1
|
Miranda S, Vermeesen R, Janssen A, Rehnberg E, Etlioglu E, Baatout S, Tabury K, Baselet B. Effects of simulated space conditions on CD4+ T cells: a multi modal analysis. Front Immunol 2024; 15:1443936. [PMID: 39286254 PMCID: PMC11402665 DOI: 10.3389/fimmu.2024.1443936] [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: 06/04/2024] [Accepted: 08/08/2024] [Indexed: 09/19/2024] Open
Abstract
Introduction The immune system is an intricate network of cellular components that safeguards against pathogens and aberrant cells, with CD4+ T cells playing a central role in this process. Human space travel presents unique health challenges, such as heavy ion ionizing radiation, microgravity, and psychological stress, which can collectively impede immune function. The aim of this research was to examine the consequences of simulated space stressors on CD4+ T cell activation, cytokine production, and gene expression. Methods CD4+ T cells were obtained from healthy individuals and subjected to Fe ion particle radiation, Photon irradiation, simulated microgravity, and hydrocortisone, either individually or in different combinations. Cytokine levels for Th1 and Th2 cells were determined using multiplex Luminex assays, and RNA sequencing was used to investigate gene expression patterns and identify essential genes and pathways impacted by these stressors. Results Simulated microgravity exposure resulted in an apparent Th1 to Th2 shift, evidenced on the level of cytokine secretion as well as altered gene expression. RNA sequencing analysis showed that several gene pathways were altered, particularly in response to Fe ions irradiation and simulated microgravity exposures. Individually, each space stressor caused differential gene expression, while the combination of stressors revealed complex interactions. Discussion The research findings underscore the substantial influence of the space exposome on immune function, particularly in the regulation of T cell responses. Future work should focus expanding the limited knowledge in this field. Comprehending these modifications will be essential for devising effective strategies to safeguard the health of astronauts during extended space missions. Conclusion The effects of simulated space stressors on CD4+ T cell function are substantial, implying that space travel poses a potential threat to immune health. Additional research is necessary to investigate the intricate relationship between space stressors and to develop effective countermeasures to mitigate these consequences.
Collapse
Affiliation(s)
- Silvana Miranda
- Radiobiology Unit, Institute for Nuclear Medical Applications, Belgian Nuclear Research Centre SCK CEN, Mol, Belgium
- Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Randy Vermeesen
- Radiobiology Unit, Institute for Nuclear Medical Applications, Belgian Nuclear Research Centre SCK CEN, Mol, Belgium
| | - Ann Janssen
- Radiobiology Unit, Institute for Nuclear Medical Applications, Belgian Nuclear Research Centre SCK CEN, Mol, Belgium
| | - Emil Rehnberg
- Radiobiology Unit, Institute for Nuclear Medical Applications, Belgian Nuclear Research Centre SCK CEN, Mol, Belgium
- Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Emre Etlioglu
- Radiobiology Unit, Institute for Nuclear Medical Applications, Belgian Nuclear Research Centre SCK CEN, Mol, Belgium
| | - Sarah Baatout
- Radiobiology Unit, Institute for Nuclear Medical Applications, Belgian Nuclear Research Centre SCK CEN, Mol, Belgium
- Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Kevin Tabury
- Radiobiology Unit, Institute for Nuclear Medical Applications, Belgian Nuclear Research Centre SCK CEN, Mol, Belgium
- Department of Biomedical Engineering, College of Engineering and Computing, University of South Carolina, Columbia, SC, United States
| | - Bjorn Baselet
- Radiobiology Unit, Institute for Nuclear Medical Applications, Belgian Nuclear Research Centre SCK CEN, Mol, Belgium
| |
Collapse
|
2
|
Wu YQ, Ding KX, Lv ZC, Cao ZY, Zhao K, Gao HY, Sun HY, Li JJ, Li SY, Zhao XW, Xue Y, Xiang SS, Zheng XF, Yang XM, Li CY. Single-Cell Transcriptomics Reveals Early Effects of Ionizing Radiation on Bone Marrow Mononuclear Cells in Mice. Int J Mol Sci 2024; 25:9287. [PMID: 39273235 PMCID: PMC11395520 DOI: 10.3390/ijms25179287] [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: 07/01/2024] [Revised: 08/10/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
Ionizing radiation exposure can cause damage to diverse tissues and organs, with the hematopoietic system being the most sensitive. However, limited information is available regarding the radiosensitivity of various hematopoietic cell populations in the bone marrow due to the high heterogeneity of the hematopoietic system. In this study, we observed that granulocyte-macrophage progenitors, hematopoietic stem/progenitor cells, and B cells within the bone marrow showed the highest sensitivity, exhibiting a rapid decrease in cell numbers following irradiation. Nonetheless, neutrophils, natural killer (NK) cells, T cells, and dendritic cells demonstrated a certain degree of radioresistance, with neutrophils exhibiting the most pronounced resistance. By employing single-cell transcriptome sequencing, we investigated the early responsive genes in various cell types following irradiation, revealing that distinct gene expression profiles emerged between radiosensitive and radioresistant cells. In B cells, radiation exposure led to a specific upregulation of genes associated with mitochondrial respiratory chain complexes, suggesting a connection between these complexes and cell radiosensitivity. In neutrophils, radiation exposure resulted in fewer gene alterations, indicating their potential for distinct mechanisms in radiation resistance. Collectively, this study provides insights into the molecular mechanism for the heterogeneity of radiosensitivity among the various bone marrow hematopoietic cell populations.
Collapse
Affiliation(s)
- Yun-Qiang Wu
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Ke-Xin Ding
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Zhi-Chun Lv
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Zheng-Yue Cao
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Ke Zhao
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Hui-Ying Gao
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Hui-Ying Sun
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Jing-Jing Li
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Si-Yu Li
- School of Basic Medicine, Anhui Medical University, Hefei 230032, China
| | - Xiong-Wei Zhao
- School of Basic Medicine, Anhui Medical University, Hefei 230032, China
| | - Yang Xue
- School of Life Sciences, Hebei University, No. 180 Wusi Dong Road, Lian Chi District, Baoding 071000, China
| | - Shen-Si Xiang
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xiao-Fei Zheng
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xiao-Ming Yang
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Chang-Yan Li
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing 100850, China
- School of Basic Medicine, Anhui Medical University, Hefei 230032, China
- School of Life Sciences, Hebei University, No. 180 Wusi Dong Road, Lian Chi District, Baoding 071000, China
| |
Collapse
|
3
|
Humayun A, Lin LYT, Li HH, Fornace AJ. FAILLA MEMORIAL LECTURE How We Got Here: One Laboratory's Odyssey in the Field of Radiation-Inducible Genes. Radiat Res 2024; 201:617-627. [PMID: 38573158 DOI: 10.1667/rade-23-00205.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 03/11/2024] [Indexed: 04/05/2024]
Abstract
This review focuses on early discoveries that contributed to our understanding and the scope of transcriptional responses after radiation damage. Before the development of modern approaches to assess overall global transcriptomic responses, the idea that mammalian cells could respond to DNA-damaging agents in a manner analogous to bacteria was not generally accepted. To investigate this possibility, the development of technology to identify differentially expressed low-abundance transcripts substantially facilitated our appreciation that DNA damaging agents like UV radiation and subsequently ionizing radiation did in fact produce robust transcriptional responses. Here we focus on our identification and characterization of radiation-inducible genes, and how even early studies on stress gene signaling highlighted the broad scope of transcriptional responses to radiation damage. Since then, the central role of transcriptional responses to radiation injury in maintaining genome integrity has been highlighted in many processes, including cell cycle checkpoint control, resistance to cancer by p53 and other key factors, cell senescence, and metabolism.
Collapse
Affiliation(s)
- Arslon Humayun
- Department of Oncology, Lombardi Comprehensive Cancer Center, Washington, DC
| | | | - Heng-Hong Li
- Department of Oncology, Lombardi Comprehensive Cancer Center, Washington, DC
| | - Albert J Fornace
- Department of Oncology, Lombardi Comprehensive Cancer Center, Washington, DC
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC
| |
Collapse
|
4
|
Shen LF, Fu ZM, Zhou SH. The role of radiotherapy in tumor immunity and the potential of PET/CT in detecting the expression of PD-1/PD-L1. Jpn J Radiol 2024; 42:347-353. [PMID: 37953364 DOI: 10.1007/s11604-023-01507-x] [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: 08/24/2023] [Accepted: 10/17/2023] [Indexed: 11/14/2023]
Abstract
Upregulation of PD-1/PD-L1 allows cancer cells to escape from host immune systems by functionally inactivating T-cell immune surveillance. Clinical blockade strategies have resulted in an increased prevalence of patients with late-stage cancers. However, many cancer patients had limited or no response to current immunotherapeutic strategies. Therefore, how to improve the sensitivity of immunotherapy has become the focus of attention of many scholars. Radiotherapy plays a role in the recruitment of T cells in the tumor microenvironment, increases CD4 + and CD8 + T cells, and increases PD-L1 expression, resulting in the synergistically enhanced antitumor effect of irradiation and PD-L1 blockade. Radiotherapy can cause changes in tumor metabolism, especially glucose metabolism. Tumor glycolysis and tumor immune evasion are interdependent, glycolytic activity enhances PD-L1 expression on tumor cells and thus promotes anti-PD-L1 immunotherapy response. Therefore, the mechanism of radiotherapy affecting tumor immunity may be partly through intervention of tumor glucose metabolism. Furthermore, some authors had found that the uptake of 2'-deoxy-2'-[18F]fluoro-D-glucose(18F-FDG) was correlated with PD-1/PD-L1 expression. Positron emission tomography/computed tomography (PET/CT) is a non-invasive detection method for PD-1/PD-L1 expression and has several potential advantages over immunohistochemical (IHC), PET/CT can dynamically reflect the expression of PD-1/PD-L1 inside the tumor and further guide clinical treatment.
