive care, suggests sargramostim may be effective in treating humans exposed to acute, high-dose whole-body, ionizing radiation in a scenario such as a mass casualty event.
Collapse
Affiliation(s)
| | | | | | - Donghui Zhang
- Global Biostatistics and Programming, Sanofi, Bridgewater, New Jersey
| | | | | | | | | | - Joan M Keutzer
- Global Rare Diseases, Sanofi Genzyme, Cambridge, Massachusetts
| | - John McManus
- Partner Therapeutics, Inc, Lexington, Massachusetts
| | | |
Collapse
|
75
|
Cheda A, Nowosielska EM, Gebicki J, Marcinek A, Chlopicki S, Janiak MK. A derivative of vitamin B 3 applied several days after exposure reduces lethality of severely irradiated mice. Sci Rep 2021; 11:7922. [PMID: 33846380 PMCID: PMC8041812 DOI: 10.1038/s41598-021-86870-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 03/22/2021] [Indexed: 12/02/2022] Open
Abstract
Most, if not all, of the hitherto tested substances exert more or less pronounced pro-survival effects when applied before or immediately after the exposure to high doses of ionizing radiation. In the present study we demonstrate for the first time that 1-methyl nicotinamide (MNA), a derivative of vitamin B3, significantly (1.6 to 1.9 times) prolonged survival of BALB/c mice irradiated at LD30/30 (6.5 Gy), LD50/30 (7.0 Gy) or LD80/30 (7.5 Gy) of γ-rays when the MNA administration started as late as 7 days post irradiation. A slightly less efficient and only after the highest dose (7.5 Gy) of γ-rays was another vitamin B3 derivative, 1-methyl-3-acetylpyridine (1,3-MAP) (1.4-fold prolonged survival). These pro-survival effects did not seem to be mediated by stimulation of haematopoiesis, but might be related to anti-inflammatory and/or anti-thrombotic properties of the vitamin B3 derivatives. Our results show that MNA may represent a prototype of a radioremedial agent capable of mitigating the severity and/or progression of radiation-induced injuries when applied several hours or days after exposure to high doses of ionizing radiation.
Collapse
Affiliation(s)
- Aneta Cheda
- Department of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, 4 Kozielska St., 01-163, Warsaw, Poland.
| | - Ewa M Nowosielska
- Department of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, 4 Kozielska St., 01-163, Warsaw, Poland
| | - Jerzy Gebicki
- Institute of Applied Radiation Chemistry, Lodz University of Technology, 15 Wroblewskiego St., 93-590, Lodz, Poland
| | - Andrzej Marcinek
- Institute of Applied Radiation Chemistry, Lodz University of Technology, 15 Wroblewskiego St., 93-590, Lodz, Poland
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 14 Bobrzynskiego St., 30-348, Kraków, Poland
- Chair of Pharmacology, Jagiellonian University Medical College, Jagiellonian University, 16 Grzegorzecka St., 31-531, Kraków, Poland
| | - Marek K Janiak
- Department of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, 4 Kozielska St., 01-163, Warsaw, Poland
| |
Collapse
|
76
|
Zhu S, Liang J, Zhu F, Zhang X, Xu M, Zhao K, Zeng L, Xu K. The effects of myeloablative or non-myeloablative total body irradiations on intestinal tract in mice. Biosci Rep 2021; 41:BSR20202993. [PMID: 33605406 PMCID: PMC7926181 DOI: 10.1042/bsr20202993] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 01/25/2021] [Accepted: 02/18/2021] [Indexed: 12/12/2022] Open
Abstract
Acute radiation injury caused by high-dose radiation exposure severely impedes the application of radiotherapy in cancer management. To deeply understand the side effects of radiation on intestinal tract, an irradiation murine model was applied and evaluated. C57BL/6 mice were given 4 Gy non-myeloablative irradiation, 8 Gy myeloablative irradiation and non-irradiation (control), respectively. Results demonstrated that the 8 Gy myeloablative irradiations significantly damaged the gut barrier along with decreasing MECA32 and ZO-1. However, a slight increase in MECA32 and ZO-1 was detected in the 4 Gy non-myeloablative irradiations treatment from day 5 to day 10. Further, the irradiations affected the expression of P38 and JNK mitogen-activated protein kinase (MAPK) but not ERK1/2 MAPK signal pathway. Moreover, irradiation had adverse effects on hematopoietic system, altered the numbers and percentages of intestinal inflammatory cells. The IL-17/AhR had big increase in the gut of 4 Gy irradiation mice at day 10 compared with other groups. Both 8 Gy myeloablative and 4 Gy non-myeloablative irradiation disturbed the levels of short-chain fatty acids (SCFAs) in intestine. Meanwhile, high dosage of irradiation decreased the intestinal bacterial diversity and altered the community composition. Importantly, the fatty acids generating bacteria Bacteroidaceae and Ruminococcaceae played key roles in community distribution and SCFAs metabolism after irradiation. Collectively, the irradiation induced gut barrier damage with dosages dependent that led to the decreased p38 MAPK and increased JNK MAPK, unbalanced the mononuclear cells (MNCs) of gut, disturbed intestinal bacterial community and SCFAs level.
Collapse
Affiliation(s)
- Shengyun Zhu
- Institute of Blood Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- Department of Hematology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- The Key Laboratory of Bone Marrow Stem Cell, Jiangsu 221002, China
| | - Jing Liang
- Institute of Blood Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Feng Zhu
- Institute of Blood Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- Department of Hematology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- The Key Laboratory of Bone Marrow Stem Cell, Jiangsu 221002, China
| | - Xue Zhang
- Institute of Blood Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Mengdi Xu
- Institute of Blood Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Kai Zhao
- Institute of Blood Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- Department of Hematology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- The Key Laboratory of Bone Marrow Stem Cell, Jiangsu 221002, China
| | - Lingyu Zeng
- Institute of Blood Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- Department of Hematology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- The Key Laboratory of Bone Marrow Stem Cell, Jiangsu 221002, China
| | - Kailin Xu
- Institute of Blood Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- Department of Hematology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
- The Key Laboratory of Bone Marrow Stem Cell, Jiangsu 221002, China
| |
Collapse
|
77
|
Li Y, Singh J, Varghese R, Zhang Y, Fatanmi OO, Cheema AK, Singh VK. Transcriptome of rhesus macaque (Macaca mulatta) exposed to total-body irradiation. Sci Rep 2021; 11:6295. [PMID: 33737626 PMCID: PMC7973550 DOI: 10.1038/s41598-021-85669-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/02/2021] [Indexed: 12/13/2022] Open
Abstract
The field of biodosimetry has seen a paradigm shift towards an increased use of molecular phenotyping technologies including omics and miRNA, in addition to conventional cytogenetic techniques. Here, we have used a nonhuman primate (NHP) model to study the impact of gamma-irradiation on alterations in blood-based gene expression. With a goal to delineate radiation induced changes in gene expression, we followed eight NHPs for 60 days after exposure to 6.5 Gy gamma-radiation for survival outcomes. Analysis of differential gene expression in response to radiation exposure yielded 26,944 dysregulated genes that were not significantly impacted by sex. Further analysis showed an increased association of several pathways including IL-3 signaling, ephrin receptor signaling, ErbB signaling, nitric oxide signaling in the cardiovascular system, Wnt/β-catenin signaling, and inflammasome pathway, which were associated with positive survival outcomes in NHPs after acute exposure to radiation. This study provides novel insights into major pathways and networks involved in radiation-induced injuries that may identify biomarkers for radiation injury.
