1
|
Ancel L, Grison S, Gabillot O, Gueguen J, Svilar L, Guen BL, Gruel G, Benderitter M, Martin JC, Souidi M, Tamarat R, Flamant S, Benadjaoud MA. Metabolomics identifies plasma biomarkers of localized radiation injury. Sci Rep 2025; 15:2166. [PMID: 39819895 PMCID: PMC11739571 DOI: 10.1038/s41598-025-85717-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 01/06/2025] [Indexed: 01/19/2025] Open
Abstract
A radiological accident may result in the development of a local skin radiation injury (LRI) which may evolve, depending on the dose, from dry desquamation to deep ulceration and necrosis through unpredictable inflammatory waves. Therefore, early diagnosis of victims of LRI is crucial for improving medical care efficiency. This preclinical study aims to identify circulating metabolites as biomarkers associated with LRI using a C57BL/6J mouse model of hind limb irradiation. More precisely, two independent mice cohorts were used to conduct a broad-spectrum profiling study followed by a suspect screening analysis performed on plasma metabolites by mass spectrometry. An integrative analysis was conducted through a multi-block sparse partial least square discriminant analysis (sPLS-DA) to establish multi-scale correlations between specific metabolites levels and biological, physiological (injury severity), and functional parameters (skin perfusion). The identified biomarker signature consists in a 6-metabolite panel including putrescine, uracil, 2,3-dihydroxybenzoate, 3-hydroxybenzoate, L-alanine and pyroglutamate, that can discriminate mice according to radiation dose and injury severity. Our results demonstrate relevant molecular signature associated with LRI in mice and support the use of plasma metabolites as suitable molecular biomarkers for LRI prognosis and diagnosis.
Collapse
Affiliation(s)
- Lucie Ancel
- Institut de Radioprotection et de Sureté Nucléaire (IRSN), PSE-SANTE/SERAMED/LRAcc, 31 av de la Division Leclerc, Fontenay-aux-Roses, 92260, France
| | - Stéphane Grison
- Institut de Radioprotection et de Sureté Nucléaire (IRSN), PSE-SANTE/SESANE/LRTox, Fontenay-aux-Roses, 92260, France
| | - Olivier Gabillot
- Institut de Radioprotection et de Sureté Nucléaire (IRSN), PSE-SANTE/SERAMED/LRAcc, 31 av de la Division Leclerc, Fontenay-aux-Roses, 92260, France
| | - Jules Gueguen
- Institut de Radioprotection et de Sureté Nucléaire (IRSN), PSE-SANTE/SERAMED/LRAcc, 31 av de la Division Leclerc, Fontenay-aux-Roses, 92260, France
| | - Ljubica Svilar
- Centre Cardiovasculaire et Nutrition (C2VN), CRIBIOM, Aix Marseille Université, Marseille, 13007, France
| | - Bernard Le Guen
- Électricité de France (EDF), DPN, 1 place Pleyel, Saint Denis, 93382, France
| | - Gaëtan Gruel
- Institut de Radioprotection et de Sureté Nucléaire (IRSN), PSE-SANTE/SERAMED, Fontenay-aux-Roses, 92260, France
| | - Marc Benderitter
- Institut de Radioprotection et de Sureté Nucléaire (IRSN), PSE-SANTE, Fontenay-aux-Roses, 92260, France
| | - Jean-Charles Martin
- C2VN, INRAE, INSERM, BIOMET, Aix Marseille Université, Marseille, 13007, France
| | - Maâmar Souidi
- Institut de Radioprotection et de Sureté Nucléaire (IRSN), PSE-SANTE/SERAMED/LRAcc, 31 av de la Division Leclerc, Fontenay-aux-Roses, 92260, France
| | - Radia Tamarat
- Institut de Radioprotection et de Sureté Nucléaire (IRSN), PSE-SANTE, Fontenay-aux-Roses, 92260, France
| | - Stéphane Flamant
- Institut de Radioprotection et de Sureté Nucléaire (IRSN), PSE-SANTE/SERAMED/LRAcc, 31 av de la Division Leclerc, Fontenay-aux-Roses, 92260, France
| | - Mohamed Amine Benadjaoud
- Institut de Radioprotection et de Sureté Nucléaire (IRSN), PSE-SANTE/SERAMED/LRAcc, 31 av de la Division Leclerc, Fontenay-aux-Roses, 92260, France.
| |
Collapse
|
2
|
Pannkuk EL, Laiakis EC, Garty G, Bansal S, Jayatilake MM, Tan Y, Ponnaiya B, Wu X, Amundson SA, Brenner DJ, Fornace AJ. Impact of Partial Body Shielding from Very High Dose Rates on Untargeted Metabolomics in Biodosimetry. ACS OMEGA 2024; 9:35182-35196. [PMID: 39157112 PMCID: PMC11325421 DOI: 10.1021/acsomega.4c05688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/16/2024] [Accepted: 07/22/2024] [Indexed: 08/20/2024]
Abstract
A realistic exposure to ionizing radiation (IR) from an improvised nuclear device will likely include individuals who are partially shielded from the initial blast delivered at a very high dose rate (VHDR). As different tissues have varying levels of radiosensitivity, e.g., hematopoietic vs gastrointestinal tissues, the effects of shielding on radiation biomarkers need to be addressed. Here, we explore how biofluid (urine and serum) metabolite signatures from male and female C57BL/6 mice exposed to VHDR (5-10 Gy/s) total body irradiation (TBI, 0, 4, and 8 Gy) compare to individuals exposed to partial body irradiation (PBI) (lower body irradiated [LBI] or upper body irradiated [UBI] at an 8 Gy dose) using a data-independent acquisition untargeted metabolomics approach. Although sex differences were observed in the spatial groupings of urine signatures from TBI and PBI mice, a metabolite signature (N6,N6,N6-trimethyllysine, carnitine, propionylcarnitine, hexosamine-valine-isoleucine, taurine, and creatine) previously developed from variable dose rate experiments was able to identify individuals with high sensitivity and specificity, irrespective of radiation shielding. A panel of serum metabolites composed from previous untargeted studies on nonhuman primates had excellent performance for separating irradiated cohorts; however, a multiomic approach to complement the metabolome could increase dose estimation confidence intervals. Overall, these results support the inclusion of small-molecule markers in biodosimetry assays without substantial interference from the upper or lower body shielding.
Collapse
Affiliation(s)
- Evan L. Pannkuk
- Department
of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia 20057, United States
- Department
of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, District of Columbia 20057, United States
- Center
for Metabolomic Studies, Georgetown University, Washington, District of
Columbia 20057, United States
| | - Evagelia C. Laiakis
- Department
of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia 20057, United States
- Department
of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, District of Columbia 20057, United States
- Center
for Metabolomic Studies, Georgetown University, Washington, District of
Columbia 20057, United States
- Department
of Radiation Medicine, Georgetown University
Hospital, Washington, District of Columbia 20057, United States
| | - Guy Garty
- Radiological
Research Accelerator Facility, Columbia
University, Irvington, New York 10533, United States
- Center for
Radiological Research, Columbia University
Irving Medical Center, New York, New York 10032, United States
| | - Sunil Bansal
- Department
of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia 20057, United States
| | - Meth M. Jayatilake
- Department
of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia 20057, United States
| | - Yuewen Tan
- Radiological
Research Accelerator Facility, Columbia
University, Irvington, New York 10533, United States
| | - Brian Ponnaiya
- Radiological
Research Accelerator Facility, Columbia
University, Irvington, New York 10533, United States
- Center for
Radiological Research, Columbia University
Irving Medical Center, New York, New York 10032, United States
| | - Xuefeng Wu
- Center for
Radiological Research, Columbia University
Irving Medical Center, New York, New York 10032, United States
| | - Sally A. Amundson
- Center for
Radiological Research, Columbia University
Irving Medical Center, New York, New York 10032, United States
| | - David J. Brenner
- Center for
Radiological Research, Columbia University
Irving Medical Center, New York, New York 10032, United States
| | - Albert J. Fornace
- Department
of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia 20057, United States
- Department
of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, District of Columbia 20057, United States
- Center
for Metabolomic Studies, Georgetown University, Washington, District of
Columbia 20057, United States
- Department
of Radiation Medicine, Georgetown University
Hospital, Washington, District of Columbia 20057, United States
| |
Collapse
|
3
|
Zhang Y, Tang N, Zhou H, Zhu Y. The role of microbial metabolites in endocrine tumorigenesis: From the mechanistic insights to potential therapeutic biomarkers. Biomed Pharmacother 2024; 172:116218. [PMID: 38308969 DOI: 10.1016/j.biopha.2024.116218] [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: 09/26/2023] [Revised: 12/28/2023] [Accepted: 01/22/2024] [Indexed: 02/05/2024] Open
Abstract
Microbial metabolites have been indicated to communicate with the host's endocrine system, regulating hormone production, immune-endocrine communications, and interactions along the gut-brain axis, eventually affecting the occurrence of endocrine cancer. Furthermore, microbiota metabolites such as short-chain fatty acids (SCFAs) have been found to affect the tumor microenvironment and boost immunity against tumors. SCFAs, including butyrate and acetate, have been demonstrated to exert anti-proliferative and anti-protective activity on pancreatic cancer cells. The employing of microbial metabolic products in conjunction with radiation and chemotherapy has shown promising outcomes in terms of reducing treatment side effects and boosting effectiveness. Certain metabolites, such as valerate and butyrate, have been made known to improve the efficiency of CAR T-cell treatment, whilst others, such as indole-derived tryptophan metabolites, have been shown to inhibit tumor immunity. This review explores the intricate interplay between microbial metabolites and endocrine tumorigenesis, spanning mechanistic insights to the discovery of potential therapeutic biomarkers.
Collapse
Affiliation(s)
- Yiyi Zhang
- Department of Endocrinology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China
| | - Nie Tang
- Department of Endocrinology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China
| | - Hui Zhou
- Department of Endocrinology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China.
| | - Ying Zhu
- Department of Endocrinology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China.
| |
Collapse
|
4
|
Taliaferro LP, Agarwal RK, Coleman CN, DiCarlo AL, Hofmeyer KA, Loelius SG, Molinar-Inglis O, Tedesco DC, Satyamitra MM. Sex differences in radiation research. Int J Radiat Biol 2023; 100:466-485. [PMID: 37991728 PMCID: PMC10922591 DOI: 10.1080/09553002.2023.2283089] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/06/2023] [Accepted: 10/26/2023] [Indexed: 11/23/2023]
Abstract
PURPOSE The Sex Differences in Radiation Research workshop addressed the role of sex as a confounder in radiation research and its implication in real-world radiological and nuclear applications. METHODS In April 2022, HHS-wide partners from the Radiation and Nuclear Countermeasures Program, the Office of Research on Women's Health National Institutes of Health Office of Women's Health, U.S. Food and Drug Administration, and the Radiological and Nuclear Countermeasures Branch at the Biomedical Advanced Research and Development Authority conducted a workshop to address the scientific implication and knowledge gaps in understanding sex in basic and translational research. The goals of this workshop were to examine sex differences in 1. Radiation animal models and understand how these may affect radiation medical countermeasure development; 2. Biodosimetry and/or biomarkers used to assess acute radiation syndrome, delayed effects of acute radiation exposure, and/or predict major organ morbidities; 3. medical research that lacks representation from both sexes. In addition, regulatory policies that influence inclusion of women in research, and the gaps that exist in drug development and device clearance were discussed. Finally, real-world sex differences in human health scenarios were also considered. RESULTS This report provides an overview of the two-day workshop, and open discussion among academic investigators, industry researchers, and U.S. government representatives. CONCLUSIONS This meeting highlighted that current study designs lack the power to determine statistical significance based on sex, and much is unknown about the underlying factors that contribute to these differences. Investigators should accommodate both sexes in all stages of research to ensure that the outcome is robust, reproducible, and accurate, and will benefit public health.
Collapse
Affiliation(s)
- Lanyn P. Taliaferro
- Division of Allergy, Immunology, and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Radiation and Nuclear Countermeasures Program (RNCP), Rockville, MD, USA
| | - Rajeev K. Agarwal
- Office of Research on Women’s Health (ORWH), Office of the Director, NIH, Rockville, MD, USA
| | - C. Norman Coleman
- Radiation Research Program Division of Cancer Treatment and Diagnosis, Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute (NCI) and Administration for Strategic Preparedness and Response (ASPR), U.S. Department of Health and Human Services (HHS), Washington, DC, USA
| | - Andrea L. DiCarlo
- Division of Allergy, Immunology, and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Radiation and Nuclear Countermeasures Program (RNCP), Rockville, MD, USA
| | - Kimberly A. Hofmeyer
- Radiological and Nuclear Countermeasures Branch, Biomedical Advanced Research and Development Authority (BARDA), ASPR, HHS, Washington, DC, USA
| | - Shannon G. Loelius
- Radiological and Nuclear Countermeasures Branch, Biomedical Advanced Research and Development Authority (BARDA), ASPR, HHS, Washington, DC, USA
| | - Olivia Molinar-Inglis
- Previously RNCP, DAIT, NIAID, NIH; now Antivirals and Antitoxins Program, Division of CBRN Countermeasures, BARDA, ASPR, HHS, Washington, DC, USA
| | - Dana C. Tedesco
- Radiological and Nuclear Countermeasures Branch, Biomedical Advanced Research and Development Authority (BARDA), ASPR, HHS, Washington, DC, USA
| | - Merriline M. Satyamitra
- Division of Allergy, Immunology, and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Radiation and Nuclear Countermeasures Program (RNCP), Rockville, MD, USA
| |
Collapse
|
5
|
Li W, Cheng F, Zhang J, Li C, Yu D, Simayijiang H, Liu H, Li S, Yan J. Changes in Gut Microbiota and Metabolites in Papillary Thyroid Carcinoma Patients Following Radioactive Iodine Therapy. Int J Gen Med 2023; 16:4453-4464. [PMID: 37808207 PMCID: PMC10557971 DOI: 10.2147/ijgm.s433433] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 09/28/2023] [Indexed: 10/10/2023] Open
Abstract
Purpose Radioactive iodine therapy is administered through oral route, which is accumulated and absorbed in the intestine. However, its effects on the intestine remain unclear. In this study, we investigated the changes in the gut microbiota and metabolites following radioactive iodine therapy. Patients and Methods A total of 76 stool samples from the same 38 patients were collected at the start of radioactive iodine therapy and three days following the therapy. Stool microbiota and metabolites were detected using 16S rRNA gene sequencing and liquid chromatography-mass spectrometry. Results Enterobacteriales, Enterobacteriaceae and Escherichia-Shigella were elevated in most patients (27/38) following the therapy. The levels of 2-hydroxyundec-7-enoylcarnitine were significantly lower, whereas those of 5-dehydroavenasterol, butylisopropylamine, and salsoline-1-carboxylate were higher following the therapy. The relative abundance of Escherichia-Shigella was negatively correlated with 2-hydroxyundec-7-enoylcarnitine level (r2 = -0.661, P = 0.009). Functional pathways were predicted to be involved in amino acid and lipid metabolism following the therapy. Particularly, phenylalanine, linoleic acid, sphingolipid, purine, and alpha-linolenic acid metabolism were the main metabolic pathways. Conclusion Gut microbiota was disturbed following radioactive iodine therapy, with increased Escherichia-Shigella. Processes associated with energy production seems to be impacted following the therapy, with significantly decreased 2-hydroxyundec-7-enoylcarnitine level. Meanwhile, some metabolites and functional pathways may have a positive effect on intestinal homeostasis, and may be related to the repair and promotion of gut recovery following the therapy. This study provides a basic foundation to explore how radioactive iodine affects gut microbiota and metabolites, and how gut function is regulated in response to radioactive iodine therapy.
