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
|
Li Y, Bansal S, Singh B, Jayatilake MM, Klotzbier W, Boerma M, Lee MH, Hack J, Iwamoto KS, Schaue D, Cheema AK. Distinct Urinary Metabolite Signatures Mirror In Vivo Oxidative Stress-Related Radiation Responses in Mice. Antioxidants (Basel) 2024; 14:24. [PMID: 39857358 PMCID: PMC11763242 DOI: 10.3390/antiox14010024] [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: 11/25/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025] Open
Abstract
Exposure to ionizing radiation disrupts metabolic pathways and causes oxidative stress, which can lead to organ damage. In this study, urinary metabolites from mice exposed to high-dose and low-dose whole-body irradiation (WBI HDR, WBI LDR) or partial-body irradiation (PBI BM2.5) were analyzed using targeted and untargeted metabolomics approaches. Significant metabolic changes particularly in oxidative stress pathways were observed on Day 2 post-radiation. By Day 30, the WBI HDR group showed persistent metabolic dysregulation, while the WBI LDR and PBI BM2.5 groups were similar to control mice. Machine learning models identified metabolites that were predictive of the type of radiation exposure with high accuracy, highlighting their potential use as biomarkers for radiation damage and oxidative stress.
Collapse
Affiliation(s)
- Yaoxiang Li
- Department of Oncology, Lombardi Comprehensive Cancer Centre, Georgetown University Medical Center, Washington, DC 20057, USA; (Y.L.); (S.B.); (B.S.); (M.M.J.)
- Departments of Biochemistry, Molecular, and Cellular Biology, Georgetown University Medical Center, Washington, DC 20057, USA;
| | - Shivani Bansal
- Department of Oncology, Lombardi Comprehensive Cancer Centre, Georgetown University Medical Center, Washington, DC 20057, USA; (Y.L.); (S.B.); (B.S.); (M.M.J.)
| | - Baldev Singh
- Department of Oncology, Lombardi Comprehensive Cancer Centre, Georgetown University Medical Center, Washington, DC 20057, USA; (Y.L.); (S.B.); (B.S.); (M.M.J.)
| | - Meth M. Jayatilake
- Department of Oncology, Lombardi Comprehensive Cancer Centre, Georgetown University Medical Center, Washington, DC 20057, USA; (Y.L.); (S.B.); (B.S.); (M.M.J.)
| | - William Klotzbier
- Departments of Biochemistry, Molecular, and Cellular Biology, Georgetown University Medical Center, Washington, DC 20057, USA;
| | - Marjan Boerma
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Mi-Heon Lee
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90024, USA; (M.-H.L.); (J.H.); (K.S.I.); (D.S.)
| | - Jacob Hack
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90024, USA; (M.-H.L.); (J.H.); (K.S.I.); (D.S.)
| | - Keisuke S. Iwamoto
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90024, USA; (M.-H.L.); (J.H.); (K.S.I.); (D.S.)
| | - Dörthe Schaue
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90024, USA; (M.-H.L.); (J.H.); (K.S.I.); (D.S.)
| | - Amrita K. Cheema
- Department of Oncology, Lombardi Comprehensive Cancer Centre, Georgetown University Medical Center, Washington, DC 20057, USA; (Y.L.); (S.B.); (B.S.); (M.M.J.)
- Departments of Biochemistry, Molecular, and Cellular Biology, Georgetown University Medical Center, Washington, DC 20057, USA;
| |
Collapse
|
3
|
Maremonti E, Brede DA, Kassaye YA, Zheng K, Lee Y, Salbu B, Teien HC. Dose rate dependent genotoxic and metabolic effects predict onset of impaired development and mortality in Atlantic salmon (S. salar) embryos exposed to chronic gamma radiation. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 954:176263. [PMID: 39278484 DOI: 10.1016/j.scitotenv.2024.176263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 06/04/2024] [Accepted: 09/12/2024] [Indexed: 09/18/2024]
Abstract
Release of radionuclides to the environment from either nuclear weapon and fuel cycles or from naturally occurring radionuclides (NORM) may cause long term contamination of aquatic ecosystems and chronic exposure of living organisms to ionizing radiation, which in turn could lead to adverse effects compromising the sustainability of populations. To address the effects of chronic ionizing radiation on the development of fish, Atlantic salmon embryos were exposed from fertilization until hatching (88 days, 550 day-degree) to dose rates from 1 to 30 mGy·h-1 gamma radiation (60Co). The lowest adopted dose rate was similar to the highest doses measured in some water bodies right after the Chernobyl accident (1 mGy·h-1), however, well above current environmentally realistic scenarios (20 μGy·h-1), or the threshold assumed for significant effects on fish population (40 μGy·h-1). Dose dependent effects were observed on survival, hatching, morbidity, DNA damage, antioxidant defenses, and metabolic status. Histopathological analysis showed dose rate dependent impairment of eye and brain tissues development and establishment of epidermal mucus cell layers accompanied by increased DNA damage at doses ≥1.3 Gy (dose rates ≥1 mGy·h-1). At ≥32.8 Gy (dose rates ≥20 mGy·h-1) deformities and developmental growth defects resulted in respective 46 and 95 % pre-hatch mortality. The 10 mGy·h-1 exposure (≥ 12 Gy total dose) caused significantly increased DNA damage, impaired eye development, and both premature and delayed hatching, while no deformities or effect on survival were observed. We observed a dose rate dependent reduction from dose rate ≥ 20 mGy·h-1 (≥ 27 Gy total dose) on antioxidant SOD, catalase and glutathione reductase enzyme activities. The reduction of antioxidant enzyme activities was in line with observed developmental delay and disturbance to time of hatching. Metabolomic profiles showed a clear shift at dose rates ≥10 mGy·h-1 (≥ 12 Gy total dose) in pathways related to oxidative stress, detoxification, DNA damage and repair. Due to gamma radiation exposure, a switch of central metabolism from glycolysis, citric acid cycle and lactate production towards pentose phosphate pathway indicated a rewiring mechanism for increased production of reductive equivalents to maintain redox homeostasis at the expense of energy output and thus embryonic development.
Collapse
Affiliation(s)
- Erica Maremonti
- Centre for Environmental Radioactivity (CERAD), Faculty of Environmental Sciences and Natural Resource Management (MINA), Norwegian University of Life Sciences (NMBU), 1432 Ås, Norway.
| | - Dag Anders Brede
- Centre for Environmental Radioactivity (CERAD), Faculty of Environmental Sciences and Natural Resource Management (MINA), Norwegian University of Life Sciences (NMBU), 1432 Ås, Norway
| | - Yetneberk A Kassaye
- Centre for Environmental Radioactivity (CERAD), Faculty of Environmental Sciences and Natural Resource Management (MINA), Norwegian University of Life Sciences (NMBU), 1432 Ås, Norway
| | - Keke Zheng
- Centre for Environmental Radioactivity (CERAD), Faculty of Environmental Sciences and Natural Resource Management (MINA), Norwegian University of Life Sciences (NMBU), 1432 Ås, Norway
| | - YeonKyeong Lee
- Korea University Graduate School, Department of Plant Biotechnology, 145, Anam-ro, Seongbuk-ku, Seoul, Republic of Korea
| | - Brit Salbu
- Centre for Environmental Radioactivity (CERAD), Faculty of Environmental Sciences and Natural Resource Management (MINA), Norwegian University of Life Sciences (NMBU), 1432 Ås, Norway
| | - Hans-Christian Teien
- Centre for Environmental Radioactivity (CERAD), Faculty of Environmental Sciences and Natural Resource Management (MINA), Norwegian University of Life Sciences (NMBU), 1432 Ås, Norway
| |
Collapse
|
4
|
Narasimhamurthy RK, Venkidesh BS, Vasishta S, Joshi MB, Rao BSS, Sharan K, Dattaram Mumbrekar K. Low-Dose Radiation Induces Alterations in Fatty Acid and Tyrosine Metabolism in the Mouse Hippocampus: Insights from Integrated Multiomics. ACS Chem Neurosci 2024; 15:3311-3320. [PMID: 39185768 DOI: 10.1021/acschemneuro.4c00231] [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] [Indexed: 08/27/2024] Open
Abstract
In recent years, there has been a drastic surge in neurological disorders with sporadic cases contributing more than ever to their cause. Radiation exposure through diagnostic or therapeutic routes often results in neurological injuries that may lead to neurodegenerative pathogenesis. However, the underlying mechanisms regulating the neurological impact of exposure to near-low doses of ionizing radiation are not known. In particular, the neurological changes caused by metabolomic reprogramming have not yet been elucidated. Hence, in the present study, C57BL/6 mice were exposed to a single whole-body X-ray dose of 0.5 Gy, and 14 days post-treatment, the hippocampus was subjected to metabolomic analysis. The hippocampus of the irradiated animals showed significant alterations in 15 metabolites, which aligned with altered tyrosine, phenylalanine, and alpha-linolenic acid metabolism and the biosynthesis of unsaturated fatty acids. Furthermore, a multiomics interaction network comprising metabolomics and RNA sequencing data analysis provided insights into gene-metabolite interactions. Tyrosine metabolism was revealed to be the most altered, which was demonstrated by the interaction of several crucial genes and metabolites. The present study revealed the regulation of low-dose radiation-induced neurotoxicity at the metabolomic level and its implications for the pathogenesis of neurological disorders. The present study also provides novel insights into metabolomic pathways altered following near-low-dose IR exposure and its link with neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease.
Collapse
Affiliation(s)
- Rekha Koravadi Narasimhamurthy
- Department of Radiation Biology & Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Babu Santhi Venkidesh
- Department of Radiation Biology & Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Sampara Vasishta
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Manjunath B Joshi
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Bola Sadashiva Satish Rao
- Department of Radiation Biology & Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
- Directorate of Research, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Krishna Sharan
- Department of Radiation Therapy and Oncology, K S Hegde Medical Academy (KSHEMA), Nitte (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Kamalesh Dattaram Mumbrekar
- Department of Radiation Biology & Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| |
Collapse
|
5
|
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
|
6
|
Okunieff P, Swarts SG, Fenton B, Zhang SB, Zhang Z, Rice L, Zhou D, Carrier F, Zhang L. Radiation Biological Toximetry Using Circulating Cell-Free DNA (cfDNA) for Rapid Radiation/Nuclear Triage. Radiat Res 2024; 202:70-79. [PMID: 38661544 PMCID: PMC11346512 DOI: 10.1667/rade-23-00159.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 04/10/2024] [Indexed: 04/26/2024]
Abstract
Optimal triage biodosimetry would include risk stratification within minutes, and it would provide useful triage despite heterogeneous dosimetry, cytokine therapy, mixed radiation quality, race, and age. For regulatory approval, the U.S. Food and Drug Administration (FDA) Biodosimetry Guidance requires suitability for purpose and a validated species-independent mechanism. Circulating cell-free DNA (cfDNA) concentration assays may provide such triage information. To test this hypothesis, cfDNA concentrations were measured in unprocessed monkey plasma using a branched DNA (bDNA) technique with a laboratory developed test. The cfDNA levels, along with hematopoietic parameters, were measured over a 7-day period in Rhesus macaques receiving total body radiation doses ranging from 1 to 6.5 Gy. Low-dose irradiation (0-2 Gy) was easily distinguished from high-dose whole-body exposures (5.5 and 6.5 Gy). Fold changes in cfDNA in the monkey model were comparable to those measured in a bone marrow transplant patient receiving a supralethal radiation dose, suggesting that the lethal threshold of cfDNA concentrations may be similar across species. Average cfDNA levels were 50 ± 40 ng/mL [±1 standard deviation (SD)] pre-irradiation, 120 ± 13 ng/mL at 1 Gy; 242 ± 71 ng/mL at 2 Gy; 607 ± 54 at 5.5 Gy; and 1585 ± 351 at 6.5 Gy (±1 SD). There was an exponential increase in cfDNA concentration with radiation dose. Comparison of the monkey model with the mouse model and the Guskova model, developed using Chernobyl responder data, further demonstrated correlation across species, supporting a similar mechanism of action. The test is available commercially in a Clinical Laboratory Improvement Amendments (CLIA) ready form in the U.S. and the European Union. The remaining challenges include developing methods for further simplification of specimen processing and assay evaluation, as well as more accurate calibration of the triage category with cfDNA concentration cutoffs.
