1
|
Liu X, Song Y, Hu S, Bai Y, Zhang J, Tai G, Shao C, Pan Y. Serum amyloid A contributes to radiation-induced lung injury by activating macrophages through FPR2/Rac1/NF-κB pathway. Int J Biol Sci 2024; 20:4941-4956. [PMID: 39309438 PMCID: PMC11414394 DOI: 10.7150/ijbs.100823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 09/03/2024] [Indexed: 09/25/2024] Open
Abstract
Patients who receive thoracic radiotherapy may suffer from radiation-induced lung injury, but the treatment options are limited as the underlying mechanisms are unclear. Using a mouse model of right thorax irradiation with fractionated doses of X-rays for three consecutive days (8 Gy/per day), this study found that the thoracic irradiation (Th-IR) induced tissue injury with aberrant infiltration of macrophages, and it significantly increased the secretion of TNF-α, IL-1β, IL-6, TGF-β1 and serum amyloid A (SAA) in mice. Interestingly, SAA could activate macrophages and then induce epithelial-mesenchymal transition (EMT) of lung epithelial cells and fibrosis progression in lung tissue. Mechanistically, SAA enhanced the transient binding of FPR2 to Rac1 protein and further activated NF-κB signaling pathway in macrophages. Inhibition of FPR2 significantly reduced pulmonary fibrosis induced by SAA administration in mice. In addition, cimetidine could reduce the level of SAA release after irradiation and attenuate the lung injury induced by SAA or Th-IR. In conclusion, our results demonstrated that SAA activated macrophages via FPR2/Rac1/NF-κB pathway and might contribute to the Th-IR induced lung injury, which may provide a new strategy to attenuate radiation-induced adverse effects during radiotherapy.
Collapse
Affiliation(s)
- Xinglong Liu
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200032, China
| | - Yimeng Song
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Songling Hu
- Department of Preventive Dentistry, Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Shanghai Stomatological Hospital & School of Stomatology, Shanghai Medical College, Fudan University, Shanghai 200001, China
| | - Yang Bai
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jianghong Zhang
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Guomei Tai
- Department of Radiotherapy, Nantong Tumor Hospital and the Affiliated Tumor Hospital of Nantong University, Nantong 226631, Jiangsu Province, China
| | - Chunlin Shao
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yan Pan
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
2
|
Blakely WF, Port M, Ostheim P, Abend M. Radiation Research Society Journal-based Historical Review of the Use of Biomarkers for Radiation Dose and Injury Assessment: Acute Health Effects Predictions. Radiat Res 2024; 202:185-204. [PMID: 38936821 DOI: 10.1667/rade-24-00121.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 06/11/2024] [Indexed: 06/29/2024]
Abstract
A multiple-parameter based approach using radiation-induced clinical signs and symptoms, hematology changes, cytogenetic chromosomal aberrations, and molecular biomarkers changes after radiation exposure is used for biodosimetry-based dose assessment. In the current article, relevant milestones from Radiation Research are documented that forms the basis of the current consensus approach for diagnostics after radiation exposure. For example, in 1962 the use of cytogenetic chromosomal aberration using the lymphocyte metaphase spread dicentric assay for biodosimetry applications was first published in Radiation Research. This assay is now complimented using other cytogenetic chromosomal aberration assays (i.e., chromosomal translocations, cytokinesis-blocked micronuclei, premature chromosome condensation, γ-H2AX foci, etc.). Changes in blood cell counts represent an early-phase biomarker for radiation exposures. Molecular biomarker changes have evolved to include panels of organ-specific plasma proteomic and blood-based gene expression biomarkers for radiation dose assessment. Maturation of these assays are shown by efforts for automated processing and scoring, development of point-of-care diagnostics devices, service laboratories inter-comparison exercises, and applications for dose and injury assessments in radiation accidents. An alternative and complementary approach has been advocated with the focus to de-emphasize "dose" and instead focus on predicting acute or delayed health effects. The same biomarkers used for dose estimation (e.g., lymphocyte counts) can be used to directly predict the later developing severity degree of acute health effects without performing dose estimation as an additional or intermediate step. This review illustrates contributing steps toward these developments published in Radiation Research.
Collapse
Affiliation(s)
- William F Blakely
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Matthias Port
- Bundeswehr Institute of Radiobiology, Munich, Germany
| | | | - Michael Abend
- Bundeswehr Institute of Radiobiology, Munich, Germany
| |
Collapse
|
3
|
Yamaga S, Aziz M, Murao A, Brenner M, Wang P. DAMPs and radiation injury. Front Immunol 2024; 15:1353990. [PMID: 38333215 PMCID: PMC10850293 DOI: 10.3389/fimmu.2024.1353990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 01/15/2024] [Indexed: 02/10/2024] Open
Abstract
The heightened risk of ionizing radiation exposure, stemming from radiation accidents and potential acts of terrorism, has spurred growing interests in devising effective countermeasures against radiation injury. High-dose ionizing radiation exposure triggers acute radiation syndrome (ARS), manifesting as hematopoietic, gastrointestinal, and neurovascular ARS. Hematopoietic ARS typically presents with neutropenia and thrombocytopenia, while gastrointestinal ARS results in intestinal mucosal injury, often culminating in lethal sepsis and gastrointestinal bleeding. This deleterious impact can be attributed to radiation-induced DNA damage and oxidative stress, leading to various forms of cell death, such as apoptosis, necrosis and ferroptosis. Damage-associated molecular patterns (DAMPs) are intrinsic molecules released by cells undergoing injury or in the process of dying, either through passive or active pathways. These molecules then interact with pattern recognition receptors, triggering inflammatory responses. Such a cascade of events ultimately results in further tissue and organ damage, contributing to the elevated mortality rate. Notably, infection and sepsis often develop in ARS cases, further increasing the release of DAMPs. Given that lethal sepsis stands as a major contributor to the mortality in ARS, DAMPs hold the potential to function as mediators, exacerbating radiation-induced organ injury and consequently worsening overall survival. This review describes the intricate mechanisms underlying radiation-induced release of DAMPs. Furthermore, it discusses the detrimental effects of DAMPs on the immune system and explores potential DAMP-targeting therapeutic strategies to alleviate radiation-induced injury.
Collapse
Affiliation(s)
- Satoshi Yamaga
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Departments of Surgery and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| | - Atsushi Murao
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Max Brenner
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Departments of Surgery and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Departments of Surgery and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| |
Collapse
|
4
|
Bai H, Wang J, Wang Q, Chen Y, Miao G, Zhang T, Hua J, Zhang Y, He J, Ding N, Zhou H, Sui L, Wei W. Identification of the Kupffer cell-derived circulating IGFBP-3 as a universal radiation biomarker for heavy ion, proton, and X-ray exposure. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 265:115526. [PMID: 37769581 DOI: 10.1016/j.ecoenv.2023.115526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 09/11/2023] [Accepted: 09/24/2023] [Indexed: 10/03/2023]
Abstract
The minimally invasive biomarkers that can facilitate a rapid dose assessment are valuable for the early medical treatment when accidental or occupational radiation exposure happens. Our previous proteomic research identified one kind of circulating protein, Insulin-like Growth Factor Binding Protein 3 (IGFBP-3), which showed a significant increase after total body exposure of mice to carbon ions and X-rays. However, several critical issues such as the responses to diverse radiation, the origin and underlying mechanism in radiation response obstruct the utilization of circulating IGFBP-3 as a reliable radiation biomarker. In this study, mice were subjected to total or partial body irradiation with carbon ions, protons or X-rays, or treated with chloroform as a comparison. The level of IGFBP-3 in serum and different organs were measured via Enzyme Linked Immunosorbent Assay (ELISA), Western blot (WB) and Immunohistochemistry (IHC). A significant increase of IGFBP-3 was discovered in serum and liver tissue post-irradiation with three kinds of radiation, but absent when challenged with chloroform. Likewise, a similar response was also observed in blood samples from patients receiving radiotherapy. Moreover, the effect of radiation on three main hepatic cells was investigated, the findings indicated that IGFBP-3 could be detected in the culture medium of Kupffer cells (MKC) alone and was elevated in cells and cultured medium of MKC post-irradiation. Additionally, we observed a co-expression effect between P53 and IGFBP-3 in liver tissues and MKC post-irradiation. Along with down-regulation of Trp53 by siRNA, the response of IGFBP-3 to radiation was attenuated. The present study demonstrated that circulating IGFBP-3 could be a promising universal biomarker for complex environmental radiation exposure, and the upregulation of IGFBP-3 is attributed to the MKC in a P53-dependent manner. Circulating IGFBP-3 assays would offer rapid, convenient and effective dose and toxicity assessment methods in occupational exposure or radiation disaster management.