Collapse
Affiliation(s)
- Wanting Li
- Shanxi Key Laboratory of Forensic Medicine, Shanxi Medical University, Jinzhong, People’s Republic of China
| | - Feng Cheng
- Shanxi Key Laboratory of Forensic Medicine, Shanxi Medical University, Jinzhong, People’s Republic of China
| | - Jun Zhang
- Shanxi Key Laboratory of Forensic Medicine, Shanxi Medical University, Jinzhong, People’s Republic of China
| | - Caihong Li
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Daijing Yu
- Shanxi Key Laboratory of Forensic Medicine, Shanxi Medical University, Jinzhong, People’s Republic of China
| | - Halimureti Simayijiang
- Shanxi Key Laboratory of Forensic Medicine, Shanxi Medical University, Jinzhong, People’s Republic of China
| | - Haiyan Liu
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Sijin Li
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Jiangwei Yan
- Shanxi Key Laboratory of Forensic Medicine, Shanxi Medical University, Jinzhong, People’s Republic of China
| |
Collapse
|
6
|
Gu L, Wang W, Gu Y, Cao J, Wang C. Metabolomic Signatures Associated with Radiation-Induced Lung Injury by Correlating Lung Tissue to Plasma in a Rat Model. Metabolites 2023; 13:1020. [PMID: 37755300 PMCID: PMC10536118 DOI: 10.3390/metabo13091020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/07/2023] [Accepted: 09/14/2023] [Indexed: 09/28/2023] Open
Abstract
The lung has raised significant concerns because of its radiosensitivity. Radiation-induced lung injury (RILI) has a serious impact on the quality of patients' lives and limits the effect of radiotherapy on chest tumors. In clinical practice, effective drug intervention for RILI remains to be fully elucidated. Therefore, an in-depth understanding of the biological characteristics is essential to reveal the mechanisms underlying the complex biological processes and discover novel therapeutic targets in RILI. In this study, Wistar rats received 0, 10, 20 or 35 Gy whole-thorax irradiation (WTI). Lung and plasma samples were collected within 5 days post-irradiation. Then, these samples were processed using liquid chromatography-mass spectrometry (LC-MS). A panel of potential plasma metabolic markers was selected by correlation analysis between the lung tissue and plasma metabolic features, followed by the evaluation of radiation injury levels within 5 days following whole-thorax irradiation (WTI). In addition, the multiple metabolic dysregulations primarily involved amino acids, bile acids and lipid and fatty acid β-oxidation-related metabolites, implying disturbances in the urea cycle, intestinal flora metabolism and mitochondrial dysfunction. In particular, the accumulation of long-chain acylcarnitines (ACs) was observed as early as 2 d post-WTI by dynamic plasma metabolic data analysis. Our findings indicate that plasma metabolic markers have the potential for RILI assessment. These results reveal metabolic characteristics following WTI and provide new insights into therapeutic interventions for RILI.
Collapse
Affiliation(s)
| | | | | | - Jianping Cao
- State Key Laboratory of Radiation Medicine and Protection, 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, China; (L.G.); (W.W.); (Y.G.)
| | - Chang Wang
- State Key Laboratory of Radiation Medicine and Protection, 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, China; (L.G.); (W.W.); (Y.G.)
| |
Collapse
|
7
|
Pannkuk EL, Laiakis EC, Garty G, Ponnaiya B, Wu X, Shuryak I, Ghandhi SA, Amundson SA, Brenner DJ, Fornace AJ. Variable Dose Rates in Realistic Radiation Exposures: Effects on Small Molecule Markers of Ionizing Radiation in the Murine Model. Radiat Res 2023; 200:1-12. [PMID: 37212727 PMCID: PMC10410530 DOI: 10.1667/rade-22-00211.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 04/27/2023] [Indexed: 05/23/2023]
Abstract
Novel biodosimetry assays for use in preparedness and response to potential malicious attacks or nuclear accidents would ideally provide accurate dose reconstruction independent of the idiosyncrasies of a complex exposure to ionizing radiation. Complex exposures will consist of dose rates spanning the low dose rates (LDR) to very high-dose rates (VHDR) that need to be tested for assay validation. Here, we investigate how a range of relevant dose rates affect metabolomic dose reconstruction at potentially lethal radiation exposures (8 Gy in mice) from an initial blast or subsequent fallout exposures compared to zero or sublethal exposures (0 or 3 Gy in mice) in the first 2 days, which corresponds to an integral time individuals will reach medical facilities after a radiological emergency. Biofluids (urine and serum) were collected from both male and female 9-10-week-old C57BL/6 mice at 1 and 2 days postirradiation (total doses of 0, 3 or 8 Gy) after a VHDR of 7 Gy/s. Additionally, samples were collected after a 2-day exposure consisting of a declining dose rate (1 to 0.004 Gy/min) recapitulating the 7:10 rule-of-thumb time dependency of nuclear fallout. Overall similar perturbations were observed in both urine and serum metabolite concentrations irrespective of sex or dose rate, with the exception of xanthurenic acid in urine (female specific) and taurine in serum (VHDR specific). In urine, we developed identical multiplex metabolite panels (N6, N6,N6-trimethyllysine, carnitine, propionylcarnitine, hexosamine-valine-isoleucine, and taurine) that could identify individuals receiving potentially lethal levels of radiation from the zero or sublethal cohorts with excellent sensitivity and specificity, with creatine increasing model performance at day 1. In serum, individuals receiving a 3 or 8 Gy exposure could be identified from their pre-irradiation samples with excellent sensitivity and specificity, however, due to a lower dose response the 3 vs. 8 Gy groups could not be distinguished from each other. Together with previous results, these data indicate that dose-rate-independent small molecule fingerprints have potential in novel biodosimetry assays.
Collapse
Affiliation(s)
- Evan L. Pannkuk
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC
- Center for Metabolomic Studies, Georgetown University, Washington, DC
| | - Evagelia C. Laiakis
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC
- Center for Metabolomic Studies, Georgetown University, Washington, DC
| | - Guy Garty
- Radiological Research Accelerator Facility, Columbia University, Irvington, New York
- Center for Radiological Research, Columbia University Irving Medical Center, New York, New York
| | - Brian Ponnaiya
- Center for Radiological Research, Columbia University Irving Medical Center, New York, New York
| | - Xuefeng Wu
- Center for Radiological Research, Columbia University Irving Medical Center, New York, New York
| | - Igor Shuryak
- Center for Radiological Research, Columbia University Irving Medical Center, New York, New York
| | - Shanaz A. Ghandhi
- Center for Radiological Research, Columbia University Irving Medical Center, New York, New York
| | - Sally A. Amundson
- Center for Radiological Research, Columbia University Irving Medical Center, New York, New York
| | - David J. Brenner
- Center for Radiological Research, Columbia University Irving Medical Center, New York, New York
| | - Albert J. Fornace
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC
- Center for Metabolomic Studies, Georgetown University, Washington, DC
| |
Collapse
|
8
|
Liu HX, Zhao H, Xi C, Li S, Ma LP, Lu X, Yan J, Tian XL, Gao L, Tian M, Liu QJ. CPT1 Mediated Ionizing Radiation-Induced Intestinal Injury Proliferation via Shifting FAO Metabolism Pathway and Activating the ERK1/2 and JNK Pathway. Radiat Res 2022; 198:488-507. [PMID: 36351324 DOI: 10.1667/rade-21-00174.1] [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: 09/15/2021] [Accepted: 07/07/2022] [Indexed: 06/16/2023]
Abstract
The intestinal compensatory proliferative potential is a key influencing factor for susceptibility to radiation-induced intestinal injury. Studies indicated that the carnitine palmitoyltransferase 1 (CPT1) mediated fatty acid β-oxidation (FAO) plays a crucial role in promoting the survival and proliferation of tumor cells. Here, we aimed to explore the effect of 60Co gamma rays on CPT1 mediated FAO in the radiation-induced intestinal injury models, and investigate the role of CPT1 mediated FAO in the survival and proliferation of intestinal cells after irradiation. We detected the changed of FAO in the plasma and small intestine of Sprague Dawley (SD) rats at 24 h after 60Co gamma irradiation (0, 5 and 10 Gy), using target metabolomics, qRT-PCR, immunohistochemistry (IHC), western blot (WB) and related enzymatic activity kits. We then analyzed the FAO changes in radiation-induced intestinal injury models regardless of ex vivo (mice enteroids), or in vitro (normal human intestinal epithelial cell lines, HIEC-6). HIEC-6 cells were transduced with lentivirus vector GV392 and treated with puromycin for obtaining CPT1 stable knockout cell lines, named CPT1 KO. CPT1 enzymatic activities of HIEC-6 cells and mice enteroids were also inhibited by pharmaceutical inhibitor ST1326 and Etomoxir (ETO), to study the function of CPT1 in the survival and proliferation of HIEC-6 cells after 60Co gamma irradiation. We found that CPT1 mediated FAO was altered in the small intestine of the SD rats after irradiation, especially, the expression level and enzymatic activity of CPT1 were significantly increased. Similarly, the expression levels of CPT1 were also remarkably enhanced in mice enteroids and HIEC-6 cells after irradiation. CPT1 inhibition decreased the proliferation of the HIEC-6 cells and mice enteroids after irradiation partially by reducing the extracellular signal-regulated kinase (ERK1/2) and c-Jun N-terminal kinase (JNK) pathways activation, CPT1 inhibition also reduced the proliferation of mice enteroids after irradiation partially by down-regulating the Wnt/β-catenin signaling activity. In conclusion, our study indicated that CPT1 plays a crucial role in promoting intestinal epithelial cell proliferation after irradiation.
Collapse
Affiliation(s)
- Hai-Xiang Liu
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, People's Republic of China
| | - Hua Zhao
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, People's Republic of China
| | - Cong Xi
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, People's Republic of China
| | - Shuang Li
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, People's Republic of China
| | - Li-Ping Ma
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, People's Republic of China
| | - Xue Lu
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, People's Republic of China
| | - Juan Yan
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, People's Republic of China
| | - Xue-Lei Tian
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, People's Republic of China
| | - Ling Gao
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, People's Republic of China
| | - Mei Tian
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, People's Republic of China
| | - Qing-Jie Liu
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, People's Republic of China
| |
Collapse
|
9
|
Azimzadeh O, Moertl S, Ramadan R, Baselet B, Laiakis EC, Sebastian S, Beaton D, Hartikainen JM, Kaiser JC, Beheshti A, Salomaa S, Chauhan V, Hamada N. Application of radiation omics in the development of adverse outcome pathway networks: an example of radiation-induced cardiovascular disease. Int J Radiat Biol 2022; 98:1722-1751. [PMID: 35976069 DOI: 10.1080/09553002.2022.2110325] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Epidemiological studies have indicated that exposure of the heart to doses of ionizing radiation as low as 0.5 Gy increases the risk of cardiac morbidity and mortality with a latency period of decades. The damaging effects of radiation to myocardial and endothelial structures and functions have been confirmed radiobiologically at high dose, but much less is known at low dose. Integration of radiation biology and epidemiology data is a recommended approach to improve the radiation risk assessment process. The adverse outcome pathway (AOP) framework offers a comprehensive tool to compile and translate mechanistic information into pathological endpoints which may be relevant for risk assessment at the different levels of a biological system. Omics technologies enable the generation of large volumes of biological data at various levels of complexity, from molecular pathways to functional organisms. Given the quality and quantity of available data across levels of biology, omics data can be attractive sources of information for use within the AOP framework. It is anticipated that radiation omics studies could improve our understanding of the molecular mechanisms behind the adverse effects of radiation on the cardiovascular system. In this review, we explored the available omics studies on radiation-induced cardiovascular disease (CVD) and their applicability to the proposed AOP for CVD. RESULTS The results of 80 omics studies published on radiation-induced CVD over the past 20 years have been discussed in the context of the AOP of CVD proposed by Chauhan et al. Most of the available omics data on radiation-induced CVD are from proteomics, transcriptomics, and metabolomics, whereas few datasets were available from epigenomics and multi-omics. The omics data presented here show great promise in providing information for several key events of the proposed AOP of CVD, particularly oxidative stress, alterations of energy metabolism, extracellular matrix and vascular remodeling. CONCLUSIONS The omics data presented here shows promise to inform the various levels of the proposed AOP of CVD. However, the data highlight the urgent need of designing omics studies to address the knowledge gap concerning different radiation scenarios, time after exposure and experimental models. This review presents the evidence to build a qualitative omics-informed AOP and provides views on the potential benefits and challenges in using omics data to assess risk-related outcomes.
Collapse
Affiliation(s)
- Omid Azimzadeh
- Federal Office for Radiation Protection (BfS), Section Radiation Biology, 85764 Neuherberg, Germany
| | - Simone Moertl
- Federal Office for Radiation Protection (BfS), Section Radiation Biology, 85764 Neuherberg, Germany
| | - Raghda Ramadan
- Institute for Environment, Health and Safety, Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Bjorn Baselet
- Institute for Environment, Health and Safety, Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Evagelia C Laiakis
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA.,Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, DC 20057, USA
| | | | | | - Jaana M Hartikainen
- School of Medicine, Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland, Kuopio, Finland
| | - Jan Christian Kaiser
- Helmholtz Zentrum München, Institute of Radiation Medicine (HMGU-IRM), 85764 Neuherberg, Germany
| | - Afshin Beheshti
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Sisko Salomaa
- Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| | - Vinita Chauhan
- Environmental Health Science Research Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Nobuyuki Hamada
- Biology and Environmental Chemistry Division, Sustainable System Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), Komae, Tokyo 201-8511, Japan
| |
Collapse
|
10
|
Geng F, Chen J, Tang S, Azzam E, Zhang J, Zhang S. Additional Evidence for Commonalities between COVID-19 and Radiation Injury: Novel Insight into COVID-19 Candidate Drugs. Radiat Res 2022; 198:306-317. [PMID: 35834824 DOI: 10.1667/rade-22-00058.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/14/2022] [Indexed: 11/03/2022]
Abstract
COVID-19 is a challenge to biosecurity and public health. The speed of vaccine development lags behind that of virus evolution and mutation. To date, no agent has been demonstrated to be fully effective against COVID-19. Therefore, it remains of great urgency to rapidly develop promising therapeutic and diagnostic candidates. Intriguingly, mounting evidence hints at parallel etiologies between SARS-CoV-2 infection and radiation injury. Herein, from the perspectives of immunogenic pathway activation and metabolic alterations, we provide novel evidence of commonalities between these two pathological conditions based on the most recent findings. Since numerous agents have been developed to prevent or reverse radiation injury in the past 70 years to ensure nuclear safety, we also advocate investigating the promising function of radioprotectors and radiomitigators against COVID-19 in clinical settings.