Collapse
Affiliation(s)
- Paul Okunieff
- Department of Radiation Oncology, University of Florida, Gainesville, Florida
| | - Steven G. Swarts
- Department of Radiation Oncology, University of Florida, Gainesville, Florida
| | - Bruce Fenton
- School of Medicine and Dentistry, University of Rochester Medical Center, Rochester, New York
| | - Steven B. Zhang
- Department of Radiation Oncology, University of Florida, Gainesville, Florida
| | - Zhenhuan Zhang
- Department of Radiation Oncology, University of Florida, Gainesville, Florida
| | - Lori Rice
- Department of Radiation Oncology, University of Florida, Gainesville, Florida
| | - Daohong Zhou
- Department of Biochemistry and Structural Biology, Center for Innovative Drug Discovery (CIDD), University of Texas Health San Antonio, Texas
| | - France Carrier
- Department of Radiation Oncology, School of Medicine, University of Maryland, Baltimore, Maryland
| | - Lurong Zhang
- Department of Radiation Oncology, University of Florida, Gainesville, Florida
- First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| |
Collapse
|
7
|
Chen S, Xie DF, Li S, Luo J, Han Y, Guo H, Gao S, Huang X, Guan H, Huang R, Zhou PK. TAB182 regulates glycolytic metabolism by controlling LDHA transcription to impact tumor radiosensitivity. Cell Death Dis 2024; 15:209. [PMID: 38480704 PMCID: PMC10937931 DOI: 10.1038/s41419-024-06588-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 02/29/2024] [Accepted: 03/04/2024] [Indexed: 03/17/2024]
Abstract
Metabolic reprogramming, a hallmark of cancer, is closely associated with tumor development and progression. Changes in glycolysis play a crucial role in conferring radiation resistance to tumor cells. How radiation changes the glycolysis status of cancer cells is still unclear. Here we revealed the role of TAB182 in regulating glycolysis and lactate production in cellular response to ionizing radiation. Irradiation can significantly stimulate the production of TAB182 protein, and inhibiting TAB182 increases cellular radiosensitivity. Proteomic analysis indicated that TAB182 influences several vital biological processes, including multiple metabolic pathways. Knockdown of TAB182 results in decreased lactate production and increased pyruvate and ATP levels in cancer cells. Moreover, knocking down TAB182 reverses radiation-induced metabolic changes, such as radioresistant-related lactate production. TAB182 is necessary for activating LDHA transcription by affecting transcription factors SP1 and c-MYC; its knockdown attenuates the upregulation of LDHA by radiation, subsequently suppressing lactate production. Targeted suppression of TAB182 significantly enhances the sensitivity of murine xenograft tumors to radiotherapy. These findings advance our understanding of glycolytic metabolism regulation in response to ionizing radiation, which may offer significant implications for developing new strategies to overcome tumor radioresistance.
Collapse
Affiliation(s)
- Shi Chen
- School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan Province, 421001, P. R. China
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Da-Fei Xie
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Saiyu Li
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
- School of Life Sciences, Hebei University, Baoding, Hebei Province, 071002, P. R. China
| | - Jinhua Luo
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan, 410078, P. R. China
| | - Yang Han
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Hejiang Guo
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Shuaining Gao
- School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan Province, 421001, P. R. China
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Xin Huang
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Hua Guan
- School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan Province, 421001, P. R. China.
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China.
| | - Ruixue Huang
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan, 410078, P. R. China.
| | - Ping-Kun Zhou
- School of Public Health, Hengyang Medical School, University of South China, Hengyang, Hunan Province, 421001, P. R. China.
- Department of Radiation Biology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China.
| |
Collapse
|
8
|
Lv M, Shao S, Du Y, Zhuang X, Wang X, Qiao T. Plasma Lipidomics Profiling to Identify the Biomarkers of Diagnosis and Radiotherapy Response for Advanced Non-Small-Cell Lung Cancer Patients. J Lipids 2024; 2024:6730504. [PMID: 38312939 PMCID: PMC10838201 DOI: 10.1155/2024/6730504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/21/2023] [Accepted: 01/10/2024] [Indexed: 02/06/2024] Open
Abstract
Background Advanced lung cancer that contributes to a heavy burden on medical institutions is the leading cause of cancer-related death and is often accompanied by metabolic disorders. In this study, we aimed to explore the biomarkers of diagnosis and radiotherapy response in non-small-cell lung cancer (NSCLC) patients by plasma lipidomics analysis. Method Using triple-quadrupole mass spectrometer analysis, our research characterized the plasma lipid metabolomics profile of 25 healthy controls and 31 advanced NSCLC patients in each of three different radiotherapy phases. Results The results showed altered lipid elements and concentrations among NSCLC patients with different radiotherapy phases, NSCLC subtypes, and different radiotherapeutic responses. We found that compared to the healthy controls, myelin-associated glycoprotein (MAG), phosphatidylinositol (PI), and phosphatidylserine (PS) were mainly and significantly altered lipid elements (> twofold, and p < 0.05) among NSCLC patients with different radiotherapy phases. Through comparison of lipid elements between bad and good responses of NSCLC patients with radiotherapy, the obviously declined phosphatidylglycerol (PG 18 : 0/14 : 0, 18 : 1/18 : 3, and 18 : 0/20 : 1) or markedly elevated PI (20 : 0/22 : 5 and 18 : 2/22 : 4) and phosphatidic acid (PA 14 : 0/20 : 4, 14 : 0/20 : 3, and 18 : 2/22 : 4) could indicate poor therapeutic response for NSCLC patients. The results of ROC curve analysis suggested that PG (18 : 0/20 : 1 and 18 : 0/14 : 0) could clearly predict the radiotherapeutic response for NSCLC patients, and PS (18 : 0/20 : 0) and cholesterol were the first two lipid components with the most potential for the diagnosis of advanced NSCLC. Conclusion Our results indicated that plasma lipidomics profiling might have a vital value to uncover the heterogeneity of lipid metabolism in healthy people and advanced NSCLC patients with different radiotherapy phase, and further to screen out radiotherapeutic response-specific biomarkers.
Collapse
Affiliation(s)
- Minghe Lv
- Center for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Jinshan District, Shanghai 201508, China
- Department of Radiotherapy, Shuguang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Pudong New Area, Shanghai 201203, China
| | - Shali Shao
- Center for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Jinshan District, Shanghai 201508, China
| | - Yajing Du
- Center for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Jinshan District, Shanghai 201508, China
| | - Xibing Zhuang
- Center for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Jinshan District, Shanghai 201508, China
| | - Xiangdong Wang
- Center for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Jinshan District, Shanghai 201508, China
| | - Tiankui Qiao
- Center for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Jinshan District, Shanghai 201508, China
| |
Collapse
|
9
|
Carpenter AD, Fatanmi OO, Wise SY, Petrus SA, Tyburski JB, Cheema AK, Singh VK. Metabolomic Changes in Plasma of Preterminal Stage of Rhesus Nonhuman Primates Exposed to Lethal Dose of Radiation. Metabolites 2023; 14:18. [PMID: 38248821 PMCID: PMC10819041 DOI: 10.3390/metabo14010018] [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: 11/20/2023] [Revised: 12/22/2023] [Accepted: 12/25/2023] [Indexed: 01/23/2024] Open
Abstract
Ionizing radiation exposure is known to induce molecular and cellular injury, inflicting a cascade of potentially catastrophic events leading to tissue and organ damage. Metabolomic analysis allows for the identification and quantification of small molecules downstream of genomic changes induced by radiation exposure. We aimed to characterize metabolomic changes that underscore the prefinal stage of lethally irradiated rhesus nonhuman primates (NHPs). Peripheral blood was drawn at baseline, post-exposure, as well as at the preterminal stage in NHPs (immediately prior to death in moribund NHPs) that did not survive exposure with 7.2 Gy total-body radiation (LD70/60). Herein, we analyzed global metabolomic changes using ultra-performance liquid chromatography (UPLC) quadrupole time-of-flight mass spectrometry (QTOF-MS) in plasma samples of NHPs collected at various timepoints in relation to irradiation. The overall goal was to identify metabolic shifts present immediately prior to death. Our findings showed radiation induced significant time-dependent metabolic perturbations when compared to pre-irradiation profiles, particularly in glycerophospholipid metabolism and steroid hormone biosynthesis and metabolism pathways. These findings provide valuable insights for identifying biomarkers for lethality, which may be helpful for triage during a mass casualty scenario.
Collapse
Affiliation(s)
- Alana D. Carpenter
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (A.D.C.)
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Oluseyi O. Fatanmi
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (A.D.C.)
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Stephen Y. Wise
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (A.D.C.)
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Sarah A. Petrus
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (A.D.C.)
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | | | - Amrita K. Cheema
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
- Department of Biochemistry, Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Vijay K. Singh
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (A.D.C.)
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| |
Collapse
|
10
|
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
|
11
|
Chakraborty N, Holmes-Hampton GP, Gautam A, Kumar R, Hritzo B, Legesse B, Dimitrov G, Ghosh SP, Hammamieh R. Early to sustained impacts of lethal radiation on circulating miRNAs in a minipig model. Sci Rep 2023; 13:18496. [PMID: 37898651 PMCID: PMC10613244 DOI: 10.1038/s41598-023-45250-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 10/17/2023] [Indexed: 10/30/2023] Open
Abstract
Early diagnosis of lethal radiation is imperative since its intervention time windows are considerably short. Hence, ideal diagnostic candidates of radiation should be easily accessible, enable to inform about the stress history and objectively triage subjects in a time-efficient manner. Therefore, the small molecules such as metabolites and microRNAs (miRNAs) from plasma are legitimate biomarker candidate for lethal radiation. Our objectives were to comprehend the radiation-driven molecular pathogenesis and thereby determine biomarkers of translational potential. We investigated an established minipig model of LD70/45 total body irradiation (TBI). In this pilot study, plasma was collected pre-TBI and at multiple time points post-TBI. The majority of differentially expressed miRNAs and metabolites were perturbed immediately after TBI that potentially underlined the severity of its acute impact. The integrative network analysis of miRNA and metabolites showed a cohesive response; the early and consistent perturbations of networks were linked to cancer and the shift in musculoskeletal atrophy synchronized with the comorbidity-networks associated with inflammation and bioenergy synthesis. Subsequent comparative pipeline delivered 92 miRNAs, which demonstrated sequential homology between human and minipig, and potentially similar responses to lethal radiation across these two species. This panel promised to retrospectively inform the time since the radiation occurred; thereby could facilitate knowledge-driven interventions.
Collapse
Affiliation(s)
- Nabarun Chakraborty
- Medical Readiness Systems Biology, CMPN, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | - Gregory P Holmes-Hampton
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, 20889, USA
| | - Aarti Gautam
- Medical Readiness Systems Biology, CMPN, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | - Raina Kumar
- The Geneva Foundation, US Army Center for Environmental Health Research, Fort Detrick, MD, 21702-5010, USA
| | - Bernadette Hritzo
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, 20889, USA
| | - Betre Legesse
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, 20889, USA
| | - George Dimitrov
- Medical Readiness Systems Biology, CMPN, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- The Geneva Foundation, US Army Center for Environmental Health Research, Fort Detrick, MD, 21702-5010, USA
| | - Sanchita P Ghosh
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, 20889, USA.
| | - Rasha Hammamieh
- Medical Readiness Systems Biology, CMPN, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| |
Collapse
|
12
|
Wang X, Yuan R, Miao L, Li X, Guo Y, Tian H. Protective mechanism of a novel aminothiol compound on radiation-induced intestinal injury. Int J Radiat Biol 2023; 99:259-269. [PMID: 35583501 DOI: 10.1080/09553002.2022.2074163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE With the development of nuclear technology and radiotherapy, the risk of radiation injury has been increasing. Therefore, it is important to find an effective radiation-protective agent. In this study, we designed and synthesized a novel compound called compound 8, of which the radioprotective effect and mechanism were studied. MATERIALS AND METHODS Before being exposed to ionizing radiation, mice were pretreated with compound 8. The 30-day mortality assay, hematoxylin-eosin staining, and immunohistochemistry staining assay were performed to evaluate the anti-radiation effect of the compound 8. TUNEL and immunofluorescence assays were conducted to study the anti-radiation mechanism of compound 8. RESULTS Compared to the IR + vehicle group, the 30-day survival rate of mice treated with 25 mg/kg of compound 8 was significantly improved after 8 Gy total body irradiation. In the morphological study of the small intestine, we found that compound 8 could maintain crypt-villus structures in the irradiated mice. Further immunohistochemical staining displayed that compound 8 could improve the survival of Lgr5+ cells, ki67+ cells, and lysozyme+ cells. The results of TUNEL and immunofluorescence assays showed that compound 8 could decrease the expression of apoptosis-related caspase-8/-9, γ-H2AX, Bax, and p53. CONCLUSIONS These results indicate that compound 8 exerts its effects by maintaining structure and function of small intestine. It also reduces DNA damage, promotes crypt proliferation and differentiation. Moreover, it may enhance the anti-apoptotic ability of small intestinal tissue by inhibiting the activation of p53 and blocking the caspase cascade reaction. Compound 8 can protect the intestinal tract from post-radiation damage, it is thus a new and effective protective agent of radiation.
