1
|
Andriotty M, Wang CKC, Kapadia A, McCord RP, Agasthya G. Integrating chromosome conformation and DNA repair in a computational framework to assess cell radiosensitivity . Phys Med Biol 2024; 69:245017. [PMID: 39569898 DOI: 10.1088/1361-6560/ad94c6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 11/19/2024] [Indexed: 11/22/2024]
Abstract
Objective.The arrangement of chromosomes in the cell nucleus has implications for cell radiosensitivity. The development of new tools to utilize Hi-C chromosome conformation data in nanoscale radiation track structure simulations allows forin silicoinvestigation of this phenomenon. We have developed a framework employing Hi-C-based cell nucleus models in Monte Carlo radiation simulations, in conjunction with mechanistic models of DNA repair, to predict not only the initial radiation-induced DNA damage, but also the repair outcomes resulting from this damage, allowing us to investigate the role chromosome conformation plays in the biological outcome of radiation exposure.Approach.In this study, we used this framework to generate cell nucleus models based on Hi-C data from fibroblast and lymphoblastoid cells and explore the effects of cell type-specific chromosome structure on radiation response. The models were used to simulate external beam irradiation including DNA damage and subsequent DNA repair. The kinetics of the simulated DNA repair were compared with previous results.Main results.We found that the fibroblast models resulted in a higher rate of inter-chromosome misrepair than the lymphoblastoid model, despite having similar amounts of initial DNA damage and total misrepairs for each irradiation scenario.Significance.This framework represents a step forward in radiobiological modeling and simulation allowing for more realistic investigation of radiosensitivity in different types of cells.
Collapse
Affiliation(s)
- Matthew Andriotty
- Oak Ridge National Laboratory, Oak Ridge, TN, United States of America
- Georgia Institute of Technology, Atlanta, GA, United States of America
| | - C-K Chris Wang
- Georgia Institute of Technology, Atlanta, GA, United States of America
| | - Anuj Kapadia
- Oak Ridge National Laboratory, Oak Ridge, TN, United States of America
| | | | - Greeshma Agasthya
- Oak Ridge National Laboratory, Oak Ridge, TN, United States of America
| |
Collapse
|
2
|
Nagashima H, Komatsu K, Tauchi H. Possible existence of dose-rate threshold for mutation induction by chronic low-dose-rate gamma-rays. RADIATION PROTECTION DOSIMETRY 2024; 200:1615-1619. [PMID: 39540484 DOI: 10.1093/rpd/ncae059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/18/2024] [Accepted: 02/29/2024] [Indexed: 11/16/2024]
Abstract
To assess the biological effects of low-dose and low-dose-rate radiation, we established a sensitive assay system for detecting somatic mutations in hypoxanthine-phosphoribosyltransferase 1 (HPRT1) gene. In this study, we investigated the dose-rate effects of mutagenesis by gamma irradiation at dose-rates of 6.6, 20 and 200 mGy d-1. We identified a potential inflection point in the gamma-induced mutant frequency, which ranged between 6.6 and 20 mGy d-1. In addition, the mutant spectrum was not different from that of the non-irradiated control at all dose-rates. Compared with previous studies with low-concentration HTO exposure, mutant frequencies were similar, but mutant spectrum showed different trends, especially at high-dose-rates (200 mGy d-1). These observations indicate the presence of potential mechanistic differences in mutagenic events between tritium beta and gamma-rays.
Collapse
Affiliation(s)
- Haruki Nagashima
- Institute for Environmental Science, 1-7 Ienomae, Obuchi, Rokkasho, Kamikita, Aomori 039-3212, Japan
| | - Kenshi Komatsu
- Radiation Biology Center, Kyoto University, Yoshida-Konoe Cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiroshi Tauchi
- Institute for Environmental Science, 1-7 Ienomae, Obuchi, Rokkasho, Kamikita, Aomori 039-3212, Japan
- Department of Biological Sciences, Ibaraki University, 2-1-1 Bunkyo, Mito, Ibaraki 310-8512, Japan
| |
Collapse
|
3
|
Seane EN, Nair S, Vandevoorde C, Bisio A, Joubert A. Multi-Target Inhibitor CUDC-101 Impairs DNA Damage Repair and Enhances Radiation Response in Triple-Negative Breast Cell Line. Pharmaceuticals (Basel) 2024; 17:1467. [PMID: 39598379 PMCID: PMC11597529 DOI: 10.3390/ph17111467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Since the discovery that Histone deacetylase inhibitors (HDCAi) could enhance radiation response, a number of HDACi, mainly pan-HDAC inhibitors, have been studied either as monotherapy or in combination with X-ray irradiation or chemotherapeutic drugs in the management of breast cancer. However, studies on the combination of HDACi and proton radiation remain limited. CUDC-101 is a multitarget inhibitor of Histone deacetylases (HDACs), epidermal growth factor receptor (EGFR), and human epidermal growth factor receptor 2 (HER-2). In this paper, the effectiveness of CUDC-101 in enhancing radiation response to both proton and X-ray irradiation was studied. METHODS MCF-7, MDA-MB-231, and MCF-10A cell lines were pre-treated with CUDC-101 and exposed to 148 MeV protons, and X-rays were used as reference radiation. Colony survival, γ-H2AX foci, apoptosis, and cell cycle analysis assays were performed. RESULTS γ-H2AX foci assays showed increased sensitivity to CUDC-101 in the MDA-MB-231 cell line compared to the MCF-7 cell line. In both cell lines, induction of apoptosis was enhanced in CUDC-101 pre-treated cells compared to radiation (protons or X-rays) alone. Increased apoptosis was also noted in CUDC-101 pre-treated cells in the MCF-10A cell line. Cell cycle analysis showed increased G2/M arrest by CUDC-101 mono-treatment as well as combination of CUDC-101 and X-ray irradiation in the MDA-MB-231 cell line. CONCLUSIONS CUDC-101 effectively enhances response to both proton and X-ray irradiation, in the triple-negative MDA-MB-231 cell line. This enhancement was most notable when CUDC-101 was combined with proton irradiation. This study highlights that CUDC-101 holds potential in the management of triple-negative breast cancer as monotherapy or in combination with protons or X-ray irradiation.
Collapse
Affiliation(s)
- Elsie Neo Seane
- Department of Radiography, School of Health Care Sciences, Faculty of Health Sciences, University of Pretoria, Pretoria 0028, South Africa
- Department of Medical Imaging and Therapeutic Sciences, Faculty of Health and Wellness, Cape Peninsula University of Technology, Bellville 7535, South Africa
- Separate Sector Cyclotron (SSC) Laboratory, Radiation Biophysics Division, iThemba LABS, Cape Town 7530, South Africa;
| | - Shankari Nair
- Separate Sector Cyclotron (SSC) Laboratory, Radiation Biophysics Division, iThemba LABS, Cape Town 7530, South Africa;
| | - Charlot Vandevoorde
- Department of Biophysics, GSI Helmholtzzentrum für Schwerionenforschung, 64291 Darmstadt, Germany;
| | - Alessandra Bisio
- Department of Cellular, Computational and Integrative Biology, Via Sommarive, 9, Povo, 38123 Trento, Italy;
| | - Anna Joubert
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0028, South Africa;
| |
Collapse
|
4
|
Valente D, Gentileschi MP, Valenti A, Burgio M, Soddu S, Bruzzaniti V, Guerrisi A, Verdina A. Cumulative Dose from Recurrent CT Scans: Exploring the DNA Damage Response in Human Non-Transformed Cells. Int J Mol Sci 2024; 25:7064. [PMID: 39000171 PMCID: PMC11241671 DOI: 10.3390/ijms25137064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
Recurrent computed tomography (CT) examination has become a common diagnostic procedure for several diseases and injuries. Though each singular CT scan exposes individuals at low doses of low linear energy transfer (LET) radiation, the cumulative dose received from recurrent CT scans poses an increasing concern for potential health risks. Here, we evaluated the biological effects of recurrent CT scans on the DNA damage response (DDR) in human fibroblasts and retinal pigment epithelial cells maintained in culture for five months and subjected to four CT scans, one every four weeks. DDR kinetics and eventual accumulation of persistent-radiation-induced foci (P-RIF) were assessed by combined immunofluorescence for γH2AX and 53BP1, i.e., γH2AX/53BP1 foci. We found that CT scan repetitions significantly increased both the number and size of γH2AX/53BP1 foci. In particular, after the third CT scan, we observed the appearance of giant foci that might result from the overlapping of individual small foci and that do not associate with irreversible growth arrest, as shown by DNA replication in the foci-carrying cells. Whether these giant foci represent coalescence of unrepaired DNA damage as reported following single exposition to high doses of high LET radiation is still unclear. However, morphologically, these giant foci resemble the recently described compartmentalization of damaged DNA that should facilitate the repair of DNA double-strand breaks but also increase the risk of chromosomal translocations. Overall, these results indicate that for a correct evaluation of the damage following recurrent CT examinations, it is necessary to consider the size and composition of the foci in addition to their number.
Collapse
Affiliation(s)
- Davide Valente
- Unit of Cellular Networks and Molecular Therapeutic Targets, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (D.V.); (M.P.G.); (S.S.)
- Institute of Molecular Biology and Pathology (IBPM), National Research Council (CNR), c/o Sapienza University, 00185 Rome, Italy
| | - Maria Pia Gentileschi
- Unit of Cellular Networks and Molecular Therapeutic Targets, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (D.V.); (M.P.G.); (S.S.)
| | - Alessandro Valenti
- Unit of Radiology and Diagnostic Imaging, Department of Clinical and Dermatological Research, IRCCS San Gallicano Dermatological Institute, 00144 Rome, Italy; (A.V.); (M.B.)
| | - Massimo Burgio
- Unit of Radiology and Diagnostic Imaging, Department of Clinical and Dermatological Research, IRCCS San Gallicano Dermatological Institute, 00144 Rome, Italy; (A.V.); (M.B.)
| | - Silvia Soddu
- Unit of Cellular Networks and Molecular Therapeutic Targets, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (D.V.); (M.P.G.); (S.S.)
| | - Vicente Bruzzaniti
- Unit of Medical Physics and Expert Systems, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Antonino Guerrisi
- Unit of Radiology and Diagnostic Imaging, Department of Clinical and Dermatological Research, IRCCS San Gallicano Dermatological Institute, 00144 Rome, Italy; (A.V.); (M.B.)
| | - Alessandra Verdina
- Unit of Cellular Networks and Molecular Therapeutic Targets, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (D.V.); (M.P.G.); (S.S.)
| |
Collapse
|
5
|
Poignant F, Pariset E, Plante I, Ponomarev AL, Evain T, Viger L, Slaba TC, Blattnig SR, Costes SV. DNA break clustering as a predictor of cell death across various radiation qualities: influence of cell size, cell asymmetry, and beam orientation. Integr Biol (Camb) 2024; 16:zyae015. [PMID: 39299711 DOI: 10.1093/intbio/zyae015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 08/23/2024] [Indexed: 09/22/2024]
Abstract
Cosmic radiation, composed of high charge and energy (HZE) particles, causes cellular DNA damage that can result in cell death or mutation that can evolve into cancer. In this work, a cell death model is applied to several cell lines exposed to HZE ions spanning a broad range of linear energy transfer (LET) values. We hypothesize that chromatin movement leads to the clustering of multiple double strand breaks (DSB) within one radiation-induced foci (RIF). The survival probability of a cell population is determined by averaging the survival probabilities of individual cells, which is function of the number of pairwise DSB interactions within RIF. The simulation code RITCARD was used to compute DSB. Two clustering approaches were applied to determine the number of RIF per cell. RITCARD outputs were combined with experimental data from four normal human cell lines to derive the model parameters and expand its predictions in response to ions with LET ranging from ~0.2 keV/μm to ~3000 keV/μm. Spherical and ellipsoidal nuclear shapes and two ion beam orientations were modeled to assess the impact of geometrical properties on cell death. The calculated average number of RIF per cell reproduces the saturation trend for high doses and high-LET values that is usually experimentally observed. The cell survival model generates the recognizable bell shape of LET dependence for the relative biological effectiveness (RBE). At low LET, smaller nuclei have lower survival due to increased DNA density and DSB clustering. At high LET, nuclei with a smaller irradiation area-either because of a smaller size or a change in beam orientation-have a higher survival rate due to a change in the distribution of DSB/RIF per cell. If confirmed experimentally, the geometric characteristics of cells would become a significant factor in predicting radiation-induced biological effects. Insight Box: High-charge and energy (HZE) ions are characterized by dense linear energy transfer (LET) that induce unique spatial distributions of DNA damage in cell nuclei that result in a greater biological effect than sparsely ionizing radiation like X-rays. HZE ions are a prominent component of galactic cosmic ray exposure during human spaceflight and specific ions are being used for radiotherapy. Here, we model DNA damage clustering at sub-micrometer scale to predict cell survival. The model is in good agreement with experimental data for a broad range of LET. Notably, the model indicates that nuclear geometry and ion beam orientation affect DNA damage clustering, which reveals their possible role in mediating cell radiosensitivity.
