1
|
Park S, Park JH, Ryu SH, Yeom J, Ryu JW, Park EY, Choi KC, Heo SH, Kim KH, Ha CH, Chang SK, Lee SW. Radiation-Induced Phosphorylation of Serine 360 of SMC1 in Human Peripheral Blood Mononuclear Cells. Radiat Res 2019; 191:262-270. [PMID: 30702968 DOI: 10.1667/rr15179.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
In the event of a mass casualty radiation scenario, biodosimetry has the potential to quantify individual exposures for triaging and providing dose-appropriate medical intervention. Structural maintenance of chromosomes 1 (SMC1) is phosphorylated in response to ionizing radiation. The goal of this study was to develop a new biodosimetry method using SMC1 phosphorylation as a measure of exposure to radiation. In the initial experiments, two normal human cell lines (WI-38VA-13 and HaCaT) and four lymphoblastoid cell lines were irradiated, and the levels of SMC1 phosphorylation at Ser-360 and Ser-957 were assessed using Western blotting. Subsequently, similar experiments were performed using peripheral blood mononuclear cells (PBMCs) obtained from 20 healthy adults. Phosphorylation of SMC1 at Ser-957 and Ser-360 was increased by exposure in a dose-dependent manner, peaked at 1-3 h postirradiation and then decreased gradually. Ser-360 was identified as a new phosphorylation site and was more sensitive to radiation than Ser-957, especially at doses below 1 Gy. Our results demonstrate a robust ex vivo response of phospho-SMC1-(Ser-360) to ionizing radiation in human PBMCs. Detection of phosphorylation at Ser-360 in SMC1 could be used as a marker of radiation exposure. Our findings suggest that it is feasible to measure blood cell-based changes in the phosphorylation level of a protein as an ex vivo radiation exposure detection method, even after low-dose exposure.
Collapse
Affiliation(s)
- Sunmin Park
- a Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jin-Hong Park
- a Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seung-Hee Ryu
- a Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jeonghun Yeom
- c Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Je-Won Ryu
- d Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Eun-Young Park
- a Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Kyung-Chul Choi
- b Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seung-Ho Heo
- d Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Kang Hyun Kim
- d Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Chang Hoon Ha
- b Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sei-Kyung Chang
- e Department of Radiation Oncology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Sang-Wook Lee
- a Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
2
|
Zhao M, Lau KK, Zhou X, Wu J, Yang J, Wang C. Urinary metabolic signatures and early triage of acute radiation exposure in rat model. MOLECULAR BIOSYSTEMS 2017; 13:756-766. [PMID: 28225098 DOI: 10.1039/c6mb00785f] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
After a large-scale radiological accident, early-response biomarkers to assess radiation exposure over a broad dose range are not only the basis of rapid radiation triage, but are also the key to the rational use of limited medical resources and to the improvement of treatment efficiency. Because of its high throughput, rapid assays and minimally invasive sample collection, metabolomics has been applied to research into radiation exposure biomarkers in recent years. Due to the complexity of radiobiological effects, most of the potential biomarkers are both dose-dependent and time-dependent. In reality, it is very difficult to find a single biomarker that is both sensitive and specific in a given radiation exposure scenario. Therefore, a multi-parameters approach for radiation exposure assessment is more realistic in real nuclear accidents. In this study, untargeted metabolomic profiling based on gas chromatography-mass spectrometry (GC-MS) and targeted amino acid profiling based on LC-MS/MS were combined to investigate early urinary metabolite responses within 48 h post-exposure in a rat model. A few of the key early-response metabolites for radiation exposure were identified, which revealed the most relevant metabolic pathways. Furthermore, a panel of potential urinary biomarkers was selected through a multi-criteria approach and applied to early triage following irradiation. Our study suggests that it is feasible to use a multi-parameters approach to triage radiation damage, and the urinary excretion levels of the relevant metabolites provide insights into radiation damage and repair.