Collapse
Affiliation(s)
- Yaoxiang Li
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Jatinder Singh
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine "America's Medical School", Uniformed Serices University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, USA.,Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Rency Varghese
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Yubo Zhang
- Department of Biochemistry and Molecular Medicine, George Washington University, Washington, DC, USA
| | - Oluseyi O Fatanmi
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine "America's Medical School", Uniformed Serices University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, USA.,Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Amrita K Cheema
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA.,Department of Biochemistry, Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC, USA
| | - Vijay K Singh
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine "America's Medical School", Uniformed Serices University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, USA. .,Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| |
Collapse
|
78
|
Mechanisms of radiation-induced endothelium damage: Emerging models and technologies. Radiother Oncol 2021; 158:21-32. [PMID: 33581220 DOI: 10.1016/j.radonc.2021.02.007] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 12/24/2022]
Abstract
Radiation-induced endothelial/vascular injury is a major complicating factor in radiotherapy and a leading cause of morbidity and mortality in nuclear or radiological catastrophes. Exposure of tissue to ionizing radiation (IR) leads to the release of oxygen radicals and proteases that result in loss of endothelial barrier function and leukocyte dysfunction leading to tissue injury and organ damage. Microvascular endothelial cells are particularly sensitive to IR and radiation-induced alterations in endothelial cell function are thought to be a critical factor in organ damage through endothelial cell activation, enhanced leukocyte-endothelial cell interactions, increased barrier permeability and initiation of apoptotic pathways. These radiation-induced inflammatory responses are important in early and late radiation pathologies in various organs. A better understanding of mechanisms of radiation-induced endothelium dysfunction is therefore vital, as radiobiological response of endothelium is of major importance for medical management and therapeutic development for radiation injuries. In this review, we summarize the current knowledge of cellular and molecular mechanisms of radiation-induced endothelium damage and their impact on early and late radiation injury. Furthermore, we review established and emerging in vivo and in vitro models that have been developed to study the mechanisms of radiation-induced endothelium damage and to design, develop and rapidly screen therapeutics for treatment of radiation-induced vascular damage. Currently there are no specific therapeutics available to protect against radiation-induced loss of endothelial barrier function, leukocyte dysfunction and resulting organ damage. Developing therapeutics to prevent endothelium dysfunction and normal tissue damage during radiotherapy can serve as the urgently needed medical countermeasures.
Collapse
|
79
|
Kenchegowda D, Seed TM, Singh VK. What are the practical, ethical, and pathobiological considerations in the use of minipigs as an animal model in drug discovery for acute radiation syndrome and delayed effects of acute radiation exposure? Expert Opin Drug Discov 2021; 16:119-124. [PMID: 32892657 DOI: 10.1080/17460441.2020.1811671] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 08/14/2020] [Indexed: 02/02/2023]
Affiliation(s)
- Doreswamy Kenchegowda
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences , Bethesda, MD, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences , Bethesda, MD, USA
| | | | - 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 , Bethesda, MD, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences , Bethesda, MD, USA
| |
Collapse
|
80
|
Rozhdestvensky LM. Challenges in the Design of Russian Radiation Protection Means in the Crisis Period: The Search for Key Directions of Development. BIOL BULL+ 2021. [DOI: 10.1134/s1062359020120080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
81
|
Hu D, Zhang Y, Cao R, Hao Y, Yang X, Tian T, Zhang J. The protective effects of granulocyte-macrophage colony-stimulating factor against radiation-induced lung injury. Transl Lung Cancer Res 2021; 9:2440-2459. [PMID: 33489805 PMCID: PMC7815363 DOI: 10.21037/tlcr-20-1272] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Radiation-induced lung injury (RILI) is a common complication of thoracic cancer radiation therapy. Currently, there is no effective treatment for RILI. RILI is associated with chronic inflammation, this injury is perpetuated by the stimulation of chemokines and proinflammatory cytokines. Recent studies have demonstrated that granulocyte-macrophage colony-stimulating factor (GM-CSF) plays a pivotal role in inflammation and fibrosis. This study aimed to investigate the protective effect of GM-CSF against the development of RILI in lung tissue. Method First, a single fraction of radiation at a dose of 16 Gy was targeted at the entire thorax of wild-type (WT) C57BL/6 mice and GM-CSF–/– mice to induce RILI. Second, we detected the radioprotective effects of GM-CSF by measuring the inflammatory biomarkers and fibrosis alteration on radiated lung tissues. Furthermore, we investigated the potential mechanism of GM-CSF protective effects in RILI. Results The GM-CSF–/– mice sustained more severe RILI than the WT mice. RILI was significantly alleviated by GM-CSF treatment. Intraperitoneally administered GM-CSF significantly inhibited inflammatory cytokine production and decreased epithelial-mesenchymal transition (EMT) in the RILI mouse model. Conclusions GM-CSF was shown to be an important modulator of RILI through regulating inflammatory cytokines, which provides a new strategy for the prevention and treatment of RILI.
Collapse
Affiliation(s)
- Dan Hu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China.,Department of Physiology, Jeonbuk National University Medical School, Jeonju, Korea
| | - Yan Zhang
- School of Medicine, Shandong University, Jinan, China
| | - Ruiqi Cao
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yuying Hao
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Xiaoye Yang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Tiantian Tian
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Jiandong Zhang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| |
Collapse
|
82
|
Singh VK, Seed TM. BIO 300: a promising radiation countermeasure under advanced development for acute radiation syndrome and the delayed effects of acute radiation exposure. Expert Opin Investig Drugs 2021; 29:429-441. [PMID: 32450051 DOI: 10.1080/13543784.2020.1757648] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION There are no radioprotectors currently approved by the United States Food and Drug Administration (US FDA) for either the hematopoietic acute radiation syndrome (H-ARS) or for the acute radiation gastrointestinal syndrome (GI-ARS). There are currently, however, three US FDA-approved medicinals that serve to mitigate acute irradiation-associated hematopoietic injury. AREA COVERED We present the current status of a promising radiation countermeasure, BIO 300 (a genistein-based agent), that has been extensively investigated in murine models of H-ARS and models of the delayed effects of acute radiation exposure (DEARE) and is currently being evaluated in large animal models. It is also being developed for the prevention of radiation-induced toxicities associated with solid tumor radiotherapy and is the subject of two active Investigational New Drug (IND) applications. We have included a listing and brief review of significant investigations of this promising medical countermeasure. EXPERT OPINION BIO 300 is a leading radioprotector under advanced development for H-ARS and DEARE, as well as for select oncologic indication(s). Efficacy following oral administration (po), lack of clinical side effects, storage at ambient temperature, and intended dual use makes BIO 300 an ideal candidate for military and civilian use as well as for storage in the Strategic National Stockpile.