Collapse
Affiliation(s)
- Floriane Poignant
- Analytical Mechanics Associates Inc., 21 Enterprise Parkway, Hampton, VA 23666, United States
| | - Eloise Pariset
- NASA Ames Research Center, MS:288/2, Mountain View, CA 94035, United States
- Universities Space Research Association, 615 National Avenue, Mountain View, CA 94043, United States
| | - Ianik Plante
- KBR, 2400 NASA Parkway, Houston, TX 77058, United States
| | | | - Trevor Evain
- Life Sciences Division, Lawrence Berkeley National Laboratory, 717 Potter Street, Berkeley, CA 94720, United States
| | - Louise Viger
- Life Sciences Division, Lawrence Berkeley National Laboratory, 717 Potter Street, Berkeley, CA 94720, United States
| | - Tony C Slaba
- NASA Langley Research Center, 1 Nasa Drive, Hampton, VA 23666, United States
| | - Steve R Blattnig
- NASA Langley Research Center, 1 Nasa Drive, Hampton, VA 23666, United States
| | - Sylvain V Costes
- NASA Ames Research Center, MS:288/2, Mountain View, CA 94035, United States
| |
Collapse
|
6
|
Scherthan H, Geiger B, Ridinger D, Müller J, Riccobono D, Bestvater F, Port M, Hausmann M. Nano-Architecture of Persistent Focal DNA Damage Regions in the Minipig Epidermis Weeks after Acute γ-Irradiation. Biomolecules 2023; 13:1518. [PMID: 37892200 PMCID: PMC10605239 DOI: 10.3390/biom13101518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/22/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
Exposure to high acute doses of ionizing radiation (IR) can induce cutaneous radiation syndrome. Weeks after such radiation insults, keratinocyte nuclei of the epidermis exhibit persisting genomic lesions that present as focal accumulations of DNA double-strand break (DSB) damage marker proteins. Knowledge about the nanostructure of these genomic lesions is scarce. Here, we compared the chromatin nano-architecture with respect to DNA damage response (DDR) factors in persistent genomic DNA damage regions and healthy chromatin in epidermis sections of two minipigs 28 days after lumbar irradiation with ~50 Gy γ-rays, using single-molecule localization microscopy (SMLM) combined with geometric and topological mathematical analyses. SMLM analysis of fluorochrome-stained paraffin sections revealed, within keratinocyte nuclei with perisitent DNA damage, the nano-arrangements of pATM, 53BP1 and Mre11 DDR proteins in γ-H2AX-positive focal chromatin areas (termed macro-foci). It was found that persistent macro-foci contained on average ~70% of 53BP1, ~23% of MRE11 and ~25% of pATM single molecule signals of a nucleus. MRE11 and pATM fluorescent tags were organized in focal nanoclusters peaking at about 40 nm diameter, while 53BP1 tags formed nanoclusters that made up super-foci of about 300 nm in size. Relative to undamaged nuclear chromatin, the enrichment of DDR protein signal tags in γ-H2AX macro-foci was on average 8.7-fold (±3) for 53BP1, 3.4-fold (±1.3) for MRE11 and 3.6-fold (±1.8) for pATM. The persistent macro-foci of minipig epidermis displayed a ~2-fold enrichment of DDR proteins, relative to DSB foci of lymphoblastoid control cells 30 min after 0.5 Gy X-ray exposure. A lasting accumulation of damage signaling and sensing molecules such as pATM and 53BP1, as well as the DSB end-processing protein MRE11 in the persistent macro-foci suggests the presence of diverse DNA damages which pose an insurmountable problem for DSB repair.
Collapse
Affiliation(s)
- Harry Scherthan
- Bundeswehr Institute for Radiobiology Affiliated to the University of Ulm, Neuherbergstr. 11, D-80937 München, Germany (M.P.)
| | - Beatrice Geiger
- Kirchhoff-Institute for Physics, Heidelberg University, Im Neuenheimer Feld 227, D-69120 Heidelberg, Germany (D.R.)
| | - David Ridinger
- Kirchhoff-Institute for Physics, Heidelberg University, Im Neuenheimer Feld 227, D-69120 Heidelberg, Germany (D.R.)
| | - Jessica Müller
- Bundeswehr Institute for Radiobiology Affiliated to the University of Ulm, Neuherbergstr. 11, D-80937 München, Germany (M.P.)
| | - Diane Riccobono
- Département des Effets Biologiques des Rayonnements, French Armed Forces Biomedical Research Institute, UMR 1296, BP 73, 91223 Brétigny-sur-Orge, France;
| | - Felix Bestvater
- Core Facility Light Microscopy, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany;
| | - Matthias Port
- Bundeswehr Institute for Radiobiology Affiliated to the University of Ulm, Neuherbergstr. 11, D-80937 München, Germany (M.P.)
| | - Michael Hausmann
- Kirchhoff-Institute for Physics, Heidelberg University, Im Neuenheimer Feld 227, D-69120 Heidelberg, Germany (D.R.)
| |
Collapse
|
7
|
Grissi C, Taverna Porro M, Perona M, Atia M, Negrin L, Moreno MS, Sacanell J, Olivera MS, Del Grosso M, Durán H, Ibañez IL. Superparamagnetic iron oxide nanoparticles induce persistent large foci of DNA damage in human melanoma cells post-irradiation. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2023:10.1007/s00411-023-01037-0. [PMID: 37452828 DOI: 10.1007/s00411-023-01037-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 06/25/2023] [Indexed: 07/18/2023]
Abstract
The synergy of superparamagnetic iron oxide nanoparticles (SPIONs) and ionizing radiation (IR), attributed to reactive oxygen species (ROS) and DNA double-strand breaks (DSBs) increase, was widely investigated in different cancers, but scarcely in melanoma. Herein, SPIONs were evaluated as radiosensitizers in A-375 human melanoma cells. Moreover, the effect of the combined treatment of SPIONs and gamma irradiation (SPIONs-IR) was assessed at the DNA level, where DSBs induction and their repair capacity were studied. SPIONs were synthesized, stabilized by poly(ethylene glycol) methyl ether and physicochemically characterized by high resolution-transmission electron microscopy (HR-TEM), X-ray diffraction and magnetometry and dynamic light scattering. The obtained nanoparticles showing superparamagnetic behavior and low dispersion in shape and sizes were tested in A-375 cells. The intracellular internalization of SPIONs was verified by HR-TEM and quantified by inductively coupled plasma atomic emission spectroscopy. Cells treated with SPIONs exhibited high ROS levels without associated cytotoxicity. Next, a significant radiosensitization in SPIONs-IR vs. control (IR) cells was demonstrated at 1 Gy of gamma radiation. Furthermore, a decreased DSBs repair capacity in SPIONs-IR vs. IR-treated cells was evidenced by the size increase of persistent phosphorylated H2AX foci at 24 h post-irradiation. In conclusion, these nanoparticles show the potential to radiosensitize melanoma cells by the induction of unrepairable DNA damage.
Collapse
Affiliation(s)
- Cecilia Grissi
- Subgerencia de Tecnología y Aplicaciones de Aceleradores, Gerencia de Investigación y Aplicaciones, Comisión Nacional de Energía Atómica (CNEA), Instituto de Nanociencia y Nanotecnología (INN), CNEA - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Nodo Constituyentes, Av. General Paz, 1499 (B1650KNA), San Martín, Buenos Aires, Argentina
| | - Marisa Taverna Porro
- Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) - Universidad de Buenos Aires, Junín 954 (C1113AAD), Ciudad Autónoma de Buenos Aires, Argentina
- Escuela de Ciencia y Tecnología, Universidad Nacional de San Martín, Campus Miguelete (B1650KNA), San Martín, Provincia de Buenos Aires, Argentina
| | - Marina Perona
- División Bioquímica Nuclear, Departamento de Radiobiología, Comisión Nacional de Energía Atómica (CNEA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Av. General Paz 1499 (B1650KNA), San Martín, Buenos Aires, Argentina
| | - Mariel Atia
- Subgerencia de Tecnología y Aplicaciones de Aceleradores, Gerencia de Investigación y Aplicaciones, Comisión Nacional de Energía Atómica (CNEA), Instituto de Nanociencia y Nanotecnología (INN), CNEA - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Nodo Constituyentes, Av. General Paz, 1499 (B1650KNA), San Martín, Buenos Aires, Argentina
| | - Lara Negrin
- Laboratorio de Radiobiología y Biodosimetría, Centro Atómico Bariloche, Comisión Nacional de Energía Atómica (CNEA), Centro de Medicina Nuclear y Radioterapia - Instituto de Tecnologías Nucleares Para La Salud (INTECNUS), Av. Bustillo Km. 9,5 (R8402AGP), S.C. de Bariloche, Río Negro, Argentina
| | - M Sergio Moreno
- Instituto de Nanociencia y Nanotecnología (INN), Comisión Nacional de Energía Atómica (CNEA) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Nodo Bariloche, Centro Atómico Bariloche, Av. Bustillo Km. 9,5 (R8402AGP), S.C. de Bariloche, Río Negro, Argentina
| | - Joaquín Sacanell
- Departamento de Física de la Materia Condensada, Comisión Nacional de Energía Atómica (CNEA), Instituto de Nanociencia y Nanotecnología (INN), CNEA - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Nodo Constituyentes, Av. General Paz 1499 (B1650KNA), San Martín, Buenos Aires, Argentina
| | - María Silvina Olivera
- Departamento Coordinación BNCT, Comisión Nacional de Energía Atómica (CNEA), Centro Atómico Constituyentes, Av. General Paz 1499 (B1650KNA), San Martín, Buenos Aires, Argentina
| | - Mariela Del Grosso
- Subgerencia de Tecnología y Aplicaciones de Aceleradores, Gerencia de Investigación y Aplicaciones, Comisión Nacional de Energía Atómica (CNEA), Instituto de Nanociencia y Nanotecnología (INN), CNEA - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Nodo Constituyentes, Av. General Paz, 1499 (B1650KNA), San Martín, Buenos Aires, Argentina
| | - Hebe Durán
- Subgerencia de Tecnología y Aplicaciones de Aceleradores, Gerencia de Investigación y Aplicaciones, Comisión Nacional de Energía Atómica (CNEA), Instituto de Nanociencia y Nanotecnología (INN), CNEA - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Nodo Constituyentes, Av. General Paz, 1499 (B1650KNA), San Martín, Buenos Aires, Argentina.