Collapse
Affiliation(s)
- Mingxiao Zhao
- School of Radiation Medicine and Protection, Medical College of Soochow University, School for Radiological and Interdisciplinary Sciences (RAD-X), Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Suzhou Industrial Park Ren'ai Road 199, Suzhou 215123, P. R. China.
| | - Kim Kt Lau
- Department of Applied Chemistry, Xi'an Jiaotong-Liverpool University, Suzhou Industrial Park Ren'ai Road 111, Suzhou 215123, P. R. China
| | - Xian Zhou
- School of Radiation Medicine and Protection, Medical College of Soochow University, School for Radiological and Interdisciplinary Sciences (RAD-X), Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Suzhou Industrial Park Ren'ai Road 199, Suzhou 215123, P. R. China.
| | - Jianfang Wu
- Department of Applied Chemistry, Xi'an Jiaotong-Liverpool University, Suzhou Industrial Park Ren'ai Road 111, Suzhou 215123, P. R. China
| | - Jun Yang
- Department of Entomology and Nematology, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA.
| | - Chang Wang
- School of Radiation Medicine and Protection, Medical College of Soochow University, School for Radiological and Interdisciplinary Sciences (RAD-X), Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Suzhou Industrial Park Ren'ai Road 199, Suzhou 215123, P. R. China.
| |
Collapse
|
3
|
Park JG, Paul S, Briones N, Zeng J, Gillis K, Wallstrom G, LaBaer J, Amundson SA. Developing Human Radiation Biodosimetry Models: Testing Cross-Species Conversion Approaches Using an Ex Vivo Model System. Radiat Res 2017; 187:708-721. [PMID: 28328310 DOI: 10.1667/rr14655.1] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In the event of a large-scale radiation exposure, accurate and quick assessment of radiation dose received would be critical for triage and medical treatment of large numbers of potentially exposed individuals. Current methods of biodosimetry, such as the dicentric chromosome assay, are time consuming and require sophisticated equipment and highly trained personnel. Therefore, scalable biodosimetry approaches, including gene expression profiles in peripheral blood cells, are being investigated. Due to the limited availability of appropriate human samples, biodosimetry development has relied heavily on mouse models, which are not directly applicable to human response. Therefore, to explore the feasibility of using non-human primate (NHP) models to build and test a biodosimetry algorithm for use in humans, we irradiated ex vivo peripheral blood samples from both humans and rhesus macaques with doses of 0, 2, 5, 6 and 7 Gy, and compared the gene expression profiles 24 h later using Agilent human microarrays. Among the dose-responsive genes in human and using non-human primate, 52 genes showed highly correlated expression patterns between the species, and were enriched in p53/DNA damage response, apoptosis and cell cycle-related genes. When these interspecies-correlated genes were used to build biodosimetry models with using NHP data, the mean prediction accuracy on non-human primate samples was about 90% within 1 Gy of delivered dose in leave-one-out cross-validation. However, tests on human samples suggested that human gene expression values may need to be adjusted prior to application of the NHP model. A "multi-gene" approach utilizing all gene values for cross-species conversion and applying the converted values on the NHP biodosimetry models, gave a leave-one-out cross-validation prediction accuracy for human samples highly comparable (up to 94%) to that for non-human primates. Overall, this study demonstrates that a robust NHP biodosimetry model can be built using interspecies-correlated genes, and that, by using multiple regression-based cross-species conversion of expression values, absorbed dose in human samples can be accurately predicted by the NHP model.