Collapse
Affiliation(s)
- Hao Bai
- Key Laboratory of Space Radiobiology of Gansu Province & CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jufang Wang
- Key Laboratory of Space Radiobiology of Gansu Province & CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiaojuan Wang
- Department of Nuclear Physics, China Institute of Atomic Energy, Beijing 102413, China; National Innovation Center of Radiation Application, Beijing 102413, China
| | - Yaxiong Chen
- Key Laboratory of Space Radiobiology of Gansu Province & CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guoying Miao
- Department of Radiotherapy, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China
| | - Tongshan Zhang
- Key Laboratory of Space Radiobiology of Gansu Province & CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Junrui Hua
- Key Laboratory of Space Radiobiology of Gansu Province & CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Yanan Zhang
- Key Laboratory of Space Radiobiology of Gansu Province & CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Jinpeng He
- Key Laboratory of Space Radiobiology of Gansu Province & CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Nan Ding
- Key Laboratory of Space Radiobiology of Gansu Province & CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Heng Zhou
- Key Laboratory of Space Radiobiology of Gansu Province & CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Li Sui
- Department of Nuclear Physics, China Institute of Atomic Energy, Beijing 102413, China; National Innovation Center of Radiation Application, Beijing 102413, China.
| | - Wenjun Wei
- Key Laboratory of Space Radiobiology of Gansu Province & CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
5
|
Mukherjee P, Kumar K, Babu B, Purkayastha J, Chandna S. Alterations in the expression pattern of RBC membrane associated proteins (RMAPs) in whole body γ-irradiated Sprague Dawley rats. Int J Radiat Biol 2023; 99:1724-1737. [PMID: 37315317 DOI: 10.1080/09553002.2023.2219726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 05/24/2023] [Accepted: 05/24/2023] [Indexed: 06/16/2023]
Abstract
PURPOSE Peripheral blood serum/plasma proteins are frequently studied for their potential use as radiation exposure biomarkers. Here we report RBC membrane associated proteins (RMAPs), which show alterations in expression level following whole-body γ-irradiation of rats at sub-lethal/lethal doses. MATERIALS AND METHODS RBCs from peripheral blood of Sprague Dawley rats were segregated using the Ficoll-Hypaque method, and membrane fractions were hypotonically isolated at various time points (6 h, 24 h, 48 h) after γ-irradiation at 2 Gy, 5 Gy, and 7.5 Gy doses. Following purification of proteins from these fractions, two-dimensional electrophoresis (2-DE) was carried out. Treatment induced differentially expressed protein spots (≥2 fold increase/decrease) were picked up, trypsinized, and identified using LC-MS/MS analysis. Western immunoblots using protein specific antibodies were used to confirm the results. Gene ontology and interactions of these proteins were also studied. RESULTS From a number of differentially expressed radiation-responsive 2-DE protein spots detected, eight were identified unequivocally using LC-MS/MS. Out of these, actin, cytoplasmic 1 (ACTB) showed detectable yet insignificant variation (<50%) in expression. In contrast, peroxiredoxin-2 (PRDX2) and 26S proteasome regulatory subunit RPN11 (PSMD14) were the two most prominently over-expressed proteins. Five more proteins, namely tropomyosin alpha-3 chain (TPM3), exosome component 6 (EXOSC6), isoform 4 of tropomyosin alpha-1 chain (TPM1), serum albumin (ALB), and the 55 kDa erythrocyte membrane protein (P55) showed distinct alteration in their expression at different time-points and doses. ALB, EXOSC6, and PSMD14 were the most responsive at 2 Gy, albeit at different time-points. While EXOSC6 and PSMD14 showed maximum over-expression (5-12 fold) at 6 h post-irradiation, ALB expression increased progressively (4 up to 7 fold) from 6 h to 48 h. TPM1 showed over-expression (2-3 fold) at all doses and time-points tested. TPM3 showed a dose-dependent response at all time-points studied; with no variation at 2 Gy, ∼2 fold increase at 5 Gy, and 3-6 fold at the highest dose used (7.5 Gy). The p55 protein was over-expressed (∼2.5 fold) only transiently at 24 h following the lethal (7.5 Gy) dose. CONCLUSION This is the first study to report γ-radiation induced alterations in the RBC membrane associated proteins. We are further evaluating the potential of these proteins as radiation biomarkers. Due to the abundance and easy use of RBCs, this approach can prove very useful for detecting ionizing radiation exposure.
Collapse
Affiliation(s)
- Prabuddho Mukherjee
- Division of Molecular & Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences, DRDO, Brig. S K Majumdar Marg, Timarpur, Delhi, India
| | - Kamendra Kumar
- Division of Molecular & Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences, DRDO, Brig. S K Majumdar Marg, Timarpur, Delhi, India
| | - Bincy Babu
- Division of Molecular & Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences, DRDO, Brig. S K Majumdar Marg, Timarpur, Delhi, India
| | - Jubilee Purkayastha
- Division of Molecular & Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences, DRDO, Brig. S K Majumdar Marg, Timarpur, Delhi, India
| | - Sudhir Chandna
- Division of Molecular & Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences, DRDO, Brig. S K Majumdar Marg, Timarpur, Delhi, India
| |
Collapse
|
6
|
Sproull M, Kawai T, Krauze A, Shankavaram U, Camphausen K. Prediction of Total-Body and Partial-Body Exposures to Radiation Using Plasma Proteomic Expression Profiles. Radiat Res 2022; 198:573-581. [PMID: 36136739 PMCID: PMC9896586 DOI: 10.1667/rade-22-00074.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 08/18/2022] [Indexed: 02/05/2023]
Abstract
There is a need to identify new biomarkers of radiation exposure for not only systemic total-body irradiation (TBI) but also to characterize partial-body irradiation and organ specific radiation injury. In the current study, we sought to develop novel biodosimetry models of radiation exposure using TBI and organ specific partial-body irradiation to only the brain, lung or gut using a multivariate proteomics approach. Subset panels of significantly altered proteins were selected to build predictive models of radiation exposure in a variety of sample cohort configurations relevant to practical field application of biodosimetry diagnostics during future radiological or nuclear event scenarios. Female C57BL/6 mice, 8-15 weeks old, received a single total-body or partial-body dose of 2 or 8 Gy TBI or 2 or 8 Gy to only the lung or gut, or 2, 8 or 16 Gy to only the brain using a Pantak X-ray source. Plasma was collected by cardiac puncture at days 1, 3 and 7 postirradiation for total-body exposures and only the lung and brain exposures, and at days 3, 7 and 14 postirradiation for gut exposures. Plasma was then screened using the aptamer-based SOMAscan proteomic assay technology, for changes in expression of 1,310 protein analytes. A subset panel of protein biomarkers which demonstrated significant changes (P < 0.01) in expression after irradiation were used to build predictive models of radiation exposure using different sample cohorts. Model 1 compared controls vs. all pooled irradiated samples, which included TBI and all organ specific partial irradiation. Model 2 compared controls vs. TBI vs. partial irradiation (with all organ specific partial exposure pooled within the partial-irradiated group), and model 3 compared controls vs. each individual organ specific partial-body exposure separately (brain, gut and lung). Detectable values were obtained for all 1,310 proteins included in the SOMAscan assay for all samples. Each model algorithm built using a unique sample cohort was validated with a training set of samples and tested with a separate new sample series. Overall predictive accuracies of 89%, 78% and 55% resulted for models 1-3, respectively, representing novel predictive panels of radiation responsive proteomic biomarkers. Though relatively high overall predictive accuracies were achieved for models 1 and 2, all three models showed limited accuracy at differentiating between the controls and partial-irradiated body samples. In our study we were able to identify novel panels of radiation responsive proteins useful for predicting radiation exposure and to create predictive models of partial-body exposure including organ specific radiation exposures. This proof-of-concept study also illustrates the inherent physiological limitations of distinguishing between small-body exposures and the unirradiated using proteomic biomarkers of radiation exposure. As use of biodosimetry diagnostics in future mass casualty settings will be complicated by the heterogeneity of partial-body exposure received in the field, further work remains in adapting these diagnostic tools for practical use.