- Escuela de Ciencia y Tecnología, Universidad Nacional de San Martín, Campus Miguelete (B1650KNA), San Martín, Provincia de Buenos Aires, Argentina.
| | - Irene L Ibañez
- Subgerencia de Tecnología y Aplicaciones de Aceleradores, Gerencia de Investigación y Aplicaciones, Comisión Nacional de Energía Atómica (CNEA), Instituto de Nanociencia y Nanotecnología (INN), CNEA - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Nodo Constituyentes, Av. General Paz, 1499 (B1650KNA), San Martín, Buenos Aires, Argentina.
| |
Collapse
|
8
|
Chaudhary P, Milluzzo G, McIlvenny A, Ahmed H, McMurray A, Maiorino C, Polin K, Romagnani L, Doria D, McMahon SJ, Botchway SW, Rajeev PP, Prise KM, Borghesi M. Cellular irradiations with laser-driven carbon ions at ultra-high dose rates. Phys Med Biol 2023; 68. [PMID: 36625355 DOI: 10.1088/1361-6560/aca387] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 11/16/2022] [Indexed: 01/11/2023]
Abstract
Objective.Carbon is an ion species of significant radiobiological interest, particularly in view of its use in cancer radiotherapy, where its large Relative Biological Efficiency is often exploited to overcome radio resistance. A growing interest in highly pulsed carbon delivery has arisen in the context of the development of the FLASH radiotherapy approach, with recent studies carried out at dose rates of 40 Gy s-1. Laser acceleration methods, producing ultrashort ion bursts, can now enable the delivery of Gy-level doses of carbon ions at ultra-high dose rates (UHDRs), exceeding 109Gy s-1. While studies at such extreme dose rate have been carried out so far using low LET particles such as electrons and protons, the radiobiology of high-LET, UHDR ions has not yet been explored. Here, we report the first application of laser-accelerated carbon ions generated by focussing 1020W cm-2intense lasers on 10-25 nm carbon targets, to irradiate radioresistant patient-derived Glioblastoma stem like cells (GSCs).Approach.We exposed GSCs to 1 Gy of 9.5 ± 0.5 MeV/n carbon ions delivered in a single ultra-short (∼400-picosecond) pulse, at a dose rate of 2 × 109Gy s-1, generated using the ASTRA GEMINI laser of the Central Laser Facility at the Rutherford Appleton Laboratory, Didcot, Oxfordshire, UK. We quantified carbon ion-induced DNA double strand break (DSB) damage using the 53BP1 foci formation assay and used 225 kVp x-rays as a reference radiation.Main Results.Laser-accelerated carbon ions induced complex DNA DSB damage, as seen through persistent 53BP1 foci (11.5 ± 0.4 foci/cell/Gy) at 24 h and significantly larger foci (1.69 ± 0.07μm2) than x-rays induced ones (0.63 ± 0.02μm2). The relative foci induction value for laser-driven carbon ions relative to conventional x-rays was 3.2 ± 0.3 at 24 h post-irradiation also confirming the complex nature of the induced damage.Significance.Our study demonstrates the feasibility of radiobiology investigations at unprecedented dose rates using laser-accelerated high-LET carbon ions in clinically relevant models.
Collapse
Affiliation(s)
- Pankaj Chaudhary
- The Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Lisburn Road, Belfast, BT9 7AE, Northern Ireland, United Kingdom.,Centre for Light-Matter Interactions, School of Mathematics and Physics, Queen's University Belfast, BT7 1NN, Northern Ireland, United Kingdom
| | - Giuliana Milluzzo
- Centre for Light-Matter Interactions, School of Mathematics and Physics, Queen's University Belfast, BT7 1NN, Northern Ireland, United Kingdom.,Laboratori Nazionali del Sud, Istituto Nazionale di Fisica Nucleare,, via S Sofia 62, I-95123 Catania, Sicily, Italy
| | - Aodhan McIlvenny
- Centre for Light-Matter Interactions, School of Mathematics and Physics, Queen's University Belfast, BT7 1NN, Northern Ireland, United Kingdom
| | - Hamad Ahmed
- Centre for Light-Matter Interactions, School of Mathematics and Physics, Queen's University Belfast, BT7 1NN, Northern Ireland, United Kingdom.,Experimental Science Group, Central Laser Facility, Rutherford Appleton Laboratory, Didcot, Oxford, OX11 0QX, England, United Kingdom
| | - Aaron McMurray
- Centre for Light-Matter Interactions, School of Mathematics and Physics, Queen's University Belfast, BT7 1NN, Northern Ireland, United Kingdom
| | - Carla Maiorino
- The Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Lisburn Road, Belfast, BT9 7AE, Northern Ireland, United Kingdom.,Laboratori Nazionali del Sud, Istituto Nazionale di Fisica Nucleare,, via S Sofia 62, I-95123 Catania, Sicily, Italy.,Extreme Light Infrastructure (ELI-NP) and Horia Hulubei National Institute for R & D in Physics and Nuclear Engineering (IFIN-HH), Str. Reactorului No. 30, 077125 Bucharest, Magurele, Romania.,University College Cork, College of Medicine and Health, Discipline of Diagnostic Radiography and Radiation Therapy, Brookfield Health Sciences Complex, Brookfield College Road, T12AK54, Cork, United Kingdom
| | - Kathryn Polin
- Centre for Light-Matter Interactions, School of Mathematics and Physics, Queen's University Belfast, BT7 1NN, Northern Ireland, United Kingdom
| | - Lorenzo Romagnani
- Centre for Light-Matter Interactions, School of Mathematics and Physics, Queen's University Belfast, BT7 1NN, Northern Ireland, United Kingdom.,Laboratoire LULI, École Polytechnique, Route de Saclay, F-91128 Palaiseau, Paris, France
| | - Domenico Doria
- Centre for Light-Matter Interactions, School of Mathematics and Physics, Queen's University Belfast, BT7 1NN, Northern Ireland, United Kingdom.,Extreme Light Infrastructure (ELI-NP) and Horia Hulubei National Institute for R & D in Physics and Nuclear Engineering (IFIN-HH), Str. Reactorului No. 30, 077125 Bucharest, Magurele, Romania
| | - Stephen J McMahon
- The Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Lisburn Road, Belfast, BT9 7AE, Northern Ireland, United Kingdom
| | - Stanley W Botchway
- Research Complex at Harwell & Central Laser facility, Rutherford Appleton Laboratory, Didcot, Oxford, OX11 0QX, England, United Kingdom
| | - Pattathil P Rajeev
- Experimental Science Group, Central Laser Facility, Rutherford Appleton Laboratory, Didcot, Oxford, OX11 0QX, England, United Kingdom
| | - Kevin M Prise
- The Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Lisburn Road, Belfast, BT9 7AE, Northern Ireland, United Kingdom
| | - Marco Borghesi
- Centre for Light-Matter Interactions, School of Mathematics and Physics, Queen's University Belfast, BT7 1NN, Northern Ireland, United Kingdom
| |
Collapse
|
9
|
Abramenkovs A, Hariri M, Spiegelberg D, Nilsson S, Stenerlöw B. Ra-223 induces clustered DNA damage and inhibits cell survival in several prostate cancer cell lines. Transl Oncol 2022; 26:101543. [PMID: 36126563 PMCID: PMC9489499 DOI: 10.1016/j.tranon.2022.101543] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 09/05/2022] [Accepted: 09/13/2022] [Indexed: 10/24/2022] Open
Abstract
The bone-seeking radiopharmaceutical Xofigo (Radium-223 dichloride) has demonstrated both extended survival and palliative effects in treatment of bone metastases in prostate cancer. The alpha-particle emitter Ra-223, targets regions undergoing active bone remodeling and strongly binds to bone hydroxyapatite (HAp). However, the toxicity mechanism and properties of Ra-223 binding to hydroxyapatite are not fully understood. By exposing 2D and 3D (spheroid) prostate cancer cell models to free and HAp-bound Ra-223 we here studied cell toxicity, apoptosis and formation and repair of DNA double-strand breaks (DSBs). The rapid binding with a high affinity of Ra-223 to bone-like HAp structures was evident (KD= 19.2 × 10-18 M) and almost no dissociation was detected within 24 h. Importantly, there was no significant uptake of Ra-223 in cells. The Ra-223 alpha-particle decay produced track-like distributions of the DNA damage response proteins 53BP1 and ɣH2AX induced high amounts of clustered DSBs in prostate cancer cells and activated DSB repair through non-homologous end-joining (NHEJ). Ra-223 inhibited growth of prostate cancer cells, independent of cell type, and induced high levels of apoptosis. In summary, we suggest the high cell killing efficacy of the Ra-223 was attributed to the clustered DNA damaged sites induced by α-particles.
Collapse
Affiliation(s)
- Andris Abramenkovs
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala SE-75185, Sweden
| | - Mehran Hariri
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala SE-75185, Sweden.
| | - Diana Spiegelberg
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala SE-75185, Sweden; Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Sten Nilsson
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - Bo Stenerlöw
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala SE-75185, Sweden.
| |
Collapse
|
10
|
Abu Shqair A, Lee US, Kim EH. Computational modelling of γ-H2AX foci formation in human cells induced by alpha particle exposure. Sci Rep 2022; 12:14360. [PMID: 35999233 PMCID: PMC9399106 DOI: 10.1038/s41598-022-17830-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 08/01/2022] [Indexed: 11/09/2022] Open
Abstract
In cellular experiments, radiation-induced DNA damage can be quantified by counting the number of γ-H2AX foci in cell nucleus by using an immunofluorescence microscope. Quantification of DNA damage carries uncertainty, not only due to lack of full understanding the biological processes but also limitations in measurement techniques. The causes of limited certainty include the possibility of expressing foci in varying sizes responding individual DSBs and the overlapping of foci on the two-dimensional (2D) immunofluorescence microscopy image of γ-H2AX foci, especially when produced due to high-LET radiation exposure. There have been discussions on those limitations, but no successful studies to overcome them. In this paper, a practical modelling has been developed to simulate the occurrences of double-strand breaks (DSBs) and the formations of γ-H2AX foci in response to individual DSB formations, in cell nucleus due to exposure to alpha particles. Cell irradiation and DSB production were simulated using a user-written code that utilizes Geant4-DNA physics models. A C + + code was used to simulate the formation γ-H2AX foci, which were spatially correlated to the loci of DBSs, and to calculate the number of individual foci from the observed 2D image of the cell nucleus containing the overlapping γ-H2AX foci. The average size of focal images was larger from alpha particle exposure than that from X-ray exposure, whereas the number of separate focal images were comparable except at doses up to 0.5 Gy. About 40% of separate focal images consisted of overlapping γ-H2AX foci at 1 Gy of alpha particle exposure. The foci overlapping ratios were obtained by simulation for individual size groups of focal images at varying doses. The size distributions of foci at varying doses were determined with experimentally obtained separate focal images. The correction factor for foci number was calculated using the foci overlapping ratio and foci size distribution, which are specific to dose from alpha particle exposure. The number of individual foci formations induced by applying the correction factor to the experimentally observed number of focal images better reflected the quality of alpha particles in causing DNA damage. Consequently, the conventional γ-H2AX assay can be better implemented by employing this computational modelling of γ-H2AX foci formation.