Collapse
Affiliation(s)
- Jin G Park
- a Biodesign Center for Personalized Diagnostic, Biodesign Institute, Arizona State University, Arizona
| | - Sunirmal Paul
- d Center for Radiological Research, Columbia University Medical Center, New York
| | - Natalia Briones
- a Biodesign Center for Personalized Diagnostic, Biodesign Institute, Arizona State University, Arizona
| | - Jia Zeng
- a Biodesign Center for Personalized Diagnostic, Biodesign Institute, Arizona State University, Arizona.,b Department of Biomedical Informatics, Arizona State University, Arizona
| | - Kristin Gillis
- a Biodesign Center for Personalized Diagnostic, Biodesign Institute, Arizona State University, Arizona
| | - Garrick Wallstrom
- a Biodesign Center for Personalized Diagnostic, Biodesign Institute, Arizona State University, Arizona.,b Department of Biomedical Informatics, Arizona State University, Arizona
| | - Joshua LaBaer
- a Biodesign Center for Personalized Diagnostic, Biodesign Institute, Arizona State University, Arizona.,c School of Molecular Sciences, Arizona State University, Arizona
| | - Sally A Amundson
- d Center for Radiological Research, Columbia University Medical Center, New York
| |
Collapse
|
4
|
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
|
5
|
Dynamic changes in the proteome of human peripheral blood mononuclear cells with low dose ionizing radiation. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2016; 797:9-20. [DOI: 10.1016/j.mrgentox.2016.01.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 12/30/2015] [Accepted: 01/05/2016] [Indexed: 02/07/2023]
|
6
|
Johnston ML, Young EF, Shepard KL. Whole-blood immunoassay for γH2AX as a radiation biodosimetry assay with minimal sample preparation. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2015; 54:365-372. [PMID: 25935208 DOI: 10.1007/s00411-015-0595-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 04/16/2015] [Indexed: 06/04/2023]
Abstract
The current state of the art in high-throughput minimally invasive radiation biodosimetry involves the collection of samples in the field and analysis at a centralized facility. We have developed a simple biological immunoassay for radiation exposure that could extend this analysis out of the laboratory into the field. Such a forward placed assay would facilitate triage of a potentially exposed population. The phosphorylation and localization of the histone H2AX at double-stranded DNA breaks has already been proven to be an adequate surrogate assay for reporting DNA damage proportional to radiation dose. Here, we develop an assay for phosphorylated H2AX directed against minimally processed sample lysates. We conduct preliminary verification of H2AX phosphorylation using irradiated mouse embryo fibroblast cultures. Additional dosimetry is performed using human blood samples irradiated ex vivo. The assay reports H2AX phosphorylation in human blood samples in response to ionizing radiation over a range of 0-5 Gy in a linear fashion, without requiring filtering, enrichment, or purification of the blood sample.
Collapse
Affiliation(s)
- Matthew L Johnston
- Bialanx, Inc., 511 Avenue of the Americas, Suite 267, New York, NY, USA,
| | | | | |
Collapse
|
7
|
Paul S, Smilenov LB, Elliston CD, Amundson SA. Radiation Dose-Rate Effects on Gene Expression in a Mouse Biodosimetry Model. Radiat Res 2015; 184:24-32. [PMID: 26114327 DOI: 10.1667/rr14044.1] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
In the event of a nuclear accident or radiological terrorist attack, there will be a pressing need for biodosimetry to triage a large, potentially exposed population and to assign individuals to appropriate treatment. Exposures from fallout are likely, resulting in protracted dose delivery that would, in turn, impact the extent of injury. Biodosimetry approaches that can distinguish such low-dose-rate (LDR) exposures from acute exposures have not yet been developed. In this study, we used the C57BL/6 mouse model in an initial investigation of the impact of low-dose-rate delivery on the transcriptomic response in blood. While a large number of the same genes responded to LDR and acute radiation exposures, for many genes the magnitude of response was lower after LDR exposures. Some genes, however, were differentially expressed (P < 0.001, false discovery rate <5%) in mice exposed to LDR compared with mice exposed to acute radiation. We identified a set of 164 genes that correctly classified 97% of the samples in this experiment as exposed to acute or LDR radiation using a support vector machine algorithm. Gene expression is a promising approach to radiation biodosimetry, enhanced greatly by this first demonstration of its potential for distinguishing between acute and LDR exposures. Further development of this aspect of radiation biodosimetry, either as part of a complete gene expression biodosimetry test or as an adjunct to other methods, could provide vital triage information in a mass radiological casualty event.