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 , Bethesda, MD, USA.,Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences , Bethesda, MD, USA
| | | |
Collapse
|
83
|
Liu G, Zeng Y, Lv T, Mao T, Wei Y, Jia S, Gou Y, Tao L. High-throughput preparation of radioprotective polymers via Hantzsch's reaction for in vivo X-ray damage determination. Nat Commun 2020; 11:6214. [PMID: 33277480 PMCID: PMC7718248 DOI: 10.1038/s41467-020-20027-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 11/09/2020] [Indexed: 02/07/2023] Open
Abstract
Radioprotectors for acute injuries caused by large doses of ionizing radiation are vital to national security, public health and future development of humankind. Here, we develop a strategy to explore safe and efficient radioprotectors by combining Hantzsch's reaction, high-throughput methods and polymer chemistry. A water-soluble polymer with low-cytotoxicity and an excellent anti-radiation capability has been achieved. In in vivo experiments, this polymer is even better than amifostine, which is the only approved radioprotector for clinical applications, in effectively protecting zebrafish embryos from fatally large doses of ionizing radiation (80 Gy X-ray). A mechanistic study also reveals that the radioprotective ability of this polymer originates from its ability to efficiently prevent DNA damage due to high doses of radiation. This is an initial attempt to explore polymer radioprotectors via a multi-component reaction. It allows exploiting functional polymers and provides the underlying insights to guide the design of radioprotective polymers.
Collapse
Affiliation(s)
- Guoqiang Liu
- The Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Yuan Zeng
- The Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Tong Lv
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Tengfei Mao
- The Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, 100084, China
- Science and Technology on Advanced Ceramic Fibers and Composites Laboratory, National University of Defense Technology, Changsha, 410073, China
| | - Yen Wei
- The Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Shunji Jia
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yanzi Gou
- Science and Technology on Advanced Ceramic Fibers and Composites Laboratory, National University of Defense Technology, Changsha, 410073, China
| | - Lei Tao
- The Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
84
|
Filimonova MV, Makarchuk VM, Shevchenko LI, Saburova AS, Surinova VI, Izmestieva OS, Lychagin AA, Saburov VO, Shegay PV, Kaprin AD, Ivanov SA, Filimonov AS. Radioprotective Activity of the Nitric Oxide Synthase Inhibitor T1023. Toxicological and Biochemical Properties, Cardiovascular and Radioprotective Effects. Radiat Res 2020; 194:532-543. [PMID: 34609510 DOI: 10.1667/rade-20-00046.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 08/03/2020] [Indexed: 11/03/2022]
Abstract
In this work, studies were performed to investigate the toxicological, biochemical, vasotropic and radiomodifying properties of the new nitric oxide synthase (NOS) inhibitor, compound T1023. Toxicological studies included the estimation of acute toxicity in mice after i.p. administration of T1023. Radiometric analysis and electron paramagnetic resonance spectroscopy were used to study NOS-inhibitory properties of T1023 in vitro and in vivo, respectively. T1023 vasoactive properties were studied in rat central hemodynamics. Radiobiological experiments were performed using endogenous and exogenous spleen colony formation as well as 30-day survival tests. The morphological changes in peripheral blood and bone marrow (BM) induced with T1023 were analyzed in mice during hematopoietic acute radiation syndrome (H-ARS). It was shown that T1023 is a sufficiently safe compound (LD10 of 317 mg/kg; LD50 of 410 mg/kg). It is an effective competitive NOS-inhibitor that is 10-to-15-fold selective to endothelial and inducible NOS (IC50 for nNOS, iNOS, eNOS: 52.3, 3.2 and 5.1 µM, respectively). Its NOS-inhibitory activity is realized in vivo and is accompanied by an increase in vascular tone. Its single i.p. administration in doses greater than 1/8 LD10 provides significant (40-50%) and long-lasting (more than 90 min) weakening of cardiac output, which can cause transient hypoxia. In radiobiological studies, T1023 proved to be a hypoxic radioprotector. Its radioprotective effect was observed only when administered prophylactically [single i.p dose, 5-120 min before total-body irradiation (TBI)] and only in doses that reduced cardiac output (1/8 LD10 and more, 40 mg/kg for mice), and was correlated in time with the dynamics of circulatory depression. Its radioprotective effect was not observed when administered in vitro and in the first 4 h after TBI. The optimal radioprotective doses of T1023 are relatively safe (1/ 5-1/4 LD10). In addition, T1023 effectively prevents H-ARS and gastrointestinal acute radiation syndrome (G-ARS) in experimental animals in vivo: dose modifying factor of 1.6-1.9. In the H-ARS mouse model, the prophylactic effect of T1023 (75 mg/kg, single i.p. injection) was accompanied by clinically significant effects. There was an express decrease in the degree of indicators of early BM devastation (by 40%) and maximal neutropenia and thrombocytopenia (2-2.5 times), in addition to a reduction in recovery time (by 30-40%). The obtained experimental results and literature data indicate that NOS inhibitors are an independent class of vasoactive radioprotectors with a specific hypoxic mechanism of action. NOS inhibitors provide new opportunities for developing effective and safe tools for the prevention of ARS.
Collapse
Affiliation(s)
- Marina V Filimonova
- A. Tsyb Medical Radiological Research Center - Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Victoria M Makarchuk
- A. Tsyb Medical Radiological Research Center - Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Ljudmila I Shevchenko
- A. Tsyb Medical Radiological Research Center - Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Alina S Saburova
- A. Tsyb Medical Radiological Research Center - Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Valentina I Surinova
- A. Tsyb Medical Radiological Research Center - Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Olga S Izmestieva
- A. Tsyb Medical Radiological Research Center - Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Anatoly A Lychagin
- A. Tsyb Medical Radiological Research Center - Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Vyacheslav O Saburov
- A. Tsyb Medical Radiological Research Center - Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Petr V Shegay
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Andrey D Kaprin
- National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Sergey A Ivanov
- A. Tsyb Medical Radiological Research Center - Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| | - Alexander S Filimonov
- A. Tsyb Medical Radiological Research Center - Branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russia
| |
Collapse
|
85
|
Comparative proteomic analysis of serum from nonhuman primates administered BIO 300: a promising radiation countermeasure. Sci Rep 2020; 10:19343. [PMID: 33168863 PMCID: PMC7653926 DOI: 10.1038/s41598-020-76494-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 10/28/2020] [Indexed: 11/12/2022] Open
Abstract
Hematopoietic acute radiation syndrome (H-ARS) and delayed effects of acute radiation exposure (DEARE) are detrimental health effects that occur after exposure to high doses of ionizing radiation. BIO 300, a synthetic genistein nanosuspension, was previously proven safe and effective against H-ARS when administered (via the oral (po) or intramuscular (im) route) prior to exposure to lethal doses of total-body radiation. In this study, we evaluated the proteomic changes in serum of nonhuman primates (NHP) after administering BIO 300 by different routes (po and im). We utilized nanoflow-ultra-performance liquid chromatography quadrupole time-of-flight mass spectrometry (NanoUPLC-MS/MS) methods for comprehensive global profiling and quantification of serum proteins. The results corroborate previous findings that suggest a very similar metabolic profile following both routes of drug administration. Furthermore, we observed minor alterations in protein levels, 2 hours after drug administration, which relates to the Cmax of BIO 300 for both routes of administration. Taken together, this assessment may provide an insight into the mechanism of radioprotection of BIO 300 and a reasonable illustration of the pharmacodynamics of this radiation countermeasure.