Collapse
Affiliation(s)
- M. Sproull
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland
| | - T Kawai
- Department of Radiology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - A Krauze
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland
| | - U Shankavaram
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland
| | - K Camphausen
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
7
|
Sproull M, Nishita D, Chang P, Moroni M, Citrin D, Shankavaram U, Camphausen K. Comparison of Proteomic Expression Profiles after Radiation Exposure across Four Different Species. Radiat Res 2022; 197:315-323. [PMID: 35073400 PMCID: PMC9053310 DOI: 10.1667/rade-21-00182.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 12/10/2021] [Indexed: 11/03/2022]
Abstract
There is a need to identify biomarkers of radiation exposure for use in development of circulating biodosimeters for radiation exposure and for clinical use as markers of radiation injury. Most research approaches for biomarker discovery rely on a single animal model. The current study sought to take advantage of a novel aptamer-based proteomic assay which has been validated for use in many species to characterize changes to the blood proteome after total-body irradiation (TBI) across four different mammalian species including humans. Plasma was collected from C57BL6 mice, Sinclair minipigs, and Rhesus non-human primates (NHPs) receiving a single dose of TBI at a range of 3.3 Gy to 4.22 Gy at 24 h postirradiation. NHP and minipig models were irradiated using a 60Co source at a dose rate of 0.6 Gy/min, the C57BL6 mouse model using an X-ray source at a dose rate of 2.28 Gy/min and clinical samples from a photon source at 10 cGy/min. Plasma was collected from human patients receiving a single dose of 2 Gy TBI collected 6 h postirradiation. Plasma was screened using the aptamer-based SomaLogic SomaScan® proteomic assay technology to evaluate changes in the expression of 1,310 protein analytes. Confirmatory analysis of protein expression of biomarker HIST1H1C, was completed using plasma from C57BL6 mice receiving a 2, 3.5 or 8 Gy TBI collected at days 1, 3, and 7 postirradiation by singleplex ELISA. Summary of key pathways with altered expression after radiation exposure across all four mammalian species was determined using Ingenuity Pathway Analysis (IPA). Detectable values were obtained for all 1,310 proteins in all samples included in the SomaScan assay. A subset panel of protein biomarkers which demonstrated significant (p < 0.05) changes in expression of at least 1.3-fold after radiation exposure were characterized for each species. IPA of significantly altered proteins yielded a variety of top disease and biofunction pathways across species with the organismal injury and abnormalities pathway held in common for all four species. The HIST1H1C protein was shown to be radiation responsive within the human, NHP and murine species within the SomaScan dataset and was shown to demonstrate dose dependent upregulation at 2, 3.5 and 8 Gy at 24 h postirradiation in a separate murine cohort by ELISA. The SomaScan proteomics platform is a useful screening tool to evaluate changes in biomarker expression across multiple mammalian species. In our study, we were able to identify a novel biomarker of radiation exposure, HIST1H1C, and characterize panels of radiation responsive proteins and functional proteomic pathways altered by radiation exposure across murine, minipig, NHP and human species. Our study demonstrates the efficacy of using a multispecies approach for biomarker discovery.
Collapse
Affiliation(s)
- Mary Sproull
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland
| | | | | | - Maria Moroni
- Armed Forces Radiobiology Research Institute, Bethesda, Maryland
| | - Deborah Citrin
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland
| | - Uma Shankavaram
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland
| | - Kevin Camphausen
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
8
|
Abend M, Blakely WF, Ostheim P, Schuele S, Port M. Early molecular markers for retrospective biodosimetry and prediction of acute health effects. JOURNAL OF RADIOLOGICAL PROTECTION : OFFICIAL JOURNAL OF THE SOCIETY FOR RADIOLOGICAL PROTECTION 2022; 42:010503. [PMID: 34492641 DOI: 10.1088/1361-6498/ac2434] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 09/07/2021] [Indexed: 06/13/2023]
Abstract
Radiation-induced biological changes occurring within hours and days after irradiation can be potentially used for either exposure reconstruction (retrospective dosimetry) or the prediction of consecutively occurring acute or chronic health effects. The advantage of molecular protein or gene expression (GE) (mRNA) marker lies in their capability for early (1-3 days after irradiation), high-throughput and point-of-care diagnosis, required for the prediction of the acute radiation syndrome (ARS) in radiological or nuclear scenarios. These molecular marker in most cases respond differently regarding exposure characteristics such as e.g. radiation quality, dose, dose rate and most importantly over time. Changes over time are in particular challenging and demand certain strategies to deal with. With this review, we provide an overview and will focus on already identified and used mRNA GE and protein markers of the peripheral blood related to the ARS. These molecules are examined in light of 'ideal' characteristics of a biomarkers (e.g. easy accessible, early response, signal persistency) and the validation degree. Finally, we present strategies on the use of these markers considering challenges as their variation over time and future developments regarding e.g. origin of samples, point of care and high-throughput diagnosis.
Collapse
Affiliation(s)
- M Abend
- Bundeswehr Institute of Radiobiology, Munich, Germany
| | - W F Blakely
- Armed Forces Radiobiology Research Institute, Bethesda, MD, United States of America
| | - P Ostheim
- Bundeswehr Institute of Radiobiology, Munich, Germany
| | - S Schuele
- Bundeswehr Institute of Radiobiology, Munich, Germany
| | - M Port
- Bundeswehr Institute of Radiobiology, Munich, Germany
| |
Collapse
|
9
|
Circulating tRNA-Derived Small RNAs as Novel Radiation Biomarkers of Heavy Ion, Proton and X-ray Exposure. Int J Mol Sci 2021; 22:ijms222413476. [PMID: 34948273 PMCID: PMC8706565 DOI: 10.3390/ijms222413476] [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: 11/06/2021] [Revised: 12/09/2021] [Accepted: 12/12/2021] [Indexed: 11/17/2022] Open
Abstract
The effective and minimally invasive radiation biomarkers are valuable for exposure scenarios in nuclear accidents or space missions. Recent studies have opened the new sight of circulating small non-coding RNA (sncRNA) as radiation biomarkers. The tRNA-derived small RNA (tsRNA) is a new class of sncRNA. It is more abundant than other kinds of sncRNAs in extracellular vesicles or blood, presenting great potential as promising biomarkers. However, the circulating tsRNAs in response to ionizing radiation have not been reported. In this research, Kunming mice were total-body exposed to 0.05-2 Gy of carbon ions, protons, or X-rays, and the RNA sequencing was performed to profile the expression of sncRNAs in serum. After conditional screening and validation, we firstly identified 5 tsRNAs including 4 tRNA-related fragments (tRFs) and 1 tRNA half (tiRNA) which showed a significant level decrease after exposure to three kinds of radiations. Moreover, the radiation responses of these 5 serum tsRNAs were reproduced in other mouse strains, and the sequences of them could be detected in serum of humans. Furthermore, we developed multi-factor models based on tsRNA biomarkers to indicate the degree of radiation exposure with high sensitivity and specificity. These findings suggest that the circulating tsRNAs can serve as new minimally invasive biomarkers and can make a triage or dose assessment from blood sample collection within 4 h in exposure scenarios.
Collapse
|
10
|
Song Y, Hu S, Zhang J, Zhu L, Zhao X, Chen Q, Zhang J, Bai Y, Pan Y, Shao C. Fractionated Irradiation of Right Thorax Induces Abscopal Damage on Bone Marrow Cells via TNF-α and SAA. Int J Mol Sci 2021; 22:9964. [PMID: 34576128 PMCID: PMC8468747 DOI: 10.3390/ijms22189964] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/09/2021] [Accepted: 09/13/2021] [Indexed: 12/25/2022] Open
Abstract
Radiation-induced abscopal effect (RIAE) outside of radiation field is becoming more attractive. However, the underlying mechanisms are still obscure. This work investigated the deleterious effect of thoracic irradiation (Th-IR) on distant bone marrow and associated signaling factors by irradiating the right thorax of mice with fractionated doses (8 Gy × 3). It was found that this localized Th-IR increased apoptosis of bone marrow cells and micronucleus formation of bone marrow polychromatic erythrocytes after irradiation. Tandem mass tagging (TMT) analysis and ELISA assay showed that the concentrations of TNF-α and serum amyloid A (SAA) in the mice were significantly increased after Th-IR. An immunohistochemistry assay revealed a robust increase in SAA expression in the liver rather than in the lungs after Th-IR. In vitro experiments demonstrated that TNF-α induced SAA expression in mouse hepatoma Hepa1-6 cells, and these two signaling factors induced DNA damage in bone marrow mesenchymal stem cells (BMSCs) by increasing reactive oxygen species (ROS). On the other hand, injection with TNF-α inhibitor before Th-IR reduced the secretion of SAA and attenuated the abscopal damage in bone marrow. ROS scavenger NAC could also mitigated Th-IR/SAA-induced bone marrow damage in mice. Our findings indicated that Th-IR triggered TNF-α release from lung, which further promoted SAA secretion from liver in a manner of cascade reaction. Consequently, these signaling factors resulted in induction of abscopal damage on bone marrow of mice.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yan Pan
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China; (Y.S.); (S.H.); (J.Z.); (L.Z.); (X.Z.); (Q.C.); (J.Z.); (Y.B.)
| | - Chunlin Shao
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China; (Y.S.); (S.H.); (J.Z.); (L.Z.); (X.Z.); (Q.C.); (J.Z.); (Y.B.)