Collapse
Affiliation(s)
- Sunirmal Paul
- a Center for Radiological Research, Columbia University Medical Center, New York, New York 10032;,b Rutgers University, Newark, New Jersey 07103; and
| | - Lubomir B Smilenov
- a Center for Radiological Research, Columbia University Medical Center, New York, New York 10032
| | - Carl D Elliston
- a Center for Radiological Research, Columbia University Medical Center, New York, New York 10032;,c Maimonides Medical Center, Brooklyn, New York 11219
| | - Sally A Amundson
- a Center for Radiological Research, Columbia University Medical Center, New York, New York 10032
| |
Collapse
|
8
|
Sproull M, Kramp T, Tandle A, Shankavaram U, Camphausen K. Serum Amyloid A as a Biomarker for Radiation Exposure. Radiat Res 2015; 184:14-23. [PMID: 26114330 DOI: 10.1667/rr13927.1] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There is a need for minimally invasive biomarkers that can accurately and quickly quantify radiation exposure. Radiation-responsive proteins have applications in clinical medicine and for mass population screenings after a nuclear or radiological incident where the level of radiation exposure and exposure pattern complicate medical triage for first responders. In this study, we evaluated the efficacy of the acute phase protein serum amyloid A (SAA) as a biomarker for radiation exposure using plasma from irradiated mice. Ten-week-old female C57BL6 mice received a 1-8 Gy single whole-body or partial-body dose from a Pantak X-ray source at a dose rate of 2.28 Gy/min. Plasma was collected by mandibular or cardiac puncture at 6, 24, 48 and 72 h or 1-3 weeks postirradiation. SAA levels were determined using a commercially available ELISA assay. Data was pooled to generate SAA μg/ml threshold values correlating plasma SAA levels with radiation dose. SAA levels were statistically significant over control at all exposures between 2 and 8 Gy at 24 h postirradiation but not at 6, 48 and 72 h or 1-3 weeks postirradiation. SAA levels at 1 Gy were not significantly elevated over control at all time points. Total-body-irradiated (TBI) SAA levels at 24 h were used to generate a dose prediction model that successfully differentiated TBI mice into dose received cohorts of control/1 Gy and ≥ 2 Gy groups with a high degree of accuracy in a blind study. Dose prediction of partial-body exposures based on the TBI model correlated increasing predictive accuracy with percentage of body exposure to radiation. Our findings indicate that plasma SAA levels might be a useful biomarker for radiation exposure in a variety of total- and partial-body irradiation settings.
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
|
9
|
Forrester HB, Sprung CN. Intragenic controls utilizing radiation-induced alternative transcript regions improves gene expression biodosimetry. Radiat Res 2014; 181:314-23. [PMID: 24625097 DOI: 10.1667/rr13501.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Ionizing-radiation exposure can be life threatening if given to the whole body. In addition, whole body radiation exposure can affect large numbers of people such as after a nuclear reactor accident, a nuclear explosion or a radiological terrorist attack. In these cases, an accurate biodosimeter is essential for triage management. One of the problems for biodosimetry in general is the interindividual variation before and after exposure, which can make it challenging to assign an accurate dose. To begin to address this challenge, lymphocyte cell lines were exposed to 0, 1, 2 and 5 Gy ionizing radiation from a ¹³⁷Cs source at a dose rate of 0.6 Gy/min. Alternative transcripts with regions showing large differential responses to ionizing radiation were determined from exon array data. Gene expression analysis was then performed on isolated mRNA using qRT-PCR with normalization to intergenic (PGK1, GAPDH) and novel intragenic regions for candidate radiation-responsive genes, PPM1D and MDM2. Our studies show that the use of a cis-associated expression reference improved the potential dose prediction approximately 2.3-8.3 fold and provided an advantage for dose prediction compared to distantly or trans-located control ionizing radiation nonresponsive genes. This approach also provides an alternative gene expression normalization method to potentially reduce interindividual variations when untreated basal gene expression levels are unavailable. Using associated noninduced regions of ionizing radiation-induced genes provides a way to estimate basal gene expression in the irradiated sample. This strategy can be utilized as a biodosimeter on its own or to enhance other gene expression candidates for biodosimetry. This normalization strategy may also be generally applicable for other quantitative PCR strategies where normalization is required for a particular response.