Collapse
Affiliation(s)
- Xinxin Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, China
| | - Renbin Yuan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, China
| | - Longfei Miao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, China
| | - Xuejiao Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, China
| | - Yuying Guo
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, China
| | - Hongqi Tian
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, China
| |
Collapse
|
13
|
Shakyawar SK, Mishra NK, Vellichirammal NN, Cary L, Helikar T, Powers R, Oberley-Deegan RE, Berkowitz DB, Bayles KW, Singh VK, Guda C. A Review of Radiation-Induced Alterations of Multi-Omic Profiles, Radiation Injury Biomarkers, and Countermeasures. Radiat Res 2023; 199:89-111. [PMID: 36368026 PMCID: PMC10279411 DOI: 10.1667/rade-21-00187.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 10/24/2022] [Indexed: 11/13/2022]
Abstract
Increasing utilization of nuclear power enhances the risks associated with industrial accidents, occupational hazards, and the threat of nuclear terrorism. Exposure to ionizing radiation interferes with genomic stability and gene expression resulting in the disruption of normal metabolic processes in cells and organs by inducing complex biological responses. Exposure to high-dose radiation causes acute radiation syndrome, which leads to hematopoietic, gastrointestinal, cerebrovascular, and many other organ-specific injuries. Altered genomic variations, gene expression, metabolite concentrations, and microbiota profiles in blood plasma or tissue samples reflect the whole-body radiation injuries. Hence, multi-omic profiles obtained from high-resolution omics platforms offer a holistic approach for identifying reliable biomarkers to predict the radiation injury of organs and tissues resulting from radiation exposures. In this review, we performed a literature search to systematically catalog the radiation-induced alterations from multi-omic studies and radiation countermeasures. We covered radiation-induced changes in the genomic, transcriptomic, proteomic, metabolomic, lipidomic, and microbiome profiles. Furthermore, we have covered promising multi-omic biomarkers, FDA-approved countermeasure drugs, and other radiation countermeasures that include radioprotectors and radiomitigators. This review presents an overview of radiation-induced alterations of multi-omics profiles and biomarkers, and associated radiation countermeasures.
Collapse
Affiliation(s)
- Sushil K Shakyawar
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Nitish K Mishra
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Neetha N Vellichirammal
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Lynnette Cary
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Tomáš Helikar
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln NE 65888, USA
| | - Robert Powers
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln NE 65888, USA
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln NE 68588, USA
| | - Rebecca E Oberley-Deegan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - David B Berkowitz
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln NE 65888, USA
| | - Kenneth W Bayles
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Vijay K Singh
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Chittibabu Guda
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Center for Biomedical Informatics Research and Innovation, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
14
|
Falcone M, Uribe AH, Papalazarou V, Newman AC, Athineos D, Stevenson K, Sauvé CEG, Gao Y, Kim JK, Del Latto M, Kierstead M, Wu C, Smith JJ, Romesser PB, Chalmers AJ, Blyth K, Maddocks ODK. Sensitisation of cancer cells to radiotherapy by serine and glycine starvation. Br J Cancer 2022; 127:1773-1786. [PMID: 36115879 PMCID: PMC9643498 DOI: 10.1038/s41416-022-01965-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 08/10/2022] [Accepted: 08/19/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Cellular metabolism is an integral component of cellular adaptation to stress, playing a pivotal role in the resistance of cancer cells to various treatment modalities, including radiotherapy. In response to radiotherapy, cancer cells engage antioxidant and DNA repair mechanisms which mitigate and remove DNA damage, facilitating cancer cell survival. Given the reliance of these resistance mechanisms on amino acid metabolism, we hypothesised that controlling the exogenous availability of the non-essential amino acids serine and glycine would radiosensitise cancer cells. METHODS We exposed colorectal, breast and pancreatic cancer cell lines/organoids to radiation in vitro and in vivo in the presence and absence of exogenous serine and glycine. We performed phenotypic assays for DNA damage, cell cycle, ROS levels and cell death, combined with a high-resolution untargeted LCMS metabolomics and RNA-Seq. RESULTS Serine and glycine restriction sensitised a range of cancer cell lines, patient-derived organoids and syngeneic mouse tumour models to radiotherapy. Comprehensive metabolomic and transcriptomic analysis of central carbon metabolism revealed that amino acid restriction impacted not only antioxidant response and nucleotide synthesis but had a marked inhibitory effect on the TCA cycle. CONCLUSION Dietary restriction of serine and glycine is a viable radio-sensitisation strategy in cancer.
Collapse
Affiliation(s)
- Mattia Falcone
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, UK
- Division of Oncogenomics, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Alejandro Huerta Uribe
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, UK
| | - Vasileios Papalazarou
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, UK
| | - Alice C Newman
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, UK
| | | | - Katrina Stevenson
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, UK
| | | | - Yajing Gao
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Jin K Kim
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael Del Latto
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Maria Kierstead
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Chao Wu
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - J Joshua Smith
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Paul B Romesser
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Early Drug Development Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Anthony J Chalmers
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, UK
| | - Karen Blyth
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, UK
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Oliver D K Maddocks
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, UK.
| |
Collapse
|
15
|
Malik S, Kaur K, Prasad S, Jha NK, Kumar V. A perspective review on medicinal plant resources for their antimutagenic potentials. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:62014-62029. [PMID: 34431051 DOI: 10.1007/s11356-021-16057-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/16/2021] [Indexed: 06/13/2023]
Abstract
Mutagens present in the environment manifest toxic effects and are considered as serious threat for human health and healthcare. Recent reports reveal that medicinal plant resources are being explored for identifying potent antimutagenic as well as cancer preventing agents. There is mounting evidence that cancer and other mutation-related diseases can be prevented with the use of medicinal pant resources including crude extracts, active fractions, phytochemicals, and pure phytomolecules. These medicinal plant resources possessing antimutagenic potentials have been shown to target molecular mechanisms underlying the mutagenic impacts. Technological advents and high-throughput screening/activity methods have revolutionized this field, though several potent plants and their active principles have been reported as effective antimutagens. The translational success rate needs to be improved, but the trends are encouraging. In this review, we present the current understandings and updates on various mutagens in the environment, toxicities related/attributed to them, the resultant mutations (and cancer), and how medicinal plants come to the rescue. A perspective review has been presented on whether and how medicinal plant resources can be an effective approach for addressing mutagens in the environment. An account of medicinal plant resources used as antimutagenic agents has been given along with the underlying mechanism of action and their therapeutic potential in various models of cancer. Recent success stories, current challenges, and future prospects are discussed.
Collapse
Affiliation(s)
- Sumira Malik
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi, Jharkhand, India
| | - Kawaljeet Kaur
- Department of Biotechnology, Modern College of Arts, Science and Commerce, Savitribai Phule Pune University, Ganeshkhind, Pune, 411016, India
| | - Shilpa Prasad
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi, Jharkhand, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, Uttar Pradesh, 201310, India
| | - Vinay Kumar
- Department of Biotechnology, Modern College of Arts, Science and Commerce, Savitribai Phule Pune University, Ganeshkhind, Pune, 411016, India.
- Department of Environmental Science, Savitribai Phule Pune University, Pune, 411007, India.
| |
Collapse
|
16
|
Wei S, Wei Y, Gong Y, Chen Y, Cui J, Li L, Yan H, Yu Y, Lin X, Li G, Yi L. Metabolomics as a valid analytical technique in environmental exposure research: application and progress. Metabolomics 2022; 18:35. [PMID: 35639180 DOI: 10.1007/s11306-022-01895-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 05/06/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND In recent years, studies have shown that exposure to environmental pollutants (e.g., radiation, heavy metal substances, air pollutants, organic pollutants) is a leading cause of human non-communicable diseases. The key to disease prevention is to clarify the harmful mechanisms and toxic effects of environmental pollutants on the body. Metabolomics is a high-sensitivity, high-throughput omics technology that can obtain detailed metabolite information of an organism. It is a crucial tool for gaining a comprehensive understanding of the pathway network regulation mechanism of the organism. Its application is widespread in many research fields such as environmental exposure assessment, medicine, systems biology, and biomarker discovery. AIM OF REVIEW Recent findings show that metabolomics can be used to obtain molecular snapshots of organisms after environmental exposure, to help understand the interaction between environmental exposure and organisms, and to identify potential biomarkers and biological mechanisms. KEY SCIENTIFIC CONCEPTS OF REVIEW This review focuses on the application of metabolomics to understand the biological effects of radiation, heavy metals, air pollution, and persistent organic pollutants exposure, and examines some potential biomarkers and toxicity mechanisms.
Collapse
Affiliation(s)
- Shuang Wei
- Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Department of Education, Institute of Cytology and Genetics, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Yuanyun Wei
- Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Department of Education, Institute of Cytology and Genetics, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Yaqi Gong
- Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Department of Education, Institute of Cytology and Genetics, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Yonglin Chen
- Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Department of Education, Institute of Cytology and Genetics, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Jian Cui
- Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Linwei Li
- Hengyang Medical School, The Second Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Hongxia Yan
- Hengyang Medical School, The Second Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Yueqiu Yu
- Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Department of Education, Institute of Cytology and Genetics, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Xiang Lin
- Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Department of Education, Institute of Cytology and Genetics, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Guoqing Li
- Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Department of Education, Institute of Cytology and Genetics, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Lan Yi
- Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Department of Education, Institute of Cytology and Genetics, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
17
|
Zhou Y, Liu J, Li X, Wang L, Hu L, Li A, Zhou J. JAC4 Protects from X-Ray Radiation-Induced Intestinal Injury by JWA-Mediated Anti-Oxidation/Inflammation Signaling. Antioxidants (Basel) 2022; 11:antiox11061067. [PMID: 35739964 PMCID: PMC9220415 DOI: 10.3390/antiox11061067] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 05/24/2022] [Accepted: 05/25/2022] [Indexed: 02/01/2023] Open
Abstract
Radiation-induced intestinal injury is one of the major side effects in patients receiving radiation therapy. There is no specific treatment for radiation-induced enteritis in the clinic. We synthesized a compound, named JAC4, which is an agonist and can increase JWA protein expression. JWA has been shown to reduce oxidative stress, DNA damage, anti-apoptosis, and anti-inflammatory; in addition, the small intestine epithelium showed dysplasia in JWA knockout mice. We hypothesized that JAC4 might exert a protective effect against radiation-induced intestinal damage. Herein, X-ray radiation models were built both in mice and in intestinal crypt epithelial cells (IEC-6). C57BL/6J mice were treated with JAC4 by gavage before abdominal irradiation (ABI); the data showed that JAC4 significantly reduced radiation-induced intestinal mucosal damage and increased the survival rate. In addition, radiation-induced oxidative stress damage and systemic inflammatory response were also mitigated by JAC4 treatment. Moreover, JAC4 treatment alleviated DNA damage, decreased cell apoptosis, and maintained intestinal epithelial cell proliferation in mice. In vitro data showed that JAC4 treatment significantly inhibited ROS formation and cell apoptosis. Importantly, all the above protective effects of JAC4 on X-ray radiation-triggered intestinal injury were no longer determined in the intestinal epithelium of JWA knockout mice. Therefore, our results provide the first evidence that JAC4 protects the intestine from radiation-induced enteritis through JWA-mediated anti-oxidation/inflammation signaling.