| |
Collapse
|
11
|
Chopra S, Moroni M, Sanjak J, MacMillan L, Hritzo B, Martello S, Bylicky M, May J, Coleman CN, Aryankalayil MJ. Whole blood gene expression within days after total-body irradiation predicts long term survival in Gottingen minipigs. Sci Rep 2021; 11:15873. [PMID: 34354115 PMCID: PMC8342483 DOI: 10.1038/s41598-021-95120-5] [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: 05/10/2021] [Accepted: 07/20/2021] [Indexed: 02/07/2023] Open
Abstract
Gottingen minipigs mirror the physiological radiation response observed in humans and hence make an ideal candidate model for studying radiation biodosimetry for both limited-sized and mass casualty incidents. We examined the whole blood gene expression profiles starting one day after total-body irradiation with increasing doses of gamma-rays. The minipigs were monitored for up to 45 days or time to euthanasia necessitated by radiation effects. We successfully identified dose- and time-agnostic (over a 1-7 day period after radiation), survival-predictive gene expression signatures derived using machine-learning algorithms with high sensitivity and specificity. These survival-predictive signatures fare better than an optimally performing dose-differentiating signature or blood cellular profiles. These findings suggest that prediction of survival is a much more useful parameter for making triage, resource-utilization and treatment decisions in a resource-constrained environment compared to predictions of total dose received. It should hopefully be possible to build such classifiers for humans in the future.
Collapse
Affiliation(s)
- Sunita Chopra
- National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD, 20892, USA
| | - Maria Moroni
- Armed Forces Radiobiological Research Institute, Bethesda, MD, 20889, USA
| | | | | | - Bernadette Hritzo
- Armed Forces Radiobiological Research Institute, Bethesda, MD, 20889, USA
| | - Shannon Martello
- National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD, 20892, USA
| | - Michelle Bylicky
- National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jared May
- National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD, 20892, USA
| | - C Norman Coleman
- National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD, 20892, USA.
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute (NCI), Bethesda, MD, 20892, USA.
| | - Molykutty J Aryankalayil
- National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD, 20892, USA.
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute (NCI), Bethesda, MD, 20892, USA.
| |
Collapse
|
12
|
Sproull M, Shankavaram U, Camphausen K. Novel Murine Biomarkers of Radiation Exposure Using An Aptamer-Based Proteomic Technology. Front Pharmacol 2021; 12:633131. [PMID: 33981223 PMCID: PMC8110031 DOI: 10.3389/fphar.2021.633131] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 04/12/2021] [Indexed: 11/13/2022] Open
Abstract
Purpose: There is a need to identify new biomarkers of radiation exposure both for use in the development of biodosimetry blood diagnostics for radiation exposure and for clinical use as markers of radiation injury. In the current study, a novel high-throughput proteomics screening approach was used to identify proteomic markers of radiation exposure in the plasma of total body irradiated mice. A subset panel of significantly altered proteins was selected to build predictive models of radiation exposure and received radiation dose useful for population screening in a future radiological or nuclear event. Methods: Female C57BL6 Mice of 8-14 weeks of age received a single total body irradiation (TBI) dose of 2, 3.5, 8 Gy or sham radiation and plasma was collected by cardiac puncture at days 1, 3, and 7 post-exposure. Plasma was then screened using the aptamer-based SOMAscan proteomic assay technology, for changes in expression of 1,310 protein analytes. A subset panel of protein biomarkers which demonstrated significant changes (p < 0.05) in expression following radiation exposure were used to build predictive models of radiation exposure and radiation dose. Results: Detectable values were obtained for all 1,310 proteins included in the SOMAscan assay. For the Control vs. Radiation model, the top predictive proteins were immunoglobulin heavy constant mu (IGHM), mitogen-activated protein kinase 14 (MAPK14), ectodysplasin A2 receptor (EDA2R) and solute carrier family 25 member 18 (SLC25A18). For the Control vs. Dose model, the top predictive proteins were cyclin dependent kinase 2/cyclin A2 (CDK2. CCNA2), E-selectin (SELE), BCL2 associated agonist of cell death (BAD) and SLC25A18. Following model validation with a training set of samples, both models tested with a new sample cohort had overall predictive accuracies of 85% and 73% for the Control vs. Radiation and Control vs. Dose models respectively. Conclusion: The SOMAscan proteomics platform is a useful screening tool to evaluate changes in biomarker expression. In our study we were able to identify a novel panel of radiation responsive proteins useful for predicting whether an animal had received a radiation exposure and to what dose they had received. Such diagnostic tools are needed for future medical management of radiation exposures.
Collapse
Affiliation(s)
| | | | - Kevin Camphausen
- Radiation Oncology Branch, National Cancer Institute, Bethesda, MD, United States
| |
Collapse
|
13
|
Rios CI, Cassatt DR, Hollingsworth BA, Satyamitra MM, Tadesse YS, Taliaferro LP, Winters TA, DiCarlo AL. Commonalities Between COVID-19 and Radiation Injury. Radiat Res 2021; 195:1-24. [PMID: 33064832 PMCID: PMC7861125 DOI: 10.1667/rade-20-00188.1] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 09/14/2020] [Indexed: 01/08/2023]
Abstract
As the multi-systemic components of COVID-19 emerge, parallel etiologies can be drawn between SARS-CoV-2 infection and radiation injuries. While some SARS-CoV-2-infected individuals present as asymptomatic, others exhibit mild symptoms that may include fever, cough, chills, and unusual symptoms like loss of taste and smell and reddening in the extremities (e.g., "COVID toes," suggestive of microvessel damage). Still others alarm healthcare providers with extreme and rapid onset of high-risk indicators of mortality that include acute respiratory distress syndrome (ARDS), multi-organ hypercoagulation, hypoxia and cardiovascular damage. Researchers are quickly refocusing their science to address this enigmatic virus that seems to unveil itself in new ways without discrimination. As investigators begin to identify early markers of disease, identification of common threads with other pathologies may provide some clues. Interestingly, years of research in the field of radiation biology documents the complex multiorgan nature of another disease state that occurs after exposure to high doses of radiation: the acute radiation syndrome (ARS). Inflammation is a key common player in COVID-19 and ARS, and drives the multi-system damage that dramatically alters biological homeostasis. Both conditions initiate a cytokine storm, with similar pro-inflammatory molecules increased and other anti-inflammatory molecules decreased. These changes manifest in a variety of ways, with a demonstrably higher health impact in patients having underlying medical conditions. The potentially dramatic human impact of ARS has guided the science that has identified many biomarkers of radiation exposure, established medical management strategies for ARS, and led to the development of medical countermeasures for use in the event of a radiation public health emergency. These efforts can now be leveraged to help elucidate mechanisms of action of COVID-19 injuries. Furthermore, this intersection between COVID-19 and ARS may point to approaches that could accelerate the discovery of treatments for both.
Collapse
Affiliation(s)
- Carmen I. Rios
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - David R. Cassatt
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - Brynn A. Hollingsworth
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - Merriline M. Satyamitra
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - Yeabsera S. Tadesse
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - Lanyn P. Taliaferro
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - Thomas A. Winters
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - Andrea L. DiCarlo
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| |
Collapse
|
14
|
A Biomarker Panel of Radiation-Upregulated miRNA as Signature for Ionizing Radiation Exposure. Life (Basel) 2020; 10:life10120361. [PMID: 33352926 PMCID: PMC7766228 DOI: 10.3390/life10120361] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/08/2020] [Accepted: 12/16/2020] [Indexed: 12/12/2022] Open
Abstract
Ionizing radiation causes serious injury to the human body and has long-time impacts on health. It is important to find optimal biomarkers for the early quick screening of exposed individuals. A series of miRNAs signatures have been developed as the new biomarkers for diagnosis, survival, and prognostic prediction of cancers. Here, we have identified the ionizing radiation-inducible miRNAs profile through microarray analysis. The biological functions were predicted for the top six upregulated miRNAs by 4 Gy γ-rays: miR-1246, miR-1307-3p, miR-3197, miR-4267, miR-5096 and miR-7641. The miRNA-gene network and target gene-pathway network analyses revealed that DNAH3 is the target gene associated with all the six miRNAs. GOLGB1 is related to 4 miRNAs and other 26 genes targeted by 3 miRNAs. The upregulation of fifteen miRNAs were further verified at 4 h and 24 h after 0 to 10 Gy irradiation in the human lymphoblastoid AHH-1 cells, and some demonstrated a dose-dependent increased. Six miRNAs, including miR-145, miR-663, miR-1273g-3p, miR-6090, miR-6727-5p and miR-7641, were validated to be dose-dependently upregulated at 4 h or 24 h post-irradiation in both AHH-1 and human peripheral blood lymphocytes irradiated ex vivo. This six-miRNA signature displays the superiority as a radiation biomarker for the translational application of screening and assessment of radiation exposed individuals.