Collapse
Affiliation(s)
- Helen B Forrester
- Centre for Innate Immunity and Infectious Diseases, MIMR-PHI Institute of Medical Research and Monash University, Victoria, Australia
| | | |
Collapse
|
10
|
Correlation of plasma FL expression with bone marrow irradiation dose. PLoS One 2013; 8:e58558. [PMID: 23505536 PMCID: PMC3591371 DOI: 10.1371/journal.pone.0058558] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 02/05/2013] [Indexed: 01/07/2023] Open
Abstract
Purpose Ablative bone marrow irradiation is an integral part of hematopoietic stem cell transplantation. These treatment regimens are based on classically held models of radiation dose and the bone marrow response. Flt-3 ligand (FL) has been suggested as a marker of hematopoiesis and bone marrow status but the kinetics of its response to bone marrow irradiation has yet to be fully characterized. In the current study, we examine plasma FL response to total body and partial body irradiation in mice and its relationship with irradiation dose, time of collection and pattern of bone marrow exposure. Materials/Methods C57BL6 mice received a single whole body or partial body irradiation dose of 1–8 Gy. Plasma was collected by mandibular or cardiac puncture at 24, 48 and 72 hr post-irradiation as well as 1–3 weeks post-irradiation. FL levels were determined via ELISA assay and used to generate two models: a linear regression model and a gated values model correlating plasma FL levels with radiation dose. Results At all doses between 1–8 Gy, plasma FL levels were greater than control and the level of FL increased proportionally to the total body irradiation dose. Differences in FL levels were statistically significant at each dose and at all time points. Partial body irradiation of the trunk areas, encompassing the bulk of the hematopoietically active bone marrow, resulted in significantly increased FL levels over control but irradiation of only the head or extremities did not. FL levels were used to generate a dose prediction model for total body irradiation. In a blinded study, the model differentiated mice into dose received cohorts of 1, 4 or 8 Gy based on plasma FL levels at 24 or 72 hrs post-irradiation. Conclusion Our findings indicate that plasma FL levels might be used as a marker of hematopoietically active bone marrow and radiation exposure in mice.
Collapse
|
11
|
Fenech M. Current status, new frontiers and challenges in radiation biodosimetry using cytogenetic, transcriptomic and proteomic technologies. RADIAT MEAS 2011. [DOI: 10.1016/j.radmeas.2011.01.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
12
|
Paul S, Barker CA, Turner HC, McLane A, Wolden SL, Amundson SA. Prediction of in vivo radiation dose status in radiotherapy patients using ex vivo and in vivo gene expression signatures. Radiat Res 2011; 175:257-65. [PMID: 21388269 DOI: 10.1667/rr2420.1] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
After a large-scale nuclear accident or an attack with an improvised nuclear device, rapid biodosimetry would be needed for triage. As a possible means to address this need, we previously defined a gene expression signature in human peripheral white blood cells irradiated ex vivo that predicts the level of radiation exposure with high accuracy. We now demonstrate this principle in vivo using blood from patients receiving total-body irradiation (TBI). Whole genome microarray analysis has identified genes responding significantly to in vivo radiation exposure in peripheral blood. A 3-nearest neighbor classifier built from the TBI patient data correctly predicted samples as exposed to 0, 1.25 or 3.75 Gy with 94% accuracy (P < 0.001) even when samples from healthy donor controls were included. The same samples were classified with 98% accuracy using a signature previously defined from ex vivo irradiation data. The samples could also be classified as exposed or not exposed with 100% accuracy. The demonstration that ex vivo irradiation is an appropriate model that can provide meaningful prediction of in vivo exposure levels, and that the signatures are robust across diverse disease states and independent sample sets, is an important advance in the application of gene expression for biodosimetry.