Collapse
Affiliation(s)
- Li-Fang Shen
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Zi-Ming Fu
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Shui-Hong Zhou
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
5
|
Zhang Y, Du X, Zhao L, Sun Y. Construction of dose prediction model and identification of sensitive genes for space radiation based on single-sample networks under spaceflight conditions. Int J Radiat Biol 2024; 100:777-790. [PMID: 38471034 DOI: 10.1080/09553002.2024.2327393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 03/01/2024] [Indexed: 03/14/2024]
Abstract
PURPOSE To identify sensitive genes for space radiation, we integrated the transcriptomic samples of spaceflight mice from GeneLab and predicted the radiation doses absorbed by individuals in space. METHODS AND MATERIALS A single-sample network (SSN) for each individual sample was constructed. Then, using machine learning and genetic algorithms, we built the regression models to predict the absorbed dose equivalent based on the topological structure of SSNs. Moreover, we analyzed the SSNs from each tissue and compared the similarities and differences among them. RESULTS Our model exhibited excellent performance with the following metrics: R 2 = 0.980 , MSE = 6.74 e - 04 , and the Pearson correlation coefficient of 0.990 (p value <.0001) between predicted and actual values. We identified 20 key genes, the majority of which had been proven to be associated with radiation. However, we uniquely established them as space radiation sensitive genes for the first time. Through further analysis of the SSNs, we discovered that the different tissues exhibited distinct mechanisms in response to space stressors. CONCLUSIONS The topology structures of SSNs effectively predicted radiation doses under spaceflight conditions, and the SSNs revealed the gene regulatory patterns within the organisms under space stressors.
Collapse
Affiliation(s)
- Yan Zhang
- Institute of Environmental Systems Biology, College of Environmental Science and Engineering, Dalian Maritime University, Dalian, Liaoning, China
| | - Xiaohui Du
- Institute of Environmental Systems Biology, College of Environmental Science and Engineering, Dalian Maritime University, Dalian, Liaoning, China
| | - Lei Zhao
- Institute of Environmental Systems Biology, College of Environmental Science and Engineering, Dalian Maritime University, Dalian, Liaoning, China
| | - Yeqing Sun
- Institute of Environmental Systems Biology, College of Environmental Science and Engineering, Dalian Maritime University, Dalian, Liaoning, China
| |
Collapse
|
6
|
Liu S, Wang W, Hu S, Jia B, Tuo B, Sun H, Wang Q, Liu Y, Sun Z. Radiotherapy remodels the tumor microenvironment for enhancing immunotherapeutic sensitivity. Cell Death Dis 2023; 14:679. [PMID: 37833255 PMCID: PMC10575861 DOI: 10.1038/s41419-023-06211-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/22/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023]
Abstract
Cancer immunotherapy has transformed traditional treatments, with immune checkpoint blockade being particularly prominent. However, immunotherapy has minimal benefit for patients in most types of cancer and is largely ineffective in some cancers (such as pancreatic cancer and glioma). A synergistic anti-tumor response may be produced through the combined application with traditional tumor treatment methods. Radiotherapy (RT) not only kills tumor cells but also triggers the pro-inflammatory molecules' release and immune cell infiltration, which remodel the tumor microenvironment (TME). Therefore, the combination of RT and immunotherapy is expected to achieve improved efficacy. In this review, we summarize the effects of RT on cellular components of the TME, including T cell receptor repertoires, different T cell subsets, metabolism, tumor-associated macrophages and other myeloid cells (dendritic cells, myeloid-derived suppressor cells, neutrophils and eosinophils). Meanwhile, non-cellular components such as lactate and extracellular vesicles are also elaborated. In addition, we discuss the impact of different RT modalities on tumor immunity and issues related to the clinical practice of combination therapy.
Collapse
Affiliation(s)
- Senbo Liu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Wenkang Wang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Shengyun Hu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Bin Jia
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Baojing Tuo
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Haifeng Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Qiming Wang
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 450001, Zhengzhou, China.
| | - Yang Liu
- Department of Radiotherapy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 450001, Zhengzhou, China.
| | - Zhenqiang Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China.
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China.
| |
Collapse
|
7
|
Guberina N, Wirsdörfer F, Stuschke M, Jendrossek V. Combined radiation- and immune checkpoint-inhibitor-induced pneumonitis – The challenge to predict and detect overlapping immune-related adverse effects from evolving laboratory biomarkers and clinical imaging. Neoplasia 2023; 39:100892. [PMID: 37011458 PMCID: PMC10124136 DOI: 10.1016/j.neo.2023.100892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 02/17/2023] [Accepted: 02/23/2023] [Indexed: 04/04/2023]
Abstract
The risk of overlapping pulmonary toxicity induced by thoracic radio(chemo)therapy and immune checkpoint inhibitor therapy in the treatment of patients suffering from non-small cell lung cancer (NSCLC) is one important challenge in successful radioimmunotherapy. In the present opinion we highlight factors that we find important to be considered before treatment initiation, during the treatment sequence, and after treatment completion combined or sequential application of radio(chemo)therapy and immune checkpoint inhibitor therapy. A major aim is to optimize the therapeutic index and to avoid immune related adverse effects. The goals in the future will be focused not only on identifying patients already in the pretreatment phase who could benefit from this complex treatment, but also in identifying patients, who are most likely to have higher grade toxicity. In this respect, proper assessment of clinical performance status, monitoring for the presence of certain comorbidities, evaluation of laboratory parameters such as TGF-α and IL-6 levels, human leukocyte antigens (HLA), and consideration of other potential biomarkers which will evolve in near future are essential. Likewise, the critical parameters must be monitored during the treatment phase and follow-up care to detect potential side effects in time. With the help of high-end imaging which is already used on a daily basis in image guided radiotherapy (IGRT) for intensity modulated radiotherapy (IMRT), its advanced form volumetric modulated arc therapy (VMAT), and adaptive radiation therapy (ART), clinically relevant changes in lung tissue can be detected at an early stage of disease. Concurrent radiotherapy and immunotherapy requires a special focus on adverse events, particularly of the lung, but, when properly approached and applied, it may offer new perspectives for patients with locally advanced NSCLC to be seriously considered as a curative option.
Collapse
|
8
|
Watowich MB, Gilbert MR, Larion M. T cell exhaustion in malignant gliomas. Trends Cancer 2023; 9:270-292. [PMID: 36681605 PMCID: PMC10038906 DOI: 10.1016/j.trecan.2022.12.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 01/21/2023]
Abstract
Despite advances in understanding tumor biology, malignant gliomas remain incurable. While immunotherapy has improved outcomes in other cancer types, comparable efficacy has not yet been demonstrated for primary cancers of the central nervous system (CNS). T cell exhaustion, defined as a progressive decrease in effector function, sustained expression of inhibitory receptors, metabolic dysfunction, and distinct epigenetic and transcriptional alterations, contributes to the failure of immunotherapy in the CNS. Herein, we describe recent advances in understanding the drivers of T cell exhaustion in the glioma microenvironment. We discuss the extrinsic and intrinsic factors that contribute to exhaustion and highlight potential avenues for reversing this phenotype. Our ability to directly target specific immunosuppressive drivers in brain cancers would be a major advance in immunotherapy.
Collapse
Affiliation(s)
- Matthew B Watowich
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mark R Gilbert
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mioara Larion
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
9
|
Pham TN, Coupey J, Candeias SM, Ivanova V, Valable S, Thariat J. Beyond lymphopenia, unraveling radiation-induced leucocyte subpopulation kinetics and mechanisms through modeling approaches. J Exp Clin Cancer Res 2023; 42:50. [PMID: 36814272 PMCID: PMC9945629 DOI: 10.1186/s13046-023-02621-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 02/13/2023] [Indexed: 02/24/2023] Open
Abstract
Leucocyte subpopulations in both lymphoid and myeloid lineages have a significant impact on antitumor immune response. While radiation-induced lymphopenia is being studied extensively, radiation effects on lymphoid and myeloid subtypes have been relatively less addressed. Interactions between leucocyte subpopulations, their specific radiation sensitivity and the specific kinetics of each subpopulation can be modeled based on both experimental data and knowledge of physiological leucocyte depletion, production, proliferation, maturation and homeostasis. Modeling approaches of the leucocyte kinetics that may be used to unravel mechanisms underlying radiation induced-leucopenia and prediction of changes in cell counts and compositions after irradiation are presented in this review. The approaches described open up new possibilities for determining the influence of irradiation parameters both on a single-time point of acute effects and the subsequent recovery of leukocyte subpopulations. Utilization of these approaches to model kinetic data in post-radiotherapy states may be a useful tool for further development of new treatment strategies or for the combination of radiotherapy and immunotherapy.