Collapse
Affiliation(s)
- Yan Zhou
- Department of Molecular Cell Biology & Toxicology, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China; (Y.Z.); (J.L.); (X.L.); (L.W.); (A.L.)
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Jingwen Liu
- Department of Molecular Cell Biology & Toxicology, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China; (Y.Z.); (J.L.); (X.L.); (L.W.); (A.L.)
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Xiong Li
- Department of Molecular Cell Biology & Toxicology, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China; (Y.Z.); (J.L.); (X.L.); (L.W.); (A.L.)
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Luman Wang
- Department of Molecular Cell Biology & Toxicology, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China; (Y.Z.); (J.L.); (X.L.); (L.W.); (A.L.)
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Lirong Hu
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co., Ltd., Nanjing 210042, China;
| | - Aiping Li
- Department of Molecular Cell Biology & Toxicology, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China; (Y.Z.); (J.L.); (X.L.); (L.W.); (A.L.)
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Jianwei Zhou
- Department of Molecular Cell Biology & Toxicology, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China; (Y.Z.); (J.L.); (X.L.); (L.W.); (A.L.)
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
- Correspondence:
| |
Collapse
|
18
|
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]
|
19
|
Korimerla N, Wahl DR. Interactions between Radiation and One-Carbon Metabolism. Int J Mol Sci 2022; 23:1919. [PMID: 35163841 PMCID: PMC8836916 DOI: 10.3390/ijms23031919] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/29/2022] [Accepted: 02/04/2022] [Indexed: 02/07/2023] Open
Abstract
Metabolic reprogramming is a hallmark of cancer. Cancer cells rewire one-carbon metabolism, a central metabolic pathway, to turn nutritional inputs into essential biomolecules required for cancer cell growth and maintenance. Radiation therapy, a common cancer therapy, also interacts and alters one-carbon metabolism. This review discusses the interactions between radiation therapy, one-carbon metabolism and its component metabolic pathways.
Collapse
Affiliation(s)
- Navyateja Korimerla
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA;
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Daniel R. Wahl
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA;
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
20
|
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
|
21
|
Padala SR, Kashyap B, Dekker H, Mikkonen JJW, Palander A, Bravenboer N, Kullaa AM. Irradiation affects the structural, cellular and molecular components of jawbones. Int J Radiat Biol 2021; 98:136-147. [PMID: 34855558 DOI: 10.1080/09553002.2022.2013568] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE Emerging evidence shows that changes in the bone and its microenvironment following radiotherapy are associated with either an inhibition or a state of low bone formation. Ionizing radiation is damaging to the jawbone as it increases the complication rate due to the development of hypovascular, hypocellular, and hypoxic tissue. This review summarizes and correlates the current knowledge on the effects of irradiation on the bone with an emphasis on jawbone, as these have been a less extensively studied area. CONCLUSIONS The stringent regulation of bone formation and bone resorption can be influenced by radiation, causing detrimental effects at structural, cellular, vascular, and molecular levels. It is also associated with a high risk of damage to surrounding healthy tissues and an increased risk of fracture. Technological advances and research on animal models as well as a few human bone tissue studies have provided novel insights into the ways in which bone can be affected by high, low and sublethal dose of radiation. The influence of radiation on bone metabolism, cellular properties, vascularity, collagen, and other factors like inflammation, reactive oxygen species are discussed.
Collapse
Affiliation(s)
- Sridhar Reddy Padala
- Institute of Dentistry, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Bina Kashyap
- Institute of Dentistry, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Hannah Dekker
- Amsterdam University Medical Centers, Academic Centre for Dentistry Amsterdam (ACTA), Department of Oral and Maxillofacial Surgery/Oral Pathology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Jopi J W Mikkonen
- Institute of Dentistry, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Anni Palander
- Institute of Dentistry, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Nathalie Bravenboer
- Amsterdam UMC, Department of Clinical Chemistry, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands.,Department of Internal Medicine, Division of Endocrinology and Center for Bone Quality, Leiden University Medical Center, Leiden, The Netherlands
| | - Arja M Kullaa
- Institute of Dentistry, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
22
|
Li W, Wang T, Zhang X, Zhu J, Li XY, Peng F, Dai J, Wang J, Zhang L, Wang Y, Chen X, Xue T, Ding C, Wang C, Jiao L. Distinct lipid profiles of radiation-induced carotid plaques from atherosclerotic carotid plaques revealed by UPLC-QTOF-MS and DESI-MSI. Radiother Oncol 2021; 167:25-33. [PMID: 34902371 DOI: 10.1016/j.radonc.2021.12.006] [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: 09/14/2021] [Revised: 11/29/2021] [Accepted: 12/03/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND PURPOSE Radiotherapy is a standard treatment for head and neck tumors that significantly increases patients' long-term survival rates. However, late cerebrovascular complications, especially carotid artery stenosis (CAS), have gained increasing attention. Investigation of biomarkers of radiation-induced CAS may help to elucidate the mechanism by which radiation induces damage to blood vessels and identify possible preventive measures against such damage. MATERIALS AND METHODS In this study, we used lipidomics strategy to characterize the lipids present in 8 radiation-induced carotid plaques (RICPs) and 12 atherosclerotic carotid plaques (ASCPs). We also used desorption electrospray ionization-mass spectrometry imaging (DESI-MSI) to map the spatial distribution of the screened lipids from 2 RICPs samples and 2 ASCPs samples. RESULTS The results showed that 31 metabolites in RICPs were significantly higher than that in ASCPs, 24 of which were triglycerides (TGs). We used four machine learning models to select potential indicators from the 31 metabolites. Six TGs [TG(17:2/17:2/18:0), TG(17:1/17:2/18:0), TG(17:0/17:2/18:0), TG(17:2/17:2/20:0), TG(17:1/17:2/20:0), TG(15:0/22:0/22:2)] were found to be the potential markers for distinguishing RICPs and ASCPs (AUC = 0.83). The DESI-MSI results suggested that the 6 TGs were localized in the collagen fiber regions and confirmed the differences of these TGs between the two kinds of plaques. CONCLUSIONS The 6 TGs primarily localized in the collagen fiber regions of plaques are likely to be potential indicators for the differentiation of RICPs from ASCPs which may have implications in the mechanisms and possible preventive measures against RICPs.
Collapse
Affiliation(s)
- Wei Li
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China; Department of Neurosurgery, Liaocheng Brain Hospital, China; Department of Interventional Neuroradiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Tao Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiao Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Junge Zhu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xu-Ying Li
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Fangda Peng
- National Center for Occupational Safety and Health, NHC (National Center for Occupational Medicine of Coal Industry, NHC), Beijing, China
| | - Jing Dai
- National Center for Occupational Safety and Health, NHC (National Center for Occupational Medicine of Coal Industry, NHC), Beijing, China
| | - Jiyue Wang
- Department of Neurosurgery, Liaocheng Brain Hospital, China
| | - Liyong Zhang
- Department of Neurosurgery, Liaocheng Brain Hospital, China
| | - Yabing Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xianyang Chen
- Zhongguancun Biological and Medical Big Data Center, Beijing, China; Bao Feng Key Laboratory of Genetics and Metabolism, Beijing, China
| | - Teng Xue
- Zhongguancun Biological and Medical Big Data Center, Beijing, China; Zhongyuanborui Key Laborotory of Genetics and Metabolism, Guangdong-Macao In-depth Cooperation Zone in Hengqin, China
| | - Chunguang Ding
- National Center for Occupational Safety and Health, NHC (National Center for Occupational Medicine of Coal Industry, NHC), Beijing, China.
| | - Chaodong Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China; National Clinical Research Center for Geriatric Diseases, Beijing, China.
| | - Liqun Jiao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China; Department of Interventional Neuroradiology, Xuanwu Hospital, Capital Medical University, Beijing, China; China International Neuroscience Institute (China-INI), Beijing, China.
| |
Collapse
|
23
|
Ferreira MR, Sands CJ, Li JV, Andreyev JN, Chekmeneva E, Gulliford S, Marchesi J, Lewis MR, Dearnaley DP. Impact of Pelvic Radiation Therapy for Prostate Cancer on Global Metabolic Profiles and Microbiota-Driven Gastrointestinal Late Side Effects: A Longitudinal Observational Study. Int J Radiat Oncol Biol Phys 2021; 111:1204-1213. [PMID: 34352290 PMCID: PMC8609156 DOI: 10.1016/j.ijrobp.2021.07.1713] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/17/2021] [Accepted: 07/26/2021] [Indexed: 12/18/2022]
Abstract
PURPOSE Radiation therapy to the prostate and pelvic lymph nodes (PLNRT) is part of the curative treatment of high-risk prostate cancer. Yet, the broader influence of radiation therapy on patient physiology is poorly understood. We conducted comprehensive global metabolomic profiling of urine, plasma, and stools sampled from patients undergoing PLNRT for high-risk prostate cancer. METHODS AND MATERIALS Samples were taken from 32 patients at 6 timepoints: baseline, 2 to 3 and 4 to 5 weeks of PLNRT; and 3, 6, and 12 months after PLNRT. We characterized the global metabolome of urine and plasma using 1H nuclear magnetic resonance spectroscopy and ultraperformance liquid chromatography-mass spectrometry, and of stools with nuclear magnetic resonance. Linear mixed-effects modeling was used to investigate metabolic changes between timepoints for each biofluid and assay and determine metabolites of interest. RESULTS Metabolites in urine, plasma and stools changed significantly after PLNRT initiation. Metabolic profiles did not return to baseline up to 1 year post-PLNRT in any biofluid. Molecules associated with cardiovascular risk were increased in plasma. Pre-PLNRT fecal butyrate levels directly associated with increasing gastrointestinal side effects, as did a sharper fall in those levels during and up to 1 year postradiation therapy, mirroring our previous results with metataxonomics. CONCLUSIONS We showed for the first time that an overall metabolic effect is observed in patients undergoing PLNRT up to 1 year posttreatment. These metabolic changes may effect on long-term morbidity after treatment, which warrants further investigation.
Collapse
Affiliation(s)
- Miguel R Ferreira
- Academic Radiotherapy Department, The Institute of Cancer Research, London, United Kingdom; Clinical Oncology Department, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Clinical Oncology Department, Guys and St Thomas NHS Foundation Trust, London, United Kingdom; School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom.
| | - Caroline J Sands
- National Phenome Centre, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Jia V Li
- Department of Metabolism, Digestion and Reproduction, Imperial College, London, United Kingdom
| | - Jervoise N Andreyev
- Gastroenterology Department, United Lincolnshire Hospitals NHS Trust, Lincolnshire, United Kingdom
| | - Elena Chekmeneva
- National Phenome Centre, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Sarah Gulliford
- Academic Radiotherapy Department, The Institute of Cancer Research, London, United Kingdom; Radiotherapy Department, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Julian Marchesi
- Department of Metabolism, Digestion and Reproduction, Imperial College, London, United Kingdom; School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Matthew R Lewis
- National Phenome Centre, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - David P Dearnaley
- Academic Radiotherapy Department, The Institute of Cancer Research, London, United Kingdom; Clinical Oncology Department, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
24
|
Laiakis EC, Canadell MP, Grilj V, Harken AD, Garty GY, Brenner DJ, Smilenov L, Fornace AJ. Small Molecule Responses to Sequential Irradiation with Neutrons and Photons for Biodosimetry Applications: An Initial Assessment. Radiat Res 2021; 196:468-477. [PMID: 33857313 PMCID: PMC9004252 DOI: 10.1667/rade-20-00032.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 09/18/2020] [Indexed: 11/03/2022]
Abstract
Mass casualty exposure scenarios from an improvised nuclear device are expected to be far more complex than simple photons. Based on the proximity to the explosion and potential shielding, a mixed field of neutrons and photons comprised of up to approximately 30% neutrons of the total dose is anticipated. This presents significant challenges for biodosimetry and for short-term and long-term medical treatment of exposed populations. In this study we employed untargeted metabolomic methods to develop a biosignature in urine and serum from C57BL/6 mice to address radiation quality issues. The signature was developed in males and applied to samples from female mice to identify potential sex differences. Thirteen urinary (primarily amino acids, vitamin products, nucleotides) and 18 serum biomarkers (primarily mitochondrial and fatty acid β oxidation intermediates) were selected and evaluated in samples from day 1 and day 7 postirradiation. Sham-irradiated groups (controls) were compared to an equitoxic dose (3 Gy X-ray equivalent) from X rays (1.2 Gy/min), neutrons (∼1 Gy/h), or neutrons-photons. Results showed a time-dependent increase in the efficiency of the signatures, with serum providing the highest levels of accuracy in distinguishing not only between exposed from non-exposed populations, but also between radiation quality (photon exposures vs. exposures with a neutron component) and in between neutron-photon exposures (5, 15 or 25% of neutrons in the total dose) for evaluating the neutron contribution. A group of metabolites known as acylcarnitines was only responsive in males, indicating the potential for different mechanisms of action in baseline levels and of neutron-photon responses between the two sexes. Our findings highlight the potential of metabolomics in developing biodosimetric methods to evaluate mixed exposures with high sensitivity and specificity.