Collapse
|
15
|
Chopra S, Moroni M, Martello S, Bylicky M, May J, Hritzo B, MacMillan L, Coleman CN, Aryankalayil MJ. Gene Expression Profiles from Heart, Lung and Liver Samples of Total-Body-Irradiated Minipigs: Implications for Predicting Radiation-Induced Tissue Toxicity. Radiat Res 2020; 194:411-430. [PMID: 32936898 DOI: 10.1667/rade-20-00123.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 08/03/2020] [Indexed: 11/03/2022]
Abstract
In the event of a major accidental or intentional radiation exposure incident, the affected population could suffer from total- or partial-body exposures to ionizing radiation with acute exposure to organs that would produce life-threatening injury. Therefore, it is necessary to identify markers capable of predicting organ-specific damage so that appropriate directed or encompassing therapies can be applied. In the current work, gene expression changes in response to total-body irradiation (TBI) were identified in heart, lungs and liver tissue of Göttingen minipigs. Animals received 1.7, 1.9, 2.1 or 2.3 Gy TBI and were followed for 45 days. Organ samples were collected at the end of day 45 or sooner if the animal displayed morbidity necessitating euthanasia. Our findings indicate that different organs respond to TBI in a very specific and distinct manner. We also found that the liver was the most affected organ in terms of gene expression changes, and that lipid metabolic pathways were the most deregulated in the liver samples of non-survivors (survival time <45 days). We identified organ-specific gene expression signatures that accurately differentiated non-survivors from survivors and control animals, irrespective of dose and time postirradiation. At what point did these radiation-induced injury markers manifest and how this information could be used for applying intervention therapies are under investigation.
Collapse
Affiliation(s)
- Sunita Chopra
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Maria Moroni
- Radiation Countermeasures Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Shannon Martello
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Michelle Bylicky
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Jared May
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Bernadette Hritzo
- Radiation Countermeasures Program, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | | | - C Norman Coleman
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland.,Radiation Research Program, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - Molykutty J Aryankalayil
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
16
|
Zhang Y, Liu J, Zhou L, Hao S, Ding Z, Xiao L, Zhou M. Exosomal Small RNA Sequencing Uncovers Dose-Specific MiRNA Markers for Ionizing Radiation Exposure. Dose Response 2020; 18:1559325820926735. [PMID: 32528236 PMCID: PMC7263154 DOI: 10.1177/1559325820926735] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 04/02/2020] [Accepted: 04/15/2020] [Indexed: 12/16/2022] Open
Abstract
Introduction: Acute exposure to ionizing radiation (IR) is hazardous or even lethal. Accurate estimation of the doses of IR exposure is critical to wisely determining the following treatments. Exosomes are nanoscale vesicles harboring biomolecules and mediate the communications among cells and tissues to influence biological processes. Screening out the microRNAs (miRNAs) contained in exosomes as biomarkers can be useful for estimating the IR exposure doses and exploring the correlation between these miRNAs and the occurrence of disease. Methods: We treated mice with 2.0, 6.5, and 8.0 Gy doses of IR and collected the mice sera at 0, 24, 48, and 72 hours after exposure. Then, the serum exosomes were isolated by ultracentrifuge and the small RNA portion was extracted for sequencing and the following bioinformatics analysis. Qualitative polymerase chain reaction was performed to validate the potential dose-specific markers. Results: Fifty-six miRNAs (31 upregulated, 25 downregulated) were differentially expressed after exposure of the above 3 IR doses and may act as common IR exposure miRNA markers. Bioinformatic analysis also identified several dosage-specific responsive miRNAs. Importantly, IR-induced miR-151-3p and miR-128-3p were significantly and stably increased at 24 hours in different mouse strains with distinct genetic background after exposed to 8.0 Gy of IR. Conclusion: Our study shows that miR-151-3p and miR-128-3p can be used as dose-specific biomarkers of 8.0 Gy IR exposure, which can be used to determine the exposure dose by detecting the amount of the 2 miRNAs in serum exosomes.
Collapse
Affiliation(s)
- Ying Zhang
- Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, Guangdong, China.,Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Jiabin Liu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Liang Zhou
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Shuai Hao
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Zhenhua Ding
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Lin Xiao
- Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, Guangdong, China
| | - Meijuan Zhou
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| |
Collapse
|
17
|
Wei W, Bai H, Feng X, Hua J, Long K, He J, Zhang Y, Ding N, Wang J, Zhou H. Serum Proteins as New Biomarkers for Whole-Body Exposure to High- and Low-LET Ionizing Radiation. Dose Response 2020; 18:1559325820914172. [PMID: 32273832 PMCID: PMC7113486 DOI: 10.1177/1559325820914172] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/29/2020] [Accepted: 02/18/2020] [Indexed: 02/02/2023] Open
Abstract
Exposure to ionizing radiation is a major threat to human health and public security. Since the inherent limitations of current methods for indicating radiation exposure, new minimally invasive biomarkers that can be easily and quickly detected at an early stage are needed for optimal medical treatment. Serum proteins are attractive biomarkers and some radiosensitive proteins have been found, but the proteins in response to low-dose and high-linear energy transfer (LET) radiation have not been reported. In this study, mice were whole body exposed to a variety doses of carbon ions and X-rays. We performed Mouse Antibody Array to detect serum proteins expression profiles at 24 hours postirradiation. After conditional screening, insulin-like growth factor-1 (IGF-1), insulin-like growth factor binding protein-1 (IGFBP-1), and IGFBP-3 were further validated using enzyme-linked immunosorbent assay. After exposure to 0.05 to 1 Gy of carbon ions and 0.5 to 4 Gy of X-rays, only IGFBP-3 showed obvious increase with increased doses, both carbon ions and X-rays. Further, IGFBP-3 was detected for observation of its time-dependent changes. The results showed the expression difference of IGFBP-3 presented from 6 to 24 hours post-irradiation by carbon ions and X-rays. Moreover, the receiver–operating characteristic analysis showed that serum IGFBP-3 is efficient to triage exposed individuals with high sensitivity and specificity. These results suggest that serum IGFBP-3 is extremely sensitive to high- and low-LET ionizing radiation and is able to respond at an early stage, which could serve as a novel minimally invasive indicator for radiation exposure.
Collapse
Affiliation(s)
- Wenjun Wei
- Key Laboratory of Space Radiobiology of Gansu Province & CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Hao Bai
- Key Laboratory of Space Radiobiology of Gansu Province & CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xiu Feng
- Key Laboratory of Space Radiobiology of Gansu Province & CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Junrui Hua
- Key Laboratory of Space Radiobiology of Gansu Province & CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Kaiqin Long
- Key Laboratory of Space Radiobiology of Gansu Province & CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Jinpeng He
- Key Laboratory of Space Radiobiology of Gansu Province & CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Yanan Zhang
- Key Laboratory of Space Radiobiology of Gansu Province & CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Nan Ding
- Key Laboratory of Space Radiobiology of Gansu Province & CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Jufang Wang
- Key Laboratory of Space Radiobiology of Gansu Province & CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Heng Zhou
- Key Laboratory of Space Radiobiology of Gansu Province & CAS Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| |
Collapse
|
18
|
Huang J, Qi Z, Chen M, Xiao T, Guan J, Zhou M, Wang Q, Lin Z, Wang Z. Serum amyloid A1 as a biomarker for radiation dose estimation and lethality prediction in irradiated mouse. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:715. [PMID: 32042731 DOI: 10.21037/atm.2019.12.27] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background Fast and reliable biomarkers are needed to distinguish whether individuals were exposed or not to radiation and assess radiation dose, and to predict the severity of radiation damage in a large-scale radiation accident. Serum amyloid A1 (SAA1) is a protein induced by multiple factors including inflammatory. Therefore, this study aimed at exploring the role of SAA1 in the radiation dose estimation and lethality prediction after radiation. Methods C57BL/6J female mice were exposed to total body irradiation (TBI) at different doses and time points and amifostine, a drug used to reduce the side effects of radiotherapy, was injected before irradiation. Patients with nasopharyngeal carcinoma subjected to radiotherapy were used as the irradiation model in humans. Results A moderate SAA1 increase was observed at 6 hours in serum samples from irradiated mice at all doses used, with a peak at 12 hours, then decreased to day 3 after exposure. A second SAA1 increase was observed from day 5 to 7, which was associated to subsequent lethality. Treatment with amifostine before irradiation could prevent mice death and inhibit the second SAA1 increase. SAA1 increase after radiation was confirmed in human serum of nasopharyngeal carcinoma patients after radiotherapy. Conclusions Serum SAA1 levels could represent a biomarker for radiation dose estimation and its second increase might be a useful lethality indicator after radiation in a mouse model.