Collapse
Affiliation(s)
- Thao-Nguyen Pham
- grid.412043.00000 0001 2186 4076Normandie Univ, UNICAEN, CNRS, ISTCT, GIP CYCERON, 14000 Caen, France ,grid.460771.30000 0004 1785 9671Laboratoire de Physique Corpusculaire UMR6534 IN2P3/ENSICAEN, Normandie Université, Caen, France
| | - Julie Coupey
- grid.412043.00000 0001 2186 4076Normandie Univ, UNICAEN, CNRS, ISTCT, GIP CYCERON, 14000 Caen, France
| | - Serge M. Candeias
- grid.457348.90000 0004 0630 1517Univ. Grenoble Alpes, CEA, CNRS, IRIG-LCBM-UMR5249, 38054 Grenoble, France
| | - Viktoriia Ivanova
- grid.412043.00000 0001 2186 4076Normandie Univ, UNICAEN, CNRS, ISTCT, GIP CYCERON, 14000 Caen, France
| | - Samuel Valable
- Normandie Univ, UNICAEN, CNRS, ISTCT, GIP CYCERON, 14000, Caen, France.
| | - Juliette Thariat
- Laboratoire de Physique Corpusculaire UMR6534 IN2P3/ENSICAEN, Normandie Université, Caen, France. .,Department of Radiation Oncology, Centre François Baclesse, Caen, Normandy, France.
| |
Collapse
|
10
|
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: 6.0] [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
|
11
|
Russ E, Davis CM, Slaven JE, Bradfield DT, Selwyn RG, Day RM. Comparison of the Medical Uses and Cellular Effects of High and Low Linear Energy Transfer Radiation. TOXICS 2022; 10:toxics10100628. [PMID: 36287908 PMCID: PMC9609561 DOI: 10.3390/toxics10100628] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 05/14/2023]
Abstract
Exposure to ionizing radiation can occur during medical treatments, from naturally occurring sources in the environment, or as the result of a nuclear accident or thermonuclear war. The severity of cellular damage from ionizing radiation exposure is dependent upon a number of factors including the absorbed radiation dose of the exposure (energy absorbed per unit mass of the exposure), dose rate, area and volume of tissue exposed, type of radiation (e.g., X-rays, high-energy gamma rays, protons, or neutrons) and linear energy transfer. While the dose, the dose rate, and dose distribution in tissue are aspects of a radiation exposure that can be varied experimentally or in medical treatments, the LET and eV are inherent characteristics of the type of radiation. High-LET radiation deposits a higher concentration of energy in a shorter distance when traversing tissue compared with low-LET radiation. The different biological effects of high and low LET with similar energies have been documented in vivo in animal models and in cultured cells. High-LET results in intense macromolecular damage and more cell death. Findings indicate that while both low- and high-LET radiation activate non-homologous end-joining DNA repair activity, efficient repair of high-LET radiation requires the homologous recombination repair pathway. Low- and high-LET radiation activate p53 transcription factor activity in most cells, but high LET activates NF-kB transcription factor at lower radiation doses than low-LET radiation. Here we review the development, uses, and current understanding of the cellular effects of low- and high-LET radiation exposure.
Collapse
Affiliation(s)
- Eric Russ
- Graduate Program of Cellular and Molecular Biology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Catherine M. Davis
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - John E. Slaven
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Dmitry T. Bradfield
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Reed G. Selwyn
- Department of Radiology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Regina M. Day
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Correspondence:
| |
Collapse
|
12
|
Tripathi AM, Khan S, Chaudhury NK. Radiomitigation by Melatonin in C57BL/6 Mice: Possible Implications as Adjuvant in Radiotherapy and Chemotherapy. In Vivo 2022; 36:1203-1221. [PMID: 35478105 DOI: 10.21873/invivo.12820] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/28/2022] [Accepted: 03/03/2022] [Indexed: 12/11/2022]
Abstract
BACKGROUND/AIM Melatonin (N-acetyl-5-methoxytryptamine), a chief secretory molecule of the pineal gland, has multiple properties, and numerous clinical investigations regarding its actions are in progress. This study investigated the radiomitigative role of melatonin in C57BL/6 mice. MATERIALS AND METHODS Melatonin (100 mg/kg) was orally administered once daily starting at 1 h on day 1 and subsequently every 24 h until day 7 after whole-body irradiation (WBI) and survival was monitored for 30 days. The bone marrow, spleen, and intestine were studied to evaluate the mitigative potential of melatonin after radiation-induced damage. RESULTS Melatonin significantly improved the survival upto 60% and 90% after 9 Gy (lethal) and 7.5 Gy (sub-lethal) WBI, respectively. Melatonin alleviated WBI-induced myelosuppression and pancytopenia, and increased white blood cell, red blood cell, platelet, and lymphocyte (CD4+ and CD8+) counts in peripheral blood. Bone marrow and spleen cellularity were restored through enhanced haematopoiesis. Melatonin ameliorated the damage in the small intestine, and promoted recovery of villi length, crypts number, and goblet cell count. CONCLUSION Melatonin mitigates the radiation-induced injury in the gastrointestinal and haematopoietic systems. The observed radiomitigative properties of melatonin can also be useful in the context of adjuvant therapy for cancer and radiotherapy.
Collapse
Affiliation(s)
- Akanchha Mani Tripathi
- Division of Radiation Biodosimetry, Institute of Nuclear Medicine and Allied Science, Defence Research & Development Organization, Delhi, India
| | - Shahanshah Khan
- Division of Radiation Biodosimetry, Institute of Nuclear Medicine and Allied Science, Defence Research & Development Organization, Delhi, India
| | - Nabo Kumar Chaudhury
- Division of Radiation Biodosimetry, Institute of Nuclear Medicine and Allied Science, Defence Research & Development Organization, Delhi, India
| |
Collapse
|
13
|
Askar MA, Guida MS, AbuNour SM, Ragab EA, Ali EN, Abdel-Magied N, Mansour NA, Elmasry SA. Nanoparticles for active combination radio mitigating agents of zinc coumarate and zinc caffeinate in a rat model. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:30233-30248. [PMID: 35000180 DOI: 10.1007/s11356-021-18411-4] [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: 01/01/2021] [Accepted: 12/26/2021] [Indexed: 06/14/2023]
Abstract
Zinc coumarate and zinc caffeinate nanoparticles (ZnCoNPs, ZnCaNPs) affect different biological processes. This study aimed to evaluate the mitigating action of ZnCoNPs in combination with ZnCaNPs against liver damage induced by gamma rays (γ-rays). Rats were exposed to 7 Gy of γ-rays and then injected intraperitoneally (i.p) with ZnCoNPs [2U/rat/day (5 mg/kg)] and ZnCaNPs [2U/rat/day (15 mg/kg)] for 7 consecutive days. The results showed that irradiated rats treated with ZnCoNPs (5 mg/kg/body weight) in combination with ZnCaNPs (15 mg/kg/body weight) for 7 days had a significant increases in body weight, antioxidant levels, T helper cell 4 (cluster of differentiation 4 (CD4)), and T cytotoxic cell 8 (cluster of differentiation 8 (CD8)), associated with a marked decrease in lipid peroxidation (LP), nitric oxide(NOx), total free radicals concentrate (TFRC), and DNA fragmentation. There were positive alterations in the morphological state, hematological parameters and the cell cycle phases. Additionally, the histopathological study demonstrated an improvement in the liver tissue of irradiated rats after treatment. Thus, ZnCoNPs and ZnCaNPs could be used as natural mitigating agents to reduce the hazards of ionizing radiation.
Collapse
Affiliation(s)
- Mostafa A Askar
- Department of Radiation Biology Research, National Centre for Radiation Research and Technology (NCRRT), Atomic Energy Authority, Cairo, Egypt
| | - Mona S Guida
- GenticUnit, Pediatric Hospital, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Seham M AbuNour
- Department of Health and Radiation Research, National Centre for Radiation Research and Technology (NCRRT), Atomic Energy Authority, Cairo, Egypt
| | - Essam A Ragab
- Department of Natural Product Research, National Centre for Radiation Research and Technology (NCRRT), Atomic Energy Authority, Cairo, Egypt
| | - Eiman N Ali
- Department of Radiation Biology Research, National Centre for Radiation Research and Technology (NCRRT), Atomic Energy Authority, Cairo, Egypt
| | - Nadia Abdel-Magied
- Department of Radiation Biology Research, National Centre for Radiation Research and Technology (NCRRT), Atomic Energy Authority, Cairo, Egypt.
| | - Nahla A Mansour
- Petrochemical Department, Egyptian Petroleum Research Institute, Nasr City, Cairo, Egypt
| | - Samir A Elmasry
- Department of Molecular Biology, Genetic Engineering & Biotechnology Research Institute, Sadat City, Egypt
| |
Collapse
|
14
|
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: 1.3] [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
|
15
|
Hussien SM. Cellular and Molecular Detection of Multi-doses of Ionizing Radiation-Induced Immunomodulatory Response. Cell Biochem Biophys 2021; 79:887-894. [PMID: 34224072 DOI: 10.1007/s12013-021-01017-5] [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] [Accepted: 06/23/2021] [Indexed: 11/26/2022]
Abstract
Ionizing radiation (IR) is used in a wide range of clinical applications. The study aims to evaluate various IR doses for their immunomodulatory responses, which can be used in multiple immunological conditions. Forty rats were exposed to whole-body gamma rays of 0, 0.25, 0.5, and 1 Gray (Gy). T-cell receptor (TCR) gene expression, serum transforming growth factor-beta, interleukin-10 (IL-10), and nitric oxide levels were measured on days 1 and 4 post irradiation. TCR activation occurred only at the genetic level, and radiation raised all measured parameters, even at low doses at α = 0.05 (P < 0.05). Except for IL-10, it shows a nearly 6% (P < 0.05) rise in early response in irradiated groups up to 0.5 Gy. At lower doses, the indirect impacts of IR were as essential as the direct impacts, and they increased over time in most measured parameters due to endogenous releases. They were having an anti-proliferative effect on the immune system. Lastly, a single acute IR dose can raise anti-inflammatory cytokines and anti-proliferative effects in the immune system, avoiding various contraindications associated with immunomodulatory drugs. More information on safety and clinical relevance is needed.