Collapse
Affiliation(s)
- Fenghao Geng
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China.,West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Jianhui Chen
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Shaokai Tang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Edouard Azzam
- Radiobiology and Health, Isotopes, Radiobiology & Environment Directorate (IRED), Canadian Nuclear Laboratories (CNL), Chalk River, ON K0J 1J0, Canada
| | - Jie Zhang
- Institute of Preventive Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Shuyu Zhang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China.,West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu 610051, China.,NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, Mianyang 621099, China
| |
Collapse
|
11
|
Hinzman CP, Jayatilake M, Bansal S, Fish BL, Li Y, Zhang Y, Bansal S, Girgis M, Iliuk A, Xu X, Fernandez JA, Griffin JH, Ballew EA, Unger K, Boerma M, Medhora M, Cheema AK. An optimized method for the isolation of urinary extracellular vesicles for molecular phenotyping: detection of biomarkers for radiation exposure. J Transl Med 2022; 20:199. [PMID: 35538547 PMCID: PMC9092707 DOI: 10.1186/s12967-022-03414-7] [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: 03/11/2022] [Accepted: 04/25/2022] [Indexed: 11/15/2022] Open
Abstract
Background Urinary extracellular vesicles (EVs) are a source of biomarkers with broad potential applications across clinical research, including monitoring radiation exposure. A key limitation to their implementation is minimal standardization in EV isolation and analytical methods. Further, most urinary EV isolation protocols necessitate large volumes of sample. This study aimed to compare and optimize isolation and analytical methods for EVs from small volumes of urine. Methods 3 EV isolation methods were compared: ultracentrifugation, magnetic bead-based, and size-exclusion chromatography from 0.5 mL or 1 mL of rat and human urine. EV yield and mass spectrometry signals (Q-ToF and Triple Quad) were evaluated from each method. Metabolomic profiling was performed on EVs isolated from the urine of rats exposed to ionizing radiation 1-, 14-, 30- or 90-days post-exposure, and human urine from patients receiving thoracic radiotherapy for the treatment of lung cancer pre- and post-treatment. Results Size-exclusion chromatography is the preferred method for EV isolation from 0.5 mL of urine. Mass spectrometry-based metabolomic analyses of EV cargo identified biochemical changes induced by radiation, including altered nucleotide, folate, and lipid metabolism. We have provided standard operating procedures for implementation of these methods in other laboratories. Conclusions We demonstrate that EVs can be isolated from small volumes of urine and analytically investigated for their biochemical contents to detect radiation induced metabolomic changes. These findings lay a groundwork for future development of methods to monitor response to radiotherapy and can be extended to an array of molecular phenotyping studies aimed at characterizing EV cargo. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03414-7.
Collapse
Affiliation(s)
- Charles P Hinzman
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC, 20007, USA
| | - Meth Jayatilake
- Department of Oncology, Georgetown University Medical Center, Washington, DC, 20007, USA
| | - Sunil Bansal
- Department of Oncology, Georgetown University Medical Center, Washington, DC, 20007, USA
| | - Brian L Fish
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Yaoxiang Li
- Department of Oncology, Georgetown University Medical Center, Washington, DC, 20007, USA
| | - Yubo Zhang
- Department of Oncology, Georgetown University Medical Center, Washington, DC, 20007, USA
| | - Shivani Bansal
- Department of Oncology, Georgetown University Medical Center, Washington, DC, 20007, USA
| | - Michael Girgis
- Department of Oncology, Georgetown University Medical Center, Washington, DC, 20007, USA
| | - Anton Iliuk
- Tymora Analytical Operations, West Lafayette, IN, 47906, USA
| | - Xiao Xu
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Jose A Fernandez
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA, 92037, USA
| | - John H Griffin
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Elizabeth A Ballew
- Department of Radiation Medicine, MedStar Georgetown University Hospital, Washington, DC, 20007, USA
| | - Keith Unger
- Department of Radiation Medicine, MedStar Georgetown University Hospital, Washington, DC, 20007, USA
| | - Marjan Boerma
- Department of Pharmaceutical Sciences, Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AK, 72205, USA
| | - Meetha Medhora
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Amrita K Cheema
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC, 20007, USA. .,Department of Oncology, Georgetown University Medical Center, Washington, DC, 20007, USA.
| |
Collapse
|
12
|
Guo L, Wu B, Wang X, Kou X, Zhu X, Fu K, Zhang Q, Hong S, Wang X. Long-term low-dose ionizing radiation induced chromosome-aberration-specific metabolic phenotype changes in radiation workers. J Pharm Biomed Anal 2022; 214:114718. [DOI: 10.1016/j.jpba.2022.114718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 03/04/2022] [Accepted: 03/05/2022] [Indexed: 10/18/2022]
|
13
|
Liu HX, Liu QJ. Logistic role of carnitine shuttle system on radiation-induced L-carnitine and acylcarnitines alteration. Int J Radiat Biol 2022; 98:1595-1608. [PMID: 35384773 DOI: 10.1080/09553002.2022.2063430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 12/13/2022]
Abstract
PURPOSE With the development of radiation metabolomics, a large number of radiation-related metabolic biomarkers have been identified and validated. The L-carnitine and acylcarnitines have the potential to be the new promising candidate indicators of radiation exposure. This review summarizes the effect of carnitine shuttle system on the profile of acylcarnitines and correlates the radiation effects on upstream regulators of carnitine shuttle system with the change characteristics of L-carnitine and acylcarnitines after irradiation across different animal models as well as a few humans. CONCLUSIONS Studies report that acylcarnitines were ubiquitously elevated after irradiation, especially the free L-carnitine and short-chain acylcarnitines (C2-C5). However, the molecular mechanism underlying acylcarnitine alterations after irradiation is not fully investigated, and further studies are needed to explore the biological effect and its mechanism. The activity of the carnitine shuttle system plays a key role in the alteration of L-carnitine and acylcarnitines, and the upstream regulators of the system are known to be affected by irradiation. These evidences indicate that that there is a logistic role of carnitine shuttle system on radiation-induced L-carnitine and acylcarnitines alteration.
Collapse
Affiliation(s)
- Hai-Xiang Liu
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, People's Republic of China
| | - Qing-Jie Liu
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, People's Republic of China
| |
Collapse
|
14
|
Strybel U, Marczak L, Zeman M, Polanski K, Mielańczyk Ł, Klymenko O, Samelak-Czajka A, Jackowiak P, Smolarz M, Chekan M, Zembala-Nożyńska E, Widlak P, Pietrowska M, Wojakowska A. Molecular Composition of Serum Exosomes Could Discriminate Rectal Cancer Patients with Different Responses to Neoadjuvant Radiotherapy. Cancers (Basel) 2022; 14:993. [PMID: 35205741 PMCID: PMC8870712 DOI: 10.3390/cancers14040993] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 11/21/2022] Open
Abstract
Identification of biomarkers that could be used for the prediction of the response to neoadjuvant radiotherapy (neo-RT) in locally advanced rectal cancer remains a challenge addressed by different experimental approaches. Exosomes and other classes of extracellular vesicles circulating in patients' blood represent a novel type of liquid biopsy and a source of cancer biomarkers. Here, we used a combined proteomic and metabolomic approach based on mass spectrometry techniques for studying the molecular components of exosomes isolated from the serum of rectal cancer patients with different responses to neo-RT. This allowed revealing several proteins and metabolites associated with common pathways relevant for the response of rectal cancer patients to neo-RT, including immune system response, complement activation cascade, platelet functions, metabolism of lipids, metabolism of glucose, and cancer-related signaling pathways. Moreover, the composition of serum-derived exosomes and a whole serum was analyzed in parallel to compare the biomarker potential of both specimens. Among proteins that the most properly discriminated good and poor responders were GPLD1 (AUC = 0.85, accuracy of 74%) identified in plasma as well as C8G (AUC = 0.91, accuracy 81%), SERPINF2 (AUC = 0.91, accuracy 79%) and CFHR3 (AUC = 0.90, accuracy 81%) identified in exosomes. We found that the proteome component of serum-derived exosomes has the highest capacity to discriminate samples of patients with different responses to neo-RT when compared to the whole plasma proteome and metabolome. We concluded that the molecular components of exosomes are associated with the response of rectal cancer patients to neo-RT and could be used for the prediction of such response.
Collapse
Affiliation(s)
- Urszula Strybel
- Department of Biomedical Proteomics, Institute of Bioorganic Chemistry Polish Academy of Sciences, 61-704 Poznan, Poland; (U.S.); (L.M.); (A.S.-C.); (P.J.)
| | - Lukasz Marczak
- Department of Biomedical Proteomics, Institute of Bioorganic Chemistry Polish Academy of Sciences, 61-704 Poznan, Poland; (U.S.); (L.M.); (A.S.-C.); (P.J.)
| | - Marcin Zeman
- Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland; (M.Z.); (M.S.); (M.C.); (E.Z.-N.); (M.P.)
| | - Krzysztof Polanski
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK;
| | - Łukasz Mielańczyk
- Department of Histology and Cell Pathology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland; (Ł.M.); (O.K.)
| | - Olesya Klymenko
- Department of Histology and Cell Pathology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland; (Ł.M.); (O.K.)
| | - Anna Samelak-Czajka
- Department of Biomedical Proteomics, Institute of Bioorganic Chemistry Polish Academy of Sciences, 61-704 Poznan, Poland; (U.S.); (L.M.); (A.S.-C.); (P.J.)
| | - Paulina Jackowiak
- Department of Biomedical Proteomics, Institute of Bioorganic Chemistry Polish Academy of Sciences, 61-704 Poznan, Poland; (U.S.); (L.M.); (A.S.-C.); (P.J.)
| | - Mateusz Smolarz
- Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland; (M.Z.); (M.S.); (M.C.); (E.Z.-N.); (M.P.)
| | - Mykola Chekan
- Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland; (M.Z.); (M.S.); (M.C.); (E.Z.-N.); (M.P.)
| | - Ewa Zembala-Nożyńska
- Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland; (M.Z.); (M.S.); (M.C.); (E.Z.-N.); (M.P.)
| | - Piotr Widlak
- Clinical Research Support Centre, Medical University of Gdańsk, 80-210 Gdańsk, Poland;
| | - Monika Pietrowska
- Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland; (M.Z.); (M.S.); (M.C.); (E.Z.-N.); (M.P.)
| | - Anna Wojakowska
- Department of Biomedical Proteomics, Institute of Bioorganic Chemistry Polish Academy of Sciences, 61-704 Poznan, Poland; (U.S.); (L.M.); (A.S.-C.); (P.J.)
| |
Collapse
|
15
|
Read GH, Bailleul J, Vlashi E, Kesarwala AH. Metabolic response to radiation therapy in cancer. Mol Carcinog 2022; 61:200-224. [PMID: 34961986 PMCID: PMC10187995 DOI: 10.1002/mc.23379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 12/01/2021] [Accepted: 12/01/2021] [Indexed: 11/11/2022]
Abstract
Tumor metabolism has emerged as a hallmark of cancer and is involved in carcinogenesis and tumor growth. Reprogramming of tumor metabolism is necessary for cancer cells to sustain high proliferation rates and enhanced demands for nutrients. Recent studies suggest that metabolic plasticity in cancer cells can decrease the efficacy of anticancer therapies by enhancing antioxidant defenses and DNA repair mechanisms. Studying radiation-induced metabolic changes will lead to a better understanding of radiation response mechanisms as well as the identification of new therapeutic targets, but there are few robust studies characterizing the metabolic changes induced by radiation therapy in cancer. In this review, we will highlight studies that provide information on the metabolic changes induced by radiation and oxidative stress in cancer cells and the associated underlying mechanisms.
Collapse
Affiliation(s)
- Graham H. Read
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Justine Bailleul
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Erina Vlashi
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California
| | - Aparna H. Kesarwala
- Department of Radiation Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
16
|
Abstract
Biological dosimetry is an internationally recognized method for quantifying and estimating radiation dose following suspected or verified excessive exposure to ionising radiation. In severe radiation accidents where a large number of people are potentially affected, it is possible to distinguish irradiated from non-irradiated people in order to initiate appropriate medical care if necessary. In addition to severe incidents caused by technical failure, environmental disasters, military actions, or criminal abuse, there are also radiation accidents in which only one or a few individuals are affected in the frame of occupational or medical exposure. The requirements for biological dosimetry are fundamentally different for these two scenarios. In particular, for large-scale radiation accidents, pre-screening methods are necessary to increase the throughput of samples for a rough first-dose categorization. The rapid development and increasing use of omics methods in research as well as in individual applications provides new opportunities for biological dosimetry. In addition to the discovery and search for new biomarkers, dosimetry assays based on omics technologies are becoming increasingly interesting and hold great potential, especially for large-scale dosimetry. In the following review, the different areas of biological dosimetry, the problems in finding suitable biomarkers, the current status of biomarker research based on omics, the potential applications of assays using omics technologies, and also the limitations for the different areas of biological dosimetry are discussed.
Collapse
|
17
|
Maan K, Baghel R, Bakhshi R, Dhariwal S, Tyagi R, Rana P. An integrative chemometric approach and correlative metabolite networking of LC-MS and 1H NMR based urine metabolomics for radiation signatures. Mol Omics 2022; 18:214-225. [PMID: 34982087 DOI: 10.1039/d1mo00399b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The increasing threat of nuclear terrorism or radiological accident has made high throughput radiation biodosimetry a requisite for the immediate response for triage. Owing to detection of subtle alterations in biological pathways before the onset of clinical conditions, metabolomics has become an important tool for studying biomarkers and the related mechanisms for radiation induced damage. Here, we have attempted to combine two detection techniques, LC-MS and 1H NMR spectroscopy, to obtain a comprehensive metabolite profile of urine at 24 h following lethal (7.5 Gy) and sub-lethal (5 Gy) irradiation in mice. Integrated data analytics using multiblock-OPLSDA (MB-OPLSDA), correlation networking and pathway analysis was used to identify metabolic disturbances associated with radiation exposure. MB-OPLSDA revealed better clustering and separation of irradiated groups compared with controls without overfitting (p-value of CV-ANOVA: 1.5 × 10-3). Metabolites identified through MB-OPLSDA, namely, taurine, creatine, citrate and 2-oxoglutarate, were found to be dose independent markers and further support and validate our earlier findings as potential radiation injury biomarkers. Integrated analysis resulted in the enhanced coverage of metabolites and better correlation networking in energy, taurine, gut flora, L-carnitine and nucleotide metabolism observed post irradiation in urine. Our study thus emphasizes the major advantage of using the two detection techniques along with integrated analysis for better detection and comprehensive understanding of disturbed metabolites in biological pathways.