Collapse
|
86
|
Royba E, Repin M, Balajee AS, Shuryak I, Pampou S, Karan C, Brenner DJ, Garty G. The RABiT-II DCA in the Rhesus Macaque Model. Radiat Res 2020; 196:501-509. [PMID: 33022052 PMCID: PMC9039759 DOI: 10.1667/rr15547.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 05/08/2020] [Indexed: 11/03/2022]
Abstract
An automated platform for cytogenetic biodosimetry, the "Rapid Automated Biodosimetry Tool II (RABiT-II)," adapts the dicentric chromosome assay (DCA) for high-throughput mass-screening of the population after a large-scale radiological event. To validate this test, the U.S. Federal Drug Administration (FDA) recommends demonstrating that the high-throughput biodosimetric assay in question correctly reports the dose in an in vivo model. Here we describe the use of rhesus macaques (Macaca mulatta) to augment human studies and validate the accuracy of the high-throughput version of the DCA. To perform analysis, we developed the 17/22-mer peptide nucleic acid (PNA) probes that bind to the rhesus macaque's centromeres. To our knowledge, these are the first custom PNA probes with high specificity that can be used for chromosome analysis in M. mulatta. The accuracy of fully-automated chromosome analysis was improved by optimizing a low-temperature telomere PNA FISH staining in multiwell plates and adding the telomere detection feature to our custom chromosome detection software, FluorQuantDic V4. The dicentric frequencies estimated from in vitro irradiated rhesus macaque samples were compared to human blood samples of individuals subjected to the same ex vivo irradiation conditions. The results of the RABiT-II DCA analysis suggest that, in the lymphocyte system, the dose responses to gamma radiation in the rhesus macaques were similar to those in humans, with small but statistically significant differences between these two model systems.
Collapse
Affiliation(s)
- Ekaterina Royba
- Center for Radiological Research, Columbia University Irving Medical Center, New York, New York 10032
| | - Mikhail Repin
- Center for Radiological Research, Columbia University Irving Medical Center, New York, New York 10032
| | - Adayabalam S. Balajee
- Radiation Emergency Assistance Center/Training Site (REAC/TS), Cytogenetic Biodosimetry Laboratory (CBL), Oak Ridge Institute for Science and Education, Oak Ridge Associated Universities, Oak Ridge, Tennessee 37830
| | - Igor Shuryak
- Center for Radiological Research, Columbia University Irving Medical Center, New York, New York 10032
| | - Sergey Pampou
- JP Sulzberger Columbia Genome Center, High-Throughput Screening Center, New York, New York 10032
| | - Charles Karan
- JP Sulzberger Columbia Genome Center, High-Throughput Screening Center, New York, New York 10032
| | - David J. Brenner
- Center for Radiological Research, Columbia University Irving Medical Center, New York, New York 10032
| | - Guy Garty
- Center for Radiological Research, Columbia University Irving Medical Center, New York, New York 10032
| |
Collapse
|
87
|
Obrador E, Salvador R, Villaescusa JI, Soriano JM, Estrela JM, Montoro A. Radioprotection and Radiomitigation: From the Bench to Clinical Practice. Biomedicines 2020; 8:E461. [PMID: 33142986 PMCID: PMC7692399 DOI: 10.3390/biomedicines8110461] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/27/2020] [Accepted: 10/29/2020] [Indexed: 02/07/2023] Open
Abstract
The development of protective agents against harmful radiations has been a subject of investigation for decades. However, effective (ideal) radioprotectors and radiomitigators remain an unsolved problem. Because ionizing radiation-induced cellular damage is primarily attributed to free radicals, radical scavengers are promising as potential radioprotectors. Early development of such agents focused on thiol synthetic compounds, e.g., amifostine (2-(3-aminopropylamino) ethylsulfanylphosphonic acid), approved as a radioprotector by the Food and Drug Administration (FDA, USA) but for limited clinical indications and not for nonclinical uses. To date, no new chemical entity has been approved by the FDA as a radiation countermeasure for acute radiation syndrome (ARS). All FDA-approved radiation countermeasures (filgrastim, a recombinant DNA form of the naturally occurring granulocyte colony-stimulating factor, G-CSF; pegfilgrastim, a PEGylated form of the recombinant human G-CSF; sargramostim, a recombinant granulocyte macrophage colony-stimulating factor, GM-CSF) are classified as radiomitigators. No radioprotector that can be administered prior to exposure has been approved for ARS. This differentiates radioprotectors (reduce direct damage caused by radiation) and radiomitigators (minimize toxicity even after radiation has been delivered). Molecules under development with the aim of reaching clinical practice and other nonclinical applications are discussed. Assays to evaluate the biological effects of ionizing radiations are also analyzed.
Collapse
Affiliation(s)
- Elena Obrador
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (E.O.); (R.S.); (J.M.E.)
| | - Rosario Salvador
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (E.O.); (R.S.); (J.M.E.)
| | - Juan I. Villaescusa
- Service of Radiological Protection, Clinical Area of Medical Image, La Fe University Hospital, 46026 Valencia, Spain;
- Biomedical Imaging Research Group GIBI230, Health Research Institute (IISLaFe), La Fe University Hospital, 46026 Valencia, Spain
| | - José M. Soriano
- Food & Health Lab, Institute of Materials Science, University of Valencia, 46980 Valencia, Spain;
- Joint Research Unit in Endocrinology, Nutrition and Clinical Dietetics, University of Valencia-Health Research Institute IISLaFe, 46026 Valencia, Spain
| | - José M. Estrela
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (E.O.); (R.S.); (J.M.E.)
| | - Alegría Montoro
- Service of Radiological Protection, Clinical Area of Medical Image, La Fe University Hospital, 46026 Valencia, Spain;
- Biomedical Imaging Research Group GIBI230, Health Research Institute (IISLaFe), La Fe University Hospital, 46026 Valencia, Spain
| |
Collapse
|
88
|
Singh VK, Seed TM. Entolimod as a radiation countermeasure for acute radiation syndrome. Drug Discov Today 2020; 26:17-30. [PMID: 33065293 DOI: 10.1016/j.drudis.2020.10.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/12/2020] [Accepted: 10/05/2020] [Indexed: 01/11/2023]
Abstract
High doses of total-body or partial-body radiation exposure can result in a life-threatening acute radiation syndrome as manifested by severe morbidity. Entolimod (CBLB502) is effective in protecting against, and mitigating the development of, the hematopoietic and gastrointestinal subsyndromes of the acute radiation syndrome in rodents and nonhuman primates. Entolimod treatment reduces radiation-induced apoptosis and accelerates the regeneration of progenitors in radiation-damaged tissues. The drug has been evaluated clinically for its pharmacokinetics (PK), toxicity, and biomarkers. The US Food and Drug Administration (FDA) has granted investigational new drug, fast-track, and orphan drug statuses to entolimod. Its safety, efficacy, and animal-to-human dose conversion data allowed its progression with a pre-emergency use authorization application submission.