Collapse
Affiliation(s)
- Ali Abu Shqair
- Department of Nuclear Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ui-Seob Lee
- Department of Nuclear Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Eun-Hee Kim
- Department of Nuclear Engineering, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
11
|
Geometrical Properties of the Nucleus and Chromosome Intermingling Are Possible Major Parameters of Chromosome Aberration Formation. Int J Mol Sci 2022; 23:ijms23158638. [PMID: 35955776 PMCID: PMC9368922 DOI: 10.3390/ijms23158638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/28/2022] [Accepted: 07/30/2022] [Indexed: 12/10/2022] Open
Abstract
Ionizing radiation causes chromosome aberrations, which are possible biomarkers to assess space radiation cancer risks. Using the Monte Carlo codes Relativistic Ion Tracks (RITRACKS) and Radiation-Induced Tracks, Chromosome Aberrations, Repair and Damage (RITCARD), we investigated how geometrical properties of the cell nucleus, irradiated with ion beams of linear energy transfer (LET) ranging from 0.22 keV/μm to 195 keV/μm, influence the yield of simple and complex exchanges. We focused on the effect of (1) nuclear volume by considering spherical nuclei of varying radii; (2) nuclear shape by considering ellipsoidal nuclei of varying thicknesses; (3) beam orientation; and (4) chromosome intermingling by constraining or not constraining chromosomes in non-overlapping domains. In general, small nuclear volumes yield a higher number of complex exchanges, as compared to larger nuclear volumes, and a higher number of simple exchanges for LET < 40 keV/μm. Nuclear flattening reduces complex exchanges for high-LET beams when irradiated along the flattened axis. The beam orientation also affects yields for ellipsoidal nuclei. Reducing chromosome intermingling decreases both simple and complex exchanges. Our results suggest that the beam orientation, the geometry of the cell nucleus, and the organization of the chromosomes within are important parameters for the formation of aberrations that must be considered to model and translate in vitro results to in vivo risks.
Collapse
|
12
|
Repair of α-particle-induced DNA damage in peripheral blood mononuclear cells after internal ex vivo irradiation with 223Ra. Eur J Nucl Med Mol Imaging 2022; 49:3981-3988. [PMID: 35759008 PMCID: PMC9525426 DOI: 10.1007/s00259-022-05860-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/30/2022] [Indexed: 11/10/2022]
Abstract
Purpose As α-emitters for radiopharmaceutical therapies are administered systemically by intravenous injection, blood will be irradiated by α-particles that induce clustered DNA double-strand breaks (DSBs). Here, we investigated the induction and repair of DSB damage in peripheral blood mononuclear cells (PBMCs) as a function of the absorbed dose to the blood following internal ex vivo irradiation with [223Ra]RaCl2. Methods Blood samples of ten volunteers were irradiated by adding [223Ra]RaCl2 solution with different activity concentrations resulting in absorbed doses to the blood of 3 mGy, 25 mGy, 50 mGy and 100 mGy. PBMCs were isolated, divided in three parts and either fixed directly (d-samples) or after 4 h or 24 h culture. After immunostaining, the induced γ-H2AX α-tracks were counted. The time-dependent decrease in α-track frequency was described with a model assuming a repair rate R and a fraction of non-repairable damage Q. Results For 25 mGy, 50 mGy and 100 mGy, the numbers of α-tracks were significantly increased compared to baseline at all time points. Compared to the corresponding d-samples, the α-track frequency decreased significantly after 4 h and after 24 h. The repair rates R were (0.24 ± 0.05) h−1 for 25 mGy, (0.16 ± 0.04) h−1 for 50 mGy and (0.13 ± 0.02) h−1 for 100 mGy, suggesting faster repair at lower absorbed doses, while Q-values were similar. Conclusion The results obtained suggest that induction and repair of the DSB damage depend on the absorbed dose to the blood. Repair rates were similar to what has been observed for irradiation with low linear energy transfer.
Collapse
|
13
|
A deep learning model (FociRad) for automated detection of γ-H2AX foci and radiation dose estimation. Sci Rep 2022; 12:5527. [PMID: 35365702 PMCID: PMC8975967 DOI: 10.1038/s41598-022-09180-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 03/18/2022] [Indexed: 11/08/2022] Open
Abstract
DNA double-strand breaks (DSBs) are the most lethal form of damage to cells from irradiation. γ-H2AX (phosphorylated form of H2AX histone variant) has become one of the most reliable and sensitive biomarkers of DNA DSBs. However, the γ-H2AX foci assay still has limitations in the time consumed for manual scoring and possible variability between scorers. This study proposed a novel automated foci scoring method using a deep convolutional neural network based on a You-Only-Look-Once (YOLO) algorithm to quantify γ-H2AX foci in peripheral blood samples. FociRad, a two-stage deep learning approach, consisted of mononuclear cell (MNC) and γ-H2AX foci detections. Whole blood samples were irradiated with X-rays from a 6 MV linear accelerator at 1, 2, 4 or 6 Gy. Images were captured using confocal microscopy. Then, dose-response calibration curves were established and implemented with unseen dataset. The results of the FociRad model were comparable with manual scoring. MNC detection yielded 96.6% accuracy, 96.7% sensitivity and 96.5% specificity. γ-H2AX foci detection showed very good F1 scores (> 0.9). Implementation of calibration curve in the range of 0-4 Gy gave mean absolute difference of estimated doses less than 1 Gy compared to actual doses. In addition, the evaluation times of FociRad were very short (< 0.5 min per 100 images), while the time for manual scoring increased with the number of foci. In conclusion, FociRad was the first automated foci scoring method to use a YOLO algorithm with high detection performance and fast evaluation time, which opens the door for large-scale applications in radiation triage.
Collapse
|
14
|
Characterization of γ-H2AX foci formation under alpha particle and X-ray exposures for dose estimation. Sci Rep 2022; 12:3761. [PMID: 35260639 PMCID: PMC8904799 DOI: 10.1038/s41598-022-07653-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 02/16/2022] [Indexed: 12/04/2022] Open
Abstract
DNA double-strand break (DSB) induction is one of the phenotypes of cellular damage from radiation exposure and is commonly quantified by γ-H2AX assay with the number of excess fluorescent foci per cell as the main component. However, the number of foci alone may not fully characterize the state of DNA damage following exposures to different radiation qualities. This study investigated the feasibility of utilizing the focus size distribution and dephosphorylation rate of γ-H2AX to identify the type of causative radiation and dose. Human lung epithelial cells and mouse vascular endothelial cells were used to observe the expression changes of γ-H2AX foci due to alpha particle and X-ray exposures. Results showed that the average number of excess foci per cell linearly increased with the dose. The focus size distribution showed a consistent pattern depending on the causative radiation type. Three criteria for the identification of causative radiation type were derived from experimental focus size distributions and were validated in blind testing with correct identification of 27 out of 32 samples. The dose could be estimated based on the proportionality constant specific to the identified radiation type with a difference of less than 15% from the actual value. The different dephosphorylation rates of γ-H2AX produced from alpha particle and X-ray exposures were effectively utilized to determine the individual dose contributions of alpha particles and X-rays under mixed beam exposure. Individual doses were estimated to have differences of less than ~ 12% from actual values.
Collapse
|
15
|
Singh G, Batzenschlager M, Tomkova D, Herzog E, Hoffmann E, Houlné G, Schmit AC, Berr A, Chabouté ME. GIP1 and GIP2 Contribute to the Maintenance of Genome Stability at the Nuclear Periphery. FRONTIERS IN PLANT SCIENCE 2022; 12:804928. [PMID: 35154196 PMCID: PMC8830487 DOI: 10.3389/fpls.2021.804928] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/20/2021] [Indexed: 06/02/2023]
Abstract
The maintenance of genetic information is important in eukaryotes notably through mechanisms occurring at the nuclear periphery where inner nuclear membrane proteins and nuclear pore-associated components are key factors regulating the DNA damage response (DDR). However, this aspect of DDR regulation is still poorly documented in plants. We addressed here how genomic stability is impaired in the gamma-tubulin complex component 3-interacting protein (gip1gip2) double mutants showing defective nuclear shaping. Using neutral comet assays for DNA double-strand breaks (DSBs) detection, we showed that GIP1 and GIP2 act redundantly to maintain genome stability. At the cellular level, γ-H2AX foci in gip1gip2 were more abundant and heterogeneous in their size compared to wild-type (WT) in root meristematic nuclei, indicative of constitutive DNA damage. This was linked to a constitutive activation of the DDR in the gip1gip2 mutant, with more emphasis on the homologous recombination (HR) repair pathway. In addition, we noticed the presence of numerous RAD51 foci which did not colocalize with γ-H2AX foci. The expression of GIP1-GFP in the double mutant rescued the cellular response to DNA damage, leading to the systematic colocalization of RAD51 and γ-H2AX foci. Interestingly, a significant proportion of RAD51 foci colocalized with GIP1-GFP at the nuclear periphery. Altogether, our data suggest that GIPs may partly contribute to the spatio-temporal recruitment of RAD51 at the nuclear periphery.
Collapse
Affiliation(s)
- Gaurav Singh
- Institut de Biologie Moléculaire des Plantes, CNRS, Université de Strasbourg, Strasbourg, France
| | | | - Denisa Tomkova
- Institut de Biologie Moléculaire des Plantes, CNRS, Université de Strasbourg, Strasbourg, France
| | - Etienne Herzog
- Institut de Biologie Moléculaire des Plantes, CNRS, Université de Strasbourg, Strasbourg, France
| | - Elise Hoffmann
- Institut de Biologie Moléculaire des Plantes, CNRS, Université de Strasbourg, Strasbourg, France
| | - Guy Houlné
- Institut de Biologie Moléculaire des Plantes, CNRS, Université de Strasbourg, Strasbourg, France
| | - Anne-Catherine Schmit
- Institut de Biologie Moléculaire des Plantes, CNRS, Université de Strasbourg, Strasbourg, France
| | - Alexandre Berr
- Institut de Biologie Moléculaire des Plantes, CNRS, Université de Strasbourg, Strasbourg, France
| | - Marie-Edith Chabouté
- Institut de Biologie Moléculaire des Plantes, CNRS, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
16
|
Penninckx S, Pariset E, Cekanaviciute E, Costes SV. Quantification of radiation-induced DNA double strand break repair foci to evaluate and predict biological responses to ionizing radiation. NAR Cancer 2021; 3:zcab046. [PMID: 35692378 PMCID: PMC8693576 DOI: 10.1093/narcan/zcab046] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 11/08/2021] [Accepted: 12/17/2021] [Indexed: 08/08/2023] Open
Abstract
Radiation-induced foci (RIF) are nuclear puncta visualized by immunostaining of proteins that regulate DNA double-strand break (DSB) repair after exposure to ionizing radiation. RIF are a standard metric for measuring DSB formation and repair in clinical, environmental and space radiobiology. The time course and dose dependence of their formation has great potential to predict in vivo responses to ionizing radiation, predisposition to cancer and probability of adverse reactions to radiotherapy. However, increasing complexity of experimentally and therapeutically setups (charged particle, FLASH …) is associated with several confounding factors that must be taken into account when interpreting RIF values. In this review, we discuss the spatiotemporal characteristics of RIF development after irradiation, addressing the common confounding factors, including cell proliferation and foci merging. We also describe the relevant endpoints and mathematical models that enable accurate biological interpretation of RIF formation and resolution. Finally, we discuss the use of RIF as a biomarker for quantification and prediction of in vivo radiation responses, including important caveats relating to the choice of the biological endpoint and the detection method. This review intends to help scientific community design radiobiology experiments using RIF as a key metric and to provide suggestions for their biological interpretation.
Collapse
Affiliation(s)
- Sébastien Penninckx
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
- Medical Physics Department, Jules Bordet Institute, Université Libre de Bruxelles, 1 Rue Héger-Bordet, 1000 Brussels, Belgium
| | - Eloise Pariset
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
- Universities Space Research Association, 615 National Avenue, Mountain View, CA 94043, USA
| | - Egle Cekanaviciute
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Sylvain V Costes
- To whom correspondence should be addressed. Tel: +1 650 604 5343;
| |
Collapse
|
17
|
Tabocchini MA. A forty-year journey from "classical" biophysics and radiobiology to hadrontherapy, space radiation and low dose rate underground radiobiology. Int J Radiat Biol 2021; 98:383-394. [PMID: 34259611 DOI: 10.1080/09553002.2021.1948142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
PURPOSE As a biologist who, since the beginning of her involvement in science, has collaborated closely with physicists, I want to share my forty years of experience describing the events that introduced me to the world of charged particle radiation biology as well as that of low doses/dose rates, with related implications in medicine and radiation protection. CONCLUSION The main features of my experience can be summarized in the development of an interdisciplinary culture and in the interest in technological advances for the study of biological responses to radiation in different scenarios, relevant for public health. Mine was a journey that began by chance, but which led me to a world that proved to be of great interest to me. With the current advances in science, the new generations of scientists have new opportunities that I wish them to face with the same interest and enthusiasm that I felt for such an interdisciplinary field as that of radiation biology.