Collapse
Affiliation(s)
- Soha M Hussien
- Immunology lecturer, Radiation Safety Department, National Center for Nuclear Safety and Radiation Control, Egyptian Atomic Energy Authority/ Egyptian Nuclear and Radiological Regulatory Authority, 3 Ahmed Elzomor Str., 11762, P.O. Box 7551, Nasr City, Cairo, Egypt.
| |
Collapse
|
16
|
Li D, Guan Y, Dong Y, Wu J, Zhang Y, Chen R, Lin YT, Fornace AJ, Li HH, Yuan Z. Radiochemotherapy upregulates expression of checkpoint receptors on circulating T cells. Int J Radiat Biol 2021; 97:1563-1568. [PMID: 34402381 DOI: 10.1080/09553002.2021.1969051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE This study assesses changes of circulating leukocyte subpopulations and the expression of checkpoint receptors in T cells in patients undergoing radiochemotherapy. MATERIALS AND METHODS Fifty-seven patients with either esophageal cancer or cervical cancer who received radiochemotherapy were recruited into this study. Serial blood collection was carried out before and during treatments. Leukocyte subpopulations and the level of PD-1 and CTLA-4 in T cells were determined by flow cytometry. The plasma concentrations of 34 human cytokines, chemokines, and growth factors were quantified. RESULTS Significant decreases of lymphocyte count and percentage of T cells and B cells were observed during radiochemotherapy. Percentages of PD-1hi and CTLA-4hi populations in T cells increased after treatments. Proportion of activated T cells showed no significant difference. No significant changes in the plasma concentrations of the 34 humoral mediators except mild decreases of six cytokines. CONCLUSION Elevated expression of PD-1 and CTLA-4 in T cells in patients receiving radiochemotherapy, which suggests that exhaustion-like T-cell dysfunction develops during cancer cytotoxic treatments.
Collapse
Affiliation(s)
- Deguan Li
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yong Guan
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yinping Dong
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Jing Wu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yuanyang Zhang
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Renxiang Chen
- Department of Oncology, Georgetown University, Washington, DC, USA
| | - Yun-Tien Lin
- Department of Oncology, Georgetown University, Washington, DC, USA
| | - Albert J Fornace
- Department of Oncology, Georgetown University, Washington, DC, USA.,Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, USA
| | - Heng-Hong Li
- Department of Oncology, Georgetown University, Washington, DC, USA.,Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, USA
| | - Zhiyong Yuan
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
17
|
Senescent T cells: a potential biomarker and target for cancer therapy. EBioMedicine 2021; 68:103409. [PMID: 34049248 PMCID: PMC8170103 DOI: 10.1016/j.ebiom.2021.103409] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/09/2021] [Accepted: 05/06/2021] [Indexed: 12/27/2022] Open
Abstract
The failure of T cells to eradicate tumour cells in the tumour microenvironment is mainly due to the dysfunction of T cells. Senescent T cells, with defects in proliferation and effector functions, accumulate in ageing, chronic viral infections, and autoimmune disorders where antigen stimulation persists. Increasing evidence suggests that inducing T cell senescence is a key strategy used by malignant tumours to evade immune surveillance. In this review, we summarize the general features, functional regulation, and signalling network of senescent T cells in tumour development and highlight their potential as prognostic biomarkers in multiple cancer treatments, including chemotherapy, radiotherapy, and immunotherapy. Moreover, we discuss possible therapeutic strategies for preventing or rejuvenating senescence in tumour-specific T cells. Understanding these critical issues may provide novel strategies to enhance cancer immunotherapy.
Collapse
|
18
|
Laiakis EC, Shuryak I, Deziel A, Wang YW, Barnette BL, Yu Y, Ullrich RL, Fornace AJ, Emmett MR. Effects of Low Dose Space Radiation Exposures on the Splenic Metabolome. Int J Mol Sci 2021; 22:3070. [PMID: 33802822 PMCID: PMC8002539 DOI: 10.3390/ijms22063070] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/14/2021] [Accepted: 03/15/2021] [Indexed: 12/12/2022] Open
Abstract
Future space missions will include a return to the Moon and long duration deep space roundtrip missions to Mars. Leaving the protection that Low Earth Orbit provides will unavoidably expose astronauts to higher cumulative doses of space radiation, in addition to other stressors, e.g., microgravity. Immune regulation is known to be impacted by both radiation and spaceflight and it remains to be seen whether prolonged effects that will be encountered in deep space can have an adverse impact on health. In this study, we investigated the effects in the overall metabolism of three different low dose radiation exposures (γ-rays, 16O, and 56Fe) in spleens from male C57BL/6 mice at 1, 2, and 4 months after exposure. Forty metabolites were identified with significant enrichment in purine metabolism, tricarboxylic acid cycle, fatty acids, acylcarnitines, and amino acids. Early perturbations were more prominent in the γ irradiated samples, while later responses shifted towards more prominent responses in groups with high energy particle irradiations. Regression analysis showed a positive correlation of the abundance of identified fatty acids with time and a negative association with γ-rays, while the degradation pathway of purines was positively associated with time. Taken together, there is a strong suggestion of mitochondrial implication and the possibility of long-term effects on DNA repair and nucleotide pools following radiation exposure.
Collapse
Affiliation(s)
- Evagelia C. Laiakis
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington, DC 20057, USA; (A.D.); (Y.-W.W.); (A.J.F.J.)
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC 20057, USA
| | - Igor Shuryak
- Center for Radiological Research, Columbia University, New York, NY 10032, USA;
| | - Annabella Deziel
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington, DC 20057, USA; (A.D.); (Y.-W.W.); (A.J.F.J.)
| | - Yi-Wen Wang
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington, DC 20057, USA; (A.D.); (Y.-W.W.); (A.J.F.J.)
| | - Brooke L. Barnette
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA; (B.L.B.); (Y.Y.); (M.R.E.)
| | - Yongjia Yu
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA; (B.L.B.); (Y.Y.); (M.R.E.)
| | | | - Albert J. Fornace
- Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington, DC 20057, USA; (A.D.); (Y.-W.W.); (A.J.F.J.)
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC 20057, USA
| | - Mark R. Emmett
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA; (B.L.B.); (Y.Y.); (M.R.E.)
- Department of Radiation Oncology, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
19
|
Chen M, Singh AK, Repasky EA. Highlighting the Potential for Chronic Stress to Minimize Therapeutic Responses to Radiotherapy through Increased Immunosuppression and Radiation Resistance. Cancers (Basel) 2020; 12:E3853. [PMID: 33419318 PMCID: PMC7767049 DOI: 10.3390/cancers12123853] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 02/07/2023] Open
Abstract
Ionizing radiation has been used in the treatment of cancer for more than 100 years. While often very effective, there is still a great effort in place to improve the efficacy of radiation therapy for controlling the progression and recurrence of tumors. Recent research has revealed the close interaction between nerves and tumor progression, especially nerves of the autonomic nervous system that are activated by a variety of stressful stimuli including anxiety, pain, sleep loss or depression, each of which is likely to be increased in cancer patients. A growing literature now points to a negative effect of chronic stressful stimuli in tumor progression. In this review article, we present data on the potential for adrenergic stress to influence the efficacy of radiation and in particular, its potential to influence the anti-tumor immune response, and the frequency of an "abscopal effect" or the shrinkage of tumors which are outside an irradiated field. We conclude that chronic stress can be a major impediment to more effective radiation therapy through mechanisms involving immunosuppression and increased resistance to radiation-induced tumor cell death. Overall, these data highlight the potential value of stress reduction strategies to improve the outcome of radiation therapy. At the same time, objective biomarkers that can accurately and objectively reflect the degree of stress in patients over prolonged periods of time, and whether it is influencing immunosuppression and radiation resistance, are also critically needed.
Collapse
Affiliation(s)
- Minhui Chen
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| | - Anurag K. Singh
- Department of Radiation Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| | - Elizabeth A. Repasky
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| |
Collapse
|
20
|
Shen LF, Zhou SH, Guo Y. Role of GLUT-1 in the Upregulation of PD-L1 Expression After Radiotherapy and Association of PD-L1 with Favourable Overall Survival in Hypopharyngeal Cancer. Onco Targets Ther 2020; 13:11221-11235. [PMID: 33173312 PMCID: PMC7648563 DOI: 10.2147/ott.s269767] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/22/2020] [Indexed: 12/18/2022] Open
Abstract
Purpose The alteration of tumor immunity after radiotherapy (RT) has been widely studied in recent years. However, the mechanism through which RT mediates tumor immunity and the involvement of glycolysis in this mediation in hypopharyngeal cancer remain unclear. This study investigated whether RT regulates programmed cell death ligand 1(PD-L1) partly via glucose transporter 1(GLUT-1) expression and whether PD-L1 expression predicts overall survival (OS) in patients with hypopharyngeal cancer. Methods The expression of PD-L1 and Glut-1, and the numbers of CD4+, CD8+ T cells were detected by immunohistochemical analysis on 47 pre-RT and 25 post-RT specimens of hypopharyngeal cancer. Changes in these indicators before and after RT were compared, and their association with the OS of patients was analyzed. Moreover, we used siRNA-GLUT-1 to inhibit GLUT-1 expression and examined whether GLUT-1 was a key factor involved in the mediation of PD-L1 expression by RT in vitro. Results In the multivariate analysis, patients with higher PD-L1 expression (p=0.037), higher CD4+ T cell infiltration (p=0.016) and earlier clinical stage (p=0.019) had favourable OS. The expression of PD-L1, and the CD4+ and CD8+ T cells was markedly increased following RT. PD-L1 expression was correlated with Glut-1 pre-RT (p=0.002), but not after RT (p=0.051). The expression of PD-L1 in FaDu cells was upregulated after RT, especially at 96h after RT in vitro. However, the expression of PD-L1 in siRNA-GLUT-1 FaDu cells was markedly decreased at 96h after RT compared with that measured in FaDu cells. Conclusion Patients with high PD-L1 expression and CD4+ T cell infiltration may have favourable OS in hypopharyngeal cancer. RT may increase PD-L1 expression and alter tumor immunity. The expression of PD-L1 was correlated with Glut-1, and inhibition of GLUT-1 expression may decrease the expression of PD-L1. GLUT-1 may participate in the alteration of tumor immunity after RT.