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, Bethesda, MD 20814, USA; Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA.
| | - Thomas M Seed
- Tech Micro Services, 4417 Maple Avenue, Bethesda, MD 20814, USA
| |
Collapse
|
89
|
Zhong Y, Pouliot M, Downey AM, Mockbee C, Roychowdhury D, Wierzbicki W, Authier S. Efficacy of delayed administration of sargramostim up to 120 hours post exposure in a nonhuman primate total body radiation model. Int J Radiat Biol 2020; 97:S100-S116. [PMID: 32960660 DOI: 10.1080/09553002.2019.1673499] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 09/09/2019] [Accepted: 09/18/2019] [Indexed: 01/07/2023]
Abstract
BACKGROUND High dose ionizing radiation exposure is associated with myelo-depression leading to pancytopenia and the expected clinical manifestations of acute radiation syndrome (ARS). Herein, we evaluated the efficacy of sargramostim (Leukine®, yeast-derived rhu GM-CSF), with regimens delivered at 48, 72, 96, or 120 h after radiation exposure. METHODS A randomized and blinded nonhuman primate (NHP) study was conducted to assess the effects of sargramostim treatment on ARS. NHPs were exposed to total body radiation (LD83/60 or lethal dose 83% by Day 60) and were randomized to groups receiving daily subcutaneous dosing of sargramostim starting from either 48, 72, 96, or 120 h post-irradiation. Additionally, separate groups receiving sargramostim treatment at 48 h post-irradiation also received prophylactic treatment with azithromycin. Sargramostim treatment of each animal continued until the preliminary absolute neutrophil count (ANC) returned to ≥1000/μL post-nadir for three consecutive days or the preliminary ANC exceeded 10,000/μL, which amounted to be an average of 15.95 days for all treatment groups. Prophylactic administration of enrofloxacin was included in the supportive care given to all animals in all groups. All animals were monitored for 60 days post-irradiation for mortality, hematological parameters, and sepsis. RESULTS Delayed sargramostim treatment at 48 h post-irradiation significantly reduced mortality (p = .0032) and improved hematological parameters including neutrophil but also lymphocyte and platelet counts. Additional delays in sargramostim administration at 72, 96, and 120 h post-irradiation were also similarly effective at enhancing the recovery of lymphocyte, neutrophil, and platelet counts compared to control. Sargramostim treatment also improved the survival of the animals when administered at up to 96 h post-irradiation. While sargramostim treatment at 48 h significantly reduced mortality associated with sepsis (p ≤ .01), the additional prophylactic treatment with azithromycin did not have clinically significant effects. CONCLUSION In a NHP ARS model, sargramostim administered starting at 48 h post-radiation was effective to improve survival, while beneficial hematological effects were observed with sargramostim initiated up to 120 h post exposure.
Collapse
|
90
|
COVID-19: Proposing a Ketone-Based Metabolic Therapy as a Treatment to Blunt the Cytokine Storm. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6401341. [PMID: 33014275 PMCID: PMC7519203 DOI: 10.1155/2020/6401341] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 06/22/2020] [Accepted: 08/17/2020] [Indexed: 02/07/2023]
Abstract
Human SARS-CoV-2 infection is characterized by a high mortality rate due to some patients developing a large innate immune response associated with a cytokine storm and acute respiratory distress syndrome (ARDS). This is characterized at the molecular level by decreased energy metabolism, altered redox state, oxidative damage, and cell death. Therapies that increase levels of (R)-beta-hydroxybutyrate (R-BHB), such as the ketogenic diet or consuming exogenous ketones, should restore altered energy metabolism and redox state. R-BHB activates anti-inflammatory GPR109A signaling and inhibits the NLRP3 inflammasome and histone deacetylases, while a ketogenic diet has been shown to protect mice from influenza virus infection through a protective γδ T cell response and by increasing electron transport chain gene expression to restore energy metabolism. During a virus-induced cytokine storm, metabolic flexibility is compromised due to increased levels of reactive oxygen species (ROS) and reactive nitrogen species (RNS) that damage, downregulate, or inactivate many enzymes of central metabolism including the pyruvate dehydrogenase complex (PDC). This leads to an energy and redox crisis that decreases B and T cell proliferation and results in increased cytokine production and cell death. It is hypothesized that a moderately high-fat diet together with exogenous ketone supplementation at the first signs of respiratory distress will increase mitochondrial metabolism by bypassing the block at PDC. R-BHB-mediated restoration of nucleotide coenzyme ratios and redox state should decrease ROS and RNS to blunt the innate immune response and the associated cytokine storm, allowing the proliferation of cells responsible for adaptive immunity. Limitations of the proposed therapy include the following: it is unknown if human immune and lung cell functions are enhanced by ketosis, the risk of ketoacidosis must be assessed prior to initiating treatment, and permissive dietary fat and carbohydrate levels for exogenous ketones to boost immune function are not yet established. The third limitation could be addressed by studies with influenza-infected mice. A clinical study is warranted where COVID-19 patients consume a permissive diet combined with ketone ester to raise blood ketone levels to 1 to 2 mM with measured outcomes of symptom severity, length of infection, and case fatality rate.