Collapse
Affiliation(s)
- Maria Antonella Tabocchini
- Istituto Nazionale di Fisica Nucleare (INFN), Rome, Italy.,Formerly: Istituto Superiore di Sanità (ISS), National Center for Innovative Technologies in Public Health, Rome, Italy
| |
Collapse
|
18
|
Rad-Bio-App: a discovery environment for biologists to explore spaceflight-related radiation exposures. NPJ Microgravity 2021; 7:15. [PMID: 33976230 PMCID: PMC8113475 DOI: 10.1038/s41526-021-00143-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 03/29/2021] [Indexed: 02/07/2023] Open
Abstract
In addition to microgravity, spaceflight simultaneously exposes biology to a suite of other stimuli. For example, in space, organisms experience ionizing radiation environments that significantly differ in both quality and quantity from those normally experienced on Earth. However, data on radiation exposure during space missions is often complex to access and to understand, limiting progress towards defining how radiation affects organisms against the unique background of spaceflight. To help address this challenge, we have developed the Rad-Bio-App. This web-accessible database imports radiation metadata from experiments archived in NASA’s GeneLab data repository, and then allows the user to explore these experiments both in the context of their radiation exposure and through their other metadata and results. Rad-Bio-App provides an easy-to-use, graphically-driven environment to enable both radiation biologists and non-specialist researchers to visualize, and understand the impact of ionizing radiation on various biological systems in the context of spaceflight.
Collapse
|
19
|
Penninckx S, Pariset E, Acuna AU, Lucas S, Costes SV. Considering Cell Proliferation to Optimize Detection of Radiation-Induced 53BP1 Positive Foci in 15 Mouse Strains Ex Vivo. Radiat Res 2021; 195:47-59. [PMID: 33181852 DOI: 10.1667/rade-20-00165.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 10/05/2020] [Indexed: 11/03/2022]
Abstract
Due to high metabolic activity, proliferating cells continuously generate free radicals, which induce DNA double-strand breaks (DSB). Fluorescently tagged nuclear foci of DNA repair protein 53 binding protein-1 (53BP1) are used as a standard metric for measuring DSB formation at baseline and in response to environmental insults such as radiation. Here we demonstrate that the background level of spontaneous 53BP1+ foci formation can be modeled mathematically as a function of cell confluence, which is a metric of their proliferation rate. This model was validated using spontaneous 53BP1+ foci data from 72 cultures of primary skin fibroblasts derived from 15 different strains of mice, showing a ∼10-fold decrease from low to full confluence that is independent of mouse strain. On the other hand, the baseline level of spontaneous 53BP1+ foci in a fully confluent cell population was strain-dependent, suggesting genomic associations, and correlated with radiation sensitivity based on previous measurements in the same cell lines. Finally, we have developed an online open-access tool to correct for the effect of cell confluence on 53BP1+ foci-based quantification of DSB. This tool provides guidelines for the number of cells required to reach statistical significance for the detection of DSB induced by low doses of ionizing radiation as a function of confluence and time postirradiation.
Collapse
Affiliation(s)
- Sébastien Penninckx
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720.,Namur Research Institute for Life Science (NARILIS - LARN), University of Namur, 5000 Namur, Belgium
| | - Eloise Pariset
- Universities Space Research Association (USRA), Mountain View, California 94043.,Space Biosciences Division, NASA Ames Research Center, Mountain View, California 94035
| | - Ana Uriarte Acuna
- Space Biosciences Division, NASA Ames Research Center, Mountain View, California 94035.,Wyle Laboratories, KBR, Inc., Houston, Texas 77002
| | - Stephane Lucas
- Namur Research Institute for Life Science (NARILIS - LARN), University of Namur, 5000 Namur, Belgium
| | - Sylvain V Costes
- Space Biosciences Division, NASA Ames Research Center, Mountain View, California 94035
| |
Collapse
|
20
|
Zaharieva E, Sasatani M, Matsumoto R, Kamiya K. Formation of DNA Damage Foci in Human and Mouse Primary Fibroblasts Chronically Exposed to Gamma Radiation at 0.1 mGy/min. Radiat Res 2021; 196:40-54. [PMID: 33857310 DOI: 10.1667/rade-20-00059.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 03/11/2021] [Indexed: 11/03/2022]
Abstract
Low-dose-rate radiation exposures and their associated cancer risk are an important concern for radiation protection today. Nevertheless, there is almost no data concerning DNA damage at dose rates below 0.1 mGy/min. In this study, we investigated the formation of DNA damage repair foci under chronic low-dose-rate irradiation relative to acute high-dose-rate irradiation and assessed the magnitude of the dose-rate effect. Four human and four mouse normal fibroblast cell models from different organs were subjected to gamma irradiation at 0.096 mGy/min or 0.81 Gy/min, and dose-response curves were established for the dose range from 0.1 to 0.8 Gy. The results indicate that prolonged low-dose-rate exposures cause modestly increased levels of DNA repair foci, with a strongly supralinear dose-response relationship, where 40-70% of the radiation effect at 1 Gy was already present at the total dose of 0.1 Gy. Thus, compared to acute irradiation, low-dose-rate exposure was 6-9 times less efficient at a total dose of 0.1 Gy, and 10-20 times less efficient at 1 Gy. Comparison between cell models revealed a certain correlation between the presence of persistent, above-background foci at 48 h after irradiation and the sensitivity to low-dose-rate radiation, suggesting that repair capacity plays an important role in the cellular response to chronic irradiation. Given the findings reported here, we propose that establishing detailed dose-response curves and accounting for the repair rates of different cell models are essential steps in elucidating dose-rate effects.
Collapse
Affiliation(s)
- Elena Zaharieva
- Department of Experimental Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Megumi Sasatani
- Department of Experimental Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Ryoga Matsumoto
- Graduate School of Medicine, Hiroshima University, Hiroshima, Japan
| | - Kenji Kamiya
- Department of Experimental Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
21
|
Ventura JA, Donoghue JF, Nowell CJ, Cann LM, Day LRJ, Smyth LML, Forrester HB, Rogers PAW, Crosbie JC. The γH2AX DSB marker may not be a suitable biodosimeter to measure the biological MRT valley dose. Int J Radiat Biol 2021; 97:642-656. [PMID: 33617395 DOI: 10.1080/09553002.2021.1893854] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 12/31/2020] [Accepted: 02/11/2021] [Indexed: 10/22/2022]
Abstract
PURPOSE γH2AX biodosimetry has been proposed as an alternative dosimetry method for microbeam radiation therapy (MRT) because conventional dosimeters, such as ionization chambers, lack the spatial resolution required to accurately measure the MRT valley dose. Here we investigated whether γH2AX biodosimetry should be used to measure the biological valley dose of MRT-irradiated mammalian cells. MATERIALS AND METHODS We irradiated human skin fibroblasts and mouse skin flaps with synchrotron MRT and broad beam (BB) radiation. BB doses of 1-5 Gy were used to generate a calibration curve in order to estimate the biological MRT valley dose using the γH2AX assay. RESULTS Our key finding was that MRT induced a non-linear dose response compared to BB, where doses 2-3 times greater showed the same level of DNA DSB damage in the valley in cell and tissue studies. This indicates that γH2AX may not be an appropriate biodosimeter to estimate the biological valley doses of MRT-irradiated samples. We also established foci yields of 5.9 ± 0 . 04 and 27.4 ± 2 . 5 foci/cell/Gy in mouse skin tissue and human fibroblasts respectively, induced by BB. Using Monte Carlo simulations, a linear dose response was seen in cell and tissue studies and produced predicted peak-to-valley dose ratios (PVDRs) of ∼30 and ∼107 for human fibroblasts and mouse skin tissue respectively. CONCLUSIONS Our report highlights novel MRT radiobiology, attempts to explain why γH2AX may not be an appropriate biodosimeter and suggests further studies aimed at revealing the biological and cellular communication mechanisms that drive the normal tissue sparing effect, which is characteristic of MRT.
Collapse
Affiliation(s)
- Jessica A Ventura
- Department of Obstetrics and Gynaecology, Royal Women's Hospital, University of Melbourne, Parkville, Australia
| | - Jacqueline F Donoghue
- Department of Obstetrics and Gynaecology, Royal Women's Hospital, University of Melbourne, Parkville, Australia
| | - Cameron J Nowell
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Leonie M Cann
- Department of Obstetrics and Gynaecology, Royal Women's Hospital, University of Melbourne, Parkville, Australia
| | - Liam R J Day
- School of Science, RMIT University, Melbourne, Australia
| | - Lloyd M L Smyth
- Department of Obstetrics and Gynaecology, Royal Women's Hospital, University of Melbourne, Parkville, Australia
| | - Helen B Forrester
- School of Science, RMIT University, Melbourne, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Monash University, Clayton, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, Australia
| | - Peter A W Rogers
- Department of Obstetrics and Gynaecology, Royal Women's Hospital, University of Melbourne, Parkville, Australia
| | | |
Collapse
|
22
|
Clinical Progress in Proton Radiotherapy: Biological Unknowns. Cancers (Basel) 2021; 13:cancers13040604. [PMID: 33546432 PMCID: PMC7913745 DOI: 10.3390/cancers13040604] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/27/2021] [Accepted: 02/01/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Proton radiation therapy is a more recent type of radiotherapy that uses proton beams instead of classical photon or X-rays beams. The clinical benefit of proton therapy is that it allows to treat tumors more precisely. As a result, proton radiotherapy induces less toxicity to healthy tissue near the tumor site. Despite the experience in the clinical use of protons, the response of cells to proton radiation, the radiobiology, is less understood. In this review, we describe the current knowledge about proton radiobiology. Abstract Clinical use of proton radiation has massively increased over the past years. The main reason for this is the beneficial depth-dose distribution of protons that allows to reduce toxicity to normal tissues surrounding the tumor. Despite the experience in the clinical use of protons, the radiobiology after proton irradiation compared to photon irradiation remains to be completely elucidated. Proton radiation may lead to differential damages and activation of biological processes. Here, we will review the current knowledge of proton radiobiology in terms of induction of reactive oxygen species, hypoxia, DNA damage response, as well as cell death after proton irradiation and radioresistance.
Collapse
|
23
|
Berrios DC, Galazka J, Grigorev K, Gebre S, Costes SV. NASA GeneLab: interfaces for the exploration of space omics data. Nucleic Acids Res 2021; 49:D1515-D1522. [PMID: 33080015 PMCID: PMC7778922 DOI: 10.1093/nar/gkaa887] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/16/2020] [Accepted: 10/06/2020] [Indexed: 12/14/2022] Open
Abstract
The mission of NASA's GeneLab database (https://genelab.nasa.gov/) is to collect, curate, and provide access to the genomic, transcriptomic, proteomic and metabolomic (so-called 'omics') data from biospecimens flown in space or exposed to simulated space stressors, maximizing their utilization. This large collection of data enables the exploration of molecular network responses to space environments using a systems biology approach. We review here the various components of the GeneLab platform, including the new data repository web interface, and the GeneLab Online Data Entry (GEODE) web portal, which will support the expansion of the database in the future to include companion non-omics assay data. We discuss our design for GEODE, particularly how it promotes investigators providing more accurate metadata, reducing the curation effort required of GeneLab staff. We also introduce here a new GeneLab Application Programming Interface (API) specifically designed to support tools for the visualization of processed omics data. We review the outreach efforts by GeneLab to utilize the spaceflight data in the repository to generate novel discoveries and develop new hypotheses, including spearheading data analysis working groups, and a high school student training program. All these efforts are aimed ultimately at supporting precision risk management for human space exploration.