Collapse
Affiliation(s)
- Kiran Maan
- Metabolomics Research Facility, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, S. K Mazumdar Road, Timarpur, Delhi-54, India. .,Department of Biomedical Sciences, Shaheed Rajguru College of Applied Sciences for Women, University of Delhi, Delhi, India
| | - Ruchi Baghel
- Metabolomics Research Facility, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, S. K Mazumdar Road, Timarpur, Delhi-54, India.
| | - Radhika Bakhshi
- Department of Biomedical Sciences, Shaheed Rajguru College of Applied Sciences for Women, University of Delhi, Delhi, India
| | - Seema Dhariwal
- Metabolomics Research Facility, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, S. K Mazumdar Road, Timarpur, Delhi-54, India.
| | - Ritu Tyagi
- Metabolomics Research Facility, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, S. K Mazumdar Road, Timarpur, Delhi-54, India.
| | - Poonam Rana
- Metabolomics Research Facility, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, S. K Mazumdar Road, Timarpur, Delhi-54, India.
| |
Collapse
|
18
|
Liu HX, Lu X, Zhao H, Li S, Gao L, Tian M, Liu QJ. Enhancement of Acylcarnitine Levels in Small Intestine of Abdominal Irradiation Rats Might Relate to Fatty Acid β-Oxidation Pathway Disequilibration. Dose Response 2022; 20:15593258221075118. [PMID: 35221822 PMCID: PMC8874182 DOI: 10.1177/15593258221075118] [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: 12/07/2021] [Accepted: 12/24/2021] [Indexed: 11/21/2022]
Abstract
Objective This study aims to analyze the alteration of carnitine profile in the small intestine of abdominal irradiation-induced intestinal injury rats and explore the possible reason for the altered carnitine profile. Methods The abdomens of 15 male Sprague Dawley (SD) rats were irradiated with 0, 10, and 15 Gy of 60Co gamma rays. The carnitine profile in the small intestine and plasma samples of SD rats at 72 h after abdominal irradiated with 0 Gy or 10 Gy of 60Co gamma rays were measured by targeted metabolomics. The changes of fatty acid β-oxidation (FAO), including the expression of carnitine palmitoyltransferase 1 (CPT1) and acyl-CoA dehydrogenases, were analyzed in the small intestine samples of SD rats after exposed to 0, 10, and 15 Gy groups. Results There were eleven acylcarnitines in the small intestine and fourteen acylcarnitines in the plasma of the rat model significantly enhanced, respectively (P < .05). The expression level and activity of CPT1 in the small intestine were remarkably increased (P < .05), and the activity of acyl-CoA dehydrogenase in the small intestine was noticeably reduced (P < .01) after abdominal irradiation. Conclusion The enhanced acylcarnitine levels in the small intestine of abdominal irradiation rats might relate to the FAO pathway disequilibration.
Collapse
Affiliation(s)
- Hai-Xiang Liu
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xue Lu
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Hua Zhao
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Shuang Li
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Ling Gao
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Mei Tian
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Qing-Jie Liu
- China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
19
|
Vinnikov V, Belyakov O. Clinical Applications of Biological Dosimetry in Patients Exposed to Low Dose Radiation Due to Radiological, Imaging or Nuclear Medicine Procedures. Semin Nucl Med 2021; 52:114-139. [PMID: 34879905 DOI: 10.1053/j.semnuclmed.2021.11.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Radiation dosimetric biomarkers have found applications beyond radiation protection area and now are actively introduced into clinical practice. Cytogenetic assays appeared to be a valuable tool for individualized quantifying radiation effects in patients, with high capability for assessing genotoxicity of various medical exposure modalities and providing meaningful radiation dose estimates for prognoses of radiation-related cancer risk. This review summarized current data on the use of biological dosimetry methods in patients undergoing various medical irradiations to low doses. The highlighted topics include basic aspects of biological dosimetry and its limitations in the range of low radiation doses, and main patterns of in vivo induction of radiation biomarkers in clinical exposure scenarios, occurring in X-ray diagnostics, computed tomography, interventional radiology, low dose radiotherapy, and nuclear medicine (internally administered 131I and other radiopharmaceuticals). Additionally, several specific issues, examined by biodosimetry techniques, are analysed, such as contrast media effect, radiation response in pediatric patients, impact of magnetic resonance imaging, evaluation of radioprotectors, detection of patients' abnormal intrinsic radiosensitivity and dose estimation in persons involved in medical radiation incidents. A prognosis of possible directions for further improvements in this area includes the automation of cytogenetic analysis, introduction of molecular biodosimeters and development of multiparametric biodosimetry platforms. A potential approach to the advanced biodosimetry of internal exposure and/or low dose external irradiation is suggested; this can be a multiparametric platform based on the combination of the γ-H2AX foci, dicentric, and translocation assays, each applied in the optimum postexposure time range, with the amalgamation of the dose estimates. The study revealed the necessity of further research, which might clarify medical radiation safety concerns for patients via using stringent biodosimetry methodology.
Collapse
Affiliation(s)
- Volodymyr Vinnikov
- International Atomic Energy Agency (IAEA), Vienna, Austria; Grigoriev Institute for Medical Radiology and Oncology (GIMRO), Kharkiv, Ukraine.
| | - Oleg Belyakov
- International Atomic Energy Agency (IAEA), Vienna, Austria
| |
Collapse
|
20
|
Wang W, Wang Y, Chen F, Zheng F. Comparison of determination of sugar-PMP derivatives by two different stationary phases and two HPLC detectors: C18 vs. amide columns and DAD vs. ELSD. J Food Compost Anal 2021. [DOI: 10.1016/j.jfca.2020.103715] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
21
|
Di Gregorio E, Miolo G, Saorin A, Muraro E, Cangemi M, Revelant A, Minatel E, Trovò M, Steffan A, Corona G. Radical Hemithoracic Radiotherapy Induces Systemic Metabolomics Changes That Are Associated with the Clinical Outcome of Malignant Pleural Mesothelioma Patients. Cancers (Basel) 2021; 13:cancers13030508. [PMID: 33572739 PMCID: PMC7866164 DOI: 10.3390/cancers13030508] [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: 12/23/2020] [Revised: 01/19/2021] [Accepted: 01/25/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Radical hemithoracic radiotherapy represents a promising new advance in the field of radiation oncology and encouraging results have been achieved in the treatment of malignant pleural mesothelioma patients. This study showed that this radiotherapy modality produces significant changes in serum metabolomics profile mainly affecting arginine and polyamine biosynthesis pathways. Interestingly, individual metabolomics alterations were found associated with the clinical overall survival outcome of the radiotherapy treatment. These results highlight metabolomics profile analysis as a powerful prognostic tool useful to better understand the mechanisms underlying the interpatients variability and to identify patients who may receive the best benefit from this specific radiotherapy treatment. Abstract Radical hemithoracic radiotherapy (RHRT) represents an advanced therapeutic option able to improve overall survival of malignant pleural mesothelioma patients. This study aims to investigate the systemic effects of this radiotherapy modality on the serum metabolome and their potential implications in determining the individual clinical outcome. Nineteen patients undergoing RHRT at the dose of 50 Gy in 25 fractions were enrolled. Serum targeted metabolomics profiles were investigated at baseline and the end of radiotherapy by liquid chromatography and tandem mass spectrometry. Univariate and multivariate OPLS-DA analyses were applied to study the serum metabolomics changes induced by RHRT while PLS regression analysis to evaluate the association between such changes and overall survival. RHRT was found to affect almost all investigated metabolites classes, in particular, the amino acids citrulline and taurine, the C14, C18:1 and C18:2 acyl-carnitines as well as the unsaturated long chain phosphatidylcholines PC ae 42:5, PC ae 44:5 and PC ae 44:6 were significantly decreased. The enrichment analysis showed arginine metabolism and the polyamine biosynthesis as the most perturbed pathways. Moreover, specific metabolic changes encompassing the amino acids and acyl-carnitines resulted in association with the clinical outcome accounting for about 60% of the interpatients overall survival variability. This study highlighted that RHRT can induce profound systemic metabolic effects some of which may have a significant prognostic value. The integration of metabolomics in the clinical assessment of the malignant pleural mesothelioma could be useful to better identify the patients who can achieve the best benefit from the RHRT treatment.
Collapse
Affiliation(s)
- Emanuela Di Gregorio
- Immunopathology and Cancer Biomarkers Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (E.D.G.); (A.S.); (E.M.); (M.C.); (A.S.)
| | - Gianmaria Miolo
- Medical Oncology and Cancer Prevention Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy;
| | - Asia Saorin
- Immunopathology and Cancer Biomarkers Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (E.D.G.); (A.S.); (E.M.); (M.C.); (A.S.)
| | - Elena Muraro
- Immunopathology and Cancer Biomarkers Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (E.D.G.); (A.S.); (E.M.); (M.C.); (A.S.)
| | - Michela Cangemi
- Immunopathology and Cancer Biomarkers Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (E.D.G.); (A.S.); (E.M.); (M.C.); (A.S.)
| | - Alberto Revelant
- Radiation Oncology Department, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (A.R.); (E.M.)
| | - Emilio Minatel
- Radiation Oncology Department, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (A.R.); (E.M.)
| | - Marco Trovò
- Radiation Oncology Department, Azienda Sanitaria Integrata, 33100 Udine, Italy;
| | - Agostino Steffan
- Immunopathology and Cancer Biomarkers Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (E.D.G.); (A.S.); (E.M.); (M.C.); (A.S.)
| | - Giuseppe Corona
- Immunopathology and Cancer Biomarkers Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (E.D.G.); (A.S.); (E.M.); (M.C.); (A.S.)
- Correspondence: ; Tel.: +39-0434-659-666
| |
Collapse
|
22
|
Obrador E, Salvador R, Villaescusa JI, Soriano JM, Estrela JM, Montoro A. Radioprotection and Radiomitigation: From the Bench to Clinical Practice. Biomedicines 2020; 8:E461. [PMID: 33142986 PMCID: PMC7692399 DOI: 10.3390/biomedicines8110461] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/27/2020] [Accepted: 10/29/2020] [Indexed: 02/07/2023] Open
Abstract
The development of protective agents against harmful radiations has been a subject of investigation for decades. However, effective (ideal) radioprotectors and radiomitigators remain an unsolved problem. Because ionizing radiation-induced cellular damage is primarily attributed to free radicals, radical scavengers are promising as potential radioprotectors. Early development of such agents focused on thiol synthetic compounds, e.g., amifostine (2-(3-aminopropylamino) ethylsulfanylphosphonic acid), approved as a radioprotector by the Food and Drug Administration (FDA, USA) but for limited clinical indications and not for nonclinical uses. To date, no new chemical entity has been approved by the FDA as a radiation countermeasure for acute radiation syndrome (ARS). All FDA-approved radiation countermeasures (filgrastim, a recombinant DNA form of the naturally occurring granulocyte colony-stimulating factor, G-CSF; pegfilgrastim, a PEGylated form of the recombinant human G-CSF; sargramostim, a recombinant granulocyte macrophage colony-stimulating factor, GM-CSF) are classified as radiomitigators. No radioprotector that can be administered prior to exposure has been approved for ARS. This differentiates radioprotectors (reduce direct damage caused by radiation) and radiomitigators (minimize toxicity even after radiation has been delivered). Molecules under development with the aim of reaching clinical practice and other nonclinical applications are discussed. Assays to evaluate the biological effects of ionizing radiations are also analyzed.
Collapse
Affiliation(s)
- Elena Obrador
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (E.O.); (R.S.); (J.M.E.)
| | - Rosario Salvador
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (E.O.); (R.S.); (J.M.E.)
| | - Juan I. Villaescusa
- Service of Radiological Protection, Clinical Area of Medical Image, La Fe University Hospital, 46026 Valencia, Spain;
- Biomedical Imaging Research Group GIBI230, Health Research Institute (IISLaFe), La Fe University Hospital, 46026 Valencia, Spain
| | - José M. Soriano
- Food & Health Lab, Institute of Materials Science, University of Valencia, 46980 Valencia, Spain;
- Joint Research Unit in Endocrinology, Nutrition and Clinical Dietetics, University of Valencia-Health Research Institute IISLaFe, 46026 Valencia, Spain
| | - José M. Estrela
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (E.O.); (R.S.); (J.M.E.)
| | - Alegría Montoro
- Service of Radiological Protection, Clinical Area of Medical Image, La Fe University Hospital, 46026 Valencia, Spain;
- Biomedical Imaging Research Group GIBI230, Health Research Institute (IISLaFe), La Fe University Hospital, 46026 Valencia, Spain
| |
Collapse
|
23
|
Satyamitra MM, Cassatt DR, Hollingsworth BA, Price PW, Rios CI, Taliaferro LP, Winters TA, DiCarlo AL. Metabolomics in Radiation Biodosimetry: Current Approaches and Advances. Metabolites 2020; 10:metabo10080328. [PMID: 32796693 PMCID: PMC7465152 DOI: 10.3390/metabo10080328] [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: 07/01/2020] [Revised: 08/01/2020] [Accepted: 08/06/2020] [Indexed: 12/11/2022] Open
Abstract
Triage and medical intervention strategies for unanticipated exposure during a radiation incident benefit from the early, rapid and accurate assessment of dose level. Radiation exposure results in complex and persistent molecular and cellular responses that ultimately alter the levels of many biological markers, including the metabolomic phenotype. Metabolomics is an emerging field that promises the determination of radiation exposure by the qualitative and quantitative measurements of small molecules in a biological sample. This review highlights the current role of metabolomics in assessing radiation injury, as well as considerations for the diverse range of bioanalytical and sampling technologies that are being used to detect these changes. The authors also address the influence of the physiological status of an individual, the animal models studied, the technology and analysis employed in interrogating response to the radiation insult, and variables that factor into discovery and development of robust biomarker signatures. Furthermore, available databases for these studies have been reviewed, and existing regulatory guidance for metabolomics are discussed, with the ultimate goal of providing both context for this area of radiation research and the consideration of pathways for continued development.
Collapse
Affiliation(s)
- Merriline M. Satyamitra
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), and National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), 5601 Fishers Lane, Rockville, MD 20852, USA; (D.R.C.); (B.A.H.); (C.I.R.); (L.P.T.); (T.A.W.); (A.L.D.)