Collapse
Affiliation(s)
- Jinfeng Huang
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China.,Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510080, China
| | - Zhenhua Qi
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Min Chen
- Department of Radiotherapy, Nanfang Hospital, Southern Medical University, Guangzhou 510080, China
| | - Ting Xiao
- Department of Radiotherapy, Nanfang Hospital, Southern Medical University, Guangzhou 510080, China
| | - Jian Guan
- Department of Radiotherapy, Nanfang Hospital, Southern Medical University, Guangzhou 510080, China
| | - Meijuan Zhou
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510080, China
| | - Qi Wang
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Zhongwu Lin
- Science Research Management Department of the Academy of Military Sciences, Beijing 100091, China
| | - Zhidong Wang
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, China
| |
Collapse
|
19
|
Rodríguez-Tomàs E, Murcia M, Arenas M, Arguís M, Gil M, Amigó N, Correig X, Torres L, Sabater S, Baiges-Gayà G, Cabré N, Luciano-Mateo F, Hernández-Aguilera A, Fort-Gallifa I, Camps J, Joven J. Serum Paraoxonase-1-Related Variables and Lipoprotein Profile in Patients with Lung or Head and Neck Cancer: Effect of Radiotherapy. Antioxidants (Basel) 2019; 8:antiox8070213. [PMID: 31295833 PMCID: PMC6680864 DOI: 10.3390/antiox8070213] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/26/2019] [Accepted: 07/05/2019] [Indexed: 12/20/2022] Open
Abstract
We investigated alterations in the levels of the antioxidant paraoxonase-1 (PON1) and the lipoprotein profile (analyzed by nuclear magnetic resonance) in patients with lung cancer (LC) or head and neck cancer (HNC), and the effects produced thereon by radiotherapy (RT). We included 33 patients with LC and 28 patients with HNC. Before irradiation, and one month after completion of RT, blood samples were obtained. The control group was composed of 50 healthy subjects. Patients had significantly lower serum PON1 activity and concentration before RT than the control group. PON1-related variables were good predictors of the presence of LC or HNC, with analytical sensitivities and specificities greater than 80%. Patients showed a significant increase in the number of particles of all subclasses of very-low-density lipoproteins (large, medium and small). However, these changes were not maintained when adjusted for age, sex, and other clinical and demographic variables. Irradiation was associated with a significant increase in PON1 concentration and, only in patients with HNC, with an increase in high-density lipoprotein-cholesterol concentration. Our results suggest that determinations of the levels of PON1-related variables may constitute good biomarkers for the evaluation of these diseases. Studies with a larger number of patients are needed to fully confirm this hypothesis.
Collapse
Affiliation(s)
- Elisabet Rodríguez-Tomàs
- Universitat Rovira i Virgili, Departament de Medicina i Cirurgia, Unitat de Recerca Biomèdica, 43201 Reus, Spain
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan de Reus, Institut d'Investigació Sanitària Pere Virgili, 43201 Reus, Spain
| | - Mauricio Murcia
- Department of Radiation Oncology, Hospital Universitari Sant Joan de Reus, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43201 Reus, Spain
| | - Meritxell Arenas
- Department of Radiation Oncology, Hospital Universitari Sant Joan de Reus, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43201 Reus, Spain.
| | - Mònica Arguís
- Department of Radiation Oncology, Hospital Universitari Sant Joan de Reus, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43201 Reus, Spain
| | | | | | - Xavier Correig
- Metabolomics Platform, CIBERDEM, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43201 Reus, Spain
| | - Laura Torres
- Department of Radiation Oncology, Hospital Universitari Sant Joan de Reus, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43201 Reus, Spain
| | - Sebastià Sabater
- Department of Radiation Oncology, Hospital Universitari Sant Joan de Reus, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, 43201 Reus, Spain
| | - Gerard Baiges-Gayà
- Universitat Rovira i Virgili, Departament de Medicina i Cirurgia, Unitat de Recerca Biomèdica, 43201 Reus, Spain
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan de Reus, Institut d'Investigació Sanitària Pere Virgili, 43201 Reus, Spain
| | - Noemí Cabré
- Universitat Rovira i Virgili, Departament de Medicina i Cirurgia, Unitat de Recerca Biomèdica, 43201 Reus, Spain
| | - Fedra Luciano-Mateo
- Universitat Rovira i Virgili, Departament de Medicina i Cirurgia, Unitat de Recerca Biomèdica, 43201 Reus, Spain
| | - Anna Hernández-Aguilera
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan de Reus, Institut d'Investigació Sanitària Pere Virgili, 43201 Reus, Spain
| | - Isabel Fort-Gallifa
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan de Reus, Institut d'Investigació Sanitària Pere Virgili, 43201 Reus, Spain
| | - Jordi Camps
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan de Reus, Institut d'Investigació Sanitària Pere Virgili, 43201 Reus, Spain.
| | - Jorge Joven
- Universitat Rovira i Virgili, Departament de Medicina i Cirurgia, Unitat de Recerca Biomèdica, 43201 Reus, Spain
- Unitat de Recerca Biomèdica, Hospital Universitari Sant Joan de Reus, Institut d'Investigació Sanitària Pere Virgili, 43201 Reus, Spain
| |
Collapse
|
20
|
Scanlon VM, Teixeira AM, Tyagi T, Zou S, Zhang PX, Booth CJ, Kowalska MA, Bao J, Hwa J, Hayes V, Marks MS, Poncz M, Krause DS. Epithelial (E)-Cadherin is a Novel Mediator of Platelet Aggregation and Clot Stability. Thromb Haemost 2019; 119:744-757. [PMID: 30861547 DOI: 10.1055/s-0039-1679908] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Cadherins play a major role in mediating cell-cell adhesion, which shares many parallels with platelet-platelet interactions during aggregate formation and clot stabilization. Platelets express epithelial (E)-cadherin, but its contribution to platelet function and/or platelet production is currently unknown. To assess the role of E-cadherin in platelet production and function in vitro and in vivo, we utilized a megakaryocyte-specific E-cadherin knockout mouse model. Loss of E-cadherin in megakaryocytes does not affect megakaryocyte maturation, platelet number or size. However, platelet dysfunction in the absence of E-cadherin is revealed when conditional knockout mice are challenged with acute antibody-mediated platelet depletion. Unlike wild-type mice that recover fully, knockout mice die within 72 hours post-antibody administration, likely from haemorrhage. Furthermore, conditional knockout mice have prolonged tail bleeding times, unstable clot formation, reduced clot retraction and reduced fibrin deposition in in vivo injury models. Murine platelet aggregation in vitro in response to thrombin and thrombin receptor activating peptide is compromised in E-cadherin null platelets, while aggregation in response to adenosine diphosphate (ADP) is not significantly different. Consistent with this, in vitro aggregation of primary human platelets in response to thrombin is decreased by an inhibitory E-cadherin antibody. Integrin activation and granule secretion in response to ADP and thrombin are not affected in E-cadherin null platelets, but Akt and glycogen synthase kinase 3β (GSK3β) activation are attenuated, suggesting a that E-cadherin contributes to aggregation, clot stabilization and retraction that is mediated by phosphoinositide 3-kinase/Akt/GSK3β signalling. In summary, E-cadherin plays a salient role in platelet aggregation and clot stability.