Collapse
Affiliation(s)
| | | | | | - Samrawit Gebre
- KBR/NASA Ames Research Center, Moffett Field, CA 94035, USA
| | | |
Collapse
|
24
|
Afshinnekoo E, Scott RT, MacKay MJ, Pariset E, Cekanaviciute E, Barker R, Gilroy S, Hassane D, Smith SM, Zwart SR, Nelman-Gonzalez M, Crucian BE, Ponomarev SA, Orlov OI, Shiba D, Muratani M, Yamamoto M, Richards SE, Vaishampayan PA, Meydan C, Foox J, Myrrhe J, Istasse E, Singh N, Venkateswaran K, Keune JA, Ray HE, Basner M, Miller J, Vitaterna MH, Taylor DM, Wallace D, Rubins K, Bailey SM, Grabham P, Costes SV, Mason CE, Beheshti A. Fundamental Biological Features of Spaceflight: Advancing the Field to Enable Deep-Space Exploration. Cell 2020; 183:1162-1184. [PMID: 33242416 PMCID: PMC8441988 DOI: 10.1016/j.cell.2020.10.050] [Citation(s) in RCA: 204] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 12/14/2022]
Abstract
Research on astronaut health and model organisms have revealed six features of spaceflight biology that guide our current understanding of fundamental molecular changes that occur during space travel. The features include oxidative stress, DNA damage, mitochondrial dysregulation, epigenetic changes (including gene regulation), telomere length alterations, and microbiome shifts. Here we review the known hazards of human spaceflight, how spaceflight affects living systems through these six fundamental features, and the associated health risks of space exploration. We also discuss the essential issues related to the health and safety of astronauts involved in future missions, especially planned long-duration and Martian missions.
Collapse
Affiliation(s)
- Ebrahim Afshinnekoo
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10021, USA; The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA; WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY 10021, USA
| | - Ryan T Scott
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Matthew J MacKay
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10021, USA; The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA; WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY 10021, USA
| | - Eloise Pariset
- Universities Space Research Association (USRA), Mountain View, CA 94043, USA; Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Egle Cekanaviciute
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Richard Barker
- Department of Botany, University of Wisconsin, Madison, WI 53706, USA
| | - Simon Gilroy
- Department of Botany, University of Wisconsin, Madison, WI 53706, USA
| | | | - Scott M Smith
- Human Health and Performance Directorate, NASA Johnson Space Center, Houston, TX 77058, USA
| | - Sara R Zwart
- Department of Preventive Medicine and Community Health, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Mayra Nelman-Gonzalez
- KBR, Human Health and Performance Directorate, NASA Johnson Space Center, Houston, TX 77058, USA
| | - Brian E Crucian
- Human Health and Performance Directorate, NASA Johnson Space Center, Houston, TX 77058, USA
| | - Sergey A Ponomarev
- Institute for the Biomedical Problems, Russian Academy of Sciences, 123007 Moscow, Russia
| | - Oleg I Orlov
- Institute for the Biomedical Problems, Russian Academy of Sciences, 123007 Moscow, Russia
| | - Dai Shiba
- JEM Utilization Center, Human Spaceflight Technology Directorate, Japan Aerospace Exploration Agency (JAXA), Ibaraki 305-8505, Japan
| | - Masafumi Muratani
- Transborder Medical Research Center, and Department of Genome Biology, Faculty of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Masayuki Yamamoto
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan; Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8573, Japan
| | - Stephanie E Richards
- Bionetics, NASA Kennedy Space Center, Kennedy Space Center, Merritt Island, FL 32899, USA
| | - Parag A Vaishampayan
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Cem Meydan
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10021, USA; The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA; WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY 10021, USA
| | - Jonathan Foox
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10021, USA; The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA; WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY 10021, USA
| | - Jacqueline Myrrhe
- European Space Agency, Research and Payloads Group, Data Exploitation and Utilisation Strategy Office, 2200 AG Noordwijk, the Netherlands
| | - Eric Istasse
- European Space Agency, Research and Payloads Group, Data Exploitation and Utilisation Strategy Office, 2200 AG Noordwijk, the Netherlands
| | - Nitin Singh
- Biotechnology and Planetary Protection Group, Jet Propulsion Laboratory, California Institute of Technology, Pasadena, CA 91109, USA
| | - Kasthuri Venkateswaran
- Biotechnology and Planetary Protection Group, Jet Propulsion Laboratory, California Institute of Technology, Pasadena, CA 91109, USA
| | - Jessica A Keune
- Space Medicine Operations Division, NASA Johnson Space Center, Houston, TX 77058, USA
| | - Hami E Ray
- ASRC Federal Space and Defense, Inc., Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Mathias Basner
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jack Miller
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA; Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Martha Hotz Vitaterna
- Center for Sleep and Circadian Biology, Northwestern University, Evanston, IL 60208, USA; Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Deanne M Taylor
- Department of Biomedical Informatics, The Children's Hospital of Philadelphia, PA 19104, USA; Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; The Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Douglas Wallace
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; The Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kathleen Rubins
- Astronaut Office, NASA Johnson Space Center, Houston, TX 77058, USA
| | - Susan M Bailey
- Department of Environmental & Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA.
| | - Peter Grabham
- Center for Radiological Research, Department of Oncology, College of Physicians and Surgeons, Columbia University, New York, NY 10027, USA.
| | - Sylvain V Costes
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA.
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10021, USA; The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA; WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY 10021, USA; The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, NY 10021, USA.
| | - Afshin Beheshti
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
25
|
DNA Damage Baseline Predicts Resilience to Space Radiation and Radiotherapy. Cell Rep 2020; 33:108434. [PMID: 33242409 PMCID: PMC7784531 DOI: 10.1016/j.celrep.2020.108434] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/22/2020] [Accepted: 11/04/2020] [Indexed: 12/11/2022] Open
Abstract
Deep space exploration will require real-time, minimally invasive monitoring of astronaut health to mitigate the potential health impairments caused by space radiation and microgravity. Genotoxic stress in humans can be monitored by quantifying the amount of DNA double-strand breaks (DSBs) in immune cells from a simple finger prick. In a cohort of 674 healthy donors, we show that the endogenous level of DSBs increases with age and with latent cytomegalovirus infection. To map the range of human responses to space radiation, we then study DSB induction and repair in immune cells from 319 healthy donors after the cells are exposed to galactic cosmic ray components and lymphocytes from 30 cancer patients after radiotherapy. Individuals with low baseline DSB have fewer clinical complications, enhanced DNA damage repair responses, and a functional dose-dependent cytokine response in healthy donor cells. This supports the use of DSB monitoring for health resilience in space.
Collapse
|
26
|
Pariset E, Penninckx S, Kerbaul CD, Guiet E, Macha AL, Cekanaviciute E, Snijders AM, Mao JH, Paris F, Costes SV. 53BP1 Repair Kinetics for Prediction of In Vivo Radiation Susceptibility in 15 Mouse Strains. Radiat Res 2020; 194:485-499. [PMID: 32991727 DOI: 10.1667/rade-20-00122.1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 08/05/2020] [Indexed: 11/03/2022]
Abstract
We present a novel mathematical formalism to predict the kinetics of DNA damage repair after exposure to both low- and high-LET radiation (X rays; 350 MeV/n 40Ar; 600 MeV/n 56Fe). Our method is based on monitoring DNA damage repair protein 53BP1 that forms radiation-induced foci (RIF) at locations of DNA double-strand breaks (DSB) in the nucleus and comparing its expression in primary skin fibroblasts isolated from 15 mice strains. We previously reported strong evidence for clustering of nearby DSB into single repair units as opposed to the classic "contact-first" model where DSB are considered immobile. Here we apply this clustering model to evaluate the number of remaining RIF over time. We also show that the newly introduced kinetic metrics can be used as surrogate biomarkers for in vivo radiation toxicity, with potential applications in radiotherapy and human space exploration. In particular, we observed an association between the characteristic time constant of RIF repair measured in vitro and survival levels of immune cells collected from irradiated mice. Moreover, the speed of DNA damage repair correlated not only with radiation-induced cellular survival in vivo, but also with spontaneous cancer incidence data collected from the Mouse Tumor Biology database, suggesting a relationship between the efficiency of DSB repair after irradiation and cancer risk.
Collapse
Affiliation(s)
- Eloise Pariset
- Universities Space Research Association (USRA), Columbia, Maryland 21046
- Space Biosciences Division, NASA Ames Research Center, Mountain View, California 94035
| | - Sébastien Penninckx
- Namur Research Institute for Life Science, University of Namur, 5000 Namur, Belgium
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720
| | | | - Elodie Guiet
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720
| | | | - Egle Cekanaviciute
- Space Biosciences Division, NASA Ames Research Center, Mountain View, California 94035
| | - Antoine M Snijders
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720
| | - Jian-Hua Mao
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720
| | - François Paris
- Université de Nantes, INSERM, CNRS, CRCINA, Nantes, France 44007
| | - Sylvain V Costes
- Space Biosciences Division, NASA Ames Research Center, Mountain View, California 94035
| |
Collapse
|
27
|
Ristic-Fira AM, Keta OD, Petković VD, Cammarata FP, Petringa G, Cirrone PG, Cuttone G, Incerti S, Petrović IM. DNA damage assessment of human breast and lung carcinoma cells irradiated with protons and carbon ions. JOURNAL OF RADIATION RESEARCH AND APPLIED SCIENCES 2020. [DOI: 10.1080/16878507.2020.1825035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
| | - Otilija D. Keta
- Vinča Institute of Nuclear Sciences, University of Belgrade, Belgrade, Serbia
| | - Vladana D. Petković
- Vinča Institute of Nuclear Sciences, University of Belgrade, Belgrade, Serbia
| | - Francesco P. Cammarata
- Istituto Nazionale Di Fisica Nucleare, Laboratori Nazionali Del Sud, Catania, Italy
- CNR-IBFM, UOS, Cefalù, Italy
| | - Giada Petringa
- Istituto Nazionale Di Fisica Nucleare, Laboratori Nazionali Del Sud, Catania, Italy
| | - Pablo G.A. Cirrone
- Istituto Nazionale Di Fisica Nucleare, Laboratori Nazionali Del Sud, Catania, Italy
| | - Giacomo Cuttone
- Istituto Nazionale Di Fisica Nucleare, Laboratori Nazionali Del Sud, Catania, Italy
| | | | - Ivan M. Petrović
- Vinča Institute of Nuclear Sciences, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
28
|
Zorin V, Grekhova A, Pustovalova M, Zorina A, Smetanina N, Vorobyeva N, Kopnin P, Gilmutdinova I, Moskalev A, Osipov AN, Leonov S. Spontaneous γH2AX foci in human dermal fibroblasts in relation to proliferation activity and aging. Aging (Albany NY) 2020; 11:4536-4546. [PMID: 31289256 PMCID: PMC6660037 DOI: 10.18632/aging.102067] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 06/26/2019] [Indexed: 12/12/2022]
Abstract
We assessed the effects of donor age on clonogenicity, proliferative potential, and spontaneous γH2AX foci in the proliferating (Ki67 +) and senescent (SA β-gal +) cultures of skin fibroblasts isolated from 34 donors of different age (23-82 years). Here, we demonstrated that neither the colony forming effectiveness of proliferating (Ki67+) fraction of the fibroblasts nor the average number of γH2AX foci of the same fraction does not depend on the age of the donor. The correlation between the number of γH2AX foci and the donor's age was reliable in quiescent (Ki67-) cells. The average number of γH2AX foci in quiescent fibroblasts of donors older than 68 years was about two times higher than in the same cells of up to 30 years old donors. The number of γH2AX foci demonstrated a statistically significant positive correlation with the fraction of proliferating cells in fibroblast cultures. On average, proliferating cells have twice as many the γH2AX foci in comparison with the quiescent cells. Within a population of proliferating (Ki67+) cells, the degree of senescence correlated with a relative declining of constitutive γH2AX foci number, whereas in the population of quiescent (Ki67-) cells, it was proportional to augmenting the number of the γH2AX foci. Our data on a statistically significant (p=0.001) correlation between the age of the donor and the number of constitutive γH2AX foci in quiescent cells, could point out the ongoing DNA-damage response due in the maintenance of the senescent state of cells.