Collapse
Affiliation(s)
- Evagelia C. Laiakis
- Department of Oncology, Lombardi Comprehensive Cancer
Center, Georgetown University, Washington, DC
- Department of Biochemistry and Molecular & Cellular
Biology, Georgetown University, Washington, DC
| | | | - Veljko Grilj
- Radiological Research Accelerator Facility, Columbia
University, Irvington, New York
| | - Andrew D. Harken
- Radiological Research Accelerator Facility, Columbia
University, Irvington, 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
| | - Lubomir Smilenov
- Center for Radiological Research, Columbia University, New
York, New York
| | - Albert J. Fornace
- Department of Oncology, Lombardi Comprehensive Cancer
Center, Georgetown University, Washington, DC
- Department of Biochemistry and Molecular & Cellular
Biology, Georgetown University, Washington, DC
| |
Collapse
|
25
|
Sakauchi K, Taira W, Otaki JM. Metabolomic Response of the Creeping Wood Sorrel Oxalis corniculata to Low-Dose Radiation Exposure from Fukushima's Contaminated Soil. Life (Basel) 2021; 11:990. [PMID: 34575139 PMCID: PMC8472241 DOI: 10.3390/life11090990] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/15/2021] [Accepted: 09/17/2021] [Indexed: 12/25/2022] Open
Abstract
The biological consequences of the Fukushima nuclear accident have been intensively studied using the pale grass blue butterfly Zizeeria maha and its host plant, the creeping wood sorrel Oxalis corniculata. Here, we performed metabolomic analyses of Oxalis leaves from Okinawa to examine the plant metabolites that were upregulated or downregulated in response to low-dose radiation exposure from Fukushima's contaminated soil. The cumulative dose of radiation to the plants was 5.7 mGy (34 μGy/h for 7 days). The GC-MS analysis revealed a systematic tendency of downregulation among the metabolites, some of which were annotated as caproic acid, nonanoic acid, azelaic acid, and oleic acid. Others were annotated as fructose, glucose, and citric acid, involved in the carbohydrate metabolic pathways. Notably, the peak annotated as lauric acid was upregulated. In contrast, the LC-MS analysis detected many upregulated metabolites, some of which were annotated as either antioxidants or stress-related chemicals involved in defense pathways. Among them, only three metabolite peaks had a single annotation, one of which was alfuzosin, an antagonist of the α1-adrenergic receptor. We conclude that this Oxalis plant responded metabolically to low-dose radiation exposure from Fukushima's contaminated soil, which may mediate the ecological "field effects" of the developmental deterioration of butterflies in Fukushima.
Collapse
Affiliation(s)
- Ko Sakauchi
- The BCPH Unit of Molecular Physiology, Department of Chemistry, Biology and Marine Science, Faculty of Science, University of the Ryukyus, Okinawa 903-0213, Japan; (K.S.); (W.T.)
| | - Wataru Taira
- The BCPH Unit of Molecular Physiology, Department of Chemistry, Biology and Marine Science, Faculty of Science, University of the Ryukyus, Okinawa 903-0213, Japan; (K.S.); (W.T.)
- Center for Research Advancement and Collaboration, University of the Ryukyus, Okinawa 903-0213, Japan
| | - Joji M. Otaki
- The BCPH Unit of Molecular Physiology, Department of Chemistry, Biology and Marine Science, Faculty of Science, University of the Ryukyus, Okinawa 903-0213, Japan; (K.S.); (W.T.)
| |
Collapse
|
26
|
Crook A, De Lima Leite A, Payne T, Bhinderwala F, Woods J, Singh VK, Powers R. Radiation exposure induces cross-species temporal metabolic changes that are mitigated in mice by amifostine. Sci Rep 2021; 11:14004. [PMID: 34234212 PMCID: PMC8263605 DOI: 10.1038/s41598-021-93401-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/24/2021] [Indexed: 11/09/2022] Open
Abstract
Exposure to acute, damaging radiation may occur through a variety of events from cancer therapy and industrial accidents to terrorist attacks and military actions. Our understanding of how to protect individuals and mitigate the effects of radiation injury or Acute Radiation Syndrome (ARS) is still limited. There are only a few Food and Drug Administration-approved therapies for ARS; whereas, amifostine is limited to treating low dose (0.7-6 Gy) radiation poisoning arising from cancer radiotherapy. An early intervention is critical to treat ARS, which necessitates identifying diagnostic biomarkers to quickly characterize radiation exposure. Towards this end, a multiplatform metabolomics study was performed to comprehensively characterize the temporal changes in metabolite levels from mice and non-human primate serum samples following γ-irradiation. The metabolomic signature of amifostine was also evaluated in mice as a model for radioprotection. The NMR and mass spectrometry metabolomics analysis identified 23 dysregulated pathways resulting from the radiation exposure. These metabolomic alterations exhibited distinct trajectories within glucose metabolism, phospholipid biosynthesis, and nucleotide metabolism. A return to baseline levels with amifostine treatment occurred for these pathways within a week of radiation exposure. Together, our data suggests a unique physiological change that is independent of radiation dose or species. Furthermore, a metabolic signature of radioprotection was observed through the use of amifostine prophylaxis of ARS.
Collapse
Affiliation(s)
- Alexandra Crook
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304, USA
| | - Aline De Lima Leite
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304, USA
| | - Thomas Payne
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304, USA
| | - Fatema Bhinderwala
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304, USA
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304, USA
| | - Jade Woods
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304, USA
| | - Vijay K Singh
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, USUHS, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
- Armed Forces Radiobiology Research Institute, USUHS, Bethesda, MD, 20814, USA.
| | - Robert Powers
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304, USA.
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, 68588-0304, USA.
| |
Collapse
|
27
|
Singh VK, Seed TM, Cheema AK. Metabolomics-based predictive biomarkers of radiation injury and countermeasure efficacy: current status and future perspectives. Expert Rev Mol Diagn 2021; 21:641-654. [PMID: 34024238 DOI: 10.1080/14737159.2021.1933448] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION There is an urgent need for specific and sensitive bioassays to augment biodosimetric assessments of unwanted and excessive radiation exposures that originate from unexpected nuclear/radiological events, including nuclear accidents, acts of terrorism, or the use of a radiological dispersal device. If sufficiently intense, such ionizing radiation exposures are likely to impact normal metabolic processes within the cells and organs of the body, thus inducing multifaceted biological responses. AREAS COVERED This review covers the application of metabolomics, an emerging and promising technology based on quantitative and qualitative determinations of small molecules in biological samples for the rapid assessment of an individual's exposure to ionizing radiation. Recent advancements in the analytics of high-resolution chromatography, mass spectrometry, and bioinformatics have led to untargeted (global) and targeted (quantitative phase) approaches to identify biomarkers of radiation injury and countermeasure efficacy. Biomarkers are deemed essential for both assessing the radiation exposure levels and for extrapolative processes involved in determining scaling factors of a given radiation countering medicinal between experimental animals and humans. EXPERT OPINION The discipline of metabolomics appears to be highly informative in assessing radiation exposure levels and for identifying biomarkers of radiation injury and countermeasure efficacy.
Collapse
Affiliation(s)
- Vijay K Singh
- Division of Radioprotectants,Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Serices University of the Health Sciences, Bethesda, MD, USA.,Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | | | - Amrita K Cheema
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA.,Department of Biochemistry, Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
28
|
Linking Serine/Glycine Metabolism to Radiotherapy Resistance. Cancers (Basel) 2021; 13:cancers13061191. [PMID: 33801846 PMCID: PMC8002185 DOI: 10.3390/cancers13061191] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Hyperactivation of the de novo serine/glycine biosynthesis across different cancer types and its critical contribution in tumor initiation, progression, and therapy resistance indicate the relevance of serine/glycine metabolism-targeted therapies as therapeutic intervention in cancer. In this review, we specifically focus on the contribution of the de novo serine/glycine biosynthesis pathway to radioresistance. We provide a future perspective on how de novo serine/glycine biosynthesis inhibition and serine-free diets may improve the outcome of radiotherapy. Future research in this field is needed to better understand serine/glycine metabolic reprogramming of cancer cells in response to radiation and the influence of this pathway in the tumor microenvironment, which may provide the rationale for the optimal combination therapies. Abstract The activation of de novo serine/glycine biosynthesis in a subset of tumors has been described as a major contributor to tumor pathogenesis, poor outcome, and treatment resistance. Amplifications and mutations of de novo serine/glycine biosynthesis enzymes can trigger pathway activation; however, a large group of cancers displays serine/glycine pathway overexpression induced by oncogenic drivers and unknown regulatory mechanisms. A better understanding of the regulatory network of de novo serine/glycine biosynthesis activation in cancer might be essential to unveil opportunities to target tumor heterogeneity and therapy resistance. In the current review, we describe how the activation of de novo serine/glycine biosynthesis in cancer is linked to treatment resistance and its implications in the clinic. To our knowledge, only a few studies have identified this pathway as metabolic reprogramming of cancer cells in response to radiation therapy. We propose an important contribution of de novo serine/glycine biosynthesis pathway activation to radioresistance by being involved in cancer cell viability and proliferation, maintenance of cancer stem cells (CSCs), and redox homeostasis under hypoxia and nutrient-deprived conditions. Current approaches for inhibition of the de novo serine/glycine biosynthesis pathway provide new opportunities for therapeutic intervention, which in combination with radiotherapy might be a promising strategy for tumor control and ultimately eradication. Further research is needed to gain molecular and mechanistic insight into the activation of this pathway in response to radiation therapy and to design sophisticated stratification methods to select patients that might benefit from serine/glycine metabolism-targeted therapies in combination with radiotherapy.
Collapse
|
29
|
Zhao H, Xi C, Tian M, Lu X, Cai TJ, Li S, Tian XL, Gao L, Liu HX, Liu KH, Liu QJ. Identification of Potential Radiation Responsive Metabolic Biomarkers in Plasma of Rats Exposed to Different Doses of Cobalt-60 Gamma Rays. Dose Response 2021; 18:1559325820979570. [PMID: 33402881 PMCID: PMC7745571 DOI: 10.1177/1559325820979570] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/27/2020] [Accepted: 11/16/2020] [Indexed: 11/21/2022] Open
Abstract
Metabolomics has great potential to process accessible biofluids through high-throughput and quantitative analysis for radiation biomarker screening. This study focused on the potential radiation responsive metabolites in rat plasma and the dose-response relationships. In the discovery stage, 20 male Sprague–Dawley rats were exposed to 0, 1, 3 and 5 Gy of cobalt-60 gamma rays at a dose rate of 1 Gy/min. Plasma samples were collected at 72 h after exposure and analyzed using liquid chromatography mass spectrometry based on non-targeted metabolomics. In the verification stage, 50 additional rats were exposed to 0, 1, 2, 3, 5 and 8 Gy of gamma rays. The concentrations of candidate metabolites were then analyzed using targeted metabolomics methods. Fifteen candidate radiation responsive metabolites were identified as potential radiation metabolite biomarkers. Metabolic pathways, such as linoleic acid metabolism and glycerophospholipid metabolism pathways, were changed after irradiation. Six radiation responsive metabolites, including LysoPC(20:2), LysoPC(20:3), PC(18:0/22:5), L-palmitoylcarnitine, N-acetylornithine and butyrylcarnitine, had good dose-response relationships (R2 > 0.80). The area under the curve of the panel of the 6 radiation responsive metabolites was 0.923. The radiation exposure metabolomics biomarkers and dose-response curves may have potential for rapid dose assessment and triage in nuclear and radiation accidents.