Collapse
|
91
|
Verma S, Gupta ML, Kumar K. A combined prophylactic modality of podophyllotoxin and rutin alleviates radiation induced injuries to the lymphohematopoietic system of mice by modulating cytokines, cell cycle progression, and apoptosis. Free Radic Res 2020; 54:497-516. [PMID: 32746646 DOI: 10.1080/10715762.2020.1805447] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The present study was conceptualized to delineate radioprotective efficacy of a formulation G-003M (a combination of podophyllotoxin and rutin) against radiation-induced damage to the lymphohematopoietic system of mice. C57BL/6J mice, treated with G-003M 1 h prior to 9 Gy lethal dose, were assessed for reactive oxygen species (ROS)/nitric oxide (NO) generation, antioxidant alterations, Annexin V/PI and TUNEL staining for apoptosis, modulation of apoptotic proteins, cell proliferation, histological alterations in thymus and cell cycle arrest in bone marrow cells. Induction of granulocyte colony-stimulating factor (G-CSF), granulocytes macrophage colony-stimulating factor (GM-CSF), interleukin-IL-6, IL-10, IL-1α, and IL-1β in response to G-003M was also evaluated in different groups of mice. Haematopoietic reconstitution with G-003M was explored by examining endogenous spleen colony-forming units (CFU-S) in irradiated animals. G-003M significantly inhibited ROS/NO, malondialdehyde (MDA) and restored cellular antioxidant glutathione in the thymus of irradiated animals. G-003M pre-treatment significantly (p < 0.001) restrained apoptosis in thymocytes via upregulation of Bcl2 and down-regulation of Bax, p53 and caspase-3. Stimulation of cell proliferation and inhibition of apoptosis by G-003M, restored architecture of thymus in irradiated animals within 30 days as evaluated by histological analysis. G-003M arrested cells at the G2/M phase by inducing reversible cell cycle arrest. Peak expression of G-CSF (45-fold) and IL-6 (60-fold) as well as moderate induction of GM-CSF, IL-10, IL-1α by G-003M helped in haematopoietic recovery of irradiated mice. A higher number of endogenous CFU-S in G-003M pre-treated irradiated mice suggested haematopoietic recovery. Data obtained from the current study affirms that G-003M can be proved as a potential radioprotective agent against radiation damage.
Collapse
Affiliation(s)
- Savita Verma
- Division of Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, Delhi, India
| | - Manju Lata Gupta
- Division of Radioprotective Drug Development Research, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, Delhi, India
| | - Kamal Kumar
- Division of Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, Delhi, India
| |
Collapse
|
92
|
Mitigative efficacy of the clinical dosage administration of granulocyte colony-stimulating factor and romiplostim in mice with severe acute radiation syndrome. Stem Cell Res Ther 2020; 11:339. [PMID: 32746943 PMCID: PMC7398212 DOI: 10.1186/s13287-020-01861-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/12/2020] [Accepted: 07/28/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND It has been reported that the high-dosage administration of domestically approved pharmaceutical drugs, especially granulocyte colony-stimulating factor (G-CSF) and romiplostim (RP), is a rapid and appropriate medical treatment for preventing severe acute radiation syndrome (ARS) of victims exposed to lethal total-body irradiation (TBI). However, it remains unclear whether or not the clinical dosage administration of these drugs can ameliorate TBI-induced ARS and related high mortality in order to find various drug treatment options and less toxic optimum protocol depending on the situation surrounding the radiological accidents. METHODS We assessed the clinical dosage administration in combination with G-CSF and RP as intraperitoneal injection in C57BL/6 J mice exposed to more than 7-Gy lethal dose of X-ray TBI for the survival study evaluated by the log-rank test. Bone marrow and splenic cells were collected on the 21st day, when 1 week has passed from last administration, to detect the level of cell apoptosis, intracellular reactive oxygen species (ROS), and nuclear factor erythroid 2-related factor 2 (Nrf2)-related anti-oxidative gene expressions, and enzyme-linked immune sorbent assay using sera was performed for cell senescence and inflammation status analyzed with one-way ANOVA and Tukey-Kramer or Bonferroni/Dunn multiple comparison tests. RESULTS The combined once-daily administration of 10 μg/kg G-CSF for 4 times and 10 μg/kg RP once a week for 3 times improve the 30-day survival rate of lethal TBI mice compared with untreated TBI mice, accompanied by a gradual increase in the body weight and hematopoietic cell numbers. The radio-mitigative effect is probably attributed to the scavenging of ROS and the reduction in cell apoptosis. These changes were associated with the upregulation of Nrf2 and its downstream anti-oxidative targets in TBI mice. Furthermore, this combination modulated TBI-induced cell senescence an d inflammation markers. CONCLUSIONS This study suggested that the clinical dosage administration in combination with G-CSF and RP may also have radio-mitigative effects on mice exposed to lethal TBI and may be a potent therapeutic agent for mitigating radiation-induced severe ARS.
Collapse
|
93
|
Xing S, Shen X, Yang JK, Wang XR, Ou HL, Zhang XW, Xiong GL, Shan YJ, Cong YW, Luo QL, Yu ZY. Single-Dose Administration of Recombinant Human Thrombopoietin Mitigates Total Body Irradiation-Induced Hematopoietic System Injury in Mice and Nonhuman Primates. Int J Radiat Oncol Biol Phys 2020; 108:1357-1367. [PMID: 32758640 DOI: 10.1016/j.ijrobp.2020.07.2325] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 07/15/2020] [Accepted: 07/29/2020] [Indexed: 02/03/2023]
Abstract
PURPOSE Recombinant human thrombopoietin (rhTPO) has been evaluated as a therapeutic intervention for radiation-induced myelosuppression. However, the immunogenicity induced by a repeated-dosing strategy raises concerns about the therapeutic use of rhTPO. In this study, single-dose administration of rhTPO was evaluated for efficacy in the hematopoietic response and survival effect on mice and nonhuman primates exposed to total body irradiation (TBI). METHODS AND MATERIALS Survival of lethally (9.0 Gy) irradiated C57BL/6J male mice was observed for 30 days after irradiation. Hematologic evaluations were performed on C57BL/6J male mice given a sublethal dose of radiation (6.5 Gy). Furthermore, in sublethally irradiated mice, we performed bone marrow (BM) histologic evaluation and evaluated BM-derived clonogenic activity. Next, the proportion and number of hematopoietic stem cells (HSCs) were analyzed. Competitive repopulation experiments were conducted to assess the multilineage engraftment of irradiated HSCs after BM transplantation. Flow cytometry was used to evaluate DNA damage, cell apoptosis, and cell cycle stage in HSCs after irradiation. Finally, we evaluated the efficacy of a single dose of rhTPO administered after 7 Gy TBI in male and female rhesus monkeys. RESULTS A single administration of rhTPO 2 hours after irradiation significantly mitigated TBI-induced death in mice. rhTPO promoted multilineage hematopoietic recovery, increasing peripheral blood cell counts, BM cellularity, and BM colony-forming ability. rhTPO administration led to an accelerated recovery of BM HSC frequency and multilineage engraftment after transplantation. rhTPO treatment reduced radiation-induced DNA damage and apoptosis and promoted HSC proliferation after TBI. Notably, a single administration of rhTPO significantly promoted multilineage hematopoietic recovery and improved survival in nonhuman primates after TBI. CONCLUSIONS These findings indicate that early intervention with a single administration of rhTPO may represent a promising and effective radiomitigative strategy for victims of radiation disasters.