Collapse
Affiliation(s)
- Vadim Zorin
- Human Stem Cells Institute, Moscow 119333, Russia
| | - Anna Grekhova
- State Research Center - Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Moscow 123098, Russia.,Emanuel Institute for Biochemical Physics, Russian Academy of Sciences, Moscow 119991, Russia
| | - Margarita Pustovalova
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region 141700, Russia
| | - Alla Zorina
- Human Stem Cells Institute, Moscow 119333, Russia
| | - Nadezhda Smetanina
- State Research Center - Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Moscow 123098, Russia
| | - Natalia Vorobyeva
- State Research Center - Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Moscow 123098, Russia.,Semenov Institute of Chemical Physics, Russian Academy of Sciences, Moscow 119991, Russia
| | - Pavel Kopnin
- N.N. Blokhin National Medical Research Oncology Center, Ministry of Health of Russia, Moscow 115478, Russia
| | - Ilmira Gilmutdinova
- FSBI "National Medical Research Center for Rehabilitation and Balneology", Ministry of Health of Russia, Moscow 121099, Russia
| | - Alexey Moskalev
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region 141700, Russia.,Laboratory of Molecular Radiobiology and Gerontology, Institute of Biology of Komi Science Center of Ural Division of Russian Academy of Sciences, Syktyvkar, Russia.,Laboratory of Post-Genomic Research, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Andreyan N Osipov
- State Research Center - Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Moscow 123098, Russia.,Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region 141700, Russia.,Semenov Institute of Chemical Physics, Russian Academy of Sciences, Moscow 119991, Russia
| | - Sergey Leonov
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region 141700, Russia.,Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region 142290, Russia
| |
Collapse
|
29
|
Liddle P, Jara-Wilde J, Lafon-Hughes L, Castro I, Härtel S, Folle G. dSTORM microscopy evidences in HeLa cells clustered and scattered γH2AX nanofoci sensitive to ATM, DNA-PK, and ATR kinase inhibitors. Mol Cell Biochem 2020; 473:77-91. [PMID: 32638256 DOI: 10.1007/s11010-020-03809-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 06/18/2020] [Indexed: 12/13/2022]
Abstract
In response to DNA double-strand breaks (DSB), histone H2AX is phosphorylated around the lesion by a feed forward signal amplification loop, originating γH2AX foci detectable by immunofluorescence and confocal microscopy as elliptical areas of uniform intensity. We exploited the significant increase in resolution (~ × 10) provided by single-molecule localization microscopy (SMLM) to investigate at nanometer scale the distribution of γH2AX signals either endogenous (controls) or induced by the radiomimetic bleomycin (BLEO) in HeLa cells. In both conditions, clustered substructures (nanofoci) confined to γH2AX foci and scattered nanofoci throughout the remnant nuclear area were detected. SR-Tesseler software (Voronoï tessellation-based segmentation) was combined with a custom Python script to first separate clustered nanofoci inside γH2AX foci from scattered nanofoci, and then to perform a cluster analysis upon each nanofoci type. Compared to controls, γH2AX foci in BLEO-treated nuclei presented on average larger areas (0.41 versus 0.19 µm2), more nanofoci per focus (22.7 versus 13.2) and comparable nanofoci densities (~ 60 nanofoci/µm2). Scattered γH2AX nanofoci were equally present (~ 3 nanofoci/µm2), suggesting an endogenous origin. BLEO-treated cells were challenged with specific inhibitors of canonical H2AX kinases, namely: KU-55933, VE-821 and NU-7026 for ATM, ATR and DNA-PK, respectively. Under treatment with pooled inhibitors, clustered nanofoci vanished from super-resolution images while scattered nanofoci decreased (~ 50%) in density. Residual scattered nanofoci could reflect, among other alternatives, H2AX phosphorylation mediated by VRK1, a recently described non-canonical H2AX kinase. In addition to H2AX findings, an analytical approach to quantify clusters of highly differing density from SMLM data is put forward.
Collapse
Affiliation(s)
- Pablo Liddle
- Departamento de Genética, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay.
| | - Jorge Jara-Wilde
- SCIAN-Lab, Biomedical Neuroscience Institute (BNI), Santiago, Chile.,Departamento de Ciencias de la Computación, Universidad de Chile, Santiago, Chile
| | - Laura Lafon-Hughes
- Departamento de Genética, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Iván Castro
- SCIAN-Lab, Biomedical Neuroscience Institute (BNI), Santiago, Chile
| | - Steffen Härtel
- SCIAN-Lab, Biomedical Neuroscience Institute (BNI), Santiago, Chile.,Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Gustavo Folle
- Departamento de Genética, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| |
Collapse
|
30
|
Quan Y, Tan Z, Yang Y, Deng B, Mu L. Prolonged effect associated with inflammatory response observed after exposure to low dose of tritium β-rays. Int J Radiat Biol 2020; 96:972-979. [PMID: 32394793 DOI: 10.1080/09553002.2020.1767817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Background: The value of relative biological effectiveness of tritium increases at low dose domain, which results in the suspicion of weighting factor of 1 for tritium after low dose exposure. Thus, present study was carried out to analyze the differences in the cellular responses at early and late period between low dose of tritium β-rays and γ-rays radiation.Methods: MCF-10A cells were exposed to low dose of tritium β-rays or γ-rays, then cellular behaviors, such as DNA double strand breaks (DSBs), apoptosis, reactive oxygen species (ROS) level and inflammatory relevant gene expression were analyzed at early and late period post-irradiation.Results: At early period the elimination of DSB foci produced by HTO is longer than γ-rays. High ROS level and a continual change of cell cycle distribution are observed in HTO radiation group. Based on the results of RNA sequencing, Ingenuity Pathway Analysis (IPA) indicates TNFR1 signaling and production of nitric oxide and ROS are activated as an acute response at 24 h post radiation. Moreover, it also shows a disturbance in cholesterol biosynthesis. The results of 30 days point that there is a lasting active inflammatory response, accompanying with a persistent high expression of relevant cytokines, such as TNF and IL1R.Conclusion: Compared to an acute response induced by γ-rays, a persistent inflammatory response exists in HTO-irradiated cells when cultured for 30 days, which might be related to accumulation of tritium in the form of organically bound tritium (OBT) in cellular DNA or lipids.
Collapse
Affiliation(s)
- Yi Quan
- China Academy of Engineering Physics, Institute of Nuclear Physics and Chemistry, Mianyang, Sichuan, China
| | - Zhaoyi Tan
- China Academy of Engineering Physics, Institute of Nuclear Physics and Chemistry, Mianyang, Sichuan, China
| | - Yang Yang
- China Academy of Engineering Physics, Institute of Nuclear Physics and Chemistry, Mianyang, Sichuan, China
| | - Bing Deng
- China Academy of Engineering Physics, Institute of Nuclear Physics and Chemistry, Mianyang, Sichuan, China
| | - Long Mu
- China Academy of Engineering Physics, Institute of Nuclear Physics and Chemistry, Mianyang, Sichuan, China
| |
Collapse
|
31
|
Alpha Radiation as a Way to Target Heterochromatic and Gamma Radiation-Exposed Breast Cancer Cells. Cells 2020; 9:cells9051165. [PMID: 32397212 PMCID: PMC7291130 DOI: 10.3390/cells9051165] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/19/2020] [Accepted: 05/06/2020] [Indexed: 12/21/2022] Open
Abstract
Compact chromatin is linked to a poor tumour prognosis and resistance to radiotherapy from photons. We investigated DNA damage induction and repair in the context of chromatin structure for densely ionising alpha radiation as well as its therapeutic potential. Chromatin opening by histone deacetylase inhibitor trichostatin A (TSA) pretreatment reduced clonogenic survival and increased γH2AX foci in MDA-MB-231 cells, indicative of increased damage induction by free radicals using gamma radiation. In contrast, TSA pretreatment tended to improve survival after alpha radiation while γH2AX foci were similar or lower; therefore, an increased DNA repair is suggested due to increased access of repair proteins. MDA-MB-231 cells exposed to fractionated gamma radiation (2 Gy × 6) expressed high levels of stem cell markers, elevated heterochromatin H3K9me3 marker, and a trend towards reduced clonogenic survival in response to alpha radiation. There was a higher level of H3K9me3 at baseline, and the ratio of DNA damage induced by alpha vs. gamma radiation was higher in the aggressive MDA-MB-231 cells compared to hormone receptor-positive MCF7 cells. We demonstrate that heterochromatin structure and stemness properties are induced by fractionated radiation exposure. Gamma radiation-exposed cells may be targeted using alpha radiation, and we provide a mechanistic basis for the involvement of chromatin in these effects.
Collapse
|
32
|
Grekhova AK, Pustovalova MV, Eremin PS, Ozerov IV, Maksimova OA, Gordeev AV, Vorobyeva NY, Osipov AN. Evaluation of the Contribution of Homologous Recombination in DNA Double-Strand Break Repair in Human Fibroblasts after Exposure to Low and Intermediate Doses of X-ray Radiation. BIOL BULL+ 2020. [DOI: 10.1134/s1062359019110037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
33
|
Hohmann T, Kessler J, Vordermark D, Dehghani F. Evaluation of machine learning models for automatic detection of DNA double strand breaks after irradiation using a γH2AX foci assay. PLoS One 2020; 15:e0229620. [PMID: 32101565 PMCID: PMC7043763 DOI: 10.1371/journal.pone.0229620] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 02/10/2020] [Indexed: 02/06/2023] Open
Abstract
Ionizing radiation induces amongst other the most critical type of DNA damage: double-strand breaks (DSBs). Efficient repair of such damage is crucial for cell survival and genomic stability. The analysis of DSB associated foci assays is often performed manually or with automatic systems. Manual evaluation is time consuming and subjective, while most automatic approaches are prone to changes in experimental conditions or to image artefacts. Here, we examined multiple machine learning models, namely a multi-layer perceptron classifier (MLP), linear support vector machine classifier (SVM), complement naive bayes classifier (cNB) and random forest classifier (RF), to correctly classify γH2AX foci in manually labeled images containing multiple types of artefacts. All models yielded reasonable agreements to the manual rating on the training images (Matthews correlation coefficient >0.4). Afterwards, the best performing models were applied on images obtained under different experimental conditions. Thereby, the MLP model produced the best results with an F1 Score >0.9. As a consequence, we have demonstrated that the used approach is sufficient to mimic manual counting and is robust against image artefacts and changes in experimental conditions.
Collapse
Affiliation(s)
- Tim Hohmann
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Germany
- * E-mail:
| | - Jacqueline Kessler
- Department of Radiotherapy, Martin Luther University Halle-Wittenberg, Germany
| | - Dirk Vordermark
- Department of Radiotherapy, Martin Luther University Halle-Wittenberg, Germany
| | - Faramarz Dehghani
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Germany
| |
Collapse
|
34
|
Cornforth MN. Occam's broom and the dirty DSB: cytogenetic perspectives on cellular response to changes in track structure and ionization density. Int J Radiat Biol 2020; 97:1099-1108. [PMID: 31971454 DOI: 10.1080/09553002.2019.1704302] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/30/2019] [Accepted: 11/11/2019] [Indexed: 12/19/2022]
Abstract
Given equal doses, it is well-known that densely ionizing radiations are more potent in causing a number of biological effects compared to sparsely ionizing radiations, such as x- or gamma rays. According to classical models of radiation action, this results from differences in the spatial distribution of lesions along charged particle tracks. In recent years investigators have been barraged with the alternative narrative that this is instead due to 'qualitative' differences in the types of molecular lesions that each type of radiation produces. The present review discusses, mainly from a cytogenetic perspective, the merits and shortcomings of these seemingly contradictory viewpoints. There may be a kernel of truth to the idea that qualitative differences in the types of molecular lesions produced at the nanometer level affect RBE/LET relationships, but to ignore the fact that such differences result from longer-range spatial distributions of lesions produced along charged particle tracks is an unjustifiably narrow stance tantamount to employing Occam's Broom. Not only are such spatial considerations indispensable in explaining the impact of ionization density upon higher-order biological endpoints, particularly chromosome aberrations, the explanations they provide render arguments based principally on the quality of IR damage largely superfluous.