Collapse
Affiliation(s)
- 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
| | - 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
| | - 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
| | - Tian-Jing Cai
- 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
| | - 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
| | - 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
| | - Ke-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, People's Republic of China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, People's Republic of China.,University of Chinese Academy of Sciences, 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
|
30
|
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
|
31
|
Tyagi R, Maan K, Khushu S, Rana P. Urine metabolomics based prediction model approach for radiation exposure. Sci Rep 2020; 10:16063. [PMID: 32999294 PMCID: PMC7527994 DOI: 10.1038/s41598-020-72426-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 08/13/2020] [Indexed: 01/21/2023] Open
Abstract
The radiological incidents and terrorism have demanded the need for the development of rapid, precise, and non-invasive technique for detection and quantification of exposed dose of radiation. Though radiation induced metabolic markers have been thoroughly investigated, but reproducibility still needs to be elucidated. The present study aims at assessing the reliability and reproducibility of markers using nuclear magnetic resonance (NMR) spectroscopy and further deriving a logistic regression model based on these markers. C57BL/6 male mice (8-10 weeks) whole body γ-irradiated and sham irradiated controls were used. Urine samples collected at 24 h post dose were investigated using high resolution NMR spectroscopy and the datasets were analyzed using multivariate analysis. Fifteen distinguishable metabolites and 3 metabolic pathways (TCA cycle, taurine and hypotaurine metabolism, primary bile acid biosynthesis) were found to be amended. ROC curve and logistic regression was used to establish a diagnostic model as Logit (p) = log (p/1 - p) = -0.498 + 13.771 (tau) - 3.412 (citrate) - 34.461 (α-KG) + 515.183 (fumarate) with a sensitivity and specificity of 1.00 and 0.964 respectively. The findings demonstrate the proof of concept and the potential of NMR based metabolomics to establish a prediction model that can be implemented as a promising mass screening tool during triage.
Collapse
Affiliation(s)
- Ritu Tyagi
- Metabolomics Research Facility, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, S. K Mazumdar Road, Timarpur, Delhi, 110054, India
| | - Kiran Maan
- Metabolomics Research Facility, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, S. K Mazumdar Road, Timarpur, Delhi, 110054, India
| | - Subash Khushu
- NMR Research Centre, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, Delhi, India
| | - Poonam Rana
- Metabolomics Research Facility, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, S. K Mazumdar Road, Timarpur, Delhi, 110054, India.
| |
Collapse
|
32
|
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
|
33
|
Salah M, Osuga S, Nakahana M, Irino Y, Shinohara M, Shimizu Y, Mukumoto N, Akasaka H, Nakaoka A, Miyawaki D, Ishihara T, Yoshida K, Okamoto Y, Sasaki R. Elucidation of gastrointestinal dysfunction in response to irradiation using metabolomics. Biochem Biophys Rep 2020; 23:100789. [PMID: 32775703 PMCID: PMC7393574 DOI: 10.1016/j.bbrep.2020.100789] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 07/01/2020] [Accepted: 07/02/2020] [Indexed: 11/29/2022] Open
Abstract
Gastrointestinal toxicity is frequently observed secondary to accidental or therapeutic radiation exposure. However, the variation in the intestinal metabolites after abdominal radiation exposure remains ambiguous. In the present study, C57BL/6 mice were exposed to 0, 2, and 20 Gy irradiation dose. The Head and chest of each mouse were covered with a lead shield before x-ray irradiation. 24 h post-irradiation treatment, intestinal tissue of each mouse was excised and prepared for metabolites measurement using gas chromatography-mass spectrometry (GC-MS). Our comprehensive analysis of metabolites in the intestinal tissues detected 44 metabolites after irradiation, including amino acids, carbohydrates, organic acids, and sugars. Amino acid levels in the intestinal tissue gradually rose, dependent on the radiation dose, perhaps as an indication of oxidative stress. Our findings raise the possibility that amino acid metabolism may be a potential target for the development of treatments to alleviate or mitigate the harmful effects of oxidative stress-related gastrointestinal toxicity due to radiation exposure. Gastrointestinal damage frequently results from radiation exposure. We analyzed the metabolic profile after local irradiation to the intestine. Amino acid levels in the intestinal tissue rose dependent on the radiation dose. Amino acid metabolism may be a good target for future therapies.
Collapse
Affiliation(s)
- Mohammed Salah
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan.,Department of Biochemistry, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Saki Osuga
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Makiko Nakahana
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Yasuhiro Irino
- Division of Evidence-based Laboratory Medicine, Kobe University Graduate School of Medicine, Japan
| | - Masakazu Shinohara
- Division of Epidemiology and the Integrated Center for Mass Spectrometry, Kobe University Graduate School of Medicine, Japan.,The Integrated Center for Mass Spectrometry, Kobe University Graduate School of Medicine, Japan
| | - Yasuyuki Shimizu
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Naritoshi Mukumoto
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Hiroaki Akasaka
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Ai Nakaoka
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Daisuke Miyawaki
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Takeaki Ishihara
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Kenji Yoshida
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| | - Yoshiaki Okamoto
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan.,Department of Radiation Therapy, Osaka Police Hospital, Osaka, Japan
| | - Ryohei Sasaki
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Kobe, Hyogo, Japan
| |
Collapse
|
34
|
Serum Metabolomic Alterations Associated with Cesium-137 Internal Emitter Delivered in Various Dose Rates. Metabolites 2020; 10:metabo10070270. [PMID: 32629836 PMCID: PMC7407308 DOI: 10.3390/metabo10070270] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/24/2020] [Accepted: 06/29/2020] [Indexed: 01/08/2023] Open
Abstract
Our laboratory and others have use radiation metabolomics to assess responses in order to develop biomarkers reflecting exposure and level of injury. To expand the types of exposure and compare to previously published results, metabolomic analysis has been carried out using serum samples from mice exposed to 137Cs internal emitters. Animals were injected intraperitoneally with 137CsCl solutions of varying radioactivity, and the absorbed doses were calculated. To determine the dose rate effect, serum samples were collected at 2, 3, 5, 7, and 14 days after injection. Based on the time for each group receiving the cumulative dose of 4 Gy, the dose rate for each group was determined. The dose rates analyzed were 0.16 Gy/day (low), 0.69 Gy/day (medium), and 1.25 Gy/day (high). The results indicated that at a cumulative dose of 4 Gy, the low dose rate group had the least number of statistically significantly differential spectral features. Some identified metabolites showed common changes for different dose rates. For example, significantly altered levels of oleamide and sphingosine 1-phosphate were seen in all three groups. On the other hand, the intensity of three amino acids, Isoleucine, Phenylalanine and Arginine, significantly decreased only in the medium dose rate group. These findings have the potential to be used in assessing the exposure and the biological effects of internal emitters.
Collapse
|
35
|
Koelmel JP, Napolitano MP, Ulmer CZ, Vasiliou V, Garrett TJ, Yost RA, Prasad MNV, Godri Pollitt KJ, Bowden JA. Environmental lipidomics: understanding the response of organisms and ecosystems to a changing world. Metabolomics 2020; 16:56. [PMID: 32307636 DOI: 10.1007/s11306-020-01665-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 03/13/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Understanding the interaction between organisms and the environment is important for predicting and mitigating the effects of global phenomena such as climate change, and the fate, transport, and health effects of anthropogenic pollutants. By understanding organism and ecosystem responses to environmental stressors at the molecular level, mechanisms of toxicity and adaptation can be determined. This information has important implications in human and environmental health, engineering biotechnologies, and understanding the interaction between anthropogenic induced changes and the biosphere. One class of molecules with unique promise for environmental science are lipids; lipids are highly abundant and ubiquitous across nearly all organisms, and lipid profiles often change drastically in response to external stimuli. These changes allow organisms to maintain essential biological functions, for example, membrane fluidity, as they adapt to a changing climate and chemical environment. Lipidomics can help scientists understand the historical and present biofeedback processes in climate change and the biogeochemical processes affecting nutrient cycles. Lipids can also be used to understand how ecosystems respond to historical environmental changes with lipid signatures dating back to hundreds of millions of years, which can help predict similar changes in the future. In addition, lipids are direct targets of environmental stressors, for example, lipids are easily prone to oxidative damage, which occurs during exposure to most toxins. AIM OF REVIEW This is the first review to summarize the current efforts to comprehensively measure lipids to better understand the interaction between organisms and their environment. This review focuses on lipidomic applications in the arenas of environmental toxicology and exposure assessment, xenobiotic exposures and health (e.g., obesity), global climate change, and nutrient cycles. Moreover, this review summarizes the use of and the potential for lipidomics in engineering biotechnologies for the remediation of persistent compounds and biofuel production. KEY SCIENTIFIC CONCEPT With the preservation of certain lipids across millions of years and our ever-increasing understanding of their diverse biological roles, lipidomic-based approaches provide a unique utility to increase our understanding of the contemporary and historical interactions between organisms, ecosystems, and anthropogenically-induced environmental changes.
Collapse
Affiliation(s)
- Jeremy P Koelmel
- Department of Chemistry, University of Florida, 125 Buckman Drive, Gainesville, FL, 32611, USA
- Department of Environmental Health Sciences, School of Public Health, Yale University, New Haven, CT, 06510, USA
| | - Michael P Napolitano
- CSS, Inc., under contract to National Oceanic and Atmospheric Administration, National Centers for Coastal Ocean Science, Hollings Marine Laboratory, 331 Fort Johnson Road, Charleston, SC, 29412, USA
| | - Candice Z Ulmer
- National Institute of Standards and Technology, Hollings Marine Laboratory, 331 Ft. Johnson Road, Charleston, SC, 29412, USA
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, School of Public Health, Yale University, New Haven, CT, 06510, USA
| | - Timothy J Garrett
- Department of Chemistry, University of Florida, 125 Buckman Drive, Gainesville, FL, 32611, USA
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Richard A Yost
- Department of Chemistry, University of Florida, 125 Buckman Drive, Gainesville, FL, 32611, USA
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - M N V Prasad
- Department of Plant Sciences, University of Hyderabad, Hyderabad, Telangana, 500046, India
| | - Krystal J Godri Pollitt
- Department of Environmental Health Sciences, School of Public Health, Yale University, New Haven, CT, 06510, USA
| | - John A Bowden
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, 1333 Center Drive, Gainesville, FL, 32610, USA.
| |
Collapse
|
36
|
Chakraborty N, Gautam A, Holmes-Hampton GP, Kumar VP, Biswas S, Kumar R, Hamad D, Dimitrov G, Olabisi AO, Hammamieh R, Ghosh SP. microRNA and Metabolite Signatures Linked to Early Consequences of Lethal Radiation. Sci Rep 2020; 10:5424. [PMID: 32214144 PMCID: PMC7096415 DOI: 10.1038/s41598-020-62255-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 03/11/2020] [Indexed: 12/12/2022] Open
Abstract
Lethal total body irradiation (TBI) triggers multifactorial health issues in a potentially short time frame. Hence, early signatures of TBI would be of great clinical value. Our study aimed to interrogate microRNA (miRNA) and metabolites, two biomolecules available in blood serum, in order to comprehend the immediate impacts of TBI. Mice were exposed to a lethal dose (9.75 Gy) of Cobalt-60 gamma radiation and euthanized at four time points, namely, days 1, 3, 7 and 9 post-TBI. Serum miRNA libraries were sequenced using the Illumina small RNA sequencing protocol, and metabolites were screened using a mass spectrometer. The degree of early impacts of irradiation was underscored by the large number of miRNAs and metabolites that became significantly expressed during the Early phase (day 0 and 1 post-TBI). Radiation-induced inflammatory markers for bone marrow aplasia and pro-sepsis markers showed early elevation with longitudinal increment. Functional analysis integrating miRNA-protein-metabolites revealed inflammation as the overarching host response to lethal TBI. Early activation of the network linked to the synthesis of reactive oxygen species was associated with the escalated regulation of the fatty acid metabolism network. In conclusion, we assembled a list of time-informed critical markers and mechanisms of significant translational potential in the context of a radiation exposure event.