Collapse
Affiliation(s)
- Sunirmal Paul
- Center for Radiological Research, Department of Radiation Oncology, Columbia University Medical Center, New York, New York 10032, USA
| | | | | | | | | | | |
Collapse
|
13
|
Ivey RG, Moore HD, Voytovich UJ, Thienes CP, Lorentzen TD, Pogosova-Agadjanyan EL, Frayo S, Izaguirre VK, Lundberg SJ, Hedin L, Badiozamani KR, Hoofnagle AN, Stirewalt DL, Wang P, Georges GE, Gopal AK, Paulovich AG. Blood-based detection of radiation exposure in humans based on novel phospho-Smc1 ELISA. Radiat Res 2010; 175:266-81. [PMID: 21388270 DOI: 10.1667/rr2402.1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The structural maintenance of chromosome 1 (Smc1) protein is a member of the highly conserved cohesin complex and is involved in sister chromatid cohesion. In response to ionizing radiation, Smc1 is phosphorylated at two sites, Ser-957 and Ser-966, and these phosphorylation events are dependent on the ATM protein kinase. In this study, we describe the generation of two novel ELISAs for quantifying phospho-Smc1(Ser-957) and phospho-Smc1(Ser-966). Using these novel assays, we quantify the kinetic and biodosimetric responses of human cells of hematological origin, including immortalized cells, as well as both quiescent and cycling primary human PBMC. Additionally, we demonstrate a robust in vivo response for phospho-Smc1(Ser-957) and phospho-Smc1(Ser-966) in lymphocytes of human patients after therapeutic exposure to ionizing radiation, including total-body irradiation, partial-body irradiation, and internal exposure to (131)I. These assays are useful for quantifying the DNA damage response in experimental systems and potentially for the identification of individuals exposed to radiation after a radiological incident.
Collapse
Affiliation(s)
- Richard G Ivey
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, Washington 98109-1024, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Partridge MA, Chai Y, Zhou H, Hei TK. High-throughput antibody-based assays to identify and quantify radiation-responsive protein biomarkers. Int J Radiat Biol 2010; 86:321-8. [PMID: 20353341 DOI: 10.3109/09553000903564034] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE After a radiological 'dirty bomb' incident in a major metropolitan center, substantial numbers of people may be exposed to radiation. However, only a fraction of those individuals will need urgent medical attention. Consequently, a rapid screening test is needed to identify those people who require immediate treatment. MATERIAL AND METHODS Ten normal human cell lines were screened by enzyme-linked immunosorbent assay (ELISA) for the expression of a dozen secreted cytokines that have been reported to have changes in protein or mRNA levels at 1, 2, and 3 days after 0-10 Gy irradiation using (137)Cs gamma rays at 0.82 Gy min(-1). After this systematic in vitro screen, we measured changes in the level of a subset of these candidate proteins in plasma from irradiated C57BL/6 mice (n = 3 per group), comparing shams with a single radiation dose (5 Gy X-rays) at 3.7 Gy min(-1) at 6 h after irradiation. RESULTS We identified four cytokine molecules that had altered levels after radiation exposure, one of which, Interleukin (IL) 6, was consistently elevated after irradiation in vitro and in vivo. CONCLUSIONS Our findings underscore the potential for IL6 as a marker for an immunoassay-based, rapid, high-throughput biodosimeter.
Collapse
Affiliation(s)
- Michael A Partridge
- Center for Radiological Research, College of Physicians & Surgeons, Columbia University, New York, New York 10032, USA
| | | | | | | |
Collapse
|