Collapse
Affiliation(s)
- Shuang Xing
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Xing Shen
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Jin-Kun Yang
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Xin-Ru Wang
- Department of Clinical Laboratory, PLA Rocket Characteristic Medical Center, Beijing, China
| | - Hong-Ling Ou
- Department of Clinical Laboratory, PLA Rocket Characteristic Medical Center, Beijing, China
| | - Xue-Wen Zhang
- Guangdong Pharmaceutical University, Guangzhou, China
| | - Guo-Lin Xiong
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Ya-Jun Shan
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Yu-Wen Cong
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Qing-Liang Luo
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Zu-Yin Yu
- Department of Experimental Hematology and Biochemistry, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China; Guangdong Pharmaceutical University, Guangzhou, China; School of Life Science, Anhui Medical University, Hefei, China.
| |
Collapse
|
94
|
Bandekar M, Maurya DK, Sharma D, Checker R, Gota V, Mishra N, Sandur SK. Xenogeneic transplantation of human WJ-MSCs rescues mice from acute radiation syndrome via Nrf-2-dependent regeneration of damaged tissues. Am J Transplant 2020; 20:2044-2057. [PMID: 32040239 DOI: 10.1111/ajt.15819] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 01/31/2020] [Accepted: 02/02/2020] [Indexed: 01/25/2023]
Abstract
There is an unmet medical need for radiation countermeasures that can be deployed for treatment of exposed individuals during ionizing radiation (IR) accidents or terrorism. Wharton's jelly mesenchymal stem cells (WJ-MSCs) from human umbilical cord have been shown to avoid allorecognition and induce a tissue-regenerating microenvironment, which makes them an attractive candidate for mitigating IR injury. We found that WJ-MSCs protected mice from a lethal dose of IR even when transplanted up to 24 hours after irradiation, and a combination of WJ-MSCs and antibiotic (tetracycline) could further expand the window of protection offered by WJ-MSCs. This combinatorial approach mitigated IR-induced damage to the hematopoietic and gastrointestinal system. WJ-MSCs increased the serum concentration of the cytoprotective cytokines granulocyte colony-stimulating factor (G-CSF) and IL-6 in mice. Knockdown of G-CSF and IL-6 in WJ-MSCs before injection to lethally irradiated mice or transplantation of WJ-MSCs to lethally irradiated Nrf-2 knockout mice significantly nullified the therapeutic protective efficacy. Hence, WJ-MSCs could be a potential cell-based therapy for individuals accidentally exposed to radiation.
Collapse
Affiliation(s)
- Mayuri Bandekar
- Radiation Biology and Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Trombay, Mumbai, India.,University of Mumbai, Kalina, Mumbai, India
| | - Dharmendra K Maurya
- Radiation Biology and Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Trombay, Mumbai, India.,Homi Bhabha National Institute, Anushaktinagar, Mumbai, India
| | - Deepak Sharma
- Radiation Biology and Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Trombay, Mumbai, India.,Homi Bhabha National Institute, Anushaktinagar, Mumbai, India
| | - Rahul Checker
- Radiation Biology and Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Trombay, Mumbai, India.,Homi Bhabha National Institute, Anushaktinagar, Mumbai, India
| | - Vikram Gota
- Clinical Pharmacology, ACTREC, Tata Memorial Centre, Kharghar, Navi Mumbai, India
| | | | - Santosh K Sandur
- Radiation Biology and Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Trombay, Mumbai, India.,Homi Bhabha National Institute, Anushaktinagar, Mumbai, India
| |
Collapse
|
95
|
Wang Q, Lee Y, Shuryak I, Pujol Canadell M, Taveras M, Perrier JR, Bacon BA, Rodrigues MA, Kowalski R, Capaccio C, Brenner DJ, Turner HC. Development of the FAST-DOSE assay system for high-throughput biodosimetry and radiation triage. Sci Rep 2020; 10:12716. [PMID: 32728041 PMCID: PMC7392759 DOI: 10.1038/s41598-020-69460-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 07/09/2020] [Indexed: 11/09/2022] Open
Abstract
Following a large-scale radiological incident, there is a need for FDA-approved biodosimetry devices and biomarkers with the ability to rapidly determine past radiation exposure with sufficient accuracy for early population triage and medical management. Towards this goal, we have developed FAST-DOSE (Fluorescent Automated Screening Tool for Dosimetry), an immunofluorescent, biomarker-based system designed to reconstruct absorbed radiation dose in peripheral blood samples collected from potentially exposed individuals. The objective of this study was to examine the performance of the FAST-DOSE assay system to quantify intracellular protein changes in blood leukocytes for early biodosimetry triage from humanized NOD-scid-gamma (Hu-NSG) mice and non-human primates (NHPs) exposed to ionizing radiation up to 8 days after radiation exposure. In the Hu-NSG mice studies, the FAST-DOSE biomarker panel was able to generate delivered dose estimates at days 1, 2 and 3 post exposure, whereas in the NHP studies, the biomarker panel was able to successfully classify samples by dose categories below or above 2 Gy up to 8 days after total body exposure. These results suggest that the FAST-DOSE bioassay has large potential as a useful diagnostic tool for rapid and reliable screening of potentially exposed individuals to aid early triage decisions within the first week post-exposure.
Collapse
Affiliation(s)
- Qi Wang
- Center for Radiological Research, Columbia University Irving Medical Center, New York, NY, 10032, USA.
- Radiation Oncology, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| | - Younghyun Lee
- Center for Radiological Research, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Igor Shuryak
- Center for Radiological Research, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Monica Pujol Canadell
- Center for Radiological Research, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Maria Taveras
- Center for Radiological Research, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Jay R Perrier
- Center for Radiological Research, Columbia University Irving Medical Center, New York, NY, 10032, USA
- ASELL, LLC, Owings Mills, MD, 21117, USA
| | - Bezalel A Bacon
- Center for Radiological Research, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | | | | | | | - David J Brenner
- Center for Radiological Research, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Helen C Turner
- Center for Radiological Research, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
96
|
Protective Effects of Biscoclaurine Alkaloids on Leukopenia Induced by 60Co- γ Radiation. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:2162915. [PMID: 32508944 PMCID: PMC7251465 DOI: 10.1155/2020/2162915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 04/16/2020] [Accepted: 05/09/2020] [Indexed: 01/15/2023]
Abstract
Objective Leukopenia, a common complication of tumor chemoradiotherapy, contributes serious damage to the hematopoietic, gastrointestinal, and immune systems of the body and can cause delay, discontinuation, or even failure to tumor treatment, thereby greatly threatening human health. The present study aims to investigate the protective effects of biscoclaurine alkaloids (BA) on leukopenia. Methods This study was conducted on 60 Kunming mice, which were randomly divided into six groups containing 10 animals each. A hematology analyzer was used to count white blood cells (WBC) in the peripheral blood cell. Mice serum was collected, and the granulocyte-macrophage colony-stimulating factor, vascular cell adhesion molecule 1 (VCAM-1), and interferon-γ (IFN-γ) were detected by enzyme-linked immunosorbent assays. Pathological changes were detected through hematoxylin and eosin staining in the liver and spleen of mice. The spleen and liver ultrastructures were observed via electron microscopy. Results Results showed that BA ameliorated WBC, PLT reduction in the peripheral blood and significantly increased the levels of IFN-γ and VCAM-1 in mice serum. BA reduced ionizing radiation-induced injuries to spleen, mitigated the reduction of superoxide dismutase (SOD), and significantly decreased the malonaldehyde (MDA) and xanthine oxidase (XOD) levels in the liver. Conclusion BA enhanced the immune and hematopoietic functions and ameliorated the oxidative stress induced by 60Co-γ radiation, revealing its therapeutic potential both as a radioprotector and as a radiation mitigator for leukopenia induced by 60Co-γ radiation.