Collapse
Affiliation(s)
- Vanessa M Scanlon
- Department of Laboratory Medicine, Yale University, New Haven, Connecticut, United States.,The Yale Stem Cell Center, Yale University, New Haven, Connecticut, United States
| | | | - Tarun Tyagi
- Department of Internal Medicine, Yale University, New Haven, Connecticut, United States
| | - Siying Zou
- Department of Cell Biology, Yale University, New Haven, Connecticut, United States
| | - Ping-Xia Zhang
- Department of Laboratory Medicine, Yale University, New Haven, Connecticut, United States.,The Yale Stem Cell Center, Yale University, New Haven, Connecticut, United States
| | - Carmen Jane Booth
- Department of Comparative Medicine, Yale University, New Haven, Connecticut, United States
| | - M Anna Kowalska
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States.,Institute of Medical Biology, Polish Academy of Sciences, Philadelphia, Pennsylvania, United States
| | - Jialing Bao
- Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States.,Department of Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States.,University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - John Hwa
- Department of Internal Medicine, Yale University, New Haven, Connecticut, United States
| | - Vincent Hayes
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States
| | - Michael S Marks
- Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States.,Department of Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States.,University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Mortimer Poncz
- University of Pennsylvania, Philadelphia, Pennsylvania, United States.,Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States
| | - Diane S Krause
- Department of Laboratory Medicine, Yale University, New Haven, Connecticut, United States.,The Yale Stem Cell Center, Yale University, New Haven, Connecticut, United States.,Department of Pathology, Yale University, New Haven, Connecticut, United States.,Department of Cell Biology, Yale University, New Haven, Connecticut, United States
| |
Collapse
|
21
|
Aryankalayil MJ, Chopra S, Levin J, Eke I, Makinde A, Das S, Shankavaram U, Vanpouille-Box C, Demaria S, Coleman CN. Radiation-Induced Long Noncoding RNAs in a Mouse Model after Whole-Body Irradiation. Radiat Res 2018; 189:251-263. [PMID: 29309266 PMCID: PMC5967844 DOI: 10.1667/rr14891.1] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Long noncoding RNAs (lncRNAs) are emerging as key molecules in regulating many biological processes and have been implicated in development and disease pathogenesis. Biomarkers of cancer and normal tissue response to treatment are of great interest in precision medicine, as well as in public health and medical management, such as for assessment of radiation injury after an accidental or intentional exposure. Circulating and functional RNAs, including microRNAs (miRNAs) and lncRNAs, in whole blood and other body fluids are potential valuable candidates as biomarkers. Early prediction of possible acute, intermediate and delayed effects of radiation exposure enables timely therapeutic interventions. To address whether long noncoding RNAs (lncRNAs) could serve as biomarkers for radiation biodosimetry we performed whole genome transcriptome analysis in a mouse model after whole-body irradiation. Differential lncRNA expression patterns were evaluated at 16, 24 and 48 h postirradiation in total RNA isolated from whole blood of mice exposed to 1, 2, 4, 8 and 12 Gy of X rays. Sham-irradiated animals served as controls. Significant alterations in the expression patterns of lncRNAs were observed after different radiation doses at the various time points. We identified several radiation-induced lncRNAs known for DNA damage response as well as immune response. Long noncoding RNA targets of tumor protein 53 (P53), Trp53cor1, Dino, Pvt1 and Tug1 and an upstream regulator of p53, Meg3, were altered in response to radiation. Gm14005 ( Morrbid) and Tmevpg1 were regulated by radiation across all time points and doses. These two lncRNAs have important potential as blood-based radiation biomarkers; Gm14005 ( Morrbid) has recently been shown to play a key role in inflammatory response, while Tmevpg1 has been implicated in the regulation of interferon gamma. Precise molecular biomarkers, likely involving a diverse group of inducible molecules, will not only enable the development and effective use of medical countermeasures but may also be used to detect and circumvent or mitigate normal tissue injury in cancer radiotherapy.
Collapse
Affiliation(s)
| | - Sunita Chopra
- Radiation Oncology Branch, Center for Cancer Research, NMional Cancer Institute, Bethesda, Maryland
| | - Joel Levin
- Radiation Oncology Branch, Center for Cancer Research, NMional Cancer Institute, Bethesda, Maryland
| | - Iris Eke
- Radiation Oncology Branch, Center for Cancer Research, NMional Cancer Institute, Bethesda, Maryland
| | - Adeola Makinde
- Radiation Oncology Branch, Center for Cancer Research, NMional Cancer Institute, Bethesda, Maryland
| | - Shaoli Das
- Radiation Oncology Branch, Center for Cancer Research, NMional Cancer Institute, Bethesda, Maryland
| | - Uma Shankavaram
- Radiation Oncology Branch, Center for Cancer Research, NMional Cancer Institute, Bethesda, Maryland
| | | | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medicine, New York, New York
| | - C. Norman Coleman
- Radiation Oncology Branch, Center for Cancer Research, NMional Cancer Institute, Bethesda, Maryland
- Radiation Research Progrnm, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| |
Collapse
|
22
|
Zhao J, Li X, Zhao X, Wang J, Xi Q, Hu G. Study on the correlation of serum amyloid A level with overall survival and radiation pneumonitis in non-small cell lung cancer patients receiving thoracic radiotherapy. PRECISION RADIATION ONCOLOGY 2017. [DOI: 10.1002/pro6.20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Jing Zhao
- Department of Oncology; Tongji Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan Hubei China
| | - Xiaoyu Li
- Department of Oncology; Tongji Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan Hubei China
| | - Xueqi Zhao
- Department of Oncology; Tongji Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan Hubei China
| | - Jianhua Wang
- Department of Oncology; Tongji Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan Hubei China
| | - Qingsong Xi
- Department of Oncology; Tongji Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan Hubei China
| | - Guangyuan Hu
- Department of Oncology; Tongji Hospital; Tongji Medical College; Huazhong University of Science and Technology; Wuhan Hubei China
| |
Collapse
|
23
|
Sproull M, Kramp T, Tandle A, Shankavaram U, Camphausen K. Multivariate Analysis of Radiation Responsive Proteins to Predict Radiation Exposure in Total-Body Irradiation and Partial-Body Irradiation Models. Radiat Res 2017; 187:251-258. [PMID: 28118115 PMCID: PMC5385841 DOI: 10.1667/rr14558.1] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
In the event of a radiological or nuclear attack, advanced clinical countermeasures are needed for screening and medical management of the exposed population. In such a scenario, minimally invasive biomarkers that can accurately quantify radiation exposure would be useful for triage management by first responders. In this murine study, we evaluated the efficacy of a novel combination of radiation responsive proteins, Flt3 ligand (FL), serum amyloid A (SAA), matrix metalloproteinase 9 (MMP9), fibrinogen beta (FGB) and pentraxin 3 (PTX3) to predict the received dose after whole- or partial-body irradiation. Ten-week-old female C57BL6 mice received a single whole-body or partial-body dose of 18 Gy from a Pantak X-ray source at a dose rate of 2.28 Gy/min. Plasma was collected by cardiac puncture at 24, 48, 72 h and 1 week postirradiation. Plasma protein levels were determined via commercially available ELISA assay. A multivariate discriminant analysis was utilized to generate best-fit dose prediction models for whole-body exposures using the selected biomarker panel and its potential application to partial-body exposures was examined. The combination of values from FL, SAA, MMP9, FGB and PTX3 between 24 h and 1 week postirradiation yielded novel dose-response relationships. For day 1 postirradiation, the best-fit model yielded a predictive accuracy of 81% utilizing FL alone. The use of additional proteins did not enhance the model accuracy whereas, at day 2 postirradiation, the addition of PTX3 and FGB to FL increased the accuracy to 100%. At day 3 the use of FL and PTX3 yielded a predictive accuracy of 93% and at day 7 use of FL and SAA had an accuracy of 90%. Dose prediction of partial-body exposures based on the TBI model had a higher predictive accuracy when the percentage of the body exposed to radiation increased. Our findings indicate that this novel combination of radiation responsive biomarker proteins are an efficient method for predicting radiation exposure and are more accurate when used in concert compared to using any single biomarker protein alone.
Collapse
Affiliation(s)
- Mary Sproull
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland
| | - Tamalee Kramp
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland
| | - Anita Tandle
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland
| | - Uma Shankavaram
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland
| | - Kevin Camphausen
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
24
|
Hall J, Jeggo PA, West C, Gomolka M, Quintens R, Badie C, Laurent O, Aerts A, Anastasov N, Azimzadeh O, Azizova T, Baatout S, Baselet B, Benotmane MA, Blanchardon E, Guéguen Y, Haghdoost S, Harms-Ringhdahl M, Hess J, Kreuzer M, Laurier D, Macaeva E, Manning G, Pernot E, Ravanat JL, Sabatier L, Tack K, Tapio S, Zitzelsberger H, Cardis E. Ionizing radiation biomarkers in epidemiological studies - An update. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2017; 771:59-84. [PMID: 28342453 DOI: 10.1016/j.mrrev.2017.01.001] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 01/09/2017] [Indexed: 01/13/2023]
Abstract
Recent epidemiology studies highlighted the detrimental health effects of exposure to low dose and low dose rate ionizing radiation (IR): nuclear industry workers studies have shown increased leukaemia and solid tumour risks following cumulative doses of <100mSv and dose rates of <10mGy per year; paediatric patients studies have reported increased leukaemia and brain tumours risks after doses of 30-60mGy from computed tomography scans. Questions arise, however, about the impact of even lower doses and dose rates where classical epidemiological studies have limited power but where subsets within the large cohorts are expected to have an increased risk. Further progress requires integration of biomarkers or bioassays of individual exposure, effects and susceptibility to IR. The European DoReMi (Low Dose Research towards Multidisciplinary Integration) consortium previously reviewed biomarkers for potential use in IR epidemiological studies. Given the increased mechanistic understanding of responses to low dose radiation the current review provides an update covering technical advances and recent studies. A key issue identified is deciding which biomarkers to progress. A roadmap is provided for biomarker development from discovery to implementation and used to summarise the current status of proposed biomarkers for epidemiological studies. Most potential biomarkers remain at the discovery stage and for some there is sufficient evidence that further development is not warranted. One biomarker identified in the final stages of development and as a priority for further research is radiation specific mRNA transcript profiles.