Collapse
Affiliation(s)
- Michael N Cornforth
- Department of Radiation Oncology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
35
|
Deycmar S, Faccin E, Kazimova T, Knobel PA, Telarovic I, Tschanz F, Waller V, Winkler R, Yong C, Zingariello D, Pruschy M. The relative biological effectiveness of proton irradiation in dependence of DNA damage repair. Br J Radiol 2019; 93:20190494. [PMID: 31687835 DOI: 10.1259/bjr.20190494] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Clinical parameters and empirical evidence are the primary determinants for current treatment planning in radiation oncology. Personalized medicine in radiation oncology is only at the very beginning to take the genetic background of a tumor entity into consideration to define an individual treatment regimen, the total dose or the combination with a specific anticancer agent. Likewise, stratification of patients towards proton radiotherapy is linked to its physical advantageous energy deposition at the tumor site with minimal healthy tissue being co-irradiated distal to the target volume. Hence, the fact that photon and proton irradiation also induce different qualities of DNA damages, which require differential DNA damage repair mechanisms has been completely neglected so far. These subtle differences could be efficiently exploited in a personalized treatment approach and could be integrated into personalized treatment planning. A differential requirement of the two major DNA double-strand break repair pathways, homologous recombination and non-homologous end joining, was recently identified in response to proton and photon irradiation, respectively, and subsequently influence the mode of ionizing radiation-induced cell death and susceptibility of tumor cells with defects in DNA repair machineries to either quality of ionizing radiation.This review focuses on the differential DNA-damage responses and subsequent biological processes induced by photon and proton irradiation in dependence of the genetic background and discusses their impact on the unicellular level and in the tumor microenvironment and their implications for combined treatment modalities.
Collapse
Affiliation(s)
- Simon Deycmar
- Laboratory for Applied Radiobiology Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Raemistrasse 100, CH-8091 Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Geißler D, Wegmann M, Jochum T, Somma V, Sowa M, Scholz J, Fröhlich E, Hoffmann K, Niehaus J, Roggenbuck D, Resch-Genger U. An automatable platform for genotoxicity testing of nanomaterials based on the fluorometric γ-H2AX assay reveals no genotoxicity of properly surface-shielded cadmium-based quantum dots. NANOSCALE 2019; 11:13458-13468. [PMID: 31287475 DOI: 10.1039/c9nr01021a] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The large number of nanomaterial-based applications emerging in the materials and life sciences and the foreseeable increasing use of these materials require methods that evaluate and characterize the toxic potential of these nanomaterials to keep safety risks to people and environment as low as possible. As nanomaterial toxicity is influenced by a variety of parameters like size, shape, chemical composition, and surface chemistry, high throughput screening (HTS) platforms are recommended for assessing cytotoxicity. Such platforms are not yet available for genotoxicity testing. Here, we present first results obtained for application-relevant nanomaterials using an automatable genotoxicity platform that relies on the quantification of the phosphorylated histone H2AX (γ-H2AX) for detecting DNA double strand breaks (DSBs) and the automated microscope system AKLIDES® for measuring integral fluorescence intensities at different excitation wavelengths. This platform is used to test the genotoxic potential of 30 nm-sized citrate-stabilized gold nanoparticles (Au-NPs) as well as micellar encapsulated iron oxide nanoparticles (FeOx-NPs) and different cadmium (Cd)-based semiconductor quantum dots (QDs), thereby also searching for positive and negative controls as reference materials. In addition, the influence of the QD shell composition on the genotoxic potential of these Cd-based QDs was studied, using CdSe cores as well as CdSe/CdS core/shell and CdSe/CdS/ZnS core/shell/shell QDs. Our results clearly revealed the genotoxicity of the Au-NPs and its absence in the FeOx-NPs. The genotoxicity of the Cd-QDs correlates with the shielding of their Cd-containing core, with the core/shell/shell architecture preventing genotoxicity risks. The fact that none of these nanomaterials showed cytotoxicity at the chosen particle concentrations in a conventional cell viability assay underlines the importance of genotoxicity studies to assess the hazardous potential of nanomaterials.
Collapse
Affiliation(s)
- D Geißler
- Bundesanstalt für Materialforschung und -prüfung (BAM), Division 1.2 Biophotonics, Richard-Willstätter-Str. 11, 12489 Berlin, Germany.
| | - M Wegmann
- Bundesanstalt für Materialforschung und -prüfung (BAM), Division 1.2 Biophotonics, Richard-Willstätter-Str. 11, 12489 Berlin, Germany. and MEDIPAN GmbH, Ludwig-Erhard-Ring 3, 15827 Dahlewitz, Germany
| | - T Jochum
- Fraunhofer-Zentrum für Angewandte Nanotechnologie CAN, Grindelallee 117, 20146 Hamburg, Germany
| | - V Somma
- MEDIPAN GmbH, Ludwig-Erhard-Ring 3, 15827 Dahlewitz, Germany
| | - M Sowa
- MEDIPAN GmbH, Ludwig-Erhard-Ring 3, 15827 Dahlewitz, Germany
| | - J Scholz
- MEDIPAN GmbH, Ludwig-Erhard-Ring 3, 15827 Dahlewitz, Germany
| | - E Fröhlich
- Medizinische Universität Graz, Zentrum für Medizinische Forschung (ZMF), Stiftingtalstrasse 24, 8010 Graz, Austria
| | - K Hoffmann
- Bundesanstalt für Materialforschung und -prüfung (BAM), Division 1.2 Biophotonics, Richard-Willstätter-Str. 11, 12489 Berlin, Germany.
| | - J Niehaus
- Medizinische Universität Graz, Zentrum für Medizinische Forschung (ZMF), Stiftingtalstrasse 24, 8010 Graz, Austria
| | - D Roggenbuck
- MEDIPAN GmbH, Ludwig-Erhard-Ring 3, 15827 Dahlewitz, Germany and Institute of Biotechnology, Faculty Environment and Natural Sciences, Brandenburg University of Technology, Germany
| | - U Resch-Genger
- Bundesanstalt für Materialforschung und -prüfung (BAM), Division 1.2 Biophotonics, Richard-Willstätter-Str. 11, 12489 Berlin, Germany.
| |
Collapse
|
37
|
Penninckx S, Cekanaviciute E, Degorre C, Guiet E, Viger L, Lucas S, Costes SV. Dose, LET and Strain Dependence of Radiation-Induced 53BP1 Foci in 15 Mouse Strains Ex Vivo Introducing Novel DNA Damage Metrics. Radiat Res 2019; 192:1-12. [PMID: 31081741 DOI: 10.1667/rr15338.1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
We present a comprehensive comparative analysis on the repair of radiation-induced DNA damage ex vivo in 15 strains of mice, including 5 inbred reference strains and 10 collaborative-cross strains, of both sexes, totaling 5 million skin fibroblast cells imaged by three-dimensional highthroughput conventional microscopy. Non-immortalized primary skin fibroblasts derived from 76 mice were subjected to increasing doses of both low- and high-LET radiation (X rays; 350 MeV/n 40Ar; 600 MeV/n 56Fe), which are relevant to carcinogenesis and human space exploration. Automated image quantification of 53BP1 radiation-induced foci (RIF) formation and repair during the first 4-48 h postirradiation was performed as a function of dose and LET. Since multiple DNA double-strand breaks (DSBs) are induced in a dose- and LET-dependent manner, our data suggest that when DSBs are formed within the same discrete nuclear region, referred to as the "repair domain", novel mathematical formalisms used to report RIF allowed us to conclude that multiple DSBs can be present in single RIF. Specifically, we observed that the number of RIF per Gy was lower for higher X-ray doses or higher LET particles (i.e., 600 MeV/n 56Fe), suggesting there are more DSBs per RIF when the local absorbed dose increases in the nucleus. The data also clearly show that with more DSBs per RIF, it becomes more difficult for cells to fully resolve RIF. All 15 strains showed the same dose and LET dependence, but strain differences were preserved under various experimental conditions, indicating that the number and sizes of repair domains are modulated by the genetic background of each strain.
Collapse
Affiliation(s)
- Sébastien Penninckx
- a Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720.,b Namur Research Institute for Life Science, University of Namur, 5000 Namur, Belgium
| | - Egle Cekanaviciute
- c Universities Space Research Association (USRA), Columbia, Maryland.,d Space Biosciences Division, NASA Ames Research Center, Mountain View, California 94035
| | | | - Elodie Guiet
- a Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720
| | - Louise Viger
- a Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720
| | - Stéphane Lucas
- b Namur Research Institute for Life Science, University of Namur, 5000 Namur, Belgium
| | - Sylvain V Costes
- a Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720.,d Space Biosciences Division, NASA Ames Research Center, Mountain View, California 94035
| |
Collapse
|
38
|
DNA DSB Repair Dynamics following Irradiation with Laser-Driven Protons at Ultra-High Dose Rates. Sci Rep 2019; 9:4471. [PMID: 30872656 PMCID: PMC6418121 DOI: 10.1038/s41598-019-40339-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 02/07/2019] [Indexed: 11/09/2022] Open
Abstract
Protontherapy has emerged as more effective in the treatment of certain tumors than photon based therapies. However, significant capital and operational costs make protontherapy less accessible. This has stimulated interest in alternative proton delivery approaches, and in this context the use of laser-based technologies for the generation of ultra-high dose rate ion beams has been proposed as a prospective route. A better understanding of the radiobiological effects at ultra-high dose-rates is important for any future clinical adoption of this technology. In this study, we irradiated human skin fibroblasts-AG01522B cells with laser-accelerated protons at a dose rate of 109 Gy/s, generated using the Gemini laser system at the Rutherford Appleton Laboratory, UK. We studied DNA double strand break (DSB) repair kinetics using the p53 binding protein-1(53BP1) foci formation assay and observed a close similarity in the 53BP1 foci repair kinetics in the cells irradiated with 225 kVp X-rays and ultra- high dose rate protons for the initial time points. At the microdosimetric scale, foci per cell per track values showed a good correlation between the laser and cyclotron-accelerated protons indicating similarity in the DNA DSB induction and repair, independent of the time duration over which the dose was delivered.
Collapse
|
39
|
Lopez Perez R, Nicolay NH, Wolf JC, Frister M, Schmezer P, Weber KJ, Huber PE. DNA damage response of clinical carbon ion versus photon radiation in human glioblastoma cells. Radiother Oncol 2019; 133:77-86. [PMID: 30935585 DOI: 10.1016/j.radonc.2018.12.028] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 12/14/2018] [Accepted: 12/30/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND PURPOSE Carbon ion radiotherapy is a promising therapeutic option for glioblastoma patients due to its high physical dose conformity and greater biological effectiveness than photons. However, the biological effects of carbon ion radiation are still incompletely understood. Here, we systematically compared the biological effects of clinically used carbon ion radiation to photon radiation with emphasis on DNA repair. MATERIALS AND METHODS Two human glioblastoma cell lines (U87 and LN229) were irradiated with carbon ions or photons and DNA damage response was systematically analyzed, including clonogenic survival, induction and repair of DNA double-strand breaks (DSBs), cell cycle arrest and apoptosis or autophagy. γH2AