1
|
Lyng FM, Azzam EI. Abscopal Effects, Clastogenic Effects and Bystander Effects: 70 Years of Non-Targeted Effects of Radiation. Radiat Res 2024; 202:355-367. [PMID: 38986531 DOI: 10.1667/rade-24-00040.1] [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: 01/31/2024] [Accepted: 04/29/2024] [Indexed: 07/12/2024]
Abstract
In vitro and in vivo observations accumulated over several decades have firmly shown that the biological effects of ionizing radiation can spread from irradiated cells/tissues to non-targeted cells/tissues. Redox-modulated intercellular communication mechanisms that include a role for secreted factors and gap junctions, can mediate these non-targeted effects. Clearly, the expression of such effects and their transmission to progeny cells has implications for issues related to radiation protection. Their elucidation is also relevant towards enhancing the efficacy of cancer radiotherapy and reducing its impact on the development of normal tissue toxicities. In addition, the study of non-targeted effects is pertinent to our basic understanding of intercellular communications under conditions of oxidative stress. This review will trace the history of non-targeted effects of radiation starting with early reports of abscopal effects which described radiation induced effects in tissues distant from the site of radiation exposure. A related effect involved the production of clastogenic factors in plasma following irradiation which can induce chromosome damage in unirradiated cells. Despite these early reports suggesting non-targeted effects of radiation, the classical paradigm that a direct deposition of energy in the nucleus was required still dominated. This paradigm was challenged by papers describing radiation induced bystander effects. This review will cover mechanisms of radiation-induced bystander effects and the potential impacts on radiation protection and radiation therapy.
Collapse
Affiliation(s)
- Fiona M Lyng
- Radiation and Environmental Science Centre, FOCAS Research Institute
- School of Physics, Clinical and Optometric Sciences, Technological University Dublin, Dublin, Ireland
| | - Edouard I Azzam
- Department of Radiology, Rutgers New Jersey Medical School Cancer Center, Newark, New Jersey
| |
Collapse
|
2
|
Giroud J, Bouriez I, Paulus H, Pourtier A, Debacq-Chainiaux F, Pluquet O. Exploring the Communication of the SASP: Dynamic, Interactive, and Adaptive Effects on the Microenvironment. Int J Mol Sci 2023; 24:10788. [PMID: 37445973 DOI: 10.3390/ijms241310788] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/20/2023] [Accepted: 06/25/2023] [Indexed: 07/15/2023] Open
Abstract
Cellular senescence is a complex cell state that can occur during physiological ageing or after exposure to stress signals, regardless of age. It is a dynamic process that continuously evolves in a context-dependent manner. Senescent cells interact with their microenvironment by producing a heterogenous and plastic secretome referred to as the senescence-associated secretory phenotype (SASP). Hence, understanding the cross-talk between SASP and the microenvironment can be challenging due to the complexity of signal exchanges. In this review, we first aim to update the definition of senescence and its associated biomarkers from its discovery to the present day. We detail the regulatory mechanisms involved in the expression of SASP at multiple levels and develop how SASP can orchestrate microenvironment modifications, by focusing on extracellular matrix modifications, neighboring cells' fate, and intercellular communications. We present hypotheses on how these microenvironmental events may affect dynamic changes in SASP composition in return. Finally, we discuss the various existing approaches to targeting SASP and clarify what is currently known about the biological effects of these modified SASPs on the cellular environment.
Collapse
Affiliation(s)
- Joëlle Giroud
- Laboratory of Biochemistry and Cell Biology (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur, 5000 Namur, Belgium
- University of Lille, CNRS, Inserm, Pasteur Institute of Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, 59000 Lille, France
| | - Inès Bouriez
- Laboratory of Biochemistry and Cell Biology (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur, 5000 Namur, Belgium
| | - Hugo Paulus
- Laboratory of Biochemistry and Cell Biology (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur, 5000 Namur, Belgium
| | - Albin Pourtier
- University of Lille, CNRS, Inserm, Pasteur Institute of Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, 59000 Lille, France
| | - Florence Debacq-Chainiaux
- Laboratory of Biochemistry and Cell Biology (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur, 5000 Namur, Belgium
| | - Olivier Pluquet
- University of Lille, CNRS, Inserm, Pasteur Institute of Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, 59000 Lille, France
| |
Collapse
|
3
|
Escoubas CC, Dorman LC, Nguyen PT, Lagares-Linares C, Nakajo H, Anderson SR, Cuevas B, Vainchtein ID, Silva NJ, Xiao Y, Lidsky PV, Wang EY, Taloma SE, Nakao-Inoue H, Schwer B, Andino R, Nowakowski TJ, Molofsky AV. Type I interferon responsive microglia shape cortical development and behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2021.04.29.441889. [PMID: 35233577 PMCID: PMC8887080 DOI: 10.1101/2021.04.29.441889] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Microglia are brain resident phagocytes that can engulf synaptic components and extracellular matrix as well as whole neurons. However, whether there are unique molecular mechanisms that regulate these distinct phagocytic states is unknown. Here we define a molecularly distinct microglial subset whose function is to engulf neurons in the developing brain. We transcriptomically identified a cluster of Type I interferon (IFN-I) responsive microglia that expanded 20-fold in the postnatal day 5 somatosensory cortex after partial whisker deprivation, a stressor that accelerates neural circuit remodeling. In situ, IFN-I responsive microglia were highly phagocytic and actively engulfed whole neurons. Conditional deletion of IFN-I signaling (Ifnar1fl/fl) in microglia but not neurons resulted in dysmorphic microglia with stalled phagocytosis and an accumulation of neurons with double strand DNA breaks, a marker of cell stress. Conversely, exogenous IFN-I was sufficient to drive neuronal engulfment by microglia and restrict the accumulation of damaged neurons. IFN-I deficient mice had excess excitatory neurons in the developing somatosensory cortex as well as tactile hypersensitivity to whisker stimulation. These data define a molecular mechanism through which microglia engulf neurons during a critical window of brain development. More broadly, they reveal key homeostatic roles of a canonical antiviral signaling pathway in brain development.
Collapse
Affiliation(s)
- Caroline C. Escoubas
- Department of Psychiatry and Behavioral Sciences/ Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA
| | - Leah C. Dorman
- Department of Psychiatry and Behavioral Sciences/ Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA
- Department of Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA
| | - Phi T. Nguyen
- Department of Psychiatry and Behavioral Sciences/ Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA
- Department of Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA
| | - Christian Lagares-Linares
- Department of Psychiatry and Behavioral Sciences/ Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA
| | - Haruna Nakajo
- Department of Psychiatry and Behavioral Sciences/ Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA
| | - Sarah R. Anderson
- Department of Psychiatry and Behavioral Sciences/ Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA
| | - Beatriz Cuevas
- Department of Psychiatry and Behavioral Sciences/ Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA
- Department of Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA
| | - Ilia D. Vainchtein
- Department of Psychiatry and Behavioral Sciences/ Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA
| | - Nicholas J. Silva
- Department of Psychiatry and Behavioral Sciences/ Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA
| | - Yinghong Xiao
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA
| | - Peter V. Lidsky
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA
| | - Ellen Y. Wang
- Department of Psychiatry and Behavioral Sciences/ Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA
- UCSF SRTP program, University of California, San Francisco, San Francisco, CA
| | - Sunrae E. Taloma
- Department of Psychiatry and Behavioral Sciences/ Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA
- Department of Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA
| | - Hiromi Nakao-Inoue
- Department of Psychiatry and Behavioral Sciences/ Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA
| | - Bjoern Schwer
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA
- Department of Neurosurgery, University of California, San Francisco, San Francisco, CA
| | - Raul Andino
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA
| | - Tomasz J. Nowakowski
- Department of Psychiatry and Behavioral Sciences/ Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA
- Department of Anatomy, University of California, San Francisco, San Francisco, CA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA
- Chan-Zuckerberg Biohub, San Francisco, CA
| | - Anna V. Molofsky
- Department of Psychiatry and Behavioral Sciences/ Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA
- Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
4
|
Mellone M, Piotrowska K, Venturi G, James L, Bzura A, Lopez MA, James S, Wang C, Ellis MJ, Hanley CJ, Buckingham JF, Cox KL, Hughes G, Valge-Archer V, King EV, Beers SA, Jaquet V, Jones GD, Savelyeva N, Sayan E, Parsons JL, Durant S, Thomas GJ. ATM Regulates Differentiation of Myofibroblastic Cancer-Associated Fibroblasts and Can Be Targeted to Overcome Immunotherapy Resistance. Cancer Res 2022; 82:4571-4585. [PMID: 36353752 PMCID: PMC9755965 DOI: 10.1158/0008-5472.can-22-0435] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 07/28/2022] [Accepted: 09/21/2022] [Indexed: 11/11/2022]
Abstract
Myofibroblastic cancer-associated fibroblast (myoCAF)-rich tumors generally contain few T cells and respond poorly to immune-checkpoint blockade. Although myoCAFs are associated with poor outcome in most solid tumors, the molecular mechanisms regulating myoCAF accumulation remain unclear, limiting the potential for therapeutic intervention. Here, we identify ataxia-telangiectasia mutated (ATM) as a central regulator of the myoCAF phenotype. Differentiating myofibroblasts in vitro and myoCAFs cultured ex vivo display activated ATM signaling, and targeting ATM genetically or pharmacologically could suppress and reverse differentiation. ATM activation was regulated by the reactive oxygen species-producing enzyme NOX4, both through DNA damage and increased oxidative stress. Targeting fibroblast ATM in vivo suppressed myoCAF-rich tumor growth, promoted intratumoral CD8 T-cell infiltration, and potentiated the response to anti-PD-1 blockade and antitumor vaccination. This work identifies a novel pathway regulating myoCAF differentiation and provides a rationale for using ATM inhibitors to overcome CAF-mediated immunotherapy resistance. SIGNIFICANCE ATM signaling supports the differentiation of myoCAFs to suppress T-cell infiltration and antitumor immunity, supporting the potential clinical use of ATM inhibitors in combination with checkpoint inhibition in myoCAF-rich, immune-cold tumors.
Collapse
Affiliation(s)
- Massimiliano Mellone
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Klaudia Piotrowska
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Giulia Venturi
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Lija James
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Aleksandra Bzura
- Department of Genetics and Genome Biology, Cancer Research Centre, University of Leicester, Leicester, United Kingdom
| | - Maria A. Lopez
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Sonya James
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Chuan Wang
- Department of Molecular and Clinical Cancer Medicine, Cancer Research Centre, University of Liverpool, Liverpool, United Kingdom
| | - Matthew J. Ellis
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Christopher J. Hanley
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Josephine F. Buckingham
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Kerry L. Cox
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Gareth Hughes
- Bioscience, Oncology Innovative Medicines and Early Development (IMED) Biotech Unit, AstraZeneca, Cambridge, United Kingdom
| | - Viia Valge-Archer
- Bioscience, Oncology Innovative Medicines and Early Development (IMED) Biotech Unit, AstraZeneca, Cambridge, United Kingdom
| | - Emma V. King
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Stephen A. Beers
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Vincent Jaquet
- Department of Pathology and Immunology, Centre Médical Universitaire, Genève, Switzerland
| | - George D.D. Jones
- Department of Genetics and Genome Biology, Cancer Research Centre, University of Leicester, Leicester, United Kingdom
| | - Natalia Savelyeva
- Department of Molecular and Clinical Cancer Medicine, Cancer Research Centre, University of Liverpool, Liverpool, United Kingdom
| | - Emre Sayan
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Jason L. Parsons
- Department of Molecular and Clinical Cancer Medicine, Cancer Research Centre, University of Liverpool, Liverpool, United Kingdom
| | - Stephen Durant
- Bioscience, Oncology Innovative Medicines and Early Development (IMED) Biotech Unit, AstraZeneca, Cambridge, United Kingdom
| | - Gareth J. Thomas
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
5
|
Wang H, Wang B, Wei J, Zheng Z, Su J, Bian C, Xin Y, Jiang X. Sulforaphane regulates Nrf2-mediated antioxidant activity and downregulates TGF-β1/Smad pathways to prevent radiation-induced muscle fibrosis. Life Sci 2022; 311:121197. [PMID: 36400201 DOI: 10.1016/j.lfs.2022.121197] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/01/2022] [Accepted: 11/11/2022] [Indexed: 11/17/2022]
Abstract
AIMS This study aimed to examine the efficacy of sulforaphane (SFN) in preventing radiation-induced muscle fibrosis (RIMF) and the potential role in nuclear factor erythroid 2-related factor 2 (Nrf2)-mediated antioxidant stress. MAIN METHODS The RIMF model was established by a single irradiation of the left thigh of C57BL/6 J mice, and the mice were then randomly divided into control, SFN, irradiation (IR), and IR + SFN (IR/SFN) groups. The serum and skeletal muscle were collected eight weeks after irradiation, and changes in oxidative stress and muscle fibrosis were detected. KEY FINDINGS The IR group showed a more obvious skeletal muscle fiber atrophy, significantly higher number of collagen fibers, and higher inflammatory cell infiltration compared to control group. Compared to the IR group, the IR/SFN group had orderly arranged muscle fibers, decreased collagen fibers, and infiltration of inflammatory cells. In addition, compared with the control group, the expression of oxidative stress-related indexes was significantly increased, accompanied by activation of the transforming growth factor (TGF-β)/Smad pathway and its downstream fibrogenic molecules in the skeletal muscle of the IR group. After SFN intervention, the above indices were significantly restored. Furthermore, SFN induced the upregulation of Nrf2, activation of AKT, and inhibition of GSK-3β and Fyn accumulation. SIGNIFICANCE These results revealed that Nrf2 plays a central role in protecting against RIMF. Furthermore, SFN prevents RIMF by activating Nrf2 via the AKT/GSK-3β/Fyn pathway.
Collapse
Affiliation(s)
- Huanhuan Wang
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China; Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China; NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China.
| | - Bin Wang
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China; Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China; NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China.
| | - Jinlong Wei
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China; Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China; NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China.
| | - Zhuangzhuang Zheng
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China; Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China; NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China.
| | - Jing Su
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China; Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China; NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China.
| | - Chenbin Bian
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China; Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China; NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China.
| | - Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China.
| | - Xin Jiang
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China; Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China; NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China.
| |
Collapse
|
6
|
Craig DJ, Ambrose S, Stanbery L, Walter A, Nemunaitis J. Systemic benefit of radiation therapy via abscopal effect. Front Oncol 2022; 12:987142. [PMID: 36387120 PMCID: PMC9641206 DOI: 10.3389/fonc.2022.987142] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 10/05/2022] [Indexed: 08/30/2023] Open
Abstract
Evidence of a systemic response related to localized radiation therapy (RT) in cancer management is rare. However, enhancing the immune response via immunotherapy followed by localized RT has shown evidence of tumor shrinkage to non-irradiated metastatic disease thereby inducing an "abscopal effect." Combined induction of the cGAS-STING pathway and activation of IFN-gamma signaling cascade related to RT within an activated immune environment promotes neoantigen presentation and expansion of cytotoxic effector cells enabling enhancement of systemic immune response. A proposed mechanism, case examples, and clinical trial evidence of "abscopal effect" benefit are reviewed. Results support strategic therapeutic testing to enhance "abscopal effect."
Collapse
Affiliation(s)
- Daniel J. Craig
- University of Toledo, Department of Internal Medicine, Toledo, OH, United States
| | | | - Laura Stanbery
- Medical Affairs, Gradalis, Inc., Carrollton, TX, United States
| | - Adam Walter
- Medical Affairs, Gradalis, Inc., Carrollton, TX, United States
- Gynecologic Oncology, Promedica, Toledo, OH, United States
| | - John Nemunaitis
- Medical Affairs, Gradalis, Inc., Carrollton, TX, United States
| |
Collapse
|
7
|
Non-Targeted Effects of Synchrotron Radiation: Lessons from Experiments at the Australian and European Synchrotrons. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12042079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
Studies have been conducted at synchrotron facilities in Europe and Australia to explore a variety of applications of synchrotron X-rays in medicine and biology. We discuss the major technical aspects of the synchrotron irradiation setups, paying specific attention to the Australian Synchrotron (AS) and the European Synchrotron Radiation Facility (ESRF) as those best configured for a wide range of biomedical research involving animals and future cancer patients. Due to ultra-high dose rates, treatment doses can be delivered within milliseconds, abiding by FLASH radiotherapy principles. In addition, a homogeneous radiation field can be spatially fractionated into a geometric pattern called microbeam radiotherapy (MRT); a coplanar array of thin beams of microscopic dimensions. Both are clinically promising radiotherapy modalities because they trigger a cascade of biological effects that improve tumor control, while increasing normal tissue tolerance compared to conventional radiation. Synchrotrons can deliver high doses to a very small volume with low beam divergence, thus facilitating the study of non-targeted effects of these novel radiation modalities in both in-vitro and in-vivo models. Non-targeted radiation effects studied at the AS and ESRF include monitoring cell–cell communication after partial irradiation of a cell population (radiation-induced bystander effect, RIBE), the response of tissues outside the irradiated field (radiation-induced abscopal effect, RIAE), and the influence of irradiated animals on non-irradiated ones in close proximity (inter-animal RIBE). Here we provide a summary of these experiments and perspectives on their implications for non-targeted effects in biomedical fields.
Collapse
|
8
|
Mukherjee S, Dutta A, Chakraborty A. The cross-talk between Bax, Bcl2, caspases, and DNA damage in bystander HepG2 cells is regulated by γ-radiation dose and time of conditioned media transfer. Apoptosis 2022; 27:184-205. [PMID: 35076828 DOI: 10.1007/s10495-022-01713-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2022] [Indexed: 01/25/2023]
Abstract
Although radiation-induced bystander effects have been broadly explored in various biological systems, the molecular mechanisms and the consequences of different regulatory factors (dose, time, cell type) on bystander responses are not clearly understood. This study investigates the effects of irradiated cell-conditioned media (ICCM) collected at different times post-irradiation on bystander cancer cells regarding DNA damage and apoptosis induction. Human hepatocellular carcinoma HepG2 cells were exposed to γ-ray doses of 2 Gy, 5 Gy, and 8 Gy. In the early and late stages (1 h, 2 h, and 24 h) after irradiation, the ICCM was collected and transferred to unirradiated cells. Compared to control, bystander cells showed an increased level of H2AX phosphorylation, mitochondrial membrane depolarization, and elevation of intrinsic apoptotic pathway mediators such as p53, Bax, cas9, cas-3, and PARP cleavage. These results were confirmed by phosphatidylserine (PS) externalization and scanning electron microscopic observations, suggesting a rise in bystander HepG2 cell apoptosis. Anti-apoptotic Bcl2-level and viability were lower in bystander cells compared to control. The highest effects were observed in 8 Gy γ radiation-induced bystander cells. Even though the bystander effect was persistent at all time points of the study, ICCM at the early time points (1 or 2 h) had the most significant impact on the apoptosis markers in bystander cells. Nevertheless, 24 h ICCM induced the highest increase in H2AX and p53 phosphorylation and Bax levels. The effects of ICCM of irradiated HepG2 cells were additionally studied in normal liver cells BRL-3A to simulate actual radiotherapy conditions. The outcomes suggest that the expression of the signaling mediators in bystander cells is highly dynamic. A cross-talk between those signaling mediators regulates bystander responses depending on the radiation dose and time of incubation post-irradiation.
Collapse
Affiliation(s)
- Sharmi Mukherjee
- UGC-DAE Consortium for Scientific Research, Kolkata Centre, Block-LB, Plot-8, Sector-III, Salt Lake, Kolkata, West Bengal, 700 106, India.
| | - Anindita Dutta
- UGC-DAE Consortium for Scientific Research, Kolkata Centre, Block-LB, Plot-8, Sector-III, Salt Lake, Kolkata, West Bengal, 700 106, India
| | - Anindita Chakraborty
- UGC-DAE Consortium for Scientific Research, Kolkata Centre, Block-LB, Plot-8, Sector-III, Salt Lake, Kolkata, West Bengal, 700 106, India
| |
Collapse
|
9
|
Hansda S, Ghosh R. Bystander effect of ultraviolet A radiation protects A375 melanoma cells by induction of antioxidant defense. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, TOXICOLOGY AND CARCINOGENESIS 2021; 40:46-67. [PMID: 35895930 DOI: 10.1080/26896583.2021.1994820] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Ultraviolet (UV) irradiated cells release factors that result in varied responses by non-irradiated cells via bystander effects (BE). The UV-BE is dependent on the cell types involved and on the wavelength of the radiation. Using conditioned medium from UVA-irradiated A375 human melanoma cells (UVA-CM), UVA-bystander response was evaluated on the viability of naïve A375 cells. UVA-CM treatment itself did not alter cell viability; however, UVA-CM treated bystander cells were more resistant to the lethal action of UVA, UVB, UVC or H2O2. Effects of UVA-CM on cell proliferation, mechanism of cell death, DNA damage, malondialdehyde formation, generation of reactive oxygen species (ROS) and antioxidant status were studied in A375 cells. We observed that UVA-CM triggered antioxidant defenses to elicit protective responses through elevation of antioxidant enzyme activities in cells, which persisted until 5 h after exposure to UVA-CM. This was possibly responsible for decreased generation of ROS and diminished DNA and membrane damage in cells. These bystander cells were resistant to killing when exposed to different genotoxic agents. Damaged nuclei, induction of apoptosis and autophagic death were also lowered in these cells. The influence of UVA-CM on cancer stem cells side population was assessed.Highlights:UVA radiation induced bystander effects in A375 cellsDamage by genotoxicants is suppressed due to lower ROS generation on UVA-CM treatmentUVA-CM exposure enhanced higher activities of CAT and GPxResistance to genotoxic agents in such cells was due to elevated antioxidant defenceUVA-bystander phenomenon was a protective response.
Collapse
Affiliation(s)
- Surajit Hansda
- Department of Biochemistry & Biophysics, University of Kalyani, Kalyani, India
| | - Rita Ghosh
- Department of Biochemistry & Biophysics, University of Kalyani, Kalyani, India
| |
Collapse
|
10
|
ATR-FTIR spectroscopy probing of structural alterations in the cellular membrane of abscopal liver cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2021; 1863:183726. [PMID: 34375629 DOI: 10.1016/j.bbamem.2021.183726] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 12/12/2022]
Abstract
In this study, we utilize ATR-FTIR spectroscopy to investigate the structural damages in the cell membrane lipids and proteins as a result of the oxidative stress in abscopal liver tissue of rats either whole-body, cranially or lower limb irradiated as compared with sham-irradiated group. We also question whether the original irradiation region would influence the induction of the abscopal effect. The data present compelling evidence that an abscopal effect was induced in the liver tissue following both cranial and lower limb irradiations, marked by damage in the membrane-associated lipids and proteins. Lipid damage manifestation is evident by; 1) decrease in the lipid/protein ratio. 2) Degradation of lipid as marked by the decrease in the area ratio CH 2 asymmetric/CH 3 asymmetric stretching bands. 3) Increase in the carbonyl content evident by the increase in the band area ratio of carbonyl ester/lipid. 4) Increase in the degree of methylation as indicated by the increase in the band area ratio of CH3/lipid. 5) Disorder in the phospholipid acyl chains marked by the shift in the CH2 asymmetric stretching and olefinic HCCH absorption bands. Protein damage was indicated by 1) Shifts in the position of amide I and amide II bands. 2) Decrease in the area ratio amide I/amide II. 3) Broadening in amide II band. Our data strongly suggest similar induction of the abscopal effect as a result of either cranial or lower limb irradiation, which means that the original irradiation region did not influence the induced abscopal effect in the examined system.
Collapse
|
11
|
Tremi I, Nowsheen S, Aziz K, Siva S, Ventura J, Hatzi VI, Martin OA, Georgakilas AG. Inflammation and oxidatively induced DNA damage: A synergy leading to cancer development. Cancer 2021. [DOI: 10.1016/b978-0-12-819547-5.00013-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
12
|
Potential application of γ-H2AX as a biodosimetry tool for radiation triage. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2020; 787:108350. [PMID: 34083048 DOI: 10.1016/j.mrrev.2020.108350] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 11/11/2020] [Accepted: 11/13/2020] [Indexed: 01/01/2023]
Abstract
Radiation triage and biological dosimetry are two initial steps in the medical management of exposed individuals following radiological accidents. Well established biodosimetry methods such as the dicentric (DC) assay, micronucleus (MN) assay, and fluorescence in-situ hybridization (FISH) translocation assay (for residual damage) have been used for this purpose for several decades. Recent advances in scoring methodology and networking among established laboratories have increased triage capacity; however, these methods still have limitations in analysing large sample numbers, particularly because of the ∼ 48 h minimum culture time required prior to analysis. Hence, there is a need for simple, and high throughput markers to identify exposed individuals in case of radiological/nuclear emergencies. In recent years, a few markers were identified, one being phosphorylated histone 2AX (γ-H2AX), which measured a nuclear foci or nuclear staining intensity that was found to be suitable for triage. Measurement of γ-H2AX foci formed at and around the sites of DNA double-strand breaks is a rapid and sensitive biodosimetry method which does not require culturing and is thus promising for the analysis of a large number of samples. In this review, we have summarized the recent developments of γ-H2AX assay in radiation triage and biodosimetry, focusing chiefly on: i) the importance of baseline frequency and reported values among different laboratories, ii) the influence of known and unknown variables on dose estimation, iii) quality assurance such as inter-laboratory comparison between scorers and scoring methods, and iv) current limitations and potential for future development.
Collapse
|
13
|
Helm JS, Rudel RA. Adverse outcome pathways for ionizing radiation and breast cancer involve direct and indirect DNA damage, oxidative stress, inflammation, genomic instability, and interaction with hormonal regulation of the breast. Arch Toxicol 2020. [PMID: 32399610 DOI: 10.1007/s00204-020-02752-z)] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Abstract
Knowledge about established breast carcinogens can support improved and modernized toxicological testing methods by identifying key mechanistic events. Ionizing radiation (IR) increases the risk of breast cancer, especially for women and for exposure at younger ages, and evidence overall supports a linear dose-response relationship. We used the Adverse Outcome Pathway (AOP) framework to outline and evaluate the evidence linking ionizing radiation with breast cancer from molecular initiating events to the adverse outcome through intermediate key events, creating a qualitative AOP. We identified key events based on review articles, searched PubMed for recent literature on key events and IR, and identified additional papers using references. We manually curated publications and evaluated data quality. Ionizing radiation directly and indirectly causes DNA damage and increases production of reactive oxygen and nitrogen species (RONS). RONS lead to DNA damage and epigenetic changes leading to mutations and genomic instability (GI). Proliferation amplifies the effects of DNA damage and mutations leading to the AO of breast cancer. Separately, RONS and DNA damage also increase inflammation. Inflammation contributes to direct and indirect effects (effects in cells not directly reached by IR) via positive feedback to RONS and DNA damage, and separately increases proliferation and breast cancer through pro-carcinogenic effects on cells and tissue. For example, gene expression changes alter inflammatory mediators, resulting in improved survival and growth of cancer cells and a more hospitable tissue environment. All of these events overlap at multiple points with events characteristic of "background" induction of breast carcinogenesis, including hormone-responsive proliferation, oxidative activity, and DNA damage. These overlaps make the breast particularly susceptible to ionizing radiation and reinforce that these biological activities are important characteristics of carcinogens. Agents that increase these biological processes should be considered potential breast carcinogens, and predictive methods are needed to identify chemicals that increase these processes. Techniques are available to measure RONS, DNA damage and mutation, cell proliferation, and some inflammatory proteins or processes. Improved assays are needed to measure GI and chronic inflammation, as well as the interaction with hormonally driven development and proliferation. Several methods measure diverse epigenetic changes, but it is not clear which changes are relevant to breast cancer. In addition, most toxicological assays are not conducted in mammary tissue, and so it is a priority to evaluate if results from other tissues are generalizable to breast, or to conduct assays in breast tissue. Developing and applying these assays to identify exposures of concern will facilitate efforts to reduce subsequent breast cancer risk.
Collapse
Affiliation(s)
- Jessica S Helm
- Silent Spring Institute, 320 Nevada Street, Suite 302, Newton, MA, 02460, USA
| | - Ruthann A Rudel
- Silent Spring Institute, 320 Nevada Street, Suite 302, Newton, MA, 02460, USA.
| |
Collapse
|
14
|
Helm JS, Rudel RA. Adverse outcome pathways for ionizing radiation and breast cancer involve direct and indirect DNA damage, oxidative stress, inflammation, genomic instability, and interaction with hormonal regulation of the breast. Arch Toxicol 2020; 94:1511-1549. [PMID: 32399610 PMCID: PMC7261741 DOI: 10.1007/s00204-020-02752-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 04/16/2020] [Indexed: 12/15/2022]
Abstract
Knowledge about established breast carcinogens can support improved and modernized toxicological testing methods by identifying key mechanistic events. Ionizing radiation (IR) increases the risk of breast cancer, especially for women and for exposure at younger ages, and evidence overall supports a linear dose-response relationship. We used the Adverse Outcome Pathway (AOP) framework to outline and evaluate the evidence linking ionizing radiation with breast cancer from molecular initiating events to the adverse outcome through intermediate key events, creating a qualitative AOP. We identified key events based on review articles, searched PubMed for recent literature on key events and IR, and identified additional papers using references. We manually curated publications and evaluated data quality. Ionizing radiation directly and indirectly causes DNA damage and increases production of reactive oxygen and nitrogen species (RONS). RONS lead to DNA damage and epigenetic changes leading to mutations and genomic instability (GI). Proliferation amplifies the effects of DNA damage and mutations leading to the AO of breast cancer. Separately, RONS and DNA damage also increase inflammation. Inflammation contributes to direct and indirect effects (effects in cells not directly reached by IR) via positive feedback to RONS and DNA damage, and separately increases proliferation and breast cancer through pro-carcinogenic effects on cells and tissue. For example, gene expression changes alter inflammatory mediators, resulting in improved survival and growth of cancer cells and a more hospitable tissue environment. All of these events overlap at multiple points with events characteristic of "background" induction of breast carcinogenesis, including hormone-responsive proliferation, oxidative activity, and DNA damage. These overlaps make the breast particularly susceptible to ionizing radiation and reinforce that these biological activities are important characteristics of carcinogens. Agents that increase these biological processes should be considered potential breast carcinogens, and predictive methods are needed to identify chemicals that increase these processes. Techniques are available to measure RONS, DNA damage and mutation, cell proliferation, and some inflammatory proteins or processes. Improved assays are needed to measure GI and chronic inflammation, as well as the interaction with hormonally driven development and proliferation. Several methods measure diverse epigenetic changes, but it is not clear which changes are relevant to breast cancer. In addition, most toxicological assays are not conducted in mammary tissue, and so it is a priority to evaluate if results from other tissues are generalizable to breast, or to conduct assays in breast tissue. Developing and applying these assays to identify exposures of concern will facilitate efforts to reduce subsequent breast cancer risk.
Collapse
Affiliation(s)
- Jessica S Helm
- Silent Spring Institute, 320 Nevada Street, Suite 302, Newton, MA, 02460, USA
| | - Ruthann A Rudel
- Silent Spring Institute, 320 Nevada Street, Suite 302, Newton, MA, 02460, USA.
| |
Collapse
|
15
|
Z E, R F. The Bystander Effect of Ultraviolet Radiation and Mediators. J Biomed Phys Eng 2020; 10:111-118. [PMID: 32158718 PMCID: PMC7036410 DOI: 10.31661/jbpe.v0i0.956] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Accepted: 06/19/2018] [Indexed: 01/01/2023]
Abstract
A bystander effect is biological changes in non-irradiated cells by transmitted signals from irradiated bystander cells, which causes the radiation toxic effects on the adjacent non-irradiated tissues. This phenomenon occurs by agents such as ionizing radiation, ultraviolet radiation (UVR) and chemotherapy. The bystander effect includes biological processes such as damage to DNA, cell death, chromosomal abnormalities, delay and premature mutations and micronuclei production. The most involved genes in creating this phenomenon are cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), the nuclear factor of kappa B (NFkB) and Mitogen-Activated Protein Kinases (MAPKs). Radiation generated reactive oxygen species (ROS) can damage DNA, membranes and protein buildings. Studies have shown that Vitamin C, Hesperidin, and melatonin can reduce the number of ROS and have a protective role. Silver nanoparticles (Ag NPs) are the most abundant nanoparticles produced and when they enter cells, they can create DNA damage. Studies have shown that combined treatment with UVR and silver nanoparticles could form γ-H2AX and 8-hydroxy-2'-deoxyguanosine (8-OHdG) synergistically. This article reviews the direct and the bystander effects of UVR on the nuclear DNA, the effect of radioprotectors and Ag NPs on these effects.
Collapse
Affiliation(s)
- Eftekhari Z
- MSc, Department of Radiology, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
- MSc, Student research committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fardid R
- PhD, Department of Radiology, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
- PhD, Ionizing and Non-Ionizing Radiation Protection Research Center (INIRPRC), School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
16
|
Wound fluids collected postoperatively from patients with breast cancer induce epithelial to mesenchymal transition but intraoperative radiotherapy impairs this effect by activating the radiation-induced bystander effect. Sci Rep 2019; 9:7891. [PMID: 31133667 PMCID: PMC6536501 DOI: 10.1038/s41598-019-44412-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 05/16/2019] [Indexed: 02/07/2023] Open
Abstract
Wound fluids (WF) are believed to play a role in the local recurrences by inducing an inflammatory process in scar tissue area. Given that most local relapse in primary breast cancer patients occur within the scar tissue area, researchers have investigated whether localized radiotherapy, such as intraoperative radiotherapy (IORT), could be more effective than postoperative RT in inhibiting local tumor recurrence. The epithelial-mesenchymal transition (EMT) program plays a critical role in promoting metastasis in epithelium-derived carcinoma. Given this background the main aim of the present study was to determine the mechanisms by which IORT decreases the tumorigenic potential of WF. We assumed that postoperative fluids from patients would activate the radiation-induced bystander effect (RIBE) in treated cells, thus altering the tumor microenvironment. To confirm this hypothesis, WF collected from patients after breast conserving surgery (BCS) alone, after BCS followed by IORT treatment or WF from BCS patients together with RIBE medium were incubated with MCF7 and MDA-MB-468 cells. Changes in the CSC phenotype, in EMT program and potential to migrate were performed to determine the possible role of WF on the migration of breast cancer cells. Our findings show that wound fluids stimulate the CSC phenotype and EMT program in breast cancer cell lines. This effect was partially abrogated when the cells were incubated in wound fluids collected from patients after breast-conserving surgery followed by IORT. Additionally, we confirmed the role of radiation-induced bystander effect in altering the properties of the WF to induce the CSC phenotype and EMT program.
Collapse
|
17
|
Zhang LY, Yong WX, Wang L, Zhang LX, Zhang YM, Gong HX, He JP, Liu YQ. Astragalus Polysaccharide Eases G1 Phase-Correlative Bystander Effects through Mediation of TGF-βR/MAPK/ROS Signal Pathway After Carbon Ion Irradiation in BMSCs. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2019; 47:595-612. [DOI: 10.1142/s0192415x19500319] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Although Astragalus polysaccharide (APS) has been shown to have various pharmacological effects, there have been no studies concerning the inhibitory effects of APS on the radiation-induced bystander effects (RIBE). The aim of this study was to investigate whether APS could suppress RIBE damage by inhibiting cell growth, micronucleus (MN) formation and 53BP1 foci number increased in bone marrow mesenchymal stem cells (BMSCs), named bystander cells, as well as to explore its mechanism. In this study, APS decreased proliferation and colony rate of bystander cells by inducing cell cycle arrest at G1 phase via extrinsic and intrinsic DNA damage. Regarding mechanism, APS inhibited mitogen-activated protein kinase (MAPK) signal pathway by down-regulating the expression of the key proteins, phosphorylated JNK (p-JNK), phosphorylated ERK (p-ERK) but not phosphorylated P38 (p-P38), and down-regulating their downstream function protein and molecule, cyclooxygenase-2 (COX-2) and reactive oxygen species (ROS). Moreover, in bystander cells, APS inhibits expression of transforming growth factor [Formula: see text] receptor II (TGF-[Formula: see text]R II), a cell membrane receptor, resulting in lower ROS production and secretion via TGF-[Formula: see text]R-JNK/ERK-COX-2/ROS not P38 signaling. They gave a hint that the decreased RIBE damage induced by APS treatment involved TGF-[Formula: see text]R-JNK/ERK-COX-2/ROS down-regulation.
Collapse
Affiliation(s)
- Li-Ying Zhang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine, Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou 730000, P. R. China
| | - Wen-Xing Yong
- Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou 730000, P. R. China
| | - Lei Wang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine, Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou 730000, P. R. China
| | - Li-Xin Zhang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine, Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou 730000, P. R. China
| | - Yi-Ming Zhang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine, Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou 730000, P. R. China
| | - Hong-Xia Gong
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine, Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou 730000, P. R. China
| | - Jin-Peng He
- Key Laboratory of Space Radiobiology of Gansu Province & Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, P. R. China
| | - Yong-Qi Liu
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine, Research in Gansu Colleges and Universities, Gansu University of Chinese Medicine, Lanzhou 730000, P. R. China
| |
Collapse
|
18
|
Krzywon A, Widel M. Bystander Me45 Melanoma Cells Increase Damaging Effect in UVC-irradiated Cells. Photochem Photobiol 2019; 95:1019-1028. [PMID: 30613987 DOI: 10.1111/php.13080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 12/26/2018] [Indexed: 11/30/2022]
Abstract
The aim of our study was to investigate the possible mechanism(s) of the bystander effect induced by UVC light in malignant melanoma Me45 cells that were co-incubated with irradiated cells of the same line. We have found that the UVC band effectively generated apoptosis, premature senescence, single and double DNA strand breaks and reduced clonogenic survival of bystander cells. However, in the feedback response, the bystander cells intensified damage in directly irradiated cells, especially seen at the level of apoptosis and survival of clonogenic cells. Pretreatment of bystander cells with inhibitor of inducible nitric oxide synthase blocks this signaling. It seems that the mediators of this phenomenon produced and secreted by neighboring cells are superoxide, nitric oxide and TGF-β. The reverse deleterious effect caused by cells not exposed to UVC in directly exposed cells is opposed to the protective/rescue effect exerted by the bystander cells in the case of ionizing radiation known in the literature. Whether this opposite adverse effect is a feature of only Me45 melanoma cells or whether it is a general phenomenon occurring between cells of other types exposed to ultraviolet radiation requires further research.
Collapse
Affiliation(s)
- Aleksandra Krzywon
- Biosystems Group, Faculty of Automatics, Electronics and Informatics, Institute of Automatic Control, Silesian University of Technology, Gliwice, Poland
| | - Maria Widel
- Biosystems Group, Faculty of Automatics, Electronics and Informatics, Institute of Automatic Control, Silesian University of Technology, Gliwice, Poland
| |
Collapse
|
19
|
Razdan N, Vasilopoulos T, Herbig U. Telomere dysfunction promotes transdifferentiation of human fibroblasts into myofibroblasts. Aging Cell 2018; 17:e12838. [PMID: 30244523 PMCID: PMC6260909 DOI: 10.1111/acel.12838] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 06/25/2018] [Accepted: 08/05/2018] [Indexed: 12/13/2022] Open
Abstract
Cells that had undergone telomere dysfunction-induced senescence secrete numerous cytokines and other molecules, collectively called the senescence-associated secretory phenotype (SASP). Although certain SASP factors have been demonstrated to promote cellular senescence in neighboring cells in a paracrine manner, the mechanisms leading to bystander senescence and the functional significance of these effects are currently unclear. Here, we demonstrate that TGF-β1, a component of the SASP, causes telomere dysfunction in normal somatic human fibroblasts in a Smad3/NOX4/ROS-dependent manner. Surprisingly, instead of activating cellular senescence, TGF-β1-induced telomere dysfunction caused fibroblasts to transdifferentiate into α-SMA-expressing myofibroblasts, a mesenchymal and contractile cell type that is critical for wound healing and tissue repair. Despite the presence of dysfunctional telomeres, transdifferentiated cells acquired the ability to contract collagen lattices and displayed a gene expression signature characteristic of functional myofibroblasts. Significantly, the formation of dysfunctional telomeres and downstream p53 signaling was necessary for myofibroblast transdifferentiation, as suppressing telomere dysfunction by expression of hTERT, inhibiting the signaling pathways that lead to stochastic telomere dysfunction, and suppressing p53 function prevented the generation of myofibroblasts in response to TGF-β1 signaling. Furthermore, inducing telomere dysfunction using shRNA against TRF2 also caused cells to develop features that are characteristic of myofibroblasts, even in the absence of exogenous TGF-β1. Overall, our data demonstrate that telomere dysfunction is not only compatible with cell functionality, but they also demonstrate that the generation of dysfunctional telomeres is an essential step for transdifferentiation of human fibroblasts into myofibroblasts.
Collapse
Affiliation(s)
- Neetu Razdan
- New Jersey Medical School, Cancer Institute of New Jersey-Newark; Rutgers Biomedical and Health Sciences; Newark New Jersey
- Department of Microbiology, Biochemistry and Molecular Genetics; Rutgers Biomedical and Health Sciences; Newark New Jersey
| | - Themistoklis Vasilopoulos
- New Jersey Medical School, Cancer Institute of New Jersey-Newark; Rutgers Biomedical and Health Sciences; Newark New Jersey
| | - Utz Herbig
- New Jersey Medical School, Cancer Institute of New Jersey-Newark; Rutgers Biomedical and Health Sciences; Newark New Jersey
- Department of Microbiology, Biochemistry and Molecular Genetics; Rutgers Biomedical and Health Sciences; Newark New Jersey
| |
Collapse
|
20
|
Radiation-induced muscle fibrosis rat model: establishment and valuation. Radiat Oncol 2018; 13:160. [PMID: 30157899 PMCID: PMC6114061 DOI: 10.1186/s13014-018-1104-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 08/17/2018] [Indexed: 02/06/2023] Open
Abstract
Background Lack of animal model of radiation induced muscle fibrosis, this study aimed to establish such a model by using 90 Gy single dose irradiation to mimic clinical relevance and also to explore the potential post-irradiation regenerative mechanism. Methods SD rats were randomly divided into dose investigation groups and time gradient groups. Group1–6 were irradiated with a single dose of 65Gy, 70Gy, 75Gy, 80Gy, 85Gy and 90Gy respectively, and the degree of rectus femoris fibrosis in the irradiated area was detected at 4 weeks after irradiation. Group 7–9 were irradiated with a single dose of 90Gy, and the results were detected 1, 2, 4, and 8 weeks after irradiation. Then the general condition of rats was recorded. Masson staining was used to detect muscle fibrosis. The ultrastructure of muscles was observed by electron microscope, and the expression changes of satellite cell proliferation and differentiation related genes were detected by quantitative real-time-PCR. Results A single dose of 90Gy irradiation could cause muscle fibrosis in rats. As time goes on, the severity of muscle fibrosis and the expression of TGF- β1 increased. Significant swelling of mitochondria, myofilament disarrangement and dissolution, obvious endothelial cell swelling, increased vascular permeability, decrease of blood cell, deposition of fibrosis tissue around the vessel could be found compared with the control group. At around the 4th week, the expressions of Pax7, Myf5, MyoD, MyoG, Mrf4 increased. Conclusion Irradiation of 90Gy can successfully establish the rat model of radiation-induced muscle fibrosis. This model demonstrated that regenerative process was initiated by the irradiation only at an early stage, which can serve a suitable model for investigating regenerative therapy for post-radiation muscle fibrosis.
Collapse
|
21
|
Sokolov M, Neumann R. Changes in gene expression as one of the key mechanisms involved in radiation-induced bystander effect. Biomed Rep 2018; 9:99-111. [PMID: 30013775 PMCID: PMC6036822 DOI: 10.3892/br.2018.1110] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 05/21/2018] [Indexed: 12/22/2022] Open
Abstract
The radiation-induced bystander effect (RIBE) refers to the manifestation of responses by non-targeted/non-hit cells or tissues situated in proximity to cells and tissues directly exposed to ionizing radiation (IR). The RIBE is elicited by agents and factors released by IR-hit cells. The growing body of data suggests that the underlying mechanisms of the RIBE are multifaceted depending both on the biological (characteristics of directly IR-exposed cells, bystander cells, intercellular milieu) and the physical (dose, rate and type of IR, time after exposure) factors/parameters. Although the exact identity of bystander signal(s) is yet to be identified, the published data indicate changes in gene expression for multiple types of RNA (mRNA, microRNA, mitochondrial RNA, long non-coding RNA, small nucleolar RNA) as being one of the major responses of cells and tissues in the context of the RIBE. Gene expression profiles demonstrate a high degree of variability between distinct bystander cell and tissue types. These alterations could independently, or in a signaling cascade, result in the manifestation of readily observable endpoints, including changes in viability and genomic instability. Here, the relevant publications on the gene candidates and signaling pathways involved in the RIBE are reviewed, and a framework for future studies, both in vitro and in vivo, on the genetic aspect of the RIBE is provided.
Collapse
Affiliation(s)
- Mykyta Sokolov
- Department of Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ronald Neumann
- Department of Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
22
|
Hawkins SJ, Crompton LA, Sood A, Saunders M, Boyle NT, Buckley A, Minogue AM, McComish SF, Jiménez-Moreno N, Cordero-Llana O, Stathakos P, Gilmore CE, Kelly S, Lane JD, Case CP, Caldwell MA. Nanoparticle-induced neuronal toxicity across placental barriers is mediated by autophagy and dependent on astrocytes. NATURE NANOTECHNOLOGY 2018; 13:427-433. [PMID: 29610530 DOI: 10.1038/s41565-018-0085-3] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 01/31/2018] [Indexed: 05/05/2023]
Abstract
The potential for maternal nanoparticle (NP) exposures to cause developmental toxicity in the fetus without the direct passage of NPs has previously been shown, but the mechanism remained elusive. We now demonstrate that exposure of cobalt and chromium NPs to BeWo cell barriers, an in vitro model of the human placenta, triggers impairment of the autophagic flux and release of interleukin-6. This contributes to the altered differentiation of human neural progenitor cells and DNA damage in the derived neurons and astrocytes. Crucially, neuronal DNA damage is mediated by astrocytes. Inhibiting the autophagic degradation in the BeWo barrier by overexpression of the dominant-negative human ATG4BC74A significantly reduces the levels of DNA damage in astrocytes. In vivo, indirect NP toxicity in mice results in neurodevelopmental abnormalities with reactive astrogliosis and increased DNA damage in the fetal hippocampus. Our results demonstrate the potential importance of autophagy to elicit NP toxicity and the risk of indirect developmental neurotoxicity after maternal NP exposure.
Collapse
Affiliation(s)
- Simon J Hawkins
- Regenerative Medicine Laboratory, School of Clinical Sciences, University of Bristol, Bristol, UK
- Musculoskeletal Research Unit, School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Lucy A Crompton
- Regenerative Medicine Laboratory, School of Clinical Sciences, University of Bristol, Bristol, UK
- Cell Biology Laboratories, School of Biochemistry, University of Bristol, Bristol, UK
| | - Aman Sood
- Musculoskeletal Research Unit, School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Margaret Saunders
- Cell Biology Laboratories, School of Biochemistry, University of Bristol, Bristol, UK
- Department of Medical Physics & Bioengineering, St Michael's Hospital, University Hospitals Bristol NHS Foundation Trust, Bristol, UK
| | - Noreen T Boyle
- Trinity College Institute of Neuroscience, Department of Physiology, Trinity College Dublin, Dublin, Ireland
| | - Amy Buckley
- Trinity College Institute of Neuroscience, Department of Physiology, Trinity College Dublin, Dublin, Ireland
| | - Aedín M Minogue
- Trinity College Institute of Neuroscience, Department of Physiology, Trinity College Dublin, Dublin, Ireland
| | - Sarah F McComish
- Trinity College Institute of Neuroscience, Department of Physiology, Trinity College Dublin, Dublin, Ireland
| | | | - Oscar Cordero-Llana
- Regenerative Medicine Laboratory, School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Petros Stathakos
- Regenerative Medicine Laboratory, School of Clinical Sciences, University of Bristol, Bristol, UK
- Cell Biology Laboratories, School of Biochemistry, University of Bristol, Bristol, UK
| | - Catherine E Gilmore
- Musculoskeletal Research Unit, School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Stephen Kelly
- Neuroscience Institute @JFK Medical Center, Edison, NJ, USA
| | - Jon D Lane
- Cell Biology Laboratories, School of Biochemistry, University of Bristol, Bristol, UK
| | - C Patrick Case
- Musculoskeletal Research Unit, School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Maeve A Caldwell
- Trinity College Institute of Neuroscience, Department of Physiology, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
23
|
Ventura J, Lobachevsky PN, Palazzolo JS, Forrester H, Haynes NM, Ivashkevich A, Stevenson AW, Hall CJ, Ntargaras A, Kotsaris V, Pollakis GC, Potsi G, Skordylis K, Terzoudi G, Pateras IS, Gorgoulis VG, Georgakilas AG, Sprung CN, Martin OA. Localized Synchrotron Irradiation of Mouse Skin Induces Persistent Systemic Genotoxic and Immune Responses. Cancer Res 2017; 77:6389-6399. [PMID: 29113972 DOI: 10.1158/0008-5472.can-17-1066] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 08/07/2017] [Accepted: 09/19/2017] [Indexed: 11/16/2022]
Abstract
The importance of nontargeted (systemic) effects of ionizing radiation is attracting increasing attention. Exploiting synchrotron radiation generated by the Imaging and Medical Beamline at the Australian Synchrotron, we studied radiation-induced nontargeted effects in C57BL/6 mice. Mice were locally irradiated with a synchrotron X-ray broad beam and a multiplanar microbeam radiotherapy beam. To assess the influence of the beam configurations and variations in peak dose and irradiated area in the response of normal tissues outside the irradiated field at 1 and 4 days after irradiation, we monitored oxidatively induced clustered DNA lesions (OCDL), DNA double-strand breaks (DSB), apoptosis, and the local and systemic immune responses. All radiation settings induced pronounced persistent systemic effects in mice, which resulted from even short exposures of a small irradiated area. OCDLs were elevated in a wide variety of unirradiated normal tissues. In out-of-field duodenum, there was a trend for elevated apoptotic cell death under most irradiation conditions; however, DSBs were elevated only after exposure to lower doses. These genotoxic events were accompanied by changes in plasma concentrations of macrophage-derived cytokine, eotaxin, IL10, TIMP1, VEGF, TGFβ1, and TGFβ2, along with changes in tissues in frequencies of macrophages, neutrophils, and T lymphocytes. Overall, our findings have implications for the planning of therapeutic and diagnostic radiation treatments to reduce the risk of radiation-related adverse systemic effects. Cancer Res; 77(22); 6389-99. ©2017 AACR.
Collapse
Affiliation(s)
- Jessica Ventura
- Molecular Radiation Biology Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Department of Obstetrics and Gynaecology, Royal Women's Hospital, University of Melbourne, Melbourne, Victoria, Australia
| | - Pavel N Lobachevsky
- Molecular Radiation Biology Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Jason S Palazzolo
- Molecular Radiation Biology Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Helen Forrester
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research and Monash University, Clayton, Victoria, Australia
| | - Nicole M Haynes
- Cancer Therapeutics Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Alesia Ivashkevich
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research and Monash University, Clayton, Victoria, Australia.,Radiation Oncology, Canberra Hospital, Garran, Australian Capital Territory, Australia
| | - Andrew W Stevenson
- CSIRO, Clayton, Victoria, Australia.,Australian Synchrotron, Clayton, Victoria, Australia
| | | | - Andreas Ntargaras
- Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Vasilis Kotsaris
- Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Gerasimos Ch Pollakis
- Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Gianna Potsi
- Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Konstantinos Skordylis
- Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Georgia Terzoudi
- Laboratory of Health Physics, Radiobiology and Cytogenetics, Institute of Nuclear and Radiological Sciences and Technology, Energy and Safety, National Center for Scientific Research 'Demokritos', Athens, Greece
| | - Ioannis S Pateras
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Vassilis G Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.,Biomedical Research Foundation of the Academy of Athens, Athens, Greece.,Faculty Institute for Cancer Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom
| | - Alexandros G Georgakilas
- Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Carl N Sprung
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research and Monash University, Clayton, Victoria, Australia
| | - Olga A Martin
- Molecular Radiation Biology Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia. .,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.,Division of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| |
Collapse
|
24
|
Chang WT, Liu W, Chiu YH, Chen BH, Chuang SC, Chen YC, Hsu YT, Lu MJ, Chiou SJ, Chou CK, Chiu CC. A 4-Phenoxyphenol Derivative Exerts Inhibitory Effects on Human Hepatocellular Carcinoma Cells through Regulating Autophagy and Apoptosis Accompanied by Downregulating α-Tubulin Expression. Molecules 2017; 22:molecules22050854. [PMID: 28531143 PMCID: PMC6154338 DOI: 10.3390/molecules22050854] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 05/16/2017] [Accepted: 05/16/2017] [Indexed: 12/30/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a leading cancer worldwide. Advanced HCCs are usually resistant to anticancer drugs, causing unsatisfactory chemotherapy outcomes. In this study, we showed that a 4-phenoxyphenol derivative, 4-[4-(4-hydroxyphenoxy)phenoxy]phenol (4-HPPP), exerts an inhibitory activity against two HCC cell lines, Huh7 and Ha22T. We further investigated the anti-HCC activities of 4-HPPP, including anti-proliferation and induction of apoptosis. Our results showed that higher dosage of 4-HPPP downregulates the expression of α-tubulin and causes nuclear enlargement in both the Huh-7 and Ha22T cell lines. Interestingly, the colony formation results showed a discrepancy in the inhibitory effect of 4-HPPP on HCC and rat liver epithelial Clone 9 cells, suggesting the selective cytotoxicity of 4-HPPP toward HCC cells. Furthermore, the cell proliferation and apoptosis assay results illustrated the differences between the two HCC cell lines. The results of cellular proliferation assays, including trypan blue exclusion and colony formation, revealed that 4-HPPP inhibits the growth of Huh7 cells, but exerts less cytotoxicity in Ha22T cells. Furthermore, the annexin V assay performed for detecting the apoptosis showed similar results. Western blotting results showed 4-HPPP caused the increase of pro-apoptotic factors including cleaved caspase-3, Bid and Bax in HCC cells, especially in Huh-7. Furthermore, an increase of autophagy-associated protein microtubule-associated protein-1 light chain-3B (LC3B)-II and the decrease of Beclin-1 and p62/SQSTM1 were observed following 4-HPPP treatment. Additionally, the level of γH2A histone family, member X (γH2AX), an endogenous DNA damage biomarker, was dramatically increased in Huh7 cells after 4-HPPP treatment, suggesting the involvement of DNA damage pathway in 4-HPPP-induced apoptosis. On the contrary, the western blotting results showed that treatment up-regulates pro-survival proteins, including the phosphorylation of protein kinase B (Akt) and the level of survivin on Ha22T cells, which may confer a resistance toward 4-HPPP. Notably, the blockade of extracellular signal-regulated kinases (ERK), but not Akt, enhanced the cytotoxicity of 4-HPPP against Ha22T cells, indicating the pro-survival role of ERK in 4-HPPP-induced anti-HCC effect. Our present work suggests that selective anti-HCC activity of 4-HPPP acts through induction of DNA damage. Accordingly, the combination of ERK inhibitor may significantly enhance the anti-cancer effect of 4-HPPP for those HCC cells which overexpress ERK in the future.
Collapse
Affiliation(s)
- Wen-Tsan Chang
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Wangta Liu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Yi-Han Chiu
- Department of Nursing, St. Mary's Junior College of Medicine, Nursing and Management, Yi-Lan 266, Taiwan.
| | - Bing-Hung Chen
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- The Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan.
| | - Shih-Chang Chuang
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Transplantation Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| | - Yen-Chun Chen
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Yun-Tzh Hsu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Mei-Jei Lu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Shean-Jaw Chiou
- Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Chon-Kit Chou
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Translational Research Center, Cancer Center, Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Research Center for Environment Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan.
| |
Collapse
|
25
|
Gomez-Casal R, Epperly MW, Wang H, Proia DA, Greenberger JS, Levina V. Radioresistant human lung adenocarcinoma cells that survived multiple fractions of ionizing radiation are sensitive to HSP90 inhibition. Oncotarget 2016; 6:44306-22. [PMID: 26517240 PMCID: PMC4792558 DOI: 10.18632/oncotarget.6248] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 10/14/2015] [Indexed: 12/23/2022] Open
Abstract
Despite the common usage of radiotherapy for the treatment of NSCLC, outcomes for these cancers when treated with ionizing radiation (IR) are still unsatisfactory. A better understanding of the mechanisms underlying resistance to IR is needed to design approaches to eliminate the radioresistant cells and prevent tumor recurrence and metastases. Using multiple fractions of IR we generated radioresistant cells from T2821 and T2851 human lung adenocarcinoma cells. The radioresistant phenotypes present in T2821/R and T2851/R cells include multiple changes in DNA repair genes and proteins expression, upregulation of EMT markers, alterations of cell cycle distribution, upregulation of PI3K/AKT signaling and elevated production of growth factors, cytokines, important for lung cancer progression, such as IL-6, PDGFB and SDF-1 (CXCL12). In addition to being radioresistant these cells were also found to be resistant to cisplatin. HSP90 is a molecular chaperone involved in stabilization and function of multiple client proteins implicated in NSCLC cell survival and radioresistance. We examined the effect of ganetespib, a novel HSP90 inhibitor, on T2821/R and T2851/R cell survival, migration and radioresistance. Our data indicates that ganetespib has cytotoxic activity against parental T2821 and T2851 cells and radioresistant T2821/R and T2851/R lung tumor cells. Ganetespib does not affect proliferation of normal human lung fibroblasts. Combining IR with ganetespib completely abrogates clonogenic survival of radioresistant cells. Our data show that HSP90 inhibition can potentiate the effect of radiotherapy and eliminate radioresistant and cisplatin -resistant residual cells, thus it may aid in reducing NSCLC tumor recurrence after fractionated radiotherapy.
Collapse
Affiliation(s)
- Roberto Gomez-Casal
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA.,Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael W Epperly
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA.,Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hong Wang
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA.,Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Joel S Greenberger
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA.,Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Vera Levina
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA.,Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Current address: Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| |
Collapse
|
26
|
de Araújo Farias V, O'Valle F, Lerma BA, Ruiz de Almodóvar C, López-Peñalver JJ, Nieto A, Santos A, Fernández BI, Guerra-Librero A, Ruiz-Ruiz MC, Guirado D, Schmidt T, Oliver FJ, Ruiz de Almodóvar JM. Human mesenchymal stem cells enhance the systemic effects of radiotherapy. Oncotarget 2016; 6:31164-80. [PMID: 26378036 PMCID: PMC4741595 DOI: 10.18632/oncotarget.5216] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 08/12/2015] [Indexed: 01/14/2023] Open
Abstract
The outcome of radiotherapy treatment might be further improved by a better understanding of individual variations in tumor radiosensitivity and normal tissue reactions, including the bystander effect. For many tumors, however, a definitive cure cannot be achieved, despite the availablity of more and more effective cancer treatments. Therefore, any improvement in the efficacy of radiotherapy will undoubtedly benefit a significant number of patients. Many experimental studies measure a bystander component of tumor cell death after radiotherapy, which highlights the importance of confirming these observations in a preclinical situation. Mesenchymal stem cells (MSCs) have been investigated for use in the treatment of cancers as they are able to both preferentially home onto tumors and become incorporated into their stroma. This process increases after radiation therapy. In our study we show that in vitro MSCs, when activated with a low dose of radiation, are a source of anti-tumor cytokines that decrease the proliferative activity of tumor cells, producing a potent cytotoxic synergistic effect on tumor cells. In vivo administration of unirradiated mesenchymal cells together with radiation leads to an increased efficacy of radiotherapy, thus leading to an enhancement of short and long range bystander effects on primary-irradiated tumors and distant-non-irradiated tumors. Our experiments indicate an increased cell loss rate and the decrease in the tumor cell proliferation activity as the major mechanisms underlying the delayed tumor growth and are a strong indicator of the synergistic effect between RT and MSC when they are applied together for tumor treatment in this model.
Collapse
Affiliation(s)
- Virgínea de Araújo Farias
- Instituto Universitario de Investigación en Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain.,Instituto de Parasitología y Biomedicina "López Neyra", Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Francisco O'Valle
- Instituto Universitario de Investigación en Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Borja Alonso Lerma
- Instituto Universitario de Investigación en Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | | | - Jesús J López-Peñalver
- Instituto Universitario de Investigación en Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain.,Unidad de Radiología Experimental, Centro de Instrumentación Científica, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Ana Nieto
- Instituto Universitario de Investigación en Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain.,Unidad de Experimentación Animal, Centro de Instrumentación Científica, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Ana Santos
- Instituto Universitario de Investigación en Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain.,Unidad de Microscopia, Centro de Instrumentación Científica, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Beatriz Irene Fernández
- Instituto Universitario de Investigación en Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Ana Guerra-Librero
- Instituto Universitario de Investigación en Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - María Carmen Ruiz-Ruiz
- Instituto Universitario de Investigación en Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | | | - Thomas Schmidt
- Department of General, Visceral and Transplant Surgery, University of Heidelberg, Heidelberg, Germany
| | - Francisco Javier Oliver
- Instituto de Parasitología y Biomedicina "López Neyra", Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - José Mariano Ruiz de Almodóvar
- Instituto Universitario de Investigación en Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain.,Hospital Universitario San Cecilio, Granada, Spain
| |
Collapse
|
27
|
Ojima M, Iwashita K, Kashino G, Kobashigawa S, Sasano N, Takeshita A, Ban N, Kai M. Early and Delayed Induction of DSBs by Nontargeted Effects in ICR Mouse Lymphocytes after In Vivo X Irradiation. Radiat Res 2016; 186:65-70. [PMID: 27351761 DOI: 10.1667/rr14053.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The goal of this study was to determine whether in vivo X irradiation induces nontargeted effects, such as delayed effects and bystander effects in ICR mouse lymphocytes. We first examined the generation of DNA double-strand breaks (DSBs) in lymphocytes, isolated from ICR mice exposed to 1 Gy X irradiation, by enumeration of p53 binding protein 1 (53BP1) foci, and observed that the number of 53BP1 foci reached their maximum 3 days postirradiation and decreased to background level 30 days postirradiation. However, the number of 53BP1 foci was significantly increased in lymphocytes isolated from ICR mice 90-365 days postirradiation. This result indicates that in vivo X irradiation induced delayed DSBs in ICR mouse lymphocytes. We next counted the number of 53BP1 foci in lymphocytes isolated from sham-irradiated ICR mice that had been co-cultured with lymphocytes isolated from 1 Gy X-irradiated ICR mice, and observed a significant increase in the number of 53BP1 foci 1-7 days postirradiation. This result indicates that in vivo X irradiation induced bystander effects in ICR mouse lymphocytes. These findings suggest that in vivo X irradiation induces early and delayed nontargeted effects in ICR mouse lymphocytes.
Collapse
Affiliation(s)
- Mitsuaki Ojima
- a Department of Environmental Health Science, Oita University of Nursing and Health Sciences, Oita 840-1201, Japan
| | - Keiko Iwashita
- a Department of Environmental Health Science, Oita University of Nursing and Health Sciences, Oita 840-1201, Japan
| | - Genro Kashino
- b Advanced Molecular Center, Faculty of Medicine, Oita University, Yufu 879-5593, Japan
| | - Shinko Kobashigawa
- b Advanced Molecular Center, Faculty of Medicine, Oita University, Yufu 879-5593, Japan
| | - Noriko Sasano
- a Department of Environmental Health Science, Oita University of Nursing and Health Sciences, Oita 840-1201, Japan
| | - Akiko Takeshita
- a Department of Environmental Health Science, Oita University of Nursing and Health Sciences, Oita 840-1201, Japan
| | - Nobuhiko Ban
- c Tokyo Healthcare University, Tokyo 152-8558, Japan
| | - Michiaki Kai
- a Department of Environmental Health Science, Oita University of Nursing and Health Sciences, Oita 840-1201, Japan
| |
Collapse
|
28
|
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: 2.3] [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
|
29
|
Stress-induced bystander signaling as a possible factor contributing to neuronal excitability and seizure generation/epileptogenesis. Med Hypotheses 2016; 90:57-62. [DOI: 10.1016/j.mehy.2016.03.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 03/10/2016] [Indexed: 01/23/2023]
|
30
|
Nikitaki Z, Mavragani IV, Laskaratou DA, Gika V, Moskvin VP, Theofilatos K, Vougas K, Stewart RD, Georgakilas AG. Systemic mechanisms and effects of ionizing radiation: A new 'old' paradigm of how the bystanders and distant can become the players. Semin Cancer Biol 2016; 37-38:77-95. [PMID: 26873647 DOI: 10.1016/j.semcancer.2016.02.002] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 02/01/2016] [Accepted: 02/07/2016] [Indexed: 12/26/2022]
Abstract
Exposure of cells to any form of ionizing radiation (IR) is expected to induce a variety of DNA lesions, including double strand breaks (DSBs), single strand breaks (SSBs) and oxidized bases, as well as loss of bases, i.e., abasic sites. The damaging potential of IR is primarily related to the generation of electrons, which through their interaction with water produce free radicals. In their turn, free radicals attack DNA, proteins and lipids. Damage is induced also through direct deposition of energy. These types of IR interactions with biological materials are collectively called 'targeted effects', since they refer only to the irradiated cells. Earlier and sometimes 'anecdotal' findings were pointing to the possibility of IR actions unrelated to the irradiated cells or area, i.e., a type of systemic response with unknown mechanistic basis. Over the last years, significant experimental evidence has accumulated, showing a variety of radiation effects for 'out-of-field' areas (non-targeted effects-NTE). The NTE involve the release of chemical and biological mediators from the 'in-field' area and thus the communication of the radiation insult via the so called 'danger' signals. The NTE can be separated in two major groups: bystander and distant (systemic). In this review, we have collected a detailed list of proteins implicated in either bystander or systemic effects, including the clinically relevant abscopal phenomenon, using improved text-mining and bioinformatics tools from the literature. We have identified which of these genes belong to the DNA damage response and repair pathway (DDR/R) and made protein-protein interaction (PPi) networks. Our analysis supports that the apoptosis, TLR-like and NOD-like receptor signaling pathways are the main pathways participating in NTE. Based on this analysis, we formulate a biophysical hypothesis for the regulation of NTE, based on DNA damage and apoptosis gradients between the irradiation point and various distances corresponding to bystander (5mm) or distant effects (5cm). Last but not least, in order to provide a more realistic support for our model, we calculate the expected DSB and non-DSB clusters along the central axis of a representative 200.6MeV pencil beam calculated using Monte Carlo DNA damage simulation software (MCDS) based on the actual beam energy-to-depth curves used in therapy.
Collapse
Affiliation(s)
- Zacharenia Nikitaki
- Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Zografou, 15780 Athens, Greece
| | - Ifigeneia V Mavragani
- Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Zografou, 15780 Athens, Greece
| | - Danae A Laskaratou
- Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Zografou, 15780 Athens, Greece
| | - Violeta Gika
- Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Zografou, 15780 Athens, Greece
| | - Vadim P Moskvin
- Department of Radiation Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Konstantinos Vougas
- Proteomics Research Unit, Center of Basic Research II, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Robert D Stewart
- Department of Radiation Oncology, University of Washington School of Medicine, School of Medicine, 1959 NE Pacific Street, Box 356043, Seattle, WA 98195, USA
| | - Alexandros G Georgakilas
- Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Zografou, 15780 Athens, Greece.
| |
Collapse
|
31
|
Martin OA, Yin X, Forrester HB, Sprung CN, Martin RF. Potential strategies to ameliorate risk of radiotherapy-induced second malignant neoplasms. Semin Cancer Biol 2015; 37-38:65-76. [PMID: 26721424 DOI: 10.1016/j.semcancer.2015.12.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 12/15/2015] [Accepted: 12/15/2015] [Indexed: 12/18/2022]
Abstract
This review is aimed at the issue of radiation-induced second malignant neoplasms (SMN), which has become an important problem with the increasing success of modern cancer radiotherapy (RT). It is imperative to avoid compromising the therapeutic ratio while addressing the challenge of SMN. The dilemma is illustrated by the role of reactive oxygen species in both the mechanisms of tumor cell kill and of radiation-induced carcinogenesis. We explore the literature focusing on three potential routes of amelioration to address this challenge. An obvious approach to avoiding compromise of the tumor response is the use of radioprotectors or mitigators that are selective for normal tissues. We also explore the opportunities to avoid protection of the tumor by topical/regional radioprotection of normal tissues, although this strategy limits the scope of protection. Finally, we explore the role of the bystander/abscopal phenomenon in radiation carcinogenesis, in association with the inflammatory response. Targeted and non-targeted effects of radiation are both linked to SMN through induction of DNA damage, genome instability and mutagenesis, but differences in the mechanisms and kinetics between targeted and non-targeted effects may provide opportunities to lessen SMN. The agents that could be employed to pursue each of these strategies are briefly reviewed. In many cases, the same agent has potential utility for more than one strategy. Although the parallel problem of chemotherapy-induced SMN shares common features, this review focuses on RT associated SMN. Also, we avoid the burgeoning literature on the endeavor to suppress cancer incidence by use of antioxidants and vitamins either as dietary strategies or supplementation.
Collapse
Affiliation(s)
- Olga A Martin
- Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, VIC 3002, Australia; Molecular Radiation Biology Laboratory, Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, VIC 3002, Australia; The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia.
| | - Xiaoyu Yin
- Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, VIC 3002, Australia; Molecular Radiation Biology Laboratory, Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, VIC 3002, Australia.
| | - Helen B Forrester
- Centre for Innate Immunity and Infectious Disease, Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia.
| | - Carl N Sprung
- Centre for Innate Immunity and Infectious Disease, Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia.
| | - Roger F Martin
- Molecular Radiation Biology Laboratory, Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, VIC 3002, Australia; The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
32
|
Tu W, Dong C, Konishi T, Kobayashi A, Furusawa Y, Uchihori Y, Xie Y, Dang B, Li W, Shao C. G(2)-M phase-correlative bystander effects are co-mediated by DNA-PKcs and ATM after carbon ion irradiation. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2015; 795:1-6. [PMID: 26774662 DOI: 10.1016/j.mrgentox.2015.11.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 10/30/2015] [Accepted: 11/05/2015] [Indexed: 10/22/2022]
Abstract
Accumulated evidence has shown that radiation-induced bystander effect (RIBE) may have significant implications to the efficiency of radiotherapy. Although cellular radiosensitivity relies on cell cycle status, it is largely unknown how about the relationship between RIBE and cell cycle distribution, much less the underlying mechanism. In the present study, the lung cancer A549 cells were synchronized into different cell cycle phases of G1, S and G2/M and irradiated with high linear energy transfer (LET) carbon ions. By treating nonirradiated cells with the conditioned medium from these irradiated cells, it was found that the G2-M phase cells had the largest contribution to RIBE. Meanwhile, the activity of DNA-PKcs but not ATM was increased in the synchronized G2-M phase cells in spite of both of them were activated in the asynchronous cells after carbon ion irradiation. When the G2-M phased cells were transferred with DNA-PKcs siRNA and ATM siRNA individually or treated with an inhibitor of either DNA-PKcs or ATM before carbon ion irradiation, the RIBE was effectively diminished. These results provide new evidence linking cell cycle to bystander responses and demonstrate that DNA-PKcs and ATM are two associated factors in co-regulating G2-M phase-related bystander effects.
Collapse
Affiliation(s)
- Wenzhi Tu
- Institute of Radiation Medicine, Fudan University, Shanghai 200032, China
| | - Chen Dong
- Institute of Radiation Medicine, Fudan University, Shanghai 200032, China
| | - Teruaki Konishi
- Research Development and Support Center, National Institute of Radiological Sciences, Inage, Chiba 263-8555, Japan
| | - Alisa Kobayashi
- Research Development and Support Center, National Institute of Radiological Sciences, Inage, Chiba 263-8555, Japan
| | - Yoshiya Furusawa
- Research Development and Support Center, National Institute of Radiological Sciences, Inage, Chiba 263-8555, Japan
| | - Yukio Uchihori
- Research Development and Support Center, National Institute of Radiological Sciences, Inage, Chiba 263-8555, Japan
| | - Yuexia Xie
- Institute of Radiation Medicine, Fudan University, Shanghai 200032, China; Central Laboratory of Renji Hospital, Shanghai Jiaotong University, Shanghai 200001, China
| | - Bingrong Dang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Wenjian Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Chunlin Shao
- Institute of Radiation Medicine, Fudan University, Shanghai 200032, China.
| |
Collapse
|
33
|
Zakhvataev VE. Possible scenarios of the influence of low-dose ionizing radiation on neural functioning. Med Hypotheses 2015; 85:723-35. [PMID: 26526727 DOI: 10.1016/j.mehy.2015.10.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 10/05/2015] [Accepted: 10/20/2015] [Indexed: 12/30/2022]
Abstract
Possible scenarios of the influence of ionizing radiation on neural functioning and the CNS are suggested. We argue that the radiation-induced bystander mechanisms associated with Ca(2+) flows, reactive nitrogen and oxygen species, and cytokines might lead to modulation of certain neuronal signaling pathways. The considered scenarios of conjugation of the bystander signaling and the neuronal signaling might result in modulation of certain synaptic receptors, neurogenesis, neurotransmission, channel conductance, synaptic signaling, different forms of neural plasticity, memory formation and storage, and learning. On this basis, corresponding new possible strategies for treating neurodegenerative deceases and mental disorders are proposed. The mechanisms considered might also be associated with neuronal survival and relevant to the treatment for brain injuries. At the same time, these mechanisms might be associated with detrimental effects and might facilitate the development of some neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Vladimir E Zakhvataev
- Neuroinformatics Department, Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands; Laboratory of Biological Action of Low-Intensity Factors, Siberian Federal University, 79 Svobodny pr., 660041 Krasnoyarsk, Russia.
| |
Collapse
|
34
|
Mathew A. The P5 disulfide switch: taming the aging unfolded protein response. Cell Stress Chaperones 2015; 20:743-51. [PMID: 26045202 PMCID: PMC4529870 DOI: 10.1007/s12192-015-0606-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 05/07/2015] [Accepted: 05/14/2015] [Indexed: 01/27/2023] Open
Abstract
Aging cells are characterized by a loss of proteostasis and a decreased ability to survive under environmental stress. Regulation of the UPR in aging cells has been under much scrutiny, and studies have shown that the UPR in these cells differs considerably from younger cells with regard to the induction of apoptosis and chaperone activity. The role of IRE-1 and PERK in UPR-associated apoptosis makes the regulation of these signaling cascades an important target of study. The seemingly contradictory findings regarding the role of P5 in activating and deactivating these responses warrant further investigation and may hold the key to unlocking the role of this protein in various pathological conditions. Another important target for study with regard to P5 is the effects of the localization of this protein in the mitochondria and the consequences, if any, of these effects on the activation of the UPR.
Collapse
Affiliation(s)
- Akash Mathew
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15-Datun Road, Beijing, 100101, China,
| |
Collapse
|
35
|
Yakovlev VA. Role of nitric oxide in the radiation-induced bystander effect. Redox Biol 2015; 6:396-400. [PMID: 26355395 PMCID: PMC4572387 DOI: 10.1016/j.redox.2015.08.018] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 08/27/2015] [Accepted: 08/31/2015] [Indexed: 11/27/2022] Open
Abstract
Cells that are not irradiated but are affected by “stress signal factors” released from irradiated cells are called bystander cells. These cells, as well as directly irradiated ones, express DNA damage-related proteins and display excess DNA damage, chromosome aberrations, mutations, and malignant transformation. This phenomenon has been studied widely in the past 20 years, since its first description by Nagasawa and Little in 1992, and is known as the radiation-induced bystander effect (RIBE). Several factors have been identified as playing a role in the bystander response. This review will focus on one of them, nitric oxide (NO), and its role in the stimulation and propagation of RIBE. The hydrophobic properties of NO, which permit its diffusion through the cytoplasm and plasma membranes, allow this signaling molecule to easily spread from irradiated cells to bystander cells without the involvement of gap junction intercellular communication. NO produced in irradiated tissues mediates cellular regulation through posttranslational modification of a number of regulatory proteins. The best studied of these modifications are S-nitrosylation (reversible oxidation of cysteine) and tyrosine nitration. These modifications can up- or down-regulate the functions of many proteins modulating different NO-dependent effects. These NO-dependent effects include the stimulation of genomic instability (GI) and the accumulation of DNA errors in bystander cells without direct DNA damage. Ionizing radiation stimulates generation of nitric oxide (NO). NO stimulates genomic instability by inhibiting BRCA1 protein expression. NO can diffuse and stimulate genomic instability in the bystander cells. Propagation of NO from cell-to-cell creates a “mutator fields”. Definition of the “mutator filed” is proposed.
Collapse
Affiliation(s)
- Vasily A Yakovlev
- Department of Radiation Oncology, Massey Cancer Center, Virginia Commonwealth University, VA, USA.
| |
Collapse
|
36
|
Malaquin N, Carrier-Leclerc A, Dessureault M, Rodier F. DDR-mediated crosstalk between DNA-damaged cells and their microenvironment. Front Genet 2015; 6:94. [PMID: 25815006 PMCID: PMC4357297 DOI: 10.3389/fgene.2015.00094] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 02/21/2015] [Indexed: 12/29/2022] Open
Abstract
The DNA damage response (DDR) is an evolutionarily conserved signaling cascade that senses and responds to double-strand DNA breaks by organizing downstream cellular events, ranging from appropriate DNA repair to cell cycle checkpoints. In higher organisms, the DDR prevents neoplastic transformation by directly protecting the information contained in the genome and by regulating cell fate decisions, like apoptosis and senescence, to ensure the removal of severely damaged cells. In addition to these well-studied cell-autonomous effects, emerging evidence now shows that the DDR signaling cascade can also function in a paracrine manner, thus influencing the biology of the surrounding cellular microenvironment. In this context, the DDR plays an emerging role in shaping the damaged tumor microenvironment through the regulation of tissue repair and local immune responses, thereby providing a promising avenue for novel therapeutic interventions. Additionally, while DDR-mediated extracellular signals can convey information to surrounding, undamaged cells, they can also feedback onto DNA-damaged cells to reinforce selected signaling pathways. Overall, these extracellular DDR signals can be subdivided into two time-specific waves: a rapid bystander effect occurring within a few hours of DNA damage; and a late, delayed, senescence-associated secretory phenotype generally requiring multiple days to establish. Here, we highlight and discuss examples of rapid and late DDR–mediated extracellular alarm signals.
Collapse
Affiliation(s)
- Nicolas Malaquin
- Centre de Recherche du Centre Hospitalier de l'Universite de Montréal (CRCHUM), et Institut du cancer de Montréal, Montreal, QC, Canada
| | - Audrey Carrier-Leclerc
- Centre de Recherche du Centre Hospitalier de l'Universite de Montréal (CRCHUM), et Institut du cancer de Montréal, Montreal, QC, Canada
| | - Mireille Dessureault
- Centre de Recherche du Centre Hospitalier de l'Universite de Montréal (CRCHUM), et Institut du cancer de Montréal, Montreal, QC, Canada
| | - Francis Rodier
- Centre de Recherche du Centre Hospitalier de l'Universite de Montréal (CRCHUM), et Institut du cancer de Montréal, Montreal, QC, Canada ; Département de Radiologie, Radio-Oncologie et Médicine Nucléaire, Université de Montréal Montreal, QC, Canada
| |
Collapse
|
37
|
Sánchez-Flores M, Pásaro E, Bonassi S, Laffon B, Valdiglesias V. γH2AX Assay as DNA Damage Biomarker for Human Population Studies: Defining Experimental Conditions. Toxicol Sci 2015; 144:406-13. [DOI: 10.1093/toxsci/kfv011] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
38
|
Haikerwal SJ, Hagekyriakou J, MacManus M, Martin OA, Haynes NM. Building immunity to cancer with radiation therapy. Cancer Lett 2015; 368:198-208. [PMID: 25592036 DOI: 10.1016/j.canlet.2015.01.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 01/05/2015] [Accepted: 01/06/2015] [Indexed: 12/14/2022]
Abstract
Over the last decade there has been a dramatic shift in the focus of cancer research toward understanding how the body's immune defenses can be harnessed to promote the effectiveness of cytotoxic anti-cancer therapies. The ability of ionizing radiation to elicit anti-cancer immune responses capable of controlling tumor growth has led to the emergence of promising combination-based radio-immunotherapeutic strategies for the treatment of cancer. Herein we review the immunoadjuvant properties of localized radiation therapy and discuss how technological advances in radio-oncology and developments in the field of tumor-immunotherapy have started to revolutionize the therapeutic application of radiotherapy.
Collapse
Affiliation(s)
- Suresh J Haikerwal
- Cancer Therapeutics Program, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
| | - Jim Hagekyriakou
- Department of Physical Sciences, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
| | - Michael MacManus
- Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Vic, Australia
| | - Olga A Martin
- Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Vic, Australia; Molecular Radiation Biology Laboratory, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
| | - Nicole M Haynes
- Cancer Therapeutics Program, Peter MacCallum Cancer Centre, Melbourne, Vic, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Vic, Australia.
| |
Collapse
|
39
|
Martin OA, Redon CE, Dickey JS, Nakamura AJ, Bonner WM. Para-inflammation mediates systemic DNA damage in response to tumor growth. Commun Integr Biol 2014. [DOI: 10.4161/cib.13942] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
40
|
Burdak-Rothkamm S, Smith A, Lobachevsky P, Martin R, Prise KM. Radioprotection of targeted and bystander cells by methylproamine. Strahlenther Onkol 2014; 191:248-55. [PMID: 25245467 PMCID: PMC4338360 DOI: 10.1007/s00066-014-0751-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 09/02/2014] [Indexed: 11/25/2022]
Abstract
Introduction Radioprotective agents are of interest for application in radiotherapy for cancer and in public health medicine in the context of accidental radiation exposure. Methylproamine is the lead compound of a class of radioprotectors which act as DNA binding anti-oxidants, enabling the repair of transient radiation-induced oxidative DNA lesions. This study tested methylproamine for the radioprotection of both directly targeted and bystander cells. Methods T98G glioma cells were treated with 15 μM methylproamine and exposed to 137Cs γ-ray/X-ray irradiation and He2+ microbeam irradiation. Radioprotection of directly targeted cells and bystander cells was measured by clonogenic survival or γH2AX assay. Results Radioprotection of directly targeted T98G cells by methylproamine was observed for 137Cs γ-rays and X-rays but not for He2+ charged particle irradiation. The effect of methylproamine on the bystander cell population was tested for both X-ray irradiation and He2+ ion microbeam irradiation. The X-ray bystander experiments were carried out by medium transfer from irradiated to non-irradiated cultures and three experimental designs were tested. Radioprotection was only observed when recipient cells were pretreated with the drug prior to exposure to the conditioned medium. In microbeam bystander experiments targeted and nontargeted cells were co-cultured with continuous methylproamine treatment during irradiation and postradiation incubation; radioprotection of bystander cells was observed. Discussion and conclusion Methylproamine protected targeted cells from DNA damage caused by γ-ray or X-ray radiation but not He2+ ion radiation. Protection of bystander cells was independent of the type of radiation which the donor population received.
Collapse
|
41
|
Site-directed delivery of nitric oxide to cancers. Nitric Oxide 2014; 43:8-16. [PMID: 25124221 DOI: 10.1016/j.niox.2014.07.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 07/15/2014] [Accepted: 07/18/2014] [Indexed: 01/28/2023]
Abstract
Nitric oxide (NO) is a reactive gaseous free radical which mediates numerous biological processes. At elevated levels, NO is found to be toxic to cancers and hence, a number of strategies for site-directed delivery of NO to cancers are in development during the past two decades. More recently, the focus of research has been to, in conjunction with other cancer drugs deliver NO to cancers for its secondary effects including inhibition of cellular drug efflux pumps. Among the various approaches toward site-selective delivery of exogenous NO sources, enzyme activated nitric oxide donors belonging to the diazeniumdiolate category afford unique advantages including exquisite control of rates of NO generation and selectivity of NO production. For this prodrug approach, enzymes including esterase, glutathione/glutathione S-transferase, DT-diaphorase, and nitroreductase are utilized. Here, we review the design and development of various approaches to enzymatic site-directed delivery of NO to cancers and their potential.
Collapse
|
42
|
Systemic DNA damage accumulation under in vivo tumor growth can be inhibited by the antioxidant Tempol. Cancer Lett 2014; 353:248-57. [PMID: 25069035 DOI: 10.1016/j.canlet.2014.07.030] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 07/16/2014] [Accepted: 07/16/2014] [Indexed: 12/18/2022]
Abstract
Recently we found that mice bearing subcutaneous non-metastatic tumors exhibited elevated levels of two types of complex DNA damage, i.e., double-strand breaks and oxidatively-induced clustered DNA lesions in various tissues throughout the body, both adjacent to and distant from the tumor site. This DNA damage was dependent on CCL2, a cytokine involved in the recruitment and activation of macrophages, suggesting that this systemic DNA damage was mediated via tumor-induced chronic inflammatory responses involving cytokines, activation of macrophages, and consequent free radical production. If free radicals are involved, then a diet containing an antioxidant may decrease the distant DNA damage. Here we repeated our standard protocol in cohorts of two syngeneic tumor-bearing C57BL/6NCr mice that were on a Tempol-supplemented diet. We show that double-strand break and oxidatively-induced clustered DNA lesion levels were considerably decreased, about two- to three fold, in the majority of tissues studied from the tumor-bearing mice fed the antioxidant Tempol compared to the control tumor-bearing mice. Similar results were also observed in nude mice suggesting that the Tempol effects are independent of functioning adaptive immunity. This is the first in vivo study demonstrating the effect of a dietary antioxidant on abscopal DNA damage in tissues distant from a localized source of genotoxic stress. These findings may be important for understanding the mechanisms of genomic instability and carcinogenesis caused by chronic stress-induced systemic DNA damage and for developing preventative strategies.
Collapse
|
43
|
Weigel C, Schmezer P, Plass C, Popanda O. Epigenetics in radiation-induced fibrosis. Oncogene 2014; 34:2145-55. [PMID: 24909163 DOI: 10.1038/onc.2014.145] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 04/17/2014] [Accepted: 04/23/2014] [Indexed: 02/06/2023]
Abstract
Radiotherapy is a major cancer treatment option but dose-limiting side effects such as late-onset fibrosis in the irradiated tissue severely impair quality of life in cancer survivors. Efforts to explain radiation-induced fibrosis, for example, by genetic variation remained largely inconclusive. Recently published molecular analyses on radiation response and fibrogenesis showed a prominent role of epigenetic gene regulation. This review summarizes the current knowledge on epigenetic modifications in fibrotic disease and radiation response, and it points out the important role for epigenetic mechanisms such as DNA methylation, microRNAs and histone modifications in the development of this disease. The synopsis illustrates the complexity of radiation-induced fibrosis and reveals the need for investigations to further unravel its molecular mechanisms. Importantly, epigenetic changes are long-term determinants of gene expression and can therefore support those mechanisms that induce and perpetuate fibrogenesis even in the absence of the initial damaging stimulus. Future work must comprise the interconnection of acute radiation response and long-lasting epigenetic effects in order to assess their role in late-onset radiation fibrosis. An improved understanding of the underlying biology is fundamental to better comprehend the origin of this disease and to improve both preventive and therapeutic strategies.
Collapse
Affiliation(s)
- C Weigel
- Department of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - P Schmezer
- Department of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - C Plass
- Department of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - O Popanda
- Department of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
44
|
Shapiro JA. Epigenetic control of mobile DNA as an interface between experience and genome change. Front Genet 2014; 5:87. [PMID: 24795749 PMCID: PMC4007016 DOI: 10.3389/fgene.2014.00087] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 04/01/2014] [Indexed: 12/29/2022] Open
Abstract
Mobile DNA in the genome is subject to RNA-targeted epigenetic control. This control regulates the activity of transposons, retrotransposons and genomic proviruses. Many different life history experiences alter the activities of mobile DNA and the expression of genetic loci regulated by nearby insertions. The same experiences induce alterations in epigenetic formatting and lead to trans-generational modifications of genome expression and stability. These observations lead to the hypothesis that epigenetic formatting directed by non-coding RNA provides a molecular interface between life history events and genome alteration.
Collapse
Affiliation(s)
- James A. Shapiro
- Department of Biochemistry and Molecular Biology, University of ChicagoChicago, IL, USA
| |
Collapse
|
45
|
Pereira S, Malard V, Ravanat JL, Davin AH, Armengaud J, Foray N, Adam-Guillermin C. Low doses of gamma-irradiation induce an early bystander effect in zebrafish cells which is sufficient to radioprotect cells. PLoS One 2014; 9:e92974. [PMID: 24667817 PMCID: PMC3965492 DOI: 10.1371/journal.pone.0092974] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 02/27/2014] [Indexed: 11/18/2022] Open
Abstract
The term “bystander effect” is used to describe an effect in which cells that have not been exposed to radiation are affected by irradiated cells though various intracellular signaling mechanisms. In this study we analyzed the kinetics and mechanisms of bystander effect and radioadaptation in embryonic zebrafish cells (ZF4) exposed to chronic low dose of gamma rays. ZF4 cells were irradiated for 4 hours with total doses of gamma irradiation ranging from 0.01–0.1 Gy. In two experimental conditions, the transfer of irradiated cells or culture medium from irradiated cells results in the occurrence of DNA double strand breaks in non-irradiated cells (assessed by the number of γ-H2AX foci) that are repaired at 24 hours post-irradiation whatever the dose. At low total irradiation doses the bystander effect observed does not affect DNA repair mechanisms in targeted and bystander cells. An increase in global methylation of ZF4 cells was observed in irradiated cells and bystander cells compared to control cells. We observed that pre-irradiated cells which are then irradiated for a second time with the same doses contained significantly less γ-H2AX foci than in 24 h gamma-irradiated control cells. We also showed that bystander cells that have been in contact with the pre-irradiated cells and then irradiated alone present less γ-H2AX foci compared to the control cells. This radioadaptation effect is significantly more pronounced at the highest doses. To determine the factors involved in the early events of the bystander effect, we performed an extensive comparative proteomic study of the ZF4 secretomes upon irradiation. In the experimental conditions assayed here, we showed that the early events of bystander effect are probably not due to the secretion of specific proteins neither the oxidation of these secreted proteins. These results suggest that early bystander effect may be due probably to a combination of multiple factors.
Collapse
Affiliation(s)
- Sandrine Pereira
- Institut de Radioprotection et de Sûreté Nucléaire, PRP-Environnement/SERIS, Laboratoire d’Ecotoxicologie des Radionucléides, Cadarache, St Paul Lez Durance, France
- CRCL - UMR INSERM 1052 - CNRS 5286, Equipe de Radiobiologie, Cheney A- 1éme étage, Lyon, France
- * E-mail:
| | - Véronique Malard
- CEA, DSV, IBEB, Lab Biochim System Perturb, Bagnols-sur-Cèze, France
| | - Jean-Luc Ravanat
- Laboratoire des Lésions des Acides Nucléiques, INAC/Scib UMR E3 CEA-UJF, CEA Grenoble, Grenoble, France
| | - Anne-Hélène Davin
- CEA, DSV, IBEB, Lab Biochim System Perturb, Bagnols-sur-Cèze, France
| | - Jean Armengaud
- CEA, DSV, IBEB, Lab Biochim System Perturb, Bagnols-sur-Cèze, France
| | - Nicolas Foray
- CRCL - UMR INSERM 1052 - CNRS 5286, Equipe de Radiobiologie, Cheney A- 1éme étage, Lyon, France
| | - Christelle Adam-Guillermin
- Institut de Radioprotection et de Sûreté Nucléaire, PRP-Environnement/SERIS, Laboratoire d’Ecotoxicologie des Radionucléides, Cadarache, St Paul Lez Durance, France
| |
Collapse
|
46
|
Widel M, Krzywon A, Gajda K, Skonieczna M, Rzeszowska-Wolny J. Induction of bystander effects by UVA, UVB, and UVC radiation in human fibroblasts and the implication of reactive oxygen species. Free Radic Biol Med 2014; 68:278-87. [PMID: 24373962 DOI: 10.1016/j.freeradbiomed.2013.12.021] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 12/05/2013] [Accepted: 12/18/2013] [Indexed: 01/26/2023]
Abstract
Radiation-induced bystander effects are various types of responses displayed by nonirradiated cells induced by signals transmitted from neighboring irradiated cells. This phenomenon has been well studied after ionizing radiation, but data on bystander effects after UV radiation are limited and so far have been reported mainly after UVA and UVB radiation. The studies described here were aimed at comparing the responses of human dermal fibroblasts exposed directly to UV (A, B, or C wavelength range) and searching for bystander effects induced in unexposed cells using a transwell co-incubation system. Cell survival and apoptosis were used as a measure of radiation effects. Additionally, induction of senescence in UV-exposed and bystander cells was evaluated. Reactive oxygen species (ROS), superoxide radical anions, and nitric oxide inside the cells and secretion of interleukins 6 and 8 (IL-6 and IL-8) into the medium were assayed and evaluated as potential mediators of bystander effects. All three regions of ultraviolet radiation induced bystander effects in unexposed cells, as shown by a diminution of survival and an increase in apoptosis, but the pattern of response to direct exposure and the bystander effects differed depending on the UV spectrum. Although UVA and UVB were more effective than UVC in generation of apoptosis in bystander cells, UVC induced senescence both in irradiated cells and in neighbors. The level of cellular ROS increased significantly shortly after UVA and UVB exposure, suggesting that the bystander effects may be mediated by ROS generated in cells by UV radiation. Interestingly, UVC was more effective at generation of ROS in bystanders than in directly exposed cells and induced a high yield of superoxide in exposed and bystander cells, which, however, was only weakly associated with impairment of mitochondrial membrane potential. Increasing concentration of IL-6 but not IL-8 after exposure to each of the three bands of UV points to its role as a mediator in the bystander effect. Nitric oxide appeared to play a minor role as a mediator of bystander effects in our experiments. The results demonstrating an increase in intracellular oxidation, not only in directly UV-exposed but also in neighboring cells, and generation of proinflammatory cytokines, processes entailing cell damage (decreased viability, apoptosis, senescence), suggest that all bands of UV radiation carry a potential hazard for human health, not only due to direct mechanisms, but also due to bystander effects.
Collapse
Affiliation(s)
- Maria Widel
- Biosystems Group, Institute of Automatic Control, Faculty of Automatics, Electronics, and Informatics, Silesian University of Technology, 44-100 Gliwice, Poland.
| | - Aleksandra Krzywon
- Biosystems Group, Institute of Automatic Control, Faculty of Automatics, Electronics, and Informatics, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Karolina Gajda
- Biosystems Group, Institute of Automatic Control, Faculty of Automatics, Electronics, and Informatics, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Magdalena Skonieczna
- Biosystems Group, Institute of Automatic Control, Faculty of Automatics, Electronics, and Informatics, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Joanna Rzeszowska-Wolny
- Biosystems Group, Institute of Automatic Control, Faculty of Automatics, Electronics, and Informatics, Silesian University of Technology, 44-100 Gliwice, Poland
| |
Collapse
|
47
|
Wu S, Jin C, Lu X, Yang J, Liu Q, Qi M, Lu S, Zhang L, Cai Y. Bystander effect induced by UVC radiation in Chinese hamster V79 cells. Photochem Photobiol 2014; 90:837-44. [PMID: 24517728 DOI: 10.1111/php.12255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 01/29/2014] [Indexed: 11/26/2022]
Abstract
In past decades, researches on radiation-induced bystander effect mainly focused on ionizing radiation such as α-particle, β-particle, X-ray and γ-ray. But few researches have been conducted on the ability of ultraviolet (UV) radiation-induced bystander effect, and knowledge of UVC-induced bystander effect is far limited. Here, we adopted medium transfer experiment to detect whether UVC could cause bystander effect in Chinese hamster V79 cells. We determined the cell viability, apoptosis rate, chromosome aberration and ultrastructure changes, respectively. Our results showed that: (1) the viability of UVC-irradiated V79 cells declined significantly with the dosage of UVC; (2) similar to the irradiated cells, the main death type of bystander cells cultured in irradiation conditioned medium (ICMs) was also apoptosis; (3) soluble factors secreted by UVC-irradiated cells could induce bystander effect in V79 cells; (4) cells treated with 4 h ICM collected from 90 mJ cm(-2) UVC-irradiated cells displayed the strongest response. Our data revealed that UVC could cause bystander effect through the medium soluble factors excreted from irradiated cells and this bystander effect was a novel quantitative and kinetic response. These findings might provide a foundation to further explore the exact soluble bystander factors and detailed mechanism underlying UVC-induced bystander effect.
Collapse
Affiliation(s)
- Shengwen Wu
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, Shenyang, China
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Havaki S, Kotsinas A, Chronopoulos E, Kletsas D, Georgakilas A, Gorgoulis VG. The role of oxidative DNA damage in radiation induced bystander effect. Cancer Lett 2014; 356:43-51. [PMID: 24530228 DOI: 10.1016/j.canlet.2014.01.023] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Revised: 12/08/2013] [Accepted: 01/24/2014] [Indexed: 02/06/2023]
Abstract
Ionizing radiation (IR) has been described as a double-edged sword, since it is used for diagnostic and therapeutic medical applications, and at the same time it is a well known human mutagen and carcinogen, causing wide-ranging chromosomal aberrations. It is nowadays accepted that the detrimental effects of IR are not restricted only in the irradiated cells, but also to non-irradiated bystander or even distant cells manifesting various biological effects. This review presents the role of oxidative stress in the induction of bystander effects referring to the types of the implicated oxidative DNA lesions, the contributing intercellular and intracellular stress mediators, the way they are transmitted from irradiated to bystander cells and finally, the complex role of the bystander effect in the therapeutic efficacy of radiation treatment of cancer.
Collapse
Affiliation(s)
- Sophia Havaki
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, University of Athens, Athens, Greece
| | - Athanassios Kotsinas
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, University of Athens, Athens, Greece
| | | | - Dimitris Kletsas
- Laboratory of Cell Proliferation and Ageing, Institute of Biosciences and Applications, National Center for Scientific Research Demokritos, Athens, Greece
| | - Alexandros Georgakilas
- Physics Department, School of Applied Mathematics and Physical Sciences, National Technical University of Athens (NTUA), Zografou, 15780 Athens, Greece
| | - Vassilis G Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, University of Athens, Athens, Greece; Biomedical Research Foundation, Academy of Athens, Athens, Greece; Faculty Institute for Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, M13 9WL, UK.
| |
Collapse
|
49
|
Lamkowski A, Forcheron F, Agay D, Ahmed EA, Drouet M, Meineke V, Scherthan H. DNA damage focus analysis in blood samples of minipigs reveals acute partial body irradiation. PLoS One 2014; 9:e87458. [PMID: 24498326 PMCID: PMC3911974 DOI: 10.1371/journal.pone.0087458] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 12/27/2013] [Indexed: 11/18/2022] Open
Abstract
Radiation accidents frequently involve acute high dose partial body irradiation leading to victims with radiation sickness and cutaneous radiation syndrome that implements radiation-induced cell death. Cells that are not lethally hit seek to repair ionizing radiation (IR) induced damage, albeit at the expense of an increased risk of mutation and tumor formation due to misrepair of IR-induced DNA double strand breaks (DSBs). The response to DNA damage includes phosphorylation of histone H2AX in the vicinity of DSBs, creating foci in the nucleus whose enumeration can serve as a radiation biodosimeter. Here, we investigated γH2AX and DNA repair foci in peripheral blood lymphocytes of Göttingen minipigs that experienced acute partial body irradiation (PBI) with 49 Gy (±6%) Co-60 γ-rays of the upper lumbar region. Blood samples taken 4, 24 and 168 hours post PBI were subjected to γ-H2AX, 53BP1 and MRE11 focus enumeration. Peripheral blood lymphocytes (PBL) of 49 Gy partial body irradiated minipigs were found to display 1–8 DNA damage foci/cell. These PBL values significantly deceed the high foci numbers observed in keratinocyte nuclei of the directly γ-irradiated minipig skin regions, indicating a limited resident time of PBL in the exposed tissue volume. Nonetheless, PBL samples obtained 4 h post IR in average contained 2.2% of cells displaying a pan-γH2AX signal, suggesting that these received a higher IR dose. Moreover, dispersion analysis indicated partial body irradiation for all 13 minipigs at 4 h post IR. While dose reconstruction using γH2AX DNA repair foci in lymphocytes after in vivo PBI represents a challenge, the DNA damage focus assay may serve as a rapid, first line indicator of radiation exposure. The occurrence of PBLs with pan-γH2AX staining and of cells with relatively high foci numbers that skew a Poisson distribution may be taken as indicator of acute high dose partial body irradiation, particularly when samples are available early after IR exposure.
Collapse
Affiliation(s)
- Andreas Lamkowski
- Institut für Radiobiologie der Bundeswehr in Verb. mit der Universität Ulm, München, Germany
| | - Fabien Forcheron
- Institut de Recherche Biomédicale des Armées (IRBA), Bretigny sur Orge, France
| | - Diane Agay
- Institut de Recherche Biomédicale des Armées (IRBA), Bretigny sur Orge, France
| | - Emad A. Ahmed
- Institut für Radiobiologie der Bundeswehr in Verb. mit der Universität Ulm, München, Germany
| | - Michel Drouet
- Institut de Recherche Biomédicale des Armées (IRBA), Bretigny sur Orge, France
| | - Viktor Meineke
- Institut für Radiobiologie der Bundeswehr in Verb. mit der Universität Ulm, München, Germany
| | - Harry Scherthan
- Institut für Radiobiologie der Bundeswehr in Verb. mit der Universität Ulm, München, Germany
- * E-mail:
| |
Collapse
|
50
|
Klammer H, Mladenov E, Li F, Iliakis G. Bystander effects as manifestation of intercellular communication of DNA damage and of the cellular oxidative status. Cancer Lett 2013; 356:58-71. [PMID: 24370566 DOI: 10.1016/j.canlet.2013.12.017] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 12/13/2013] [Accepted: 12/14/2013] [Indexed: 12/30/2022]
Abstract
It is becoming increasingly clear that cells exposed to ionizing radiation (IR) and other genotoxic agents (targeted cells) can communicate their DNA damage response (DDR) status to cells that have not been directly irradiated (bystander cells). The term radiation-induced bystander effects (RIBE) describes facets of this phenomenon, but its molecular underpinnings are incompletely characterized. Consequences of DDR in bystander cells have been extensively studied and include transformation and mutation induction; micronuclei, chromosome aberration and sister chromatid exchange formation; as well as modulations in gene expression, proliferation and differentiation patterns. A fundamental question arising from such observations is why targeted cells induce DNA damage in non-targeted, bystander cells threatening thus their genomic stability and risking the induction of cancer. Here, we review and synthesize available literature to gather support for a model according to which targeted cells modulate as part of DDR their redox status and use it as a source to generate signals for neighboring cells. Such signals can be either small molecules transported to adjacent non-targeted cells via gap-junction intercellular communication (GJIC), or secreted factors that can reach remote, non-targeted cells by diffusion or through the circulation. We review evidence that such signals can induce in the recipient cell modulations of redox status similar to those seen in the originating targeted cell - occasionally though self-amplifying feedback loops. The resulting increase of oxidative stress in bystander cells induces, often in conjunction with DNA replication, the observed DDR-like responses that are at times strong enough to cause apoptosis. We reason that RIBE reflect the function of intercellular communication mechanisms designed to spread within tissues, or the entire organism, information about DNA damage inflicted to individual, constituent cells. Such responses are thought to protect the organism by enhancing repair in a community of cells and by eliminating severely damaged cells.
Collapse
Affiliation(s)
- Holger Klammer
- Institute of Medical Radiation Biology, University of Duisburg-Essen Medical School, Essen, Germany
| | - Emil Mladenov
- Institute of Medical Radiation Biology, University of Duisburg-Essen Medical School, Essen, Germany
| | - Fanghua Li
- Institute of Medical Radiation Biology, University of Duisburg-Essen Medical School, Essen, Germany
| | - George Iliakis
- Institute of Medical Radiation Biology, University of Duisburg-Essen Medical School, Essen, Germany.
| |
Collapse
|