Collapse
Affiliation(s)
- Janet Hall
- Centre de Recherche en Cancérologie de Lyon, INSERM 1052, CNRS 5286, Univ Lyon, Université Claude Bernard, Lyon 1, Lyon, F-69424, France.
| | - Penny A Jeggo
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Falmer, Brighton, BN1 9RQ, United Kingdom
| | - Catharine West
- Translational Radiobiology Group, Institute of Cancer Sciences, The University of Manchester, Manchester Academic Health Science Centre, Christie Hospital, Manchester, M20 4BX, United Kingdom
| | - Maria Gomolka
- Federal Office for Radiation Protection, Department of Radiation Protection and Health, D-85764 Neuherberg, Germany
| | - Roel Quintens
- Radiobiology Unit, Belgian Nuclear Research Centre, SCK·CEN, B-2400 Mol, Belgium
| | - Christophe Badie
- Cancer Mechanisms and Biomarkers group, Radiation Effects Department, Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Chilton, Didcot, United Kingdom
| | - Olivier Laurent
- Institut de Radioprotection et de Sûreté Nucléaire, F-92260 Fontenay-aux-Roses, France
| | - An Aerts
- Radiobiology Unit, Belgian Nuclear Research Centre, SCK·CEN, B-2400 Mol, Belgium
| | - Nataša Anastasov
- Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Institute of Radiation Biology, D-85764 Neuherberg, Germany
| | - Omid Azimzadeh
- Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Institute of Radiation Biology, D-85764 Neuherberg, Germany
| | - Tamara Azizova
- Southern Urals Biophysics Institute, Clinical Department, Ozyorsk, Russia
| | - Sarah Baatout
- Radiobiology Unit, Belgian Nuclear Research Centre, SCK·CEN, B-2400 Mol, Belgium; Cell Systems and Imaging Research Group, Department of Molecular Biotechnology, Ghent University, B-9000 Ghent, Belgium
| | - Bjorn Baselet
- Radiobiology Unit, Belgian Nuclear Research Centre, SCK·CEN, B-2400 Mol, Belgium; Pole of Pharmacology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, B-1200 Brussels, Belgium
| | - Mohammed A Benotmane
- Radiobiology Unit, Belgian Nuclear Research Centre, SCK·CEN, B-2400 Mol, Belgium
| | - Eric Blanchardon
- Institut de Radioprotection et de Sûreté Nucléaire, F-92260 Fontenay-aux-Roses, France
| | - Yann Guéguen
- Institut de Radioprotection et de Sûreté Nucléaire, F-92260 Fontenay-aux-Roses, France
| | - Siamak Haghdoost
- Centre for Radiation Protection Research, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE 106 91 Stockholm, Sweden
| | - Mats Harms-Ringhdahl
- Centre for Radiation Protection Research, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE 106 91 Stockholm, Sweden
| | - Julia Hess
- Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Institute of Radiation Biology, D-85764 Neuherberg, Germany
| | - Michaela Kreuzer
- Federal Office for Radiation Protection, Department of Radiation Protection and Health, D-85764 Neuherberg, Germany
| | - Dominique Laurier
- Institut de Radioprotection et de Sûreté Nucléaire, F-92260 Fontenay-aux-Roses, France
| | - Ellina Macaeva
- Radiobiology Unit, Belgian Nuclear Research Centre, SCK·CEN, B-2400 Mol, Belgium; Cell Systems and Imaging Research Group, Department of Molecular Biotechnology, Ghent University, B-9000 Ghent, Belgium
| | - Grainne Manning
- Cancer Mechanisms and Biomarkers group, Radiation Effects Department, Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Chilton, Didcot, United Kingdom
| | - Eileen Pernot
- INSERM U897, Université de Bordeaux, F-33076 Bordeaux cedex, France
| | - Jean-Luc Ravanat
- Laboratoire des Lésions des Acides Nucléiques, Univ. Grenoble Alpes, INAC-SCIB, F-38000 Grenoble, France; Commissariat à l'Énergie Atomique, INAC-SyMMES, F-38000 Grenoble, France
| | - Laure Sabatier
- Commissariat à l'Énergie Atomique, BP6, F-92265 Fontenay-aux-Roses, France
| | - Karine Tack
- Institut de Radioprotection et de Sûreté Nucléaire, F-92260 Fontenay-aux-Roses, France
| | - Soile Tapio
- Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Institute of Radiation Biology, D-85764 Neuherberg, Germany
| | - Horst Zitzelsberger
- Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Institute of Radiation Biology, D-85764 Neuherberg, Germany
| | - Elisabeth Cardis
- Barcelona Institute of Global Health (ISGlobal), Centre for Research in Environmental Epidemiology, Radiation Programme, Barcelona Biomedical Research Park, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF) (MTD formerly), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.
| |
Collapse
|
25
|
Sproull M, Camphausen K. State-of-the-Art Advances in Radiation Biodosimetry for Mass Casualty Events Involving Radiation Exposure. Radiat Res 2016; 186:423-435. [PMID: 27710702 DOI: 10.1667/rr14452.1] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
With the possibility of large-scale terrorist attacks around the world, the need for modeling and development of new medical countermeasures for potential future chemical, biological, radiological and nuclear (CBRN) has been well established. Project Bioshield, initiated in 2004, provided a framework to develop and expedite research in the field of CBRN exposures. To respond to large-scale population exposures from a nuclear event or radiation dispersal device (RDD), new methods for determining received dose using biological modeling became necessary. The field of biodosimetry has advanced significantly beyond this original initiative, with expansion into the fields of genomics, proteomics, metabolomics and transcriptomics. Studies are ongoing to evaluate the use of lymphocyte kinetics for dose assessment, as well as the development of field-deployable EPR technology. In addition, expansion of traditional cytogenetic assessment methods through the use of automated platforms and the development of laboratory surge capacity networks have helped to advance our biodefense preparedness. In this review of the latest advances in the field of biodosimetry we evaluate our progress and identify areas that still need to be addressed to achieve true field-deployment readiness.
Collapse
Affiliation(s)
- Mary Sproull
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland
| | - Kevin Camphausen
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
26
|
Menon SS, Uppal M, Randhawa S, Cheema MS, Aghdam N, Usala RL, Ghosh SP, Cheema AK, Dritschilo A. Radiation Metabolomics: Current Status and Future Directions. Front Oncol 2016; 6:20. [PMID: 26870697 PMCID: PMC4736121 DOI: 10.3389/fonc.2016.00020] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 01/18/2016] [Indexed: 12/25/2022] Open
Abstract
Human exposure to ionizing radiation (IR) disrupts normal metabolic processes in cells and organs by inducing complex biological responses that interfere with gene and protein expression. Conventional dosimetry, monitoring of prodromal symptoms, and peripheral lymphocyte counts are of limited value as organ- and tissue-specific biomarkers for personnel exposed to radiation, particularly, weeks or months after exposure. Analysis of metabolites generated in known stress-responsive pathways by molecular profiling helps to predict the physiological status of an individual in response to environmental or genetic perturbations. Thus, a multi-metabolite profile obtained from a high-resolution mass spectrometry-based metabolomics platform offers potential for identification of robust biomarkers to predict radiation toxicity of organs and tissues resulting from exposures to therapeutic or non-therapeutic IR. Here, we review the status of radiation metabolomics and explore applications as a standalone technology, as well as its integration in systems biology, to facilitate a better understanding of the molecular basis of radiation response. Finally, we draw attention to the identification of specific pathways that can be targeted for the development of therapeutics to alleviate or mitigate harmful effects of radiation exposure.
Collapse
Affiliation(s)
- Smrithi S Menon
- Department of Oncology, Georgetown University Medical Center , Washington, DC , USA
| | - Medha Uppal
- Department of Oncology, Georgetown University Medical Center , Washington, DC , USA
| | - Subeena Randhawa
- Department of Oncology, Georgetown University Medical Center , Washington, DC , USA
| | - Mehar S Cheema
- Department of Radiation Medicine, Georgetown University Medical Center , Washington, DC , USA
| | - Nima Aghdam
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center , Washington, DC , USA
| | - Rachel L Usala
- School of Medicine, Georgetown University Medical Center , Washington, DC , USA
| | - Sanchita P Ghosh
- Armed Forces Radiobiology Research Institute , Bethesda, MD , USA
| | - Amrita K Cheema
- Department of Oncology, Georgetown University Medical Center , Washington, DC , USA
| | - Anatoly Dritschilo
- Department of Radiation Medicine, Georgetown University Medical Center , Washington, DC , USA
| |
Collapse
|