Collapse
Affiliation(s)
- Li-Fang Shen
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Shui-Hong Zhou
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Yu Guo
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
21
|
Li JJ. Mitigating Coronavirus-Induced Acute Respiratory Distress Syndrome by Radiotherapy. iScience 2020; 23:101215. [PMID: 32512383 PMCID: PMC7260547 DOI: 10.1016/j.isci.2020.101215] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/26/2020] [Accepted: 05/26/2020] [Indexed: 01/08/2023] Open
Abstract
The acute respiratory distress syndrome (ARDS) induced by SARS-CoV-2-mediated cytokine storm (CS) in lungs leads to the high mortality in COVID-19 patients. To reduce ARDS, an ideal approach is to diminish virus loading by activating immune cells for CS prevention or to suppress the overactive cytokine-releasing immune cells for CS inhibition. Here, a potential radiation-mediated CS regulation is raised by reevaluating the radiation-mediated pneumonia control in the 1920s, with the following latent advantages of lung radiotherapy (LR) in treatment of COVID-19: (1) radiation accesses poorly circulated tissue more efficiently than blood-delivered medications; (2) low-dose radiation (LDR)-mediated metabolic rewiring and immune cell activation inhibit virus loading; (3) pre-consumption of immune reserves by LDR decreases CS severity; (4) higherdose radiation (HDR) within lung-tolerable doses relieves CS by eliminating in situ overactive cytokine-releasing cells. Thus, LDR and HDR or combined with antiviral and life-supporting modalities may mitigate SARS-CoV-2 and other virus-mediated ARDS.
Collapse
Affiliation(s)
- Jian Jian Li
- Department of Radiation Oncology, NCI-designated Comprehensive Cancer Center, University of California at Davis School of Medicine, 4501 X Street, Suite G0140, Sacramento, CA 95817, USA.
| |
Collapse
|
22
|
Borgatti A, Dickerson EB, Lawrence J. Emerging therapeutic approaches for canine sarcomas: Pushing the boundaries beyond the conventional. Vet Comp Oncol 2019; 18:9-24. [PMID: 31749286 DOI: 10.1111/vco.12554] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 11/08/2019] [Accepted: 11/11/2019] [Indexed: 12/21/2022]
Abstract
Sarcomas represent a group of genomically chaotic, highly heterogenous tumours of mesenchymal origin with variable mutational load. Conventional therapy with surgery and radiation therapy is effective for managing small, low-grade sarcomas and remains the standard therapeutic approach. For advanced, high-grade, recurrent, or metastatic sarcomas, systemic chemotherapy provides minimal benefit, therefore, there is a drive to develop novel approaches. The discovery of "Coley's toxins" in the 19th century, and their use to stimulate the immune system supported the application of unconventional therapies for the treatment of sarcomas. While promising, this initial work was abandoned and treatment paradigm and disease course of sarcomas was largely unchanged for several decades. Exciting new therapies are currently changing treatment algorithms for advanced carcinomas and melanomas, and similar approaches are being applied to advance the field of sarcoma research. Recent discoveries in subtype-specific cancer biology and the identification of distinct molecular targets have led to the development of promising targeted strategies with remarkable potential to change the landscape of sarcoma therapy in dogs. The purpose of this review article is to describe the current standard of care and limitations as well as emerging approaches for sarcoma therapy that span many of the most active paradigms in oncologic research, including immunotherapies, checkpoint inhibitors, and drugs capable of cellular metabolic reprogramming.
Collapse
Affiliation(s)
- Antonella Borgatti
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota.,Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.,Clinical Investigation Center, College of Veterinary Medicine, St. Paul, Minnesota
| | - Erin B Dickerson
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota.,Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Jessica Lawrence
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota.,Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
23
|
Tharmalingam S, Sreetharan S, Brooks AL, Boreham DR. Re-evaluation of the linear no-threshold (LNT) model using new paradigms and modern molecular studies. Chem Biol Interact 2019; 301:54-67. [PMID: 30763548 DOI: 10.1016/j.cbi.2018.11.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 11/13/2018] [Accepted: 11/22/2018] [Indexed: 02/06/2023]
Abstract
The linear no-threshold (LNT) model is currently used to estimate low dose radiation (LDR) induced health risks. This model lacks safety thresholds and postulates that health risks caused by ionizing radiation is directly proportional to dose. Therefore even the smallest radiation dose has the potential to cause an increase in cancer risk. Advances in LDR biology and cell molecular techniques demonstrate that the LNT model does not appropriately reflect the biology or the health effects at the low dose range. The main pitfall of the LNT model is due to the extrapolation of mutation and DNA damage studies that were conducted at high radiation doses delivered at a high dose-rate. These studies formed the basis of several outdated paradigms that are either incorrect or do not hold for LDR doses. Thus, the goal of this review is to summarize the modern cellular and molecular literature in LDR biology and provide new paradigms that better represent the biological effects in the low dose range. We demonstrate that LDR activates a variety of cellular defense mechanisms including DNA repair systems, programmed cell death (apoptosis), cell cycle arrest, senescence, adaptive memory, bystander effects, epigenetics, immune stimulation, and tumor suppression. The evidence presented in this review reveals that there are minimal health risks (cancer) with LDR exposure, and that a dose higher than some threshold value is necessary to achieve the harmful effects classically observed with high doses of radiation. Knowledge gained from this review can help the radiation protection community in making informed decisions regarding radiation policy and limits.
Collapse
Affiliation(s)
- Sujeenthar Tharmalingam
- Northern Ontario School of Medicine, Laurentian University, 935 Ramsey Lake Rd, Sudbury, ON, P3E 2C6, Canada.
| | - Shayenthiran Sreetharan
- Department of Medical Physics and Applied Radiation Sciences, McMaster University, 1280 Main Street W, Hamilton ON, L8S 4K1, Canada
| | - Antone L Brooks
- Environmental Science, Washington State University, Richland, WA, USA
| | - Douglas R Boreham
- Northern Ontario School of Medicine, Laurentian University, 935 Ramsey Lake Rd, Sudbury, ON, P3E 2C6, Canada; Bruce Power, Tiverton, ON(3), UK.
| |
Collapse
|
24
|
Basler L, Andratschke N, Ehrbar S, Guckenberger M, Tanadini-Lang S. Modelling the immunosuppressive effect of liver SBRT by simulating the dose to circulating lymphocytes: an in-silico planning study. Radiat Oncol 2018; 13:10. [PMID: 29357886 PMCID: PMC5778751 DOI: 10.1186/s13014-018-0952-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 01/03/2018] [Indexed: 12/18/2022] Open
Abstract
Background Tumor immune-evasion and associated failure of immunotherapy can potentially be overcome by radiotherapy, which however also has detrimental effects on tumor-infiltrating and circulating lymphocytes (CL). We therefore established a model to simulate the radiation-dose delivered to CL. Methods A MATLAB-model was established to quantify the CL-dose during SBRT of liver metastases by considering the factors: hepatic blood-flow, −velocity and transition-time of individual hepatic segments, as well as probability-based recirculation. The effects of intra-hepatic tumor-location and size, fractionation and treatment planning parameters (VMAT, 3DCRT, photon-energy, dose-rate and beam-on-time) were analyzed. A threshold dose ≥0.5Gy was considered inactivating CL and CL0.5 (%) is the proportion of inactivated CL. Results Mean liver dose was mostly influenced by treatment-modality, whereas CL0.5 was mostly influenced by beam-on-time. 3DCRT and VMAT (10MV-FFF) resulted in lowest CL0.5 values of 16 and 19%. Metastasis location influenced CL0.5, with a mean of 19% for both apical and basal and 31% for the central location. PTV-volume significantly increased CL0.5 from 27 to 67% (10MV-FFF) and from 31 to 98% (6MV-FFF) for PTV-volumes ranging from 14cm3 to 268cm3. Conclusion A simulation-model was established, quantifying the strong effects of treatment-technique, tumor-location and tumor-volume on dose to CL with potential implications for immune-optimized treatment-planning in the future. Electronic supplementary material The online version of this article (doi: 10.1186/s13014-018-0952-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- L Basler
- University Hospital Zurich, Department of Radiation Oncology, University of Zurich, Rämistrasse 100, CH 8091, Zürich, Switzerland.
| | - N Andratschke
- University Hospital Zurich, Department of Radiation Oncology, University of Zurich, Rämistrasse 100, CH 8091, Zürich, Switzerland
| | - S Ehrbar
- University Hospital Zurich, Department of Radiation Oncology, University of Zurich, Rämistrasse 100, CH 8091, Zürich, Switzerland
| | - M Guckenberger
- University Hospital Zurich, Department of Radiation Oncology, University of Zurich, Rämistrasse 100, CH 8091, Zürich, Switzerland
| | - S Tanadini-Lang
- University Hospital Zurich, Department of Radiation Oncology, University of Zurich, Rämistrasse 100, CH 8091, Zürich, Switzerland
| |
Collapse
|
25
|
Li D, Chen R, Wang YW, Fornace AJ, Li HH. Prior irradiation results in elevated programmed cell death protein 1 (PD-1) in T cells. Int J Radiat Biol 2017; 94:488-494. [PMID: 29108460 DOI: 10.1080/09553002.2017.1400192] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE In this study we addressed the question whether radiation-induced adverse effects on T cell activation are associated with alterations of T cell checkpoint receptors. MATERIALS AND METHODS Expression levels of checkpoint receptors on T cell subpopulations were analyzed at multiple post-radiation time points ranging from one to four weeks in mice receiving a single fraction of 1 or 4 Gy of γ-ray. T cell activation associated metabolic changes were assessed. RESULTS Our results showed that prior irradiation resulted in significant elevated expression of programmed cell death protein 1 (PD-1) in both CD4+ and CD8+ populations, at all three post-radiation time points. T cells with elevated PD-1 mostly were either central memory or naïve cells. In addition, the feedback induction of PD-1 expression in activated T cells declined after radiation. CONCLUSION Taken together, the elevated PD-1 level observed at weeks after radiation exposure is connected to T cell dysfunction. Recent preclinical and clinical studies have showed that a combination of radiotherapy and T cell checkpoint blockade immunotherapy including targeting the programmed death-ligand 1 (PD-L1)/PD-1 axis may potentiate the antitumor response. Understanding the dynamic changes in PD-1 levels in T cells after radiation should help in the development of a more effective therapeutic strategy.