- Correspondence: ; Tel.: +1-240-669-5432
| | - David R. Cassatt
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), and National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), 5601 Fishers Lane, Rockville, MD 20852, USA; (D.R.C.); (B.A.H.); (C.I.R.); (L.P.T.); (T.A.W.); (A.L.D.)
| | - Brynn A. Hollingsworth
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), and National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), 5601 Fishers Lane, Rockville, MD 20852, USA; (D.R.C.); (B.A.H.); (C.I.R.); (L.P.T.); (T.A.W.); (A.L.D.)
| | - Paul W. Price
- Office of Regulatory Affairs, Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), 5601 Fishers Lane, Rockville, MD 20852, USA;
| | - Carmen I. Rios
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), and National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), 5601 Fishers Lane, Rockville, MD 20852, USA; (D.R.C.); (B.A.H.); (C.I.R.); (L.P.T.); (T.A.W.); (A.L.D.)
| | - Lanyn P. Taliaferro
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), and National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), 5601 Fishers Lane, Rockville, MD 20852, USA; (D.R.C.); (B.A.H.); (C.I.R.); (L.P.T.); (T.A.W.); (A.L.D.)
| | - Thomas A. Winters
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), and National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), 5601 Fishers Lane, Rockville, MD 20852, USA; (D.R.C.); (B.A.H.); (C.I.R.); (L.P.T.); (T.A.W.); (A.L.D.)
| | - Andrea L. DiCarlo
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), and National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), 5601 Fishers Lane, Rockville, MD 20852, USA; (D.R.C.); (B.A.H.); (C.I.R.); (L.P.T.); (T.A.W.); (A.L.D.)
| |
Collapse
|
24
|
Vicente E, Vujaskovic Z, Jackson IL. A Systematic Review of Metabolomic and Lipidomic Candidates for Biomarkers in Radiation Injury. Metabolites 2020; 10:E259. [PMID: 32575772 PMCID: PMC7344731 DOI: 10.3390/metabo10060259] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/09/2020] [Accepted: 06/13/2020] [Indexed: 12/16/2022] Open
Abstract
A large-scale nuclear event has the ability to inflict mass casualties requiring point-of-care and laboratory-based diagnostic and prognostic biomarkers to inform victim triage and appropriate medical intervention. Extensive progress has been made to develop post-exposure point-of-care biodosimetry assays and to identify biomarkers that may be used in early phase testing to predict the course of the disease. Screening for biomarkers has recently extended to identify specific metabolomic and lipidomic responses to radiation using animal models. The objective of this review was to determine which metabolites or lipids most frequently experienced perturbations post-ionizing irradiation (IR) in preclinical studies using animal models of acute radiation sickness (ARS) and delayed effects of acute radiation exposure (DEARE). Upon review of approximately 65 manuscripts published in the peer-reviewed literature, the most frequently referenced metabolites showing clear changes in IR induced injury were found to be citrulline, citric acid, creatine, taurine, carnitine, xanthine, creatinine, hypoxanthine, uric acid, and threonine. Each metabolite was evaluated by specific study parameters to determine whether trends were in agreement across several studies. A select few show agreement across variable animal models, IR doses and timepoints, indicating that they may be ubiquitous and appropriate for use in diagnostic or prognostic biomarker panels.
Collapse
Affiliation(s)
| | | | - Isabel L. Jackson
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (E.V.); (Z.V.)
| |
Collapse
|
25
|
Mak TD, Goudarzi M, Laiakis EC, Stein SE. Disparate Metabolomics Data Reassembler: A Novel Algorithm for Agglomerating Incongruent LC-MS Metabolomics Datasets. Anal Chem 2020; 92:5231-5239. [PMID: 32118408 PMCID: PMC10926180 DOI: 10.1021/acs.analchem.9b05763] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In the past decade, the field of LC-MS-based metabolomics has transformed from an obscure specialty into a major "-omics" platform for studying metabolic processes and biomolecular characterization. However, as a whole the field is still very fractured, as the nature of the instrumentation and the information produced by the platform essentially creates incompatible "islands" of datasets. This lack of data coherency results in the inability to accumulate a critical mass of metabolomics data that has enabled other -omics platforms to make impactful discoveries and meaningful advances. As such, we have developed a novel algorithm, called Disparate Metabolomics Data Reassembler (DIMEDR), which attempts to bridge the inconsistencies between incongruent LC-MS metabolomics datasets of the same biological sample type. A single "primary" dataset is postprocessed via traditional means of peak identification, alignment, and grouping. DIMEDR utilizes this primary dataset as a progenitor template by which data from subsequent disparate datasets are reassembled and integrated into a unified framework that maximizes spectral feature similarity across all samples. This is accomplished by a novel procedure for universal retention time correction and comparison via identification of ubiquitous features in the initial primary dataset, which are subsequently utilized as endogenous internal standards during integration. For demonstration purposes, two human and two mouse urine metabolomics datasets from four unrelated studies acquired over 4 years were unified via DIMEDR, which enabled meaningful analysis across otherwise incomparable and unrelated datasets.
Collapse
Affiliation(s)
- Tytus D. Mak
- Mass Spectrometry Data Center, National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, MD 20899-8632
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, New Research Building E504/508, 3970 Reservoir Rd, NW, Washington, DC 20057
| | - Maryam Goudarzi
- Department of Cellular & Molecular Medicine, Cleveland Clinic Lerner Research Institute, Building NN1, Room 28, 9500 Euclid Ave, Cleveland, OH 44195
| | - Evagelia C. Laiakis
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, New Research Building E504/508, 3970 Reservoir Rd, NW, Washington, DC 20057
| | - Stephen E. Stein
- Mass Spectrometry Data Center, National Institute of Standards and Technology, 100 Bureau Drive, Gaithersburg, MD 20899-8632
| |
Collapse
|
26
|
Vera NB, Coy SL, Laiakis EC, Fornace AJ, Clasquin M, Barker CA, Pfefferkorn JA, Vouros P. Quantitation of Urinary Acylcarnitines by DMS-MS/MS Uncovers the Effects of Total Body Irradiation in Cancer Patients. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2020; 31:498-507. [PMID: 32013416 PMCID: PMC7489307 DOI: 10.1021/jasms.9b00076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Acylcarnitines have been identified in human and animal metabolomic-profiling studies as urinary markers of radiation exposure, a result which is consistent with their cytoprotective effects and roles in energy metabolism. In the present work, a rapid method for quantitation of the more abundant acylcarnitines in human urine is developed using a valuable set of samples from cancer patients who received total body irradiation (TBI) at Memorial Sloan Kettering Cancer Center. The method uses solid-phase extraction (SPE) processing followed by differential mobility spectrometry (DMS with ethyl acetate modifier) tandem mass spectrometry (ESI-DMS-MS/MS) with deuterated internal standards. The analyzed human urine samples were collected from 38 individual patients at three time points over 24 h during and after the course of radiation treatment, a design allowing each patient to act as their own control and creatinine normalization. Creatinine-normalized concentrations for nine urinary acylcarnitine (acyl-CN) species are reported. Six acyl-CN species were reduced at the 6 h point. Acetylcarnitine (C2:0-CN) and valerylcarnitine (C5:0-CN) showed recovery at 24 h, but none of the other acyl-CN species showed recovery at that point. Levels of three acyl-CN species were not significantly altered by radiation. This rapid quantitative method for clinical samples covers the short- and medium-chain acylcarnitines and has the flexibility to be expanded to cover additional radiation-linked metabolites. The human data presented here indicates the utility of the current approach as a rapid, quantitative technique with potential applications by the medical community, by space research laboratories concerned with radiation exposure, and by disaster response groups.
Collapse
Affiliation(s)
- Nicholas B. Vera
- Pfizer Global Research and Development, Cambridge Laboratories, Pfizer Inc., Cambridge, Massachusetts 02139 United States
- Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115 United States
| | - Stephen L. Coy
- Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115 United States
| | - Evagelia C. Laiakis
- Department of Oncology, Georgetown University, 3700 O Street NW, Washington, D.C. 20057 United States
- Department of Biochemistry and Molecular & Cellular Oncology, Georgetown University, 3700 O Street NW, Washington D.C. 20057 United States
| | - Albert J. Fornace
- Department of Oncology, Georgetown University, 3700 O Street NW, Washington, D.C. 20057 United States
- Department of Biochemistry and Molecular & Cellular Oncology, Georgetown University, 3700 O Street NW, Washington D.C. 20057 United States
| | - Michelle Clasquin
- Pfizer Global Research and Development, Cambridge Laboratories, Pfizer Inc., Cambridge, Massachusetts 02139 United States
| | - Christopher A. Barker
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, United States
| | - Jeffrey A. Pfefferkorn
- Pfizer Global Research and Development, Cambridge Laboratories, Pfizer Inc., Cambridge, Massachusetts 02139 United States
| | - Paul Vouros
- Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115 United States
| |
Collapse
|
27
|
Shuryak I, Turner HC, Perrier JR, Cunha L, Canadell MP, Durrani MH, Harken A, Bertucci A, Taveras M, Garty G, Brenner DJ. A High Throughput Approach to Reconstruct Partial-Body and Neutron Radiation Exposures on an Individual Basis. Sci Rep 2020; 10:2899. [PMID: 32076014 PMCID: PMC7031285 DOI: 10.1038/s41598-020-59695-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 01/27/2020] [Indexed: 11/28/2022] Open
Abstract
Biodosimetry-based individualized reconstruction of complex irradiation scenarios (partial-body shielding and/or neutron + photon mixtures) can improve treatment decisions after mass-casualty radiation-related incidents. We used a high-throughput micronucleus assay with automated scanning and imaging software on ex-vivo irradiated human lymphocytes to: a) reconstruct partial-body and/or neutron exposure, and b) estimate separately the photon and neutron doses in a mixed exposure. The mechanistic background is that, compared with total-body photon irradiations, neutrons produce more heavily-damaged lymphocytes with multiple micronuclei/binucleated cell, whereas partial-body exposures produce fewer such lymphocytes. To utilize these differences for biodosimetry, we developed metrics that describe micronuclei distributions in binucleated cells and serve as predictors in machine learning or parametric analyses of the following scenarios: (A) Homogeneous gamma-irradiation, mimicking total-body exposures, vs. mixtures of irradiated blood with unirradiated blood, mimicking partial-body exposures. (B) X rays vs. various neutron + photon mixtures. The results showed high accuracies of scenario and dose reconstructions. Specifically, receiver operating characteristic curve areas (AUC) for sample classification by exposure type reached 0.931 and 0.916 in scenarios A and B, respectively. R2 for actual vs. reconstructed doses in these scenarios reached 0.87 and 0.77, respectively. These encouraging findings demonstrate a proof-of-principle for the proposed approach of high-throughput reconstruction of clinically-relevant complex radiation exposure scenarios.
Collapse
Affiliation(s)
- Igor Shuryak
- Center for Radiological Research, Columbia University Irving Medical Center, New York, NY, USA.
| | - Helen C Turner
- Center for Radiological Research, Columbia University Irving Medical Center, New York, NY, USA
| | - Jay R Perrier
- Center for Radiological Research, Columbia University Irving Medical Center, New York, NY, USA
| | - Lydia Cunha
- Center for Radiological Research, Columbia University Irving Medical Center, New York, NY, USA
| | - Monica Pujol Canadell
- Center for Radiological Research, Columbia University Irving Medical Center, New York, NY, USA
| | - Mohammad H Durrani
- Center for Radiological Research, Columbia University Irving Medical Center, New York, NY, USA
| | - Andrew Harken
- Center for Radiological Research, Columbia University Irving Medical Center, New York, NY, USA
| | - Antonella Bertucci
- Center for Radiological Research, Columbia University Irving Medical Center, New York, NY, USA
| | - Maria Taveras
- Center for Radiological Research, Columbia University Irving Medical Center, New York, NY, USA
| | - Guy Garty
- Center for Radiological Research, Columbia University Irving Medical Center, New York, NY, USA
| | - David J Brenner
- Center for Radiological Research, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
28
|
Systemic Effects of Radiotherapy and Concurrent Chemo-Radiotherapy in Head and Neck Cancer Patients-Comparison of Serum Metabolome Profiles. Metabolites 2020; 10:metabo10020060. [PMID: 32046123 PMCID: PMC7074210 DOI: 10.3390/metabo10020060] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/01/2020] [Accepted: 02/05/2020] [Indexed: 12/22/2022] Open
Abstract
Anticancer treatment induces systemic molecular changes that could be detected at the level of biofluids. Understanding how human metabolism is influenced by these treatments is crucial to predict the individual response and adjust personalized therapies. Here, we aimed to compare profiles of metabolites in serum of head and neck cancer patients treated with concurrent chemo-radiotherapy, radiotherapy alone, or induction chemotherapy. Serum samples were analyzed by a targeted quantitative approach using combined direct flow injection and liquid chromatography coupled to tandem mass spectrometry, which allowed simultaneous quantification of 149 metabolites. There were 45 metabolites whose levels were significantly changed between pretreatment and within- or post-treatment serum samples, including 38 phospholipids. Concurrent chemo-radiotherapy induced faster and stronger effects than radiotherapy alone. On the other hand, chemotherapy alone did not result in significant changes. The decreased level of total phospholipids was the most apparent effect observed during the first step of the treatment. This corresponded to the loss of patients’ body mass, yet no correlation between both parameters was observed for individual patients. We concluded that different molecular changes were measured at the level of serum metabolome in response to different treatment modalities.
Collapse
|
29
|
Kultova G, Tichy A, Rehulkova H, Myslivcova-Fucikova A. The hunt for radiation biomarkers: current situation. Int J Radiat Biol 2020; 96:370-382. [PMID: 31829779 DOI: 10.1080/09553002.2020.1704909] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Purpose: The possibility of a large-scale acute radiation exposure necessitates the development of new methods that could provide a rapid assessment of the doses received by individuals using high-throughput technologies. There is also a great interest in developing new biomarkers of dose exposure, which could be used in large molecular epidemiological studies in order to correlate estimated doses received and health effects. The goal of this review was to summarize current literature focused on biological dosimetry, namely radiation-responsive biomarkers.Methods: The studies involved in this review were thoroughly selected according to the determined criteria and PRISMA guidelines.Results: We described briefly recent advances in radiation genomics and metabolomics, giving particular emphasis to proteomic analysis. The majority of studies were performed on animal models (rats, mice, and non-human primates). They have provided much beneficial information, but the most relevant tests have been done on human (oncological) patients. By inspecting the radiaiton biodosimetry literate of the last 10 years, we identified a panel of candidate markers for each -omic approach involved.Conslusions: We reviewed different methodological approaches and various biological materials, which can be exploited for dose-effect prediction. The protein biomarkers from human plasma are ideal for this specific purpose. From a plethora of candidate markers, FDXR is a very promising transcriptomic candidate, and importantly this biomarker was also confirmed by some studies at protein level in humans.
Collapse
Affiliation(s)
- Gabriela Kultova
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Czech Republic.,Department of Biology, Faculty of Science, University of Hradec Králové, Hradec Kralove, Czech Republic
| | - Ales Tichy
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Czech Republic
| | - Helena Rehulkova
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Czech Republic
| | - Alena Myslivcova-Fucikova
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Czech Republic
| |
Collapse
|
30
|
Gao Y, Li X, Gao J, Zhang Z, Feng Y, Nie J, Zhu W, Zhang S, Cao J. Metabolomic Analysis of Radiation-Induced Lung Injury in Rats: The Potential Radioprotective Role of Taurine. Dose Response 2019; 17:1559325819883479. [PMID: 31700502 PMCID: PMC6823985 DOI: 10.1177/1559325819883479] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/16/2019] [Accepted: 09/24/2019] [Indexed: 12/17/2022] Open
Abstract
Radiation-induced lung injury is a major dose-limiting toxicity that occurs due to thoracic radiotherapy. Metabolomics is a powerful quantitative measurement of low-molecular-weight metabolites in response to environmental disturbances. However, the metabolomic profiles of radiation-induced lung injury have not been reported yet. In this study, male Sprague-Dawley rats were subjected to a single dose of 10 or 20 Gy irradiation to the right lung. One week after radiation, the obvious morphological alteration of lung tissues after radiation was observed by hematoxylin and eosin staining through a transmission electron microscope. We then analyzed the metabolites and related pathways of radiation-induced lung injury by gas chromatography-mass spectrometry, and a total of 453 metabolites were identified. Compared to the nonirradiated left lung, 19 metabolites (8 upregulated and 11 downregulated) showed a significant difference in 10 Gy irradiated lung tissues, including mucic acid, methyl-β-d-galactopyranoside, quinoline-4-carboxylic acid, and pyridoxine. There were 31 differential metabolites (16 upregulated and 15 downregulated) between 20 Gy irradiated and nonirradiated lung tissues, including taurine, piperine, 1,2,4-benzenetriol, and lactamide. The Kyoto Encyclopedia of Genes and Genomes-based pathway analysis enriched 32 metabolic pathways between the irradiated and nonirradiated lung tissues, including pyrimidine metabolism, ATP-binding cassette transporters, aminoacyl-tRNA biosynthesis, and β-alanine metabolism. Among the dysregulated metabolites, we found that taurine promoted clonogenic survival and reduced radiation-induced necrosis in human embryonic lung fibroblast (HELF) cells. This study provides evidence indicating that radiation induces metabolic alterations of the lung. These findings significantly advance our understanding of the pathophysiology of radiation-induced lung injury from the perspective of metabolism.