Collapse
|
97
|
Peng R, Zhang W, Zuo Z, Shan Y, Liu X, Tang Y, Yu Z, Wang L, Cong Y. Dimethyl sulfoxide, a potent oral radioprotective agent, confers radioprotection of hematopoietic stem and progenitor cells independent of apoptosis. Free Radic Biol Med 2020; 153:1-11. [PMID: 32222468 DOI: 10.1016/j.freeradbiomed.2020.03.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 02/28/2020] [Accepted: 03/23/2020] [Indexed: 12/17/2022]
Abstract
In mass casualty events involving radiation exposure, there is a substantial unmet need for identifying and developing an orally bioavailable agent that can be used to protect the hematopoietic stem cell pool and regenerate hematopoiesis after radiation injury. Dimethyl sulfoxide (DMSO), a free-radical scavenger, has shown therapeutic benefits in many preclinical and clinical studies. This study investigates the radioprotective effects of DMSO on oral administration. Single dose of oral DMSO administrated before irradiation conferred 100% survival of C57BL6/J mice receiving otherwise lethal as well as super-lethal radiation dose, with wide radioprotective time frame (from 15min to 4h). Oral DMSO not only protected radiation-induced acute hematopoietic stem and progenitor cell (HSPC) injury, but also ameliorated long-term BM suppression following irradiation in mice. Mechanistically, DMSO directly protected HSPC survival after irradiation in vitro and in vivo, whereas no radioprotective effect was seen in MLL-AF9-induced leukemia cells. Unexpectedly, DMSO treatment did not inhibit radiation-induced HSPC apoptosis, and the HSPC survival from Trp53-and PUMA-deficient mice after irradiation was also protected by DMSO. In conclusion, our findings demonstrate the radioprotective efficacy of oral DMSO. Given its oral efficacy and little toxicity, DMSO is an attractive candidate for human use in a wide variety of settings, including nuclear accidents and medical radiation.
Collapse
Affiliation(s)
- Renjun Peng
- Department of Pathophysiology, Beijing Institute of Radiation Medicine, Beijing Key Laboratory for Radiobiology (BKLRB), Beijing, PR China
| | - Wenting Zhang
- Department of Pathophysiology, Beijing Institute of Radiation Medicine, Beijing Key Laboratory for Radiobiology (BKLRB), Beijing, PR China
| | - Zongchao Zuo
- Department of Pathophysiology, Beijing Institute of Radiation Medicine, Beijing Key Laboratory for Radiobiology (BKLRB), Beijing, PR China
| | - Yajun Shan
- Department of Pathophysiology, Beijing Institute of Radiation Medicine, Beijing Key Laboratory for Radiobiology (BKLRB), Beijing, PR China
| | - Xiaolan Liu
- Department of Pathophysiology, Beijing Institute of Radiation Medicine, Beijing Key Laboratory for Radiobiology (BKLRB), Beijing, PR China
| | - Yingying Tang
- Department of Pathophysiology, Beijing Institute of Radiation Medicine, Beijing Key Laboratory for Radiobiology (BKLRB), Beijing, PR China
| | - Zuyin Yu
- Department of Pathophysiology, Beijing Institute of Radiation Medicine, Beijing Key Laboratory for Radiobiology (BKLRB), Beijing, PR China
| | - Limei Wang
- Department of Pathophysiology, Beijing Institute of Radiation Medicine, Beijing Key Laboratory for Radiobiology (BKLRB), Beijing, PR China.
| | - Yuwen Cong
- Department of Pathophysiology, Beijing Institute of Radiation Medicine, Beijing Key Laboratory for Radiobiology (BKLRB), Beijing, PR China.
| |
Collapse
|
98
|
Alterations in Tissue Metabolite Profiles with Amifostine-Prophylaxed Mice Exposed to Gamma Radiation. Metabolites 2020; 10:metabo10050211. [PMID: 32455594 PMCID: PMC7281564 DOI: 10.3390/metabo10050211] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 12/02/2022] Open
Abstract
Acute exposure to high-dose ionizing irradiation has the potential to severely injure the hematopoietic system and its capacity to produce vital blood cells that innately serve to ward off infections and excessive bleeding. Developing a medical radiation countermeasure that can protect individuals from the damaging effects of irradiation remains a significant, unmet need and an area of great public health interest and concern. Despite significant advancements in the field of radiation countermeasure development to find a nontoxic and effective prophylactic agent for acute radiation syndrome, no such drug has yet been approved by the Food and Drug Administration. This study focuses on examining the metabolic corrections elicited by amifostine, a potent radioprotector, on tissues of vital body organs, such as the heart, spleen, and kidney. Our findings indicate that prophylaxis with this drug offers significant protection against potentially lethal radiation injury, in part, by correction of radiation-induced metabolic pathway perturbations.
Collapse
|
99
|
Use of molecularly-cloned haematopoietic growth factors in persons exposed to acute high-dose, high-dose rate whole-body ionizing radiations. Blood Rev 2020; 45:100690. [PMID: 32273121 DOI: 10.1016/j.blre.2020.100690] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/02/2020] [Accepted: 03/30/2020] [Indexed: 11/21/2022]
Abstract
Exposure to acute, high-dose, high dose-rate whole-body ionizing radiations damages the bone marrow resulting in rapid decreases in concentrations of blood cells, especially lymphocytes, granulocytes and platelets with associated risks of infection and bleeding. In several experimental models including non-human primate radiation exposure models giving molecularly cloned haematopoietic growth factor including granulocyte/macrophage colony-stimulating factor (G/M-CSF; sargramostim) and granulocyte colony-stimulating factor (G-CSF; filgrastim and pegylated G-CSF [peg-filgrastim]) accelerates bone marrow recovery and increases survival. Based on these data these molecules are US FDA approved for treating victims of radiation and nuclear incidents, accident and events such as nuclear terrorism and are included in the US National Strategic Stockpile. We discuss the immediate medical response to these events including how to estimate radiation dose and uniformity and which interventions are appropriate in different radiation exposures settings. We also discuss similarities and differences between molecularly cloned haematopoietic growth factors.
Collapse
|
100
|
Grebenyuk AN, Gladkikh VD. Modern Condition and Prospects for the Development of Medicines towards Prevention and Early Treatment of Radiation Damage. BIOL BULL+ 2020. [DOI: 10.1134/s1062359019110141] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|