Collapse
Affiliation(s)
- Deguan Li
- a Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College , Tianjin , China
| | - Renxiang Chen
- b Department of Biochemistry and Molecular & Cellular Biology , Georgetown University , Washington , DC , USA
| | - Yi-Wen Wang
- b Department of Biochemistry and Molecular & Cellular Biology , Georgetown University , Washington , DC , USA
| | - Albert J Fornace
- b Department of Biochemistry and Molecular & Cellular Biology , Georgetown University , Washington , DC , USA.,c Department of Oncology , Georgetown University , Washington , DC , USA
| | - Heng-Hong Li
- b Department of Biochemistry and Molecular & Cellular Biology , Georgetown University , Washington , DC , USA.,c Department of Oncology , Georgetown University , Washington , DC , USA
| |
Collapse
|
26
|
Komova O, Krasavin E, Nasonova E, Mel’nikova L, Shmakova N, Cunha M, Testa E, Beuve M. Relationship between radioadaptive response and individual radiosensitivity to low doses of gamma radiation: an extended study of chromosome damage in blood lymphocytes of three donors. Int J Radiat Biol 2017; 94:54-61. [DOI: 10.1080/09553002.2018.1399226] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Olga Komova
- Laboratory of Radiation Biology, Department of Radiation Cytology, Joint Institute for Nuclear Research (JINR), Dubna, Russia
| | - Eugene Krasavin
- Laboratory of Radiation Biology, Department of Radiation Cytology, Joint Institute for Nuclear Research (JINR), Dubna, Russia
| | - Elena Nasonova
- Laboratory of Radiation Biology, Department of Radiation Cytology, Joint Institute for Nuclear Research (JINR), Dubna, Russia
| | - Larisa Mel’nikova
- Laboratory of Radiation Biology, Department of Radiation Cytology, Joint Institute for Nuclear Research (JINR), Dubna, Russia
| | - Nina Shmakova
- Laboratory of Radiation Biology, Department of Radiation Cytology, Joint Institute for Nuclear Research (JINR), Dubna, Russia
| | - Micaela Cunha
- Department of Radiation Sciences, Université de Lyon, Lyon, France
- Department of Radiation Sciences, Institut de Physique Nucléaire de Lyon, Villeurbanne, France
| | - Etienne Testa
- Department of Radiation Sciences, Université de Lyon, Lyon, France
- Department of Radiation Sciences, Institut de Physique Nucléaire de Lyon, Villeurbanne, France
| | - Michaël Beuve
- Department of Radiation Sciences, Université de Lyon, Lyon, France
- Department of Radiation Sciences, Institut de Physique Nucléaire de Lyon, Villeurbanne, France
| |
Collapse
|
27
|
Cohen-Inbar O, Shih HH, Xu Z, Schlesinger D, Sheehan JP. The effect of timing of stereotactic radiosurgery treatment of melanoma brain metastases treated with ipilimumab. J Neurosurg 2017; 127:1007-1014. [DOI: 10.3171/2016.9.jns161585] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVEMelanoma represents the third most common cause of CNS metastases. Immunotherapy has evolved as a treatment option for patients with Stage IV melanoma. Stereotactic radiosurgery (SRS) also elicits an immune response within the brain and may interact with immunotherapy. The authors report on a cohort of patients treated for brain metastases with immunotherapy and evaluate the effect of SRS timing on the intracranial response.METHODSAll consecutively treated melanoma patients receiving ipilimumab and SRS for treatment of brain metastases at the University of Virginia between 2009 and 2014 were included in this retrospective analysis; data from 46 patients harboring 232 brain metastases were reviewed. The median duration of clinical follow-up was 7.9 months (range 3–42.6 months). The median age of the patients was 63 years (range 24.3–83.6 years). Thirty-two patients received SRS before or during ipilimumab cycles (Group A), whereas 14 patients received SRS after ipilimumab treatment (Group B). Radiographic and clinical responses were assessed at approximately 3-month intervals after SRS.RESULTSThe 2 cohorts were comparable in pertinent baseline characteristics with the exception of SRS timing relative to ipilimumab. Local recurrence–free duration (LRFD) was significantly longer in Group A (median 19.6 months, range 1.1–34.7 months) than in Group B patients (median 3 months, range 0.4–20.4 months) (p = 0.002). Post-SRS perilesional edema was more significant in Group A.CONCLUSIONSThe effect of SRS and ipilimumab on LRFD seems greater when SRS is performed before or during ipilimumab treatments. The timing of immunotherapy and SRS may affect LRFD and postradiosurgical edema. The interactions between immunotherapy and SRS warrant further investigation so as to optimize the therapeutic benefits and mitigate the risks associated with multimodality, targeted therapy.
Collapse
Affiliation(s)
- Or Cohen-Inbar
- 1Department of Neurological Surgery, University of Virginia, Charlottesville, Virginia
- 2Department of Neurosurgery, Rambam Health Care Center, Haifa, Israel
| | - Han-Hsun Shih
- 1Department of Neurological Surgery, University of Virginia, Charlottesville, Virginia
- 3Department of Anesthesiology, Taichung Veterans General Hospital, Taichung City; and
- 4National Yang-Ming University, Taipei, Taiwan
| | - Zhiyuan Xu
- 1Department of Neurological Surgery, University of Virginia, Charlottesville, Virginia
| | - David Schlesinger
- 1Department of Neurological Surgery, University of Virginia, Charlottesville, Virginia
| | - Jason P. Sheehan
- 1Department of Neurological Surgery, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
28
|
Hall J, Jeggo PA, West C, Gomolka M, Quintens R, Badie C, Laurent O, Aerts A, Anastasov N, Azimzadeh O, Azizova T, Baatout S, Baselet B, Benotmane MA, Blanchardon E, Guéguen Y, Haghdoost S, Harms-Ringhdahl M, Hess J, Kreuzer M, Laurier D, Macaeva E, Manning G, Pernot E, Ravanat JL, Sabatier L, Tack K, Tapio S, Zitzelsberger H, Cardis E. Ionizing radiation biomarkers in epidemiological studies - An update. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2017; 771:59-84. [PMID: 28342453 DOI: 10.1016/j.mrrev.2017.01.001] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 01/09/2017] [Indexed: 01/13/2023]
Abstract
Recent epidemiology studies highlighted the detrimental health effects of exposure to low dose and low dose rate ionizing radiation (IR): nuclear industry workers studies have shown increased leukaemia and solid tumour risks following cumulative doses of <100mSv and dose rates of <10mGy per year; paediatric patients studies have reported increased leukaemia and brain tumours risks after doses of 30-60mGy from computed tomography scans. Questions arise, however, about the impact of even lower doses and dose rates where classical epidemiological studies have limited power but where subsets within the large cohorts are expected to have an increased risk. Further progress requires integration of biomarkers or bioassays of individual exposure, effects and susceptibility to IR. The European DoReMi (Low Dose Research towards Multidisciplinary Integration) consortium previously reviewed biomarkers for potential use in IR epidemiological studies. Given the increased mechanistic understanding of responses to low dose radiation the current review provides an update covering technical advances and recent studies. A key issue identified is deciding which biomarkers to progress. A roadmap is provided for biomarker development from discovery to implementation and used to summarise the current status of proposed biomarkers for epidemiological studies. Most potential biomarkers remain at the discovery stage and for some there is sufficient evidence that further development is not warranted. One biomarker identified in the final stages of development and as a priority for further research is radiation specific mRNA transcript profiles.
Collapse
Affiliation(s)
- Janet Hall
- Centre de Recherche en Cancérologie de Lyon, INSERM 1052, CNRS 5286, Univ Lyon, Université Claude Bernard, Lyon 1, Lyon, F-69424, France.