Collapse
Affiliation(s)
- Yiying Gao
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China
- State Key Lab of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
- Sichuan Center for Disease Control and Prevention, Sichuan, China
| | - Xugang Li
- Anshan Cancer Hospital, Anshan, China
| | | | | | - Yang Feng
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China
- State Key Lab of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Jihua Nie
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China
- State Key Lab of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Wei Zhu
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China
- State Key Lab of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Shuyu Zhang
- State Key Lab of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
- The Second Affiliated Hospital of Chengdu Medical College (China National Nuclear Corporation 416 Hospital), Chengdu, China
| | - Jianping Cao
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China
- State Key Lab of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| |
Collapse
|
31
|
Lindell Jonsson E, Erngren I, Engskog M, Haglöf J, Arvidsson T, Hedeland M, Petterson C, Laurell G, Nestor M. Exploring Radiation Response in Two Head and Neck Squamous Carcinoma Cell Lines Through Metabolic Profiling. Front Oncol 2019; 9:825. [PMID: 31544064 PMCID: PMC6728927 DOI: 10.3389/fonc.2019.00825] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 08/12/2019] [Indexed: 12/27/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth most common form of cancer worldwide. Radiotherapy, with or without surgery, represents the major approach to curative treatment. However, not all tumors are equally sensitive to irradiation. It is therefore of interest to apply newer system biology approaches (e.g., metabolic profiling) in squamous cancer cells with different radiosensitivities in order to provide new insights on the mechanisms of radiation response. In this study, two cultured HNSCC cell lines from the same donor, UM-SCC-74A and UM-SCC-74B, were first genotyped using Short Tandem Repeat (STR), and assessed for radiation response by the means of clonogenic survival and growth inhibition assays. Thereafter, cells were cultured, irradiated and collected for subsequent metabolic profiling analyses using liquid chromatography-mass spectrometry (LC-MS). STR verified the similarity of UM-SCC-74A and UM-SCC-74B cells, and three independent assays proved UM-SCC-74B to be clearly more radioresistant than UM-SCC-74A. The LC-MS metabolic profiling demonstrated significant differences in the intracellular metabolome of the two cell lines before irradiation, as well as significant alterations after irradiation. The most important differences between the two cell lines before irradiation were connected to nicotinic acid and nicotinamide metabolism and purine metabolism. In the more radiosensitive UM-SCC-74A cells, the most significant alterations after irradiation were linked to tryptophan metabolism. In the more radioresistant UM-SCC-74B cells, the major alterations after irradiation were connected to nicotinic acid and nicotinamide metabolism, purine metabolism, the methionine cycle as well as the serine, and glycine metabolism. The data suggest that the more radioresistant cell line UM-SCC-74B altered the metabolism to control redox-status, manage DNA-repair, and change DNA methylation after irradiation. This provides new insights on the mechanisms of radiation response, which may aid future identification of biomarkers associated with radioresistance of cancer cells.
Collapse
Affiliation(s)
| | - Ida Erngren
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Mikael Engskog
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Jakob Haglöf
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Torbjörn Arvidsson
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden.,Medical Product Agency, Uppsala, Sweden
| | - Mikael Hedeland
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Curt Petterson
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Göran Laurell
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Marika Nestor
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
32
|
Simón-Manso Y, Marupaka R, Yan X, Liang Y, Telu KH, Mirokhin Y, Stein SE. Mass Spectrometry Fingerprints of Small-Molecule Metabolites in Biofluids: Building a Spectral Library of Recurrent Spectra for Urine Analysis. Anal Chem 2019; 91:12021-12029. [PMID: 31424920 DOI: 10.1021/acs.analchem.9b02977] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
A large fraction of ions observed in electrospray liquid chromatography-mass spectrometry (LC-ESI-MS) experiments of biological samples remain unidentified. One of the main reasons for this is that spectral libraries of pure compounds fail to account for the complexity of the metabolite profiling of complex materials. Recently, the NIST Mass Spectrometry Data Center has been developing a novel type of searchable mass spectral library that includes all recurrent unidentified spectra found in the sample profile. These libraries, in conjunction with the NIST tandem mass spectral library, allow analysts to explore most of the chemical space accessible to LC-MS analysis. In this work, we demonstrate how these libraries can provide a reliable fingerprint of the material by applying them to a variety of urine samples, including an extremely altered urine from cancer patients undergoing total body irradiation. The same workflow is applicable to any other biological fluid. The selected class of acylcarnitines is examined in detail, and derived libraries and related software are freely available. They are intended to serve as online resources for continuing community review and improvement.
Collapse
Affiliation(s)
- Yamil Simón-Manso
- Spectrometry Data Center, Biomolecular Measurement Division , National Institute of Standards and Technology (NIST) , Gaithersburg , Maryland 20899 , United States
| | - Ramesh Marupaka
- Spectrometry Data Center, Biomolecular Measurement Division , National Institute of Standards and Technology (NIST) , Gaithersburg , Maryland 20899 , United States
| | - Xinjian Yan
- Spectrometry Data Center, Biomolecular Measurement Division , National Institute of Standards and Technology (NIST) , Gaithersburg , Maryland 20899 , United States
| | - Yuxue Liang
- Spectrometry Data Center, Biomolecular Measurement Division , National Institute of Standards and Technology (NIST) , Gaithersburg , Maryland 20899 , United States
| | - Kelly H Telu
- Spectrometry Data Center, Biomolecular Measurement Division , National Institute of Standards and Technology (NIST) , Gaithersburg , Maryland 20899 , United States
| | - Yuri Mirokhin
- Spectrometry Data Center, Biomolecular Measurement Division , National Institute of Standards and Technology (NIST) , Gaithersburg , Maryland 20899 , United States
| | - Stephen E Stein
- Spectrometry Data Center, Biomolecular Measurement Division , National Institute of Standards and Technology (NIST) , Gaithersburg , Maryland 20899 , United States
| |
Collapse
|
33
|
Taraboletti A, Goudarzi M, Kabir A, Moon BH, Laiakis EC, Lacombe J, Ake P, Shoishiro S, Brenner D, Fornace AJ, Zenhausern F. Fabric Phase Sorptive Extraction-A Metabolomic Preprocessing Approach for Ionizing Radiation Exposure Assessment. J Proteome Res 2019; 18:3020-3031. [PMID: 31090424 PMCID: PMC7437658 DOI: 10.1021/acs.jproteome.9b00142] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The modern application of mass spectrometry-based metabolomics to the field of radiation assessment and biodosimetry has allowed for the development of prompt biomarker screenings for radiation exposure. Our previous work on radiation assessment, in easily accessible biofluids (such as urine, blood, saliva), has revealed unique metabolic perturbations in response to radiation quality, dose, and dose rate. Nevertheless, the employment of swift injury assessment in the case of a radiological disaster still remains a challenge as current sample processing can be time consuming and cause sample degradation. To address these concerns, we report a metabolomics workflow using a mass spectrometry-compatible fabric phase sorptive extraction (FPSE) technique. FPSE employs a matrix coated with sol-gel poly(caprolactone-b-dimethylsiloxane-b-caprolactone) that binds both polar and nonpolar metabolites in whole blood, eliminating serum processing steps. We confirm that the FPSE preparation technique combined with liquid chromatography-mass spectrometry can distinguish radiation exposure markers such as taurine, carnitine, arachidonic acid, α-linolenic acid, and oleic acid found 24 h after 8 Gy irradiation. We also note the effect of different membrane fibers on both metabolite extraction efficiency and the temporal stabilization of metabolites in whole blood at room temperature. These findings suggest that the FPSE approach could work in future technology to triage irradiated individuals accurately, via biomarker screening, by providing a novel method to stabilize biofluids between collection and sample analysis.
Collapse
Affiliation(s)
- Alexandra Taraboletti
- Department of Oncology, Georgetown University Medical Center, Georgetown University, 3800 Reservoir Road Northwest, Washington, District of Columbia 20057, United States
| | - Maryam Goudarzi
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Georgetown University, 3800 Reservoir Road Northwest, Washington, District of Columbia 20057, United States
- Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, Ohio 44195, United States
| | - Abuzar Kabir
- International Forensic Research Institute, Department of Chemistry and Biochemistry, Florida International University, 11200 Southwest Eighth Street, Miami, Florida 33199, United States
| | - Bo-Hyun Moon
- Department of Oncology, Georgetown University Medical Center, Georgetown University, 3800 Reservoir Road Northwest, Washington, District of Columbia 20057, United States
| | - Evagelia C. Laiakis
- Department of Oncology, Georgetown University Medical Center, Georgetown University, 3800 Reservoir Road Northwest, Washington, District of Columbia 20057, United States
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Georgetown University, 3800 Reservoir Road Northwest, Washington, District of Columbia 20057, United States
| | - Jerome Lacombe
- Center for Applied NanoBiosience and Medicine, University of Arizona, 475 North Fifth Street, Phoenix, Arizona 85004, United States
| | - Pelagie Ake
- Department of Oncology, Georgetown University Medical Center, Georgetown University, 3800 Reservoir Road Northwest, Washington, District of Columbia 20057, United States
| | - Sueoka Shoishiro
- Center for Applied NanoBiosience and Medicine, University of Arizona, 475 North Fifth Street, Phoenix, Arizona 85004, United States
| | - David Brenner
- Center for Radiological Research, Columbia University, 630 West 168th Street, New York, New York 10032, United States
| | - Albert J. Fornace
- Department of Oncology, Georgetown University Medical Center, Georgetown University, 3800 Reservoir Road Northwest, Washington, District of Columbia 20057, United States
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Georgetown University, 3800 Reservoir Road Northwest, Washington, District of Columbia 20057, United States
| | - Frederic Zenhausern
- Center for Applied NanoBiosience and Medicine, University of Arizona, 475 North Fifth Street, Phoenix, Arizona 85004, United States
- Translational Genomics Research Institute, 445 North Fifth Street, Phoenix, Arizona 85004, United States
- Department of Basic Medical Sciences, College of Medicine Phoenix, 425 North Fifth Street, Phoenix, Arizona 85004, United States
| |
Collapse
|
34
|
Temporal Effects on Radiation Responses in Nonhuman Primates: Identification of Biofluid Small Molecule Signatures by Gas Chromatography⁻Mass Spectrometry Metabolomics. Metabolites 2019; 9:metabo9050098. [PMID: 31096611 PMCID: PMC6571779 DOI: 10.3390/metabo9050098] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 05/07/2019] [Accepted: 05/10/2019] [Indexed: 12/28/2022] Open
Abstract
Whole body exposure to ionizing radiation damages tissues leading to physical symptoms which contribute to acute radiation syndrome. Radiation biodosimetry aims to determine characteristic early biomarkers indicative of radiation exposure and is necessary for effective triage after an unanticipated radiological incident. Radiation metabolomics can address this aim by assessing metabolic perturbations following exposure. Gas chromatography-mass spectrometry (GC-MS) is a standardized platform ideal for compound identification. We performed GC time-of-flight MS for the global profiling of nonhuman primate urine and serum samples up to 60 d after a single 4 Gy γ-ray total body exposure. Multivariate statistical analysis showed higher group separation in urine vs. serum. We identified biofluid markers involved in amino acid, lipid, purine, and serotonin metabolism, some of which may indicate host microbiome dysbiosis. Sex differences were observed for amino acid fold changes in serum samples. Additionally, we explored mitochondrial dysfunction by tricarboxylic acid intermediate analysis in the first week with a GC tandem quadrupole MS platform. By adding this temporal component to our previous work exploring dose effects at 7 d, we observed the highest fold changes occurring at 3 d, returning closer to basal levels by 7 d. These results emphasize the utility of both MS-based metabolomics for biodosimetry and complementary analytical platforms for increased metabolome coverage.
Collapse
|
35
|
Pannkuk EL, Laiakis EC, Gill K, Jain SK, Mehta KY, Nishita D, Bujold K, Bakke J, Gahagen J, Authier S, Chang P, Fornace AJ. Liquid Chromatography-Mass Spectrometry-Based Metabolomics of Nonhuman Primates after 4 Gy Total Body Radiation Exposure: Global Effects and Targeted Panels. J Proteome Res 2019; 18:2260-2269. [PMID: 30843397 DOI: 10.1021/acs.jproteome.9b00101] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Rapid assessment of radiation signatures in noninvasive biofluids may aid in assigning proper medical treatments for acute radiation syndrome (ARS) and delegating limited resources after a nuclear disaster. Metabolomic platforms allow for rapid screening of biofluid signatures and show promise in differentiating radiation quality and time postexposure. Here, we use global metabolomics to differentiate temporal effects (1-60 d) found in nonhuman primate (NHP) urine and serum small molecule signatures after a 4 Gy total body irradiation. Random Forests analysis differentially classifies biofluid signatures according to days post 4 Gy exposure. Eight compounds involved in protein metabolism, fatty acid β oxidation, DNA base deamination, and general energy metabolism were identified in each urine and serum sample and validated through tandem MS. The greatest perturbations were seen at 1 d in urine and 1-21 d in serum. Furthermore, we developed a targeted liquid chromatography tandem mass spectrometry (LC-MS/MS) with multiple reaction monitoring (MRM) method to quantify a six compound panel (hypoxanthine, carnitine, acetylcarnitine, proline, taurine, and citrulline) identified in a previous training cohort at 7 d after a 4 Gy exposure. The highest sensitivity and specificity for classifying exposure at 7 d after a 4 Gy exposure included carnitine and acetylcarnitine in urine and taurine, carnitine, and hypoxanthine in serum. Receiver operator characteristic (ROC) curve analysis using combined compounds show excellent sensitivity and specificity in urine (area under the curve [AUC] = 0.99) and serum (AUC = 0.95). These results highlight the utility of MS platforms to differentiate time postexposure and acquire reliable quantitative biomarker panels for classifying exposed individuals.