Collapse
Affiliation(s)
- Nabarun Chakraborty
- The Geneva Foundation, Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Fort Detrick, MD, 21702-5010, USA
- Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Fort Detrick, MD, 21702-5010, USA
| | - Aarti Gautam
- Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Fort Detrick, MD, 21702-5010, USA
| | - Gregory P Holmes-Hampton
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, 20889, USA
| | - Vidya P Kumar
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, 20889, USA
| | - Shukla Biswas
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, 20889, USA
| | - Raina Kumar
- The Geneva Foundation, Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Fort Detrick, MD, 21702-5010, USA
- Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Fort Detrick, MD, 21702-5010, USA
| | - Dana Hamad
- ORISE, Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Fort Detrick, MD, 21702-5010, USA
| | - George Dimitrov
- The Geneva Foundation, Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Fort Detrick, MD, 21702-5010, USA
- Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Fort Detrick, MD, 21702-5010, USA
| | - Ayodele O Olabisi
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, 20889, USA
| | - Rasha Hammamieh
- Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Fort Detrick, MD, 21702-5010, USA
| | - Sanchita P Ghosh
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, 20889, USA.
| |
Collapse
|
37
|
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
|
38
|
de Leve S, Wirsdörfer F, Jendrossek V. The CD73/Ado System-A New Player in RT Induced Adverse Late Effects. Cancers (Basel) 2019; 11:cancers11101578. [PMID: 31623231 PMCID: PMC6827091 DOI: 10.3390/cancers11101578] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 10/11/2019] [Accepted: 10/12/2019] [Indexed: 02/06/2023] Open
Abstract
Radiotherapy (RT) is a central component of standard treatment for many cancer patients. RT alone or in multimodal treatment strategies has a documented contribution to enhanced local control and overall survival of cancer patients, and cancer cure. Clinical RT aims at maximizing tumor control, while minimizing the risk for RT-induced adverse late effects. However, acute and late toxicities of IR in normal tissues are still important biological barriers to successful RT: While curative RT may not be tolerable, sub-optimal tolerable RT doses will lead to fatal outcomes by local recurrence or metastatic disease, even when accepting adverse normal tissue effects that decrease the quality of life of irradiated cancer patients. Technical improvements in treatment planning and the increasing use of particle therapy have allowed for a more accurate delivery of IR to the tumor volume and have thereby helped to improve the safety profile of RT for many solid tumors. With these technical and physical strategies reaching their natural limits, current research for improving the therapeutic gain of RT focuses on innovative biological concepts that either selectively limit the adverse effects of RT in normal tissues without protecting the tumor or specifically increase the radiosensitivity of the tumor tissue without enhancing the risk of normal tissue complications. The biology-based optimization of RT requires the identification of biological factors that are linked to differential radiosensitivity of normal or tumor tissues, and are amenable to therapeutic targeting. Extracellular adenosine is an endogenous mediator critical to the maintenance of homeostasis in various tissues. Adenosine is either released from stressed or injured cells or generated from extracellular adenine nucleotides by the concerted action of the ectoenzymes ectoapyrase (CD39) and 5′ ectonucleotidase (NT5E, CD73) that catabolize ATP to adenosine. Recent work revealed a role of the immunoregulatory CD73/adenosine system in radiation-induced fibrotic disease in normal tissues suggesting a potential use as novel therapeutic target for normal tissue protection. The present review summarizes relevant findings on the pathologic roles of CD73 and adenosine in radiation-induced fibrosis in different organs (lung, skin, gut, and kidney) that have been obtained in preclinical models and proposes a refined model of radiation-induced normal tissue toxicity including the disease-promoting effects of radiation-induced activation of CD73/adenosine signaling in the irradiated tissue environment. However, expression and activity of the CD73/adenosine system in the tumor environment has also been linked to increased tumor growth and tumor immune escape, at least in preclinical models. Therefore, we will discuss the use of pharmacologic inhibition of CD73/adenosine-signaling as a promising strategy for improving the therapeutic gain of RT by targeting both, malignant tumor growth and adverse late effects of RT with a focus on fibrotic disease. The consideration of the therapeutic window is particularly important in view of the increasing use of RT in combination with various molecularly targeted agents and immunotherapy to enhance the tumor radiation response, as such combinations may result in increased or novel toxicities, as well as the increasing number of cancer survivors.
Collapse
Affiliation(s)
- Simone de Leve
- Institute of Cell Biology (Cancer Research), University Hospital Essen, 45122 Essen, Germany.
| | - Florian Wirsdörfer
- Institute of Cell Biology (Cancer Research), University Hospital Essen, 45122 Essen, Germany.
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University Hospital Essen, 45122 Essen, Germany.
| |
Collapse
|
39
|
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
|
40
|
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
|
41
|
Araújo S, Pagano A, Dondi D, Lazzaroni S, Pinela E, Macovei A, Balestrazzi A. Metabolic signatures of germination triggered by kinetin in Medicago truncatula. Sci Rep 2019; 9:10466. [PMID: 31320688 PMCID: PMC6639397 DOI: 10.1038/s41598-019-46866-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 07/01/2019] [Indexed: 11/09/2022] Open
Abstract
In the present work, non-targeted metabolomics was used to investigate the seed response to kinetin, a phytohormone with potential roles in seed germination, still poorly explored. The aim of this study was to elucidate the metabolic signatures of germination triggered by kinetin and explore changes in metabolome to identify novel vigor/stress hallmarks in Medicago truncatula. Exposure to 0.5 mM kinetin accelerated seed germination but impaired seedling growth. Metabolite composition was investigated in seeds imbibed with water or with 0.5 mM kinetin collected at 2 h and 8 h of imbibition, and at the radicle protrusion stage. According to Principal Component Analysis, inositol pentakisphosphate, agmatine, digalactosylglycerol, inositol hexakisphosphate, and oleoylcholine were the metabolites that mostly contributed to the separation between 2 h, 8 h and radicle protrusion stage, irrespective of the treatment applied. Overall, only 27 metabolites showed significant changes in mean relative contents triggered by kinetin, exclusively at the radicle protrusion stage. The observed metabolite depletion might associate with faster germination or regarded as a stress signature. Results from alkaline comet assay, highlighting the occurrence of DNA damage at this stage of germination, are consistent with the hypothesis that prolonged exposure to kinetin induces stress conditions leading to genotoxic injury.
Collapse
Affiliation(s)
- Susana Araújo
- Instituto de Tecnologia Química e Biológica António Xavier - Universidade Nova de Lisboa (ITQB-NOVA), Av. da República, 2780-157, Oeiras, Portugal
| | - Andrea Pagano
- Department of Biology and Biotechnology 'L. Spallanzani', via Ferrata 9, 27100, Pavia, Italy
| | - Daniele Dondi
- Department of Chemistry, Viale Taramelli 12, 27100, Pavia, Italy
| | - Simone Lazzaroni
- Department of Chemistry, Viale Taramelli 12, 27100, Pavia, Italy
| | - Eduardo Pinela
- Instituto de Tecnologia Química e Biológica António Xavier - Universidade Nova de Lisboa (ITQB-NOVA), Av. da República, 2780-157, Oeiras, Portugal
| | - Anca Macovei
- Department of Biology and Biotechnology 'L. Spallanzani', via Ferrata 9, 27100, Pavia, Italy
| | - Alma Balestrazzi
- Department of Biology and Biotechnology 'L. Spallanzani', via Ferrata 9, 27100, Pavia, Italy.
| |
Collapse
|
42
|
Laiakis EC, Canadell MP, Grilj V, Harken AD, Garty GY, Astarita G, Brenner DJ, Smilenov L, Fornace AJ. Serum lipidomic analysis from mixed neutron/X-ray radiation fields reveals a hyperlipidemic and pro-inflammatory phenotype. Sci Rep 2019; 9:4539. [PMID: 30872747 PMCID: PMC6418166 DOI: 10.1038/s41598-019-41083-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 02/27/2019] [Indexed: 01/06/2023] Open
Abstract
Heightened threats for nuclear terrorism using improvised nuclear devices (IND) necessitate the development of biodosimetry assays that could rapidly assess thousands of individuals. However, the radiation exposures from an IND may be complex due to mixed fields of neutrons and photons (γ-rays), shielding from buildings, and proximity to the epicenter among others. In this study we utilized lipidomics to analyze serum samples from mice exposed to various percentages of neutrons and X-rays to a total dose of 3 Gy. Triacylglycerides, phosphatidylserines, lysophosphatidylethanolamines, lysophosphatidylcholines (LPCs), sphingolipids, and cholesteryl esters all showed delayed increases at day 7 compared to day 1 after irradiation, while diacylglycerides decreased in mixed field exposures and phosphatidylcholines (PCs) remained largely unchanged. Individual lipid molecules with a high degree of unsaturation exhibited the highest fold changes in mixed fields compared to photons alone. More importantly, the increased ratio of LPCs to PCs of each irradiation group compared to control could be used as a radiation biomarker and highlights the existence of a pro-inflammatory phenotype. The results showed that even a small percentage of neutrons in a mixed field can lead to high biological responses with implications for accurate biodosimetry, triage and medical managements of exposed populations.
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.
| | | | - Veljko Grilj
- Radiological Research Accelerator Facility, Columbia University, Irvington, NY, USA
| | - Andrew D Harken
- Radiological Research Accelerator Facility, Columbia University, Irvington, NY, USA
| | - Guy Y Garty
- Radiological Research Accelerator Facility, Columbia University, Irvington, NY, USA
| | - Giuseppe Astarita
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, USA
| | - David J Brenner
- Center for Radiological Research, Columbia University, New York, NY, USA
| | - Lubomir Smilenov
- Center for Radiological Research, Columbia University, New York, NY, USA
| | - Albert J Fornace
- 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
|
43
|
Cheng Y, Dong Y, Hou Q, Wu J, Zhang W, Tian H, Li D. The protective effects of XH-105 against radiation-induced intestinal injury. J Cell Mol Med 2019; 23:2238-2247. [PMID: 30663222 PMCID: PMC6378229 DOI: 10.1111/jcmm.14159] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/17/2018] [Accepted: 12/24/2018] [Indexed: 12/26/2022] Open
Abstract
Radiation-induced intestinal injury is one of the major side effects in patients receiving radiation therapy. There is no specific treatment for radiation enteritis in the clinic. We designed and synthesized a new compound named XH-105, which is expected to cleave into polyphenol and aminothiol in vivo to mitigate radiation injury. In the following study, we describe the beneficial effects of XH-105 against radiation-induced intestinal injury. C57BL/6J mice were treated by gavage with XH-105 1 hour before total body irradiation (TBI), and the survival rate was monitored. Histological changes were examined, and survival of Lgr5+ intestinal stem cells Ki67+ cells, villi+ enterocytes and lysozymes was determined by immunohistochemistry. DNA damage and cellular apoptosis in intestinal tissue were also evaluated. Compared to vehicle-treated mice after TBI, XH-105 treatment significantly enhanced the survival rate, attenuated structural damage of the small intestine, decreased the apoptotic rate, reduced DNA damage, maintained cell regeneration and promoted crypt proliferation and differentiation. XH-105 also reduced the expression of Bax and p53 in the small intestine. These data suggest that XH-105 is beneficial for the protection of radiation-induced intestinal injury by inhibiting the p53-dependent apoptosis signalling pathway.