| | - Penny A Jeggo
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Falmer, Brighton, BN1 9RQ, United Kingdom
| | - Catharine West
- Translational Radiobiology Group, Institute of Cancer Sciences, The University of Manchester, Manchester Academic Health Science Centre, Christie Hospital, Manchester, M20 4BX, United Kingdom
| | - Maria Gomolka
- Federal Office for Radiation Protection, Department of Radiation Protection and Health, D-85764 Neuherberg, Germany
| | - Roel Quintens
- Radiobiology Unit, Belgian Nuclear Research Centre, SCK·CEN, B-2400 Mol, Belgium
| | - Christophe Badie
- Cancer Mechanisms and Biomarkers group, Radiation Effects Department, Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Chilton, Didcot, United Kingdom
| | - Olivier Laurent
- Institut de Radioprotection et de Sûreté Nucléaire, F-92260 Fontenay-aux-Roses, France
| | - An Aerts
- Radiobiology Unit, Belgian Nuclear Research Centre, SCK·CEN, B-2400 Mol, Belgium
| | - Nataša Anastasov
- Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Institute of Radiation Biology, D-85764 Neuherberg, Germany
| | - Omid Azimzadeh
- Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Institute of Radiation Biology, D-85764 Neuherberg, Germany
| | - Tamara Azizova
- Southern Urals Biophysics Institute, Clinical Department, Ozyorsk, Russia
| | - Sarah Baatout
- Radiobiology Unit, Belgian Nuclear Research Centre, SCK·CEN, B-2400 Mol, Belgium; Cell Systems and Imaging Research Group, Department of Molecular Biotechnology, Ghent University, B-9000 Ghent, Belgium
| | - Bjorn Baselet
- Radiobiology Unit, Belgian Nuclear Research Centre, SCK·CEN, B-2400 Mol, Belgium; Pole of Pharmacology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, B-1200 Brussels, Belgium
| | - Mohammed A Benotmane
- Radiobiology Unit, Belgian Nuclear Research Centre, SCK·CEN, B-2400 Mol, Belgium
| | - Eric Blanchardon
- Institut de Radioprotection et de Sûreté Nucléaire, F-92260 Fontenay-aux-Roses, France
| | - Yann Guéguen
- Institut de Radioprotection et de Sûreté Nucléaire, F-92260 Fontenay-aux-Roses, France
| | - Siamak Haghdoost
- Centre for Radiation Protection Research, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE 106 91 Stockholm, Sweden
| | - Mats Harms-Ringhdahl
- Centre for Radiation Protection Research, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE 106 91 Stockholm, Sweden
| | - Julia Hess
- Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Institute of Radiation Biology, D-85764 Neuherberg, Germany
| | - Michaela Kreuzer
- Federal Office for Radiation Protection, Department of Radiation Protection and Health, D-85764 Neuherberg, Germany
| | - Dominique Laurier
- Institut de Radioprotection et de Sûreté Nucléaire, F-92260 Fontenay-aux-Roses, France
| | - Ellina Macaeva
- Radiobiology Unit, Belgian Nuclear Research Centre, SCK·CEN, B-2400 Mol, Belgium; Cell Systems and Imaging Research Group, Department of Molecular Biotechnology, Ghent University, B-9000 Ghent, Belgium
| | - Grainne Manning
- Cancer Mechanisms and Biomarkers group, Radiation Effects Department, Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Chilton, Didcot, United Kingdom
| | - Eileen Pernot
- INSERM U897, Université de Bordeaux, F-33076 Bordeaux cedex, France
| | - Jean-Luc Ravanat
- Laboratoire des Lésions des Acides Nucléiques, Univ. Grenoble Alpes, INAC-SCIB, F-38000 Grenoble, France; Commissariat à l'Énergie Atomique, INAC-SyMMES, F-38000 Grenoble, France
| | - Laure Sabatier
- Commissariat à l'Énergie Atomique, BP6, F-92265 Fontenay-aux-Roses, France
| | - Karine Tack
- Institut de Radioprotection et de Sûreté Nucléaire, F-92260 Fontenay-aux-Roses, France
| | - Soile Tapio
- Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Institute of Radiation Biology, D-85764 Neuherberg, Germany
| | - Horst Zitzelsberger
- Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, Institute of Radiation Biology, D-85764 Neuherberg, Germany
| | - Elisabeth Cardis
- Barcelona Institute of Global Health (ISGlobal), Centre for Research in Environmental Epidemiology, Radiation Programme, Barcelona Biomedical Research Park, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF) (MTD formerly), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.
| |
Collapse
|
29
|
Chen R, Wang YW, Fornace AJ, Li HH. Impairment of the Intrinsic Capability of Th1 Polarization in Irradiated Mice: A Close Look at the Imbalanced Th1/Th2 Response after Irradiation. Radiat Res 2016; 186:559-567. [PMID: 27849436 DOI: 10.1667/rr14401.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Two major CD4+ T-helper (Th) lineages are Th1 and Th2, and well balanced Th1/Th2 responses are essential for immune function. In previously published studies, it was reported that radiation induces a Th1/Th2 immune imbalance toward a Th2-dominant direction, and this imbalance may contribute to postirradiation immune dysfunction. The polarization of Th cells is driven by the cytokine milieu and controlled by intracellular regulatory pathways that respond to cytokine signaling. It is widely accepted that radiation induces cytokine aberration, however, the precise alterations of cytokines in various tissue environments have been difficult to evaluate. In addition, the effects of radiation on the intrinsic functions of Th cells remain uncharacterized. Therefore, how radiation affects Th1/Th2 balance remains somewhat unclear. To address this, we investigated the changes in the polarization capability of Th cells by isolating them from mice previously exposed to radiation and assessing the cells in an established in vitro Th polarization system. Our novel results demonstrate that prior exposure to radiation led to the persistent aberration of the inherent capability of Th cells to differentiate into Th1 and Th2 lineages. The parallel changes in expression of Th1-specific master transcription factors and the key genes in metabolic reprograming indicated that radiation affects the core components in Th1 polarization. While Th1 differentiation was impaired after irradiation, little adverse effect was observed in Th2 differentiation; both of these findings contribute to the known phenotypes of Th1/Th2 imbalance caused by radiation.
Collapse
Affiliation(s)
- Renxiang Chen
- a Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, D.C. 20057
| | - Yi-Wen Wang
- a Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, D.C. 20057
| | - Albert J Fornace
- a Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, D.C. 20057.,b Department of Oncology, Georgetown University Medical Center, Washington, D.C. 20057
| | - Heng-Hong Li
- a Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, D.C. 20057.,b Department of Oncology, Georgetown University Medical Center, Washington, D.C. 20057
| |
Collapse
|
30
|
Pinto AT, Pinto ML, Velho S, Pinto MT, Cardoso AP, Figueira R, Monteiro A, Marques M, Seruca R, Barbosa MA, Mareel M, Oliveira MJ, Rocha S. Intricate Macrophage-Colorectal Cancer Cell Communication in Response to Radiation. PLoS One 2016; 11:e0160891. [PMID: 27513864 PMCID: PMC4981353 DOI: 10.1371/journal.pone.0160891] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 07/26/2016] [Indexed: 11/19/2022] Open
Abstract
Both cancer and tumour-associated host cells are exposed to ionizing radiation when a tumour is subjected to radiotherapy. Macrophages frequently constitute the most abundant tumour-associated immune population, playing a role in tumour progression and response to therapy. The present work aimed to evaluate the importance of macrophage-cancer cell communication in the cellular response to radiation. To address this question, we established monocultures and indirect co-cultures of human monocyte-derived macrophages with RKO or SW1463 colorectal cancer cells, which exhibit higher and lower radiation sensitivity, respectively. Mono- and co-cultures were then irradiated with 5 cumulative doses, in a similar fractionated scheme to that used during cancer patients' treatment (2 Gy/fraction/day). Our results demonstrated that macrophages sensitize RKO to radiation-induced apoptosis, while protecting SW1463 cells. Additionally, the co-culture with macrophages increased the mRNA expression of metabolism- and survival-related genes more in SW1463 than in RKO. The presence of macrophages also upregulated glucose transporter 1 expression in irradiated SW1463, but not in RKO cells. In addition, the influence of cancer cells on the expression of pro- and anti-inflammatory macrophage markers, upon radiation exposure, was also evaluated. In the presence of RKO or SW1463, irradiated macrophages exhibit higher levels of pro-inflammatory TNF, IL6, CCL2 and CCR7, and of anti-inflammatory CCL18. However, RKO cells induce an increase of macrophage pro-inflammatory IL1B, while SW1463 cells promote higher pro-inflammatory CXCL8 and CD80, and also anti-inflammatory VCAN and IL10 levels. Thus, our data demonstrated that macrophages and cancer cells mutually influence their response to radiation. Notably, conditioned medium from irradiated co-cultures increased non-irradiated RKO cell migration and invasion and did not impact on angiogenesis in a chicken embryo chorioallantoic membrane assay. Overall, the establishment of primary human macrophage-cancer cell co-cultures revealed an intricate cell communication in response to ionizing radiation, which should be considered when developing therapies adjuvant to radiotherapy.