Collapse
Affiliation(s)
- Evan L Pannkuk
- Department of Oncology, Lombardi Comprehensive Cancer Center , Georgetown University Medical Center , Washington, D.C. 20007 , United States
| | - Evagelia C Laiakis
- Department of Oncology, Lombardi Comprehensive Cancer Center , Georgetown University Medical Center , Washington, D.C. 20007 , United States.,Department of Biochemistry and Molecular & Cellular Biology , Georgetown University Medical Center , Washington, D.C. 20007 , United States
| | - Kirandeep Gill
- Department of Biochemistry and Molecular & Cellular Biology , Georgetown University Medical Center , Washington, D.C. 20007 , United States
| | - Shreyans K Jain
- Department of Biochemistry and Molecular & Cellular Biology , Georgetown University Medical Center , Washington, D.C. 20007 , United States
| | - Khyati Y Mehta
- Department of Oncology, Lombardi Comprehensive Cancer Center , Georgetown University Medical Center , Washington, D.C. 20007 , United States
| | - Denise Nishita
- SRI International , Menlo Park , California 94025 , United States
| | - Kim Bujold
- Citoxlab North America , Laval , QC H7V 4B3 , Canada
| | - James Bakke
- SRI International , Menlo Park , California 94025 , United States
| | - Janet Gahagen
- SRI International , Menlo Park , California 94025 , United States
| | - Simon Authier
- Citoxlab North America , Laval , QC H7V 4B3 , Canada
| | - Polly Chang
- SRI International , Menlo Park , California 94025 , United States
| | - Albert J Fornace
- Department of Oncology, Lombardi Comprehensive Cancer Center , Georgetown University Medical Center , Washington, D.C. 20007 , United States.,Department of Biochemistry and Molecular & Cellular Biology , Georgetown University Medical Center , Washington, D.C. 20007 , United States
| |
Collapse
|
36
|
Abstract
Metabolomics, the collective assessment and quantification of small molecules in a given biofluid or tissue sample, has provided new ways in evaluating an individual's exposure level to ionizing radiation or other genotoxic stressors. Protocols that are routinely utilized for the preparation of samples from rodents to patients are presented here in order to be analyzed by high-throughput liquid chromatography-mass spectrometry techniques. These protocols are based on established methods in our laboratory that have been used extensively in radiation biodosimetry through metabolomics. These protocols are focused on general profiling of samples and therefore do not concentrate on extraction of specific classes on metabolites (e.g., eicosanoids).
Collapse
Affiliation(s)
- Evagelia C Laiakis
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA.
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, USA.
| |
Collapse
|
37
|
Pannkuk EL, Laiakis EC, Garcia M, Fornace AJ, Singh VK. Nonhuman Primates with Acute Radiation Syndrome: Results from a Global Serum Metabolomics Study after 7.2 Gy Total-Body Irradiation. Radiat Res 2018; 190:576-583. [PMID: 30183511 DOI: 10.1667/rr15167.1] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Threats of nuclear terrorism coupled with potential unintentional ionizing radiation exposures have necessitated the need for large-scale response efforts of such events, including high-throughput biodosimetry for medical triage. Global metabolomics utilizing mass spectrometry (MS) platforms has proven an ideal tool for generating large compound databases with relative quantification and structural information in a short amount of time. Determining metabolite panels for biodosimetry requires experimentation to evaluate the many factors associated with compound concentrations in biofluids after radiation exposures, including temporal changes, pre-existing conditions, dietary intake, partial- vs. total-body irradiation (TBI), among others. Here, we utilize a nonhuman primate (NHP) model and identify metabolites perturbed in serum after 7.2 Gy TBI without supportive care [LD70/60, hematologic (hematopoietic) acute radiation syndrome (HARS) level H3] at 24, 36, 48 and 96 h compared to preirradiation samples with an ultra-performance liquid chromatography quadrupole time-of-flight (UPLC-QTOF) MS platform. Additionally, we document changes in cytokine levels. Temporal changes observed in serum carnitine, acylcarnitines, amino acids, lipids, deaminated purines and increases in pro-inflammatory cytokines indicate clear metabolic dysfunction after radiation exposure. Multivariate data analysis shows distinct separation from preirradiation groups and receiver operator characteristic curve analysis indicates high specificity and sensitivity based on area under the curve at all time points after 7.2 Gy irradiation. Finally, a comparison to a 6.5 Gy (LD50/60, HARS level H2) cohort after 24 h postirradiation revealed distinctly increased separations from the 7.2 Gy cohort based on multivariate data models and higher compound fold changes. These results highlight the utility of MS platforms to differentiate time and absorbed dose after a potential radiation exposure that may aid in assigning specific medical interventions and contribute as additional biodosimetry tools.
Collapse
Affiliation(s)
| | - Evagelia C Laiakis
- Departments of Oncology.,Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC
| | - Melissa Garcia
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Bethesda, Maryland
| | - Albert J Fornace
- Departments of Oncology.,Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC
| | - Vijay K Singh
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Bethesda, Maryland.,Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| |
Collapse
|
38
|
Chen Z, Coy SL, Pannkuk EL, Laiakis EC, Fornace AJ, Vouros P. Differential Mobility Spectrometry-Mass Spectrometry (DMS-MS) in Radiation Biodosimetry: Rapid and High-Throughput Quantitation of Multiple Radiation Biomarkers in Nonhuman Primate Urine. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2018; 29:1650-1664. [PMID: 29736597 PMCID: PMC6287943 DOI: 10.1007/s13361-018-1977-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 04/19/2018] [Accepted: 04/21/2018] [Indexed: 05/21/2023]
Abstract
High-throughput methods to assess radiation exposure are a priority due to concerns that include nuclear power accidents, the spread of nuclear weapon capability, and the risk of terrorist attacks. Metabolomics, the assessment of small molecules in an easily accessible sample, is the most recent method to be applied for the identification of biomarkers of the biological radiation response with a useful dose-response profile. Profiling for biomarker identification is frequently done using an LC-MS platform which has limited throughput due to the time-consuming nature of chromatography. We present here a chromatography-free simplified method for quantitative analysis of seven metabolites in urine with radiation dose-response using urine samples provided from the Pannkuk et al. (2015) study of long-term (7-day) radiation response in nonhuman primates (NHP). The stable isotope dilution (SID) analytical method consists of sample preparation by strong cation exchange-solid phase extraction (SCX-SPE) to remove interferences and concentrate the metabolites of interest, followed by differential mobility spectrometry (DMS) ion filtration to select the ion of interest and reduce chemical background, followed by mass spectrometry (overall SID-SPE-DMS-MS). Since no chromatography is used, calibration curves were prepared rapidly, in under 2 h (including SPE) for six simultaneously analyzed radiation biomarkers. The seventh, creatinine, was measured separately after 2500× dilution. Creatinine plays a dual role, measuring kidney glomerular filtration rate (GFR), and indicating kidney damage at high doses. The current quantitative method using SID-SPE-DMS-MS provides throughput which is 7.5 to 30 times higher than that of LC-MS and provides a path to pre-clinical radiation dose estimation. Graphical Abstract.
Collapse
Affiliation(s)
- Zhidan Chen
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA, 02115, USA
| | - Stephen L Coy
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA, 02115, USA.
| | - Evan L Pannkuk
- Tumor Biology Program, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20057, USA
| | - Evagelia C Laiakis
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC, 20057, USA
| | - Albert J Fornace
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC, 20057, USA
- Department of Oncology, Georgetown University Medical Center, Washington, DC, 20057, USA
| | - Paul Vouros
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA, 02115, USA.
- Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, MA, 02115, USA.
| |
Collapse
|
39
|
Vera NB, Chen Z, Pannkuk E, Laiakis EC, Fornace AJ, Erion DM, Coy SL, Pfefferkorn JA, Vouros P. Differential mobility spectrometry (DMS) reveals the elevation of urinary acetylcarnitine in non-human primates (NHPs) exposed to radiation. JOURNAL OF MASS SPECTROMETRY : JMS 2018; 53:548-559. [PMID: 29596720 PMCID: PMC6030448 DOI: 10.1002/jms.4085] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 03/08/2018] [Accepted: 03/12/2018] [Indexed: 05/21/2023]
Abstract
Acetylcarnitine has been identified as one of several urinary biomarkers indicative of radiation exposure in adult rhesus macaque monkeys (non-human primates, NHPs). Previous work has demonstrated an up-regulated dose-response profile in a balanced male/female NHP cohort. As a contribution toward the development of metabolomics-based radiation biodosimetry in human populations and other applications of acetylcarnitine screening, we have developed a quantitative, high-throughput method for the analysis of acetylcarnitine. We employed the Sciex SelexIon DMS-MS/MS QTRAP 5500 platform coupled to flow injection analysis (FIA), thereby allowing for fast analysis times of less than 0.5 minutes per injection with no chromatographic separation. Ethyl acetate is used as a DMS modifier to reduce matrix chemical background. We have measured NHP urinary acetylcarnitine from the male cohorts that were exposed to the following radiation levels: control, 2, 4, 6, 7, and 10 Gy. Biological variability, along with calibration accuracy of the FIA-DMS-MS/MS method, indicates LOQ of 20 μM, with observed biological levels on the order of 600 μM and control levels near 10 μM. There is an apparent onset of intensified response in the transition from 6 to 10 Gy. The results demonstrate that FIA-DMS-MS/MS is a rapid, quantitative technique that can be utilized for the analysis of urinary biomarker levels for radiation biodosimetry.
Collapse
Affiliation(s)
- Nicholas B Vera
- Pfizer Global Research and Development, Cambridge Laboratories, Pfizer Inc., Cambridge, MA, 02139, USA
- Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, MA, 02115, USA
| | - Zhidan Chen
- Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, MA, 02115, USA
| | - Evan Pannkuk
- Georgetown University, 3700 O Street NW, Washington, DC, 20057, USA
| | | | - Albert J Fornace
- Georgetown University, 3700 O Street NW, Washington, DC, 20057, USA
| | - Derek M Erion
- Pfizer Global Research and Development, Cambridge Laboratories, Pfizer Inc., Cambridge, MA, 02139, USA
| | - Stephen L Coy
- Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, MA, 02115, USA
| | - Jeffrey A Pfefferkorn
- Pfizer Global Research and Development, Cambridge Laboratories, Pfizer Inc., Cambridge, MA, 02139, USA
| | - Paul Vouros
- Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, MA, 02115, USA
| |
Collapse
|
40
|
Pannkuk EL, Laiakis EC, Fornace AJ, Fatanmi OO, Singh VK. A Metabolomic Serum Signature from Nonhuman Primates Treated with a Radiation Countermeasure, Gamma-tocotrienol, and Exposed to Ionizing Radiation. HEALTH PHYSICS 2018; 115:3-11. [PMID: 29787425 PMCID: PMC5967639 DOI: 10.1097/hp.0000000000000776] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The search for and development of radiation countermeasures to treat acute lethal radiation injury has been underway for the past six decades, resulting in the identification of multiple classes of radiation countermeasures. However, to date only granulocyte colony-stimulating factor (Neupogen) and PEGylated granulocyte colony-stimulating factor (Neulasta) have been approved by the U.S. Food and Drug Administration for the treatment of hematopoietic acute radiation syndrome. Gamma-tocotrienol has demonstrated radioprotective efficacy in murine and nonhuman primate models. Currently, this agent is under advanced development as a radioprotector, and the authors are trying to identify its efficacy biomarkers. In this study, global metabolomic changes were analyzed using ultraperformance liquid chromatography quadrupole time-of-flight mass spectrometry. The pilot study using 16 nonhuman primates (8 nonhuman primates each in gamma-tocotrienol- and vehicle-treated groups), with samples obtained from gamma-tocotrienol-treated and irradiated nonhuman primates, demonstrates several metabolites that are altered after irradiation, including compounds involved in fatty acid beta-oxidation, purine catabolism, and amino acid metabolism. The machine-learning algorithm, Random Forest, separated control, irradiated gamma-tocotrienol-treated, and irradiated vehicle-treated nonhuman primates at 12 h and 24 h as evident in a multidimensional scaling plot. Primary metabolites validated included carnitine/acylcarnitines, amino acids, creatine, and xanthine. Overall, gamma-tocotrienol administration reduced high fluctuations in serum metabolite levels, suggesting an overall beneficial effect on animals exposed to radiation. This initial assessment also highlights the utility of metabolomics in determining underlying physiological mechanisms responsible for the radioprotective efficacy of gamma-tocotrienol.
Collapse
Affiliation(s)
- Evan L. Pannkuk
- Tumor Biology Program, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20057, USA
| | - Evagelia C. Laiakis
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC, 20057, USA
| | - Albert J. Fornace
- Tumor Biology Program, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, 20057, USA
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC, 20057, USA
| | - Oluseyi O. Fatanmi
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Vijay K. Singh
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| |
Collapse
|
41
|
Roh C. Metabolomics in Radiation-Induced Biological Dosimetry: A Mini-Review and a Polyamine Study. Biomolecules 2018; 8:biom8020034. [PMID: 29844258 PMCID: PMC6023017 DOI: 10.3390/biom8020034] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/18/2018] [Accepted: 05/25/2018] [Indexed: 01/09/2023] Open
Abstract
In this study, we elucidate that polyamine metabolite is a powerful biomarker to study post-radiation changes. Metabolomics in radiation biodosimetry, the application of a metabolomics analysis to the field of radiobiology, promises to increase the understanding of biological responses by ionizing radiation (IR). Radiation exposure triggers a complex network of molecular and cellular responses that impacts metabolic processes and alters the levels of metabolites. Such metabolites have potential as biomarkers for radiation dosimetry. Among metabolites, polyamine is one of many potential biomarkers to estimate radiation response. In addition, this review provides an opportunity for the understanding of a radiation metabolomics in biodosimetry and a polyamine case study.
Collapse
Affiliation(s)
- Changhyun Roh
- Biotechnology Research Division, Advanced Radiation Technology Institute (ARTI), Korea Atomic Energy Research Institute (KAERI), 29, Geumgu-gil, Jeongeup-si, Jeonbuk 56212, Korea.
- Radiation Biotechnology and Applied Radioisotope Science, University of Science Technology (UST), 217 Gajeong-ro, Daejeon 34113, Korea.
| |
Collapse
|
42
|
Suresh Kumar MA, Laiakis EC, Ghandhi SA, Morton SR, Fornace AJ, Amundson SA. Gene Expression in Parp1 Deficient Mice Exposed to a Median Lethal Dose of Gamma Rays. Radiat Res 2018; 190:53-62. [PMID: 29746213 DOI: 10.1667/rr14990.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
There is a current interest in the development of biodosimetric methods for rapidly assessing radiation exposure in the wake of a large-scale radiological event. This work was initially focused on determining the exposure dose to an individual using biological indicators. Gene expression signatures show promise for biodosimetric application, but little is known about how these signatures might translate for the assessment of radiological injury in radiosensitive individuals, who comprise a significant proportion of the general population, and who would likely require treatment after exposure to lower doses. Using Parp1-/- mice as a model radiation-sensitive genotype, we have investigated the effect of this DNA repair deficiency on the gene expression response to radiation. Although Parp1 is known to play general roles in regulating transcription, the pattern of gene expression changes observed in Parp1-/- mice 24 h postirradiation to a LD50/30 was remarkably similar to that in wild-type mice after exposure to LD50/30. Similar levels of activation of both the p53 and NFκB radiation response pathways were indicated in both strains. In contrast, exposure of wild-type mice to a sublethal dose that was equal to the Parp1-/- LD50/30 resulted in a lower magnitude gene expression response. Thus, Parp1-/- mice displayed a heightened gene expression response to radiation, which was more similar to the wild-type response to an equitoxic dose than to an equal absorbed dose. Gene expression classifiers trained on the wild-type data correctly identified all wild-type samples as unexposed, exposed to a sublethal dose or exposed to an LD50/30. All unexposed samples from Parp1-/- mice were also correctly classified with the same gene set, and 80% of irradiated Parp1-/- samples were identified as exposed to an LD50/30. The results of this study suggest that, at least for some pathways that may influence radiosensitivity in humans, specific gene expression signatures have the potential to accurately detect the extent of radiological injury, rather than serving only as a surrogate of physical radiation dose.