Collapse
Affiliation(s)
- Ying Cheng
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China.,Center for Marine Bioproducts Development, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, South Australia, Australia
| | - Yinping Dong
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| | - Qinlian Hou
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| | - Jing Wu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| | - Wei Zhang
- Center for Marine Bioproducts Development, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, South Australia, Australia
| | - Hongqi Tian
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| | - Deguan Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| |
Collapse
|
44
|
Pagano A, de Sousa Araújo S, Macovei A, Dondi D, Lazzaroni S, Balestrazzi A. Metabolic and gene expression hallmarks of seed germination uncovered by sodium butyrate in Medicago truncatula. PLANT, CELL & ENVIRONMENT 2019; 42:259-269. [PMID: 29756644 DOI: 10.1111/pce.13342] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 05/03/2018] [Accepted: 05/05/2018] [Indexed: 05/20/2023]
Abstract
Because high-quality seeds are essential for successful crop production in challenging environments, understanding the molecular bases of seed vigour will lead to advances in seed technology. Histone deacetylase inhibitors, promoting histone hyperacetylation, are used as tools to explore aspects still uncovered of the abiotic stress response in plants. The aim of this work was to investigate novel signatures of seed germination in Medicago truncatula, using the histone deacetylase inhibitor sodium butyrate (NaB) as stress agent. NaB-treated and untreated seeds collected at 2 and 8 hr of imbibition and at the radicle protrusion stage underwent molecular phenotyping and nontargeted metabolome profiling. Quantitative enrichment analysis revealed the influence of NaB on seed nucleotide, amino acid, lipid, and carbohydrate metabolism. Up-regulation of antioxidant and polyamine biosynthesis genes occurred in response to NaB. DNA damage evidenced in NaB-treated seeds correlated with up-regulation of base-excision repair genes. Changes in N1 -methyladenosine and N1 -methylguanine were associated with up-regulation of MtALKBH1 (alkylation repair homolog) gene. N2 ,N2 -dimethylguanosine and 5-methylcytidine, tRNA modifications involved in the post-transcriptional regulation of DNA damage response, were also accumulated in NaB-treated seeds at the radicle protrusion stage. The observed changes in seed metabolism can provide novel potential metabolic hallmarks of germination.
Collapse
Affiliation(s)
- Andrea Pagano
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, via Ferrata 9, Pavia, 27100, Italy
| | - Susana de Sousa Araújo
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB-NOVA), Avenida da República, Estação Agronómica Nacional, Oeiras, 2780-157, Portugal
| | - Anca Macovei
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, via Ferrata 9, Pavia, 27100, Italy
| | - Daniele Dondi
- Department of Chemistry, University of Pavia, Viale Taramelli 12, Pavia, 27100, Italy
| | - Simone Lazzaroni
- Department of Chemistry, University of Pavia, Viale Taramelli 12, Pavia, 27100, Italy
| | - Alma Balestrazzi
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, via Ferrata 9, Pavia, 27100, Italy
| |
Collapse
|
45
|
Cheema AK, Hinzman CP, Mehta KY, Hanlon BK, Garcia M, Fatanmi OO, Singh VK. Plasma Derived Exosomal Biomarkers of Exposure to Ionizing Radiation in Nonhuman Primates. Int J Mol Sci 2018; 19:ijms19113427. [PMID: 30388807 PMCID: PMC6274965 DOI: 10.3390/ijms19113427] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 10/12/2018] [Accepted: 10/30/2018] [Indexed: 12/18/2022] Open
Abstract
Exposure to ionizing radiation induces a cascade of molecular events that ultimately impact endogenous metabolism. Qualitative and quantitative characterization of metabolomic profiles is a pragmatic approach to studying the risks of radiation exposure since it provides a phenotypic readout. Studies were conducted in irradiated nonhuman primates (NHP) to investigate metabolic changes in plasma and plasma-derived exosomes. Specifically, rhesus macaques (Macaca mulatta) were exposed to cobalt-60 gamma-radiation and plasma samples were collected prior to and after exposure to 5.8 Gy or 6.5 Gy radiation. Exosomes were isolated using ultracentrifugation and analyzed by untargeted profiling via ultra-performance liquid chromatography mass spectrometry (UPLC-MS) based metabolomic and lipidomic analyses, with the goal of identifying a molecular signature of irradiation. The enrichment of an exosomal fraction was confirmed using quantitative ELISA. Plasma profiling showed markers of dyslipidemia, inflammation and oxidative stress post-irradiation. Exosomal profiling, on the other hand, enabled detection and identification of low abundance metabolites that comprise exosomal cargo which would otherwise get obscured with plasma profiling. We discovered enrichment of different classes of metabolites including N-acyl-amino acids, Fatty Acid ester of Hydroxyl Fatty Acids (FAHFA’s), glycolipids and triglycerides as compared to the plasma metabolome composition with implications in mediation of systemic response to radiation induced stress signaling.
Collapse
Affiliation(s)
- Amrita K Cheema
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA.
- Department of Biochemistry, Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC 20057, USA.
| | - Charles P Hinzman
- Department of Biochemistry, Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC 20057, USA.
| | - Khyati Y Mehta
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA.
| | - Briana K Hanlon
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, USUHS, Bethesda, MD 20814, USA.
- Armed Forces Radiobiology Research Institute, USUHS, Bethesda, MD 20814, USA.
| | - Melissa Garcia
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, USUHS, Bethesda, MD 20814, USA.
- Armed Forces Radiobiology Research Institute, USUHS, Bethesda, MD 20814, USA.
| | - Oluseyi O Fatanmi
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, USUHS, Bethesda, MD 20814, USA.
- Armed Forces Radiobiology Research Institute, USUHS, Bethesda, MD 20814, USA.
| | - Vijay K Singh
- Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, USUHS, Bethesda, MD 20814, USA.
- Armed Forces Radiobiology Research Institute, USUHS, Bethesda, MD 20814, USA.
| |
Collapse
|
46
|
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
|
47
|
Kirsch DG, Diehn M, Kesarwala AH, Maity A, Morgan MA, Schwarz JK, Bristow R, Demaria S, Eke I, Griffin RJ, Haas-Kogan D, Higgins GS, Kimmelman AC, Kimple RJ, Lombaert IM, Ma L, Marples B, Pajonk F, Park CC, Schaue D, Tran PT, Willers H, Wouters BG, Bernhard EJ. The Future of Radiobiology. J Natl Cancer Inst 2018; 110:329-340. [PMID: 29126306 PMCID: PMC5928778 DOI: 10.1093/jnci/djx231] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 07/19/2017] [Accepted: 10/06/2017] [Indexed: 12/23/2022] Open
Abstract
Innovation and progress in radiation oncology depend on discovery and insights realized through research in radiation biology. Radiobiology research has led to fundamental scientific insights, from the discovery of stem/progenitor cells to the definition of signal transduction pathways activated by ionizing radiation that are now recognized as integral to the DNA damage response (DDR). Radiobiological discoveries are guiding clinical trials that test radiation therapy combined with inhibitors of the DDR kinases DNA-dependent protein kinase (DNA-PK), ataxia telangiectasia mutated (ATM), ataxia telangiectasia related (ATR), and immune or cell cycle checkpoint inhibitors. To maintain scientific and clinical relevance, the field of radiation biology must overcome challenges in research workforce, training, and funding. The National Cancer Institute convened a workshop to discuss the role of radiobiology research and radiation biologists in the future scientific enterprise. Here, we review the discussions of current radiation oncology research approaches and areas of scientific focus considered important for rapid progress in radiation sciences and the continued contribution of radiobiology to radiation oncology and the broader biomedical research community.
Collapse
Affiliation(s)
- David G Kirsch
- Department of Radiation Oncology and Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC
| | - Max Diehn
- Department of Radiation Oncology, Stanford Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA
| | | | - Amit Maity
- Department of Radiation Oncology Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Meredith A Morgan
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI
| | - Julie K Schwarz
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO
| | - Robert Bristow
- Department of Radiation Oncology, Princess Margaret Cancer Center, Toronto, ON, Canada
| | - Sandra Demaria
- Department of Radiation Oncology and Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY
| | - Iris Eke
- Radiation Oncology Branch, National Institutes of Health, Bethesda, MD
| | - Robert J Griffin
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Daphne Haas-Kogan
- Department of Radiation Oncology, Harvard Medical School, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Boston Children's Hospital, Boston, MA
| | - Geoff S Higgins
- Department of Oncology, University of Oxford, Oxford, Oxfordshire, UK
| | - Alec C Kimmelman
- Perlmutter Cancer Center and Department of Radiation Oncology, New York University Langone Medical Center, New York, NY
| | - Randall J Kimple
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Isabelle M Lombaert
- Department of Biologic and Materials Sciences, Biointerfaces Institute, School of Dentistry, University of Michigan, Ann Arbor, MI
| | - Li Ma
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Brian Marples
- Department of Radiation Oncology, University of Miami, Miami, FL
| | - Frank Pajonk
- Department of Radiation Oncology, University of California, Los Angeles, CA
| | - Catherine C Park
- David Geffen School of Medicine, University of California, Los Angeles, CA
- Department of Radiation Oncology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA
| | - Dörthe Schaue
- Division of Molecular and Cellular Oncology, University of California, Los Angeles, CA
| | - Phuoc T. Tran
- Department of Radiation Oncology and Molecular Radiation Sciences, Oncology and Urology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Henning Willers
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Brad G. Wouters
- Department of Radiation Oncology (RB), Princess Margaret Cancer Center
| | - Eric J Bernhard
- Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
48
|
Hu JM, Sun HT. Serum proton NMR metabolomics analysis of human lung cancer following microwave ablation. Radiat Oncol 2018; 13:40. [PMID: 29530051 PMCID: PMC5848604 DOI: 10.1186/s13014-018-0982-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 02/22/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND To find potential serum biomarkers of microwave ablation (MWA) for treatment of human lung cancer by 1H nuclear magnetic resonance (NMR)-based metabolomics analysis. METHODS Serum specimens collected from 43 healthy individuals, 39 patients with advanced non-small cell lung cancer (NSCLC) and 38 NSCLC patients treated with MWA, were subjected to 1H NMR-based metabolomics analysis. Partial least squares discriminant analysis was used to analyze the data. RESULTS Compared with healthy controls, NSCLC patients showed significantly elevated serum levels of lactate, alanine, glutamate, proline, glycoprotein, phenylalanine, tyrosine and tryptophan, and markedly decreased serum levels of glucose, taurine, glutamine, glycine, phosphocreatine and threonine (p < 0.05). MWA treatment reversed the metabolic profiles of NSCLC patients towards the control group. CONCLUSIONS 1H NMR-based metabolomics analysis enhanced the current understanding of the mechanisms involved in NSCLC, and uncovered the therapeutic potential of MWA for treatment of NSCLC. The above disturbed serum metabolites were proposed to be the potential biomarkers that may help to predict NSCLC and to evaluate the efficacy of MWA in the treatment of NSCLC.
Collapse
Affiliation(s)
- Jian-Ming Hu
- Thoracic surgeons, Jiangxi Provincial People’s Hospital, 152 Patriotic Road, Nanchang City, 333000 People’s Republic of China
| | - Huang-Tao Sun
- Thoracic surgeons, Jiangxi Provincial People’s Hospital, 152 Patriotic Road, Nanchang City, 333000 People’s Republic of China
| |
Collapse
|
49
|
Gil AM, Duarte D, Pinto J, Barros AS. Assessing Exposome Effects on Pregnancy through Urine Metabolomics of a Portuguese (Estarreja) Cohort. J Proteome Res 2018; 17:1278-1289. [DOI: 10.1021/acs.jproteome.7b00878] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Ana M. Gil
- CICECO
- Aveiro Institute of Materials (CICECO/UA), Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Daniela Duarte
- CICECO
- Aveiro Institute of Materials (CICECO/UA), Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Joana Pinto
- CICECO
- Aveiro Institute of Materials (CICECO/UA), Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
- UCIBIO@REQUIMTE/Laboratório
de Toxicologia, Departamento de Ciências Biológicas,
Faculdade de Farmácia, Universidade do Porto, 4050-313 Porto, Portugal
| | - António S. Barros
- CICECO
- Aveiro Institute of Materials (CICECO/UA), Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
- Department
of Cardiothoracic Surgery and Physiology, Faculty of Medicine, Porto 4200-319, Portugal
| |
Collapse
|
50
|
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
|