Collapse
Affiliation(s)
- Ana T. Pinto
- i3s-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB-Institute of Biomedical Engineering, University of Porto, Porto, Portugal
- FEUP-Faculty of Engineering, University of Porto, Porto, Portugal
| | - Marta L. Pinto
- i3s-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB-Institute of Biomedical Engineering, University of Porto, Porto, Portugal
- ICBAS-Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Sérgia Velho
- i3s-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IPATIMUP-Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
| | - Marta T. Pinto
- i3s-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IPATIMUP-Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
| | - Ana P. Cardoso
- i3s-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB-Institute of Biomedical Engineering, University of Porto, Porto, Portugal
| | - Rita Figueira
- Radiotherapy Service, Centro Hospitalar S. João, EPE, Porto, Portugal
| | - Armanda Monteiro
- Radiotherapy Service, Centro Hospitalar S. João, EPE, Porto, Portugal
| | - Margarida Marques
- Radiotherapy Service, Centro Hospitalar S. João, EPE, Porto, Portugal
| | - Raquel Seruca
- i3s-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IPATIMUP-Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
- Department of Pathology and Oncology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Mário A. Barbosa
- i3s-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB-Institute of Biomedical Engineering, University of Porto, Porto, Portugal
- ICBAS-Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Marc Mareel
- Department of Radiation Oncology and Experimental Cancer Research, Ghent University Hospital, Ghent, Belgium
| | - Maria J. Oliveira
- i3s-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB-Institute of Biomedical Engineering, University of Porto, Porto, Portugal
- Department of Pathology and Oncology, Faculty of Medicine, University of Porto, Porto, Portugal
- * E-mail:
| | - Sónia Rocha
- Centre for Gene Regulation and Expression, College of Life Sciences, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
31
|
Sridharan DM, Asaithamby A, Blattnig SR, Costes SV, Doetsch PW, Dynan WS, Hahnfeldt P, Hlatky L, Kidane Y, Kronenberg A, Naidu MD, Peterson LE, Plante I, Ponomarev AL, Saha J, Snijders AM, Srinivasan K, Tang J, Werner E, Pluth JM. Evaluating biomarkers to model cancer risk post cosmic ray exposure. LIFE SCIENCES IN SPACE RESEARCH 2016; 9:19-47. [PMID: 27345199 PMCID: PMC5613937 DOI: 10.1016/j.lssr.2016.05.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 05/11/2016] [Indexed: 06/06/2023]
Abstract
Robust predictive models are essential to manage the risk of radiation-induced carcinogenesis. Chronic exposure to cosmic rays in the context of the complex deep space environment may place astronauts at high cancer risk. To estimate this risk, it is critical to understand how radiation-induced cellular stress impacts cell fate decisions and how this in turn alters the risk of carcinogenesis. Exposure to the heavy ion component of cosmic rays triggers a multitude of cellular changes, depending on the rate of exposure, the type of damage incurred and individual susceptibility. Heterogeneity in dose, dose rate, radiation quality, energy and particle flux contribute to the complexity of risk assessment. To unravel the impact of each of these factors, it is critical to identify sensitive biomarkers that can serve as inputs for robust modeling of individual risk of cancer or other long-term health consequences of exposure. Limitations in sensitivity of biomarkers to dose and dose rate, and the complexity of longitudinal monitoring, are some of the factors that increase uncertainties in the output from risk prediction models. Here, we critically evaluate candidate early and late biomarkers of radiation exposure and discuss their usefulness in predicting cell fate decisions. Some of the biomarkers we have reviewed include complex clustered DNA damage, persistent DNA repair foci, reactive oxygen species, chromosome aberrations and inflammation. Other biomarkers discussed, often assayed for at longer points post exposure, include mutations, chromosome aberrations, reactive oxygen species and telomere length changes. We discuss the relationship of biomarkers to different potential cell fates, including proliferation, apoptosis, senescence, and loss of stemness, which can propagate genomic instability and alter tissue composition and the underlying mRNA signatures that contribute to cell fate decisions. Our goal is to highlight factors that are important in choosing biomarkers and to evaluate the potential for biomarkers to inform models of post exposure cancer risk. Because cellular stress response pathways to space radiation and environmental carcinogens share common nodes, biomarker-driven risk models may be broadly applicable for estimating risks for other carcinogens.
Collapse
Affiliation(s)
| | | | - Steve R Blattnig
- Langley Research Center, Langley Research Center (LaRC), VA, United States
| | - Sylvain V Costes
- Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | | | | | | | - Lynn Hlatky
- CCSB-Tufts School of Medicine, Boston, MA, United States
| | - Yared Kidane
- Wyle Science, Technology & Engineering Group, Houston, TX, United States
| | - Amy Kronenberg
- Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Mamta D Naidu
- CCSB-Tufts School of Medicine, Boston, MA, United States
| | - Leif E Peterson
- Houston Methodist Research Institute, Houston, TX, United States
| | - Ianik Plante
- Wyle Science, Technology & Engineering Group, Houston, TX, United States
| | - Artem L Ponomarev
- Wyle Science, Technology & Engineering Group, Houston, TX, United States
| | - Janapriya Saha
- UT Southwestern Medical Center, Dallas, TX, United States
| | | | | | - Jonathan Tang
- Exogen Biotechnology, Inc., Berkeley, CA, United States
| | | | - Janice M Pluth
- Lawrence Berkeley National Laboratory, Berkeley, CA, United States.
| |
Collapse
|
32
|
Laiakis EC, Pannkuk EL, Diaz-Rubio ME, Wang YW, Mak TD, Simbulan-Rosenthal CM, Brenner DJ, Fornace AJ. Implications of genotypic differences in the generation of a urinary metabolomics radiation signature. Mutat Res 2016; 788:41-9. [PMID: 27040378 PMCID: PMC4887295 DOI: 10.1016/j.mrfmmm.2016.03.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 03/04/2016] [Accepted: 03/24/2016] [Indexed: 02/04/2023]
Abstract
The increased threat of radiological terrorism and accidental nuclear exposures, together with increased usage of radiation-based medical procedures, has made necessary the development of minimally invasive methods for rapid identification of exposed individuals. Genetically predisposed radiosensitive individuals comprise a significant number of the population and require specialized attention and treatments after such events. Metabolomics, the assessment of the collective small molecule content in a given biofluid or tissue, has proven effective in the rapid identification of radiation biomarkers and metabolic perturbations. To investigate how the genotypic background may alter the ionizing radiation (IR) signature, we analyzed urine from Parp1(-/-) mice, as a model radiosensitive genotype, exposed to IR by utilizing the analytical power of liquid chromatography coupled with mass spectrometry (LC-MS), as urine has been thoroughly investigated in wild type (WT) mice in previous studies from our laboratory. Samples were collected at days one and three after irradiation, time points that are important for the early and efficient triage of exposed individuals. Time-dependent perturbations in metabolites were observed in the tricarboxylic acid pathway (TCA). Other differentially excreted metabolites included amino acids and metabolites associated with dysregulation of energy metabolism pathways. Time-dependent apoptotic pathway activation between WT and mutant mice following IR exposure may explain the altered excretion patterns, although the origin of the metabolites remains to be determined. This first metabolomics study in urine from radiation exposed genetic mutant animal models provides evidence that this technology can be used to dissect the effects of genotoxic agents on metabolism by assessing easily accessible biofluids and identify biomarkers of radiation exposure. Applications of metabolomics could be incorporated in the future to further elucidate the effects of IR on the metabolism of Parp1(-/-) genotype by assessing individual tissues.
Collapse
Affiliation(s)
- Evagelia C Laiakis
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington DC, USA.
| | - Evan L Pannkuk
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington DC, USA
| | - Maria Elena Diaz-Rubio
- Pediatrics, Division of Developmental Nutrition, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Yi-Wen Wang
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington DC, USA
| | - Tytus D Mak
- Mass Spectrometry Data Center, National Institute of Standards and Technology (NIST), Gaithersburg MD, USA
| | | | | | - Albert J Fornace
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington DC, USA; Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC, USA; Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah 22254, Saudi Arabia
| |
Collapse
|
33
|
Szyska M, Na IK. Bone Marrow GvHD after Allogeneic Hematopoietic Stem Cell Transplantation. Front Immunol 2016; 7:118. [PMID: 27066008 PMCID: PMC4811960 DOI: 10.3389/fimmu.2016.00118] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 03/15/2016] [Indexed: 12/15/2022] Open
Abstract
The bone marrow is the origin of all hematopoietic lineages and an important homing site for memory cells of the adaptive immune system. It has recently emerged as a graft-versus-host disease (GvHD) target organ after allogeneic stem cell transplantation (alloHSCT), marked by depletion of both hematopoietic progenitors and niche-forming cells. Serious effects on the restoration of hematopoietic function and immunological memory are common, especially in patients after myeloablative conditioning therapy. Cytopenia and durable immunodeficiency caused by the depletion of hematopoietic progenitors and destruction of bone marrow niches negatively influence the outcome of alloHSCT. The complex balance between immunosuppressive and cell-depleting treatments, GvHD and immune reconstitution, as well as the desirable graft-versus-tumor (GvT) effect remains a great challenge for clinicians.
Collapse
Affiliation(s)
- Martin Szyska
- Experimental and Clinical Research Center (ECRC) , Berlin , Germany
| | - Il-Kang Na
- Experimental and Clinical Research Center (ECRC), Berlin, Germany; Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
34
|
Menon SS, Uppal M, Randhawa S, Cheema MS, Aghdam N, Usala RL, Ghosh SP, Cheema AK, Dritschilo A. Radiation Metabolomics: Current Status and Future Directions. Front Oncol 2016; 6:20. [PMID: 26870697 PMCID: PMC4736121 DOI: 10.3389/fonc.2016.00020] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 01/18/2016] [Indexed: 12/25/2022] Open
Abstract
Human exposure to ionizing radiation (IR) disrupts normal metabolic processes in cells and organs by inducing complex biological responses that interfere with gene and protein expression. Conventional dosimetry, monitoring of prodromal symptoms, and peripheral lymphocyte counts are of limited value as organ- and tissue-specific biomarkers for personnel exposed to radiation, particularly, weeks or months after exposure. Analysis of metabolites generated in known stress-responsive pathways by molecular profiling helps to predict the physiological status of an individual in response to environmental or genetic perturbations. Thus, a multi-metabolite profile obtained from a high-resolution mass spectrometry-based metabolomics platform offers potential for identification of robust biomarkers to predict radiation toxicity of organs and tissues resulting from exposures to therapeutic or non-therapeutic IR. Here, we review the status of radiation metabolomics and explore applications as a standalone technology, as well as its integration in systems biology, to facilitate a better understanding of the molecular basis of radiation response. Finally, we draw attention to the identification of specific pathways that can be targeted for the development of therapeutics to alleviate or mitigate harmful effects of radiation exposure.
Collapse
Affiliation(s)
- Smrithi S Menon
- Department of Oncology, Georgetown University Medical Center , Washington, DC , USA
| | - Medha Uppal
- Department of Oncology, Georgetown University Medical Center , Washington, DC , USA
| | - Subeena Randhawa
- Department of Oncology, Georgetown University Medical Center , Washington, DC , USA
| | - Mehar S Cheema
- Department of Radiation Medicine, Georgetown University Medical Center , Washington, DC , USA
| | - Nima Aghdam
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center , Washington, DC , USA
| | - Rachel L Usala
- School of Medicine, Georgetown University Medical Center , Washington, DC , USA
| | - Sanchita P Ghosh
- Armed Forces Radiobiology Research Institute , Bethesda, MD , USA
| | - Amrita K Cheema
- Department of Oncology, Georgetown University Medical Center , Washington, DC , USA
| | - Anatoly Dritschilo
- Department of Radiation Medicine, Georgetown University Medical Center , Washington, DC , USA
| |
Collapse
|