Collapse
Affiliation(s)
- M A Suresh Kumar
- a Center for Radiological Research, Columbia University Medical Center, Columbia University, New York, New York
| | - Evagelia C Laiakis
- b Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, DC
| | - Shanaz A Ghandhi
- a Center for Radiological Research, Columbia University Medical Center, Columbia University, New York, New York
| | - Shad R Morton
- a Center for Radiological Research, Columbia University Medical Center, Columbia University, New York, New York
| | - Albert J Fornace
- b Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, DC
| | - Sally A Amundson
- a Center for Radiological Research, Columbia University Medical Center, Columbia University, New York, New York
| |
Collapse
|
43
|
Renal and hepatic effects following neonatal exposure to low doses of Bisphenol-A and 137Cs. Food Chem Toxicol 2018; 114:270-277. [PMID: 29477810 DOI: 10.1016/j.fct.2018.02.046] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 02/16/2018] [Accepted: 02/20/2018] [Indexed: 12/13/2022]
Abstract
137-Cesium (137Cs) is one of the most important distributed radionuclides after a nuclear accident. Humans are usually co-exposed to various environmental toxicants, being Bisphenol-A (BPA) one of them. Exposure to IR and BPA in early life is of major concern, due to the higher vulnerability of developing organs. We evaluate the renal and hepatic effects of low doses of ionizing radiation (IR) and BPA. Sixty male mice (C57BL/6J) were randomly assigned to six experimental groups (n=10) and received a single subcutaneous dose of 0.9% saline solution, 137Cs and/or BPA on postnatal day 10: control, BPA (25 μg/kgbw), Cs4000 (4000 Bq 137Cs/kgbw), Cs8000 (8000 Bq 137Cs/kgbw), BPA/Cs4000 and BPA/Cs8000. At the age of two months, urines (24h) and blood samples were collected from animals of each group to determine biochemical parameters. Finally, kidneys and liver were removed to quantify DNA damage (8-OHdG), as well as to determine CYP1A2 mRNA expression. Data suggest that both BPA and 137Cs induced renal and liver damage evidenced by oxidative stress. However, when there is a co-exposure, it seems that there are compensatory mechanisms that may reverse the damage induced by each toxic itself. Notwithstanding, more studies are necessary to better understand the synergistic mechanisms behind.
Collapse
|
44
|
Laiakis EC, Pannkuk EL, Chauthe SK, Wang YW, Lian M, Mak TD, Barker CA, Astarita G, Fornace AJ. A Serum Small Molecule Biosignature of Radiation Exposure from Total Body Irradiated Patients. J Proteome Res 2017; 16:3805-3815. [PMID: 28825479 DOI: 10.1021/acs.jproteome.7b00468] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The potential for radiological accidents and nuclear terrorism has increased the need for the development of new rapid biodosimetry methods. In addition, in a clinical setting the issue of an individual's radiosensitivity should be taken into consideration during radiotherapy. We utilized metabolomics and lipidomics to investigate changes of metabolites in serum samples following exposure to total body ionizing radiation in humans. Serum was collected prior to irradiation, at 3-8 h after a single dose of 1.25-2 Gy, and at 24 h with a total delivered dose of 2-3.75 Gy. Metabolomics revealed perturbations in glycerophosphocholine, phenylalanine, ubiquinone Q2, and oxalic acid. Alterations were observed in circulating levels of lipids from monoacylglycerol, triacylglycerol, phosphatidylcholine, and phosphatidylglycerol lipid classes. Polyunsaturated fatty acids were some of the most dysregulated lipids, with increased levels linked to proinflammatory processes. A targeted metabolomics approach for eicosanoids was also employed. The results showed a rapid response for proinflammatory eicosanoids, with a dampening of the signal at the later time point. Sex differences were observed in the markers from the untargeted approach but not the targeted method. The ability to identify and quantify small molecules in blood can therefore be utilized to monitor radiation exposure in human populations.
Collapse
Affiliation(s)
| | | | | | | | - Ming Lian
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center , New York, New York 10065, United States
| | - Tytus D Mak
- National Institute of Standards and Technology (NIST) , Gaithersburg, Maryland 20899, United States
| | - Christopher A Barker
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center , New York, New York 10065, United States
| | | | | |
Collapse
|
45
|
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: 2.1] [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
|
46
|
Bellés M, Gonzalo S, Serra N, Esplugas R, Arenas M, Domingo JL, Linares V. Environmental exposure to low-doses of ionizing radiation. Effects on early nephrotoxicity in mice. ENVIRONMENTAL RESEARCH 2017; 156:291-296. [PMID: 28371757 DOI: 10.1016/j.envres.2017.03.034] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 03/22/2017] [Accepted: 03/23/2017] [Indexed: 06/07/2023]
Abstract
Nuclear accidents of tremendous magnitude, such as those of Chernobyl (1986) and Fukushima (2011), mean that individuals living in the contaminated areas are potentially exposed to ionizing radiation (IR). However, the dose-response relationship for effects of low doses of radiation is not still established. The present study was aimed at investigating in mice the early effects of low-dose internal radiation exposure on the kidney. Adult male (C57BL/6J) mice were divided into three groups. Two groups received a single subcutaneous (s.c.) doses of cesium (137Cs) with activities of 4000 and 8000Bq/kg bw. A third group (control group) received a single s.c. injection of 0.9% saline. To evaluate acute and subacute effects, mice (one-half of each group) were euthanized at 72h and 10 days post-exposure to 137Cs, respectively. Urine samples were collected for biochemical analysis, including the measurement of F2-isoprostane (F2-IsoP) and kidney injury molecule-1 (KIM-1) levels. Moreover, the concentrations of 8-hydroxy-2'-deoxyguanosine (8-OHdG), a sensitive marker of oxidative DNA damage, were measured in renal tissue. Urinary excretion of total protein significantly increased at 72h in mice exposed to Cs4000. Uric acid and lactate dehydrogenase (LDH) decreased significantly at both times post-exposure in animals exposed to Cs8000. After 72h and 10d of exposure to Cs4000, a significant increase in the γ-glutamil transferase (GGT) and N-acetyl-β-D-glucosaminidase (NAG) activities was observed. In turn, F2-IsoP levels increased -mainly in the Cs4000 group- at 72h post-exposure. Following irradiation (137Cs), the highest level of KIM-1 was corresponded to the Cs4000 group at 72h. Likewise, the main DNA damage was detected in mice exposed to Cs4000, mainly at 10d after irradiation. The alterations observed in several biomarkers suggest an immediate renal damage following exposure to low doses of IR (given as 137Cs). Further investigations are required to clarify the mechanisms involved in the internal IR-induced nephrotoxicity.
Collapse
Affiliation(s)
- Montserrat Bellés
- Physiology Unit, School of Medicine, IISPV, Rovira i Virgili University, Reus, Spain; Laboratory of Toxicology and Environmental Health, School of Medicine, IISPV, Rovira i Virgili University, Reus, Spain
| | - Sergio Gonzalo
- Laboratory of Toxicology and Environmental Health, School of Medicine, IISPV, Rovira i Virgili University, Reus, Spain
| | - Noemí Serra
- Laboratory of Toxicology and Environmental Health, School of Medicine, IISPV, Rovira i Virgili University, Reus, Spain
| | - Roser Esplugas
- Laboratory of Toxicology and Environmental Health, School of Medicine, IISPV, Rovira i Virgili University, Reus, Spain
| | - Meritxell Arenas
- Radiation Oncology Department, Sant Joan University Hospital, IISPV, Rovira i Virgili University, Reus, Spain
| | - José Luis Domingo
- Laboratory of Toxicology and Environmental Health, School of Medicine, IISPV, Rovira i Virgili University, Reus, Spain
| | - Victoria Linares
- Physiology Unit, School of Medicine, IISPV, Rovira i Virgili University, Reus, Spain; Laboratory of Toxicology and Environmental Health, School of Medicine, IISPV, Rovira i Virgili University, Reus, Spain.
| |
Collapse
|
47
|
Laiakis EC, Wang YW, Young EF, Harken AD, Xu Y, Smilenov L, Garty GY, Brenner DJ, Fornace AJ. Metabolic Dysregulation after Neutron Exposures Expected from an Improvised Nuclear Device. Radiat Res 2017; 188:21-34. [PMID: 28475424 PMCID: PMC5714588 DOI: 10.1667/rr14656.1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The increased threat of terrorism across the globe has raised fears that certain groups will acquire and use radioactive materials to inflict maximum damage. In the event that an improvised nuclear device (IND) is detonated, a potentially large population of victims will require assessment for radiation exposure. While photons will contribute to a major portion of the dose, neutrons may be responsible for the severity of the biologic effects and cellular responses. We investigated differences in response between these two radiation types by using metabolomics and lipidomics to identify biomarkers in urine and blood of wild-type C57BL/6 male mice. Identification of metabolites was based on a 1 Gy dose of radiation. Compared to X rays, a neutron spectrum similar to that encountered in Hiroshima at 1-1.5 km from the epicenter induced a severe metabolic dysregulation, with perturbations in amino acid metabolism and fatty acid β-oxidation being the predominant ones. Urinary metabolites were able to discriminate between neutron and X rays on day 1 as well as day 7 postirradiation, while serum markers showed such discrimination only on day 1. Free fatty acids from omega-6 and omega-3 pathways were also decreased with 1 Gy of neutrons, implicating cell membrane dysfunction and impaired phospholipid metabolism, which should otherwise lead to release of those molecules in circulation. While a precise relative biological effectiveness value could not be calculated from this study, the results are consistent with other published studies showing higher levels of damage from neutrons, demonstrated here by increased metabolic dysregulation. Metabolomics can therefore aid in identifying global perturbations in blood and urine, and effectively distinguishing between neutron and photon exposures.
Collapse
Affiliation(s)
| | - Yi-Wen Wang
- Department of Infectious Diseases, The Scripps Research Institute, Jupiter, Florida
| | | | - Andrew D. Harken
- Radiological Research Accelerator Facility, Columbia University, Irvington, New York
| | - Yanping Xu
- Radiological Research Accelerator Facility, Columbia University, Irvington, New York
- Department of Physics, East Carolina University, Greenville, North Carolina
| | - Lubomir Smilenov
- Center for Radiological Research, Columbia University, New York, New York
| | - Guy Y. Garty
- Radiological Research Accelerator Facility, Columbia University, Irvington, New York
| | - David J. Brenner
- Center for Radiological Research, Columbia University, New York, New York
| | - Albert J. Fornace
- Department of Biochemistry and Molecular and Cellular Biology
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| |
Collapse
|
48
|
Pannkuk EL, Laiakis EC, Authier S, Wong K, Fornace AJ. Gas Chromatography/Mass Spectrometry Metabolomics of Urine and Serum from Nonhuman Primates Exposed to Ionizing Radiation: Impacts on the Tricarboxylic Acid Cycle and Protein Metabolism. J Proteome Res 2017; 16:2091-2100. [PMID: 28351153 PMCID: PMC5720681 DOI: 10.1021/acs.jproteome.7b00064] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Ionizing radiation (IR) directly damages cells and tissues or indirectly damages them through reactive free radicals that may lead to longer term adverse sequelae such as cancers, persistent inflammation, or possible death. Potential exposures include nuclear reactor accidents, improper disposal of equipment containing radioactive materials or medical errors, and terrorist attacks. Metabolomics (comprehensive analysis of compounds <1 kDa) by mass spectrometry (MS) has been proposed as a tool for high-throughput biodosimetry and rapid assessment of exposed dose and triage needed. While multiple studies have been dedicated to radiation biomarker discovery, many have utilized liquid chromatography (LC) MS platforms that may not detect particular compounds (e.g., small carboxylic acids or isomers) that complementary analytical tools, such as gas chromatography (GC) time-of-flight (TOF) MS, are ideal for. The current study uses global GC-TOF-MS metabolomics to complement previous LC-MS analyses on nonhuman primate biofluids (urine and serum) 7 days after exposure to 2, 4, 6, 7, and 10 Gy IR. Multivariate data analysis was used to visualize differences between control and IR exposed groups. Univariate analysis was used to determine a combined 26 biomarkers in urine and serum that significantly changed after exposure to IR. We found several metabolites involved in tricarboxylic acid cycle function, amino acid metabolism, and host microbiota that were not previously detected by global and targeted LC-MS studies.
Collapse
Affiliation(s)
- Evan L. Pannkuk
- Tumor Biology Program, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, D.C. 20057, United States
| | - Evagelia C. Laiakis
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, D.C. 20057, United States
| | - Simon Authier
- CiToxLAB North America, Laval, Quebec H7V 4B3, Canada
| | - Karen Wong
- CiToxLAB North America, Laval, Quebec H7V 4B3, Canada
| | - Albert J. Fornace
- Tumor Biology Program, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, D.C. 20057, United States
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, D.C. 20057, United States
| |
Collapse
|
49
|
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: 4.0] [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
|
50
|
Garty G, Xu Y, Elliston C, Marino SA, Randers-Pehrson G, Brenner DJ. Mice and the A-Bomb: Irradiation Systems for Realistic Exposure Scenarios. Radiat Res 2017; 187:465-475. [PMID: 28211757 DOI: 10.1667/rr008cc.1] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Validation of biodosimetry assays is normally performed with acute exposures to uniform external photon fields. Realistically, exposure to a radiological dispersal device or reactor leak will include exposure to low dose rates and likely exposure to ingested radionuclides. An improvised nuclear device will likely include a significant neutron component in addition to a mixture of high- and low-dose-rate photons and ingested radionuclides. We present here several novel irradiation systems developed at the Center for High Throughput Minimally Invasive Radiation Biodosimetry to provide more realistic exposures for testing of novel biodosimetric assays. These irradiators provide a wide range of dose rates (from Gy/s to Gy/week) as well as mixed neutron/photon fields mimicking an improvised nuclear device.
Collapse
Affiliation(s)
- Guy Garty
- a Radiological Research Accelerator Facility, Columbia University, Irvington, New York 10533; and
| | - Yanping Xu
- a Radiological Research Accelerator Facility, Columbia University, Irvington, New York 10533; and
| | - Carl Elliston
- b Center for Radiological Research, Columbia University, New York, New York 10032
| | - Stephen A Marino
- a Radiological Research Accelerator Facility, Columbia University, Irvington, New York 10533; and
| | - Gerhard Randers-Pehrson
- a Radiological Research Accelerator Facility, Columbia University, Irvington, New York 10533; and
| | - David J Brenner
- b Center for Radiological Research, Columbia University, New York, New York 10032
| |
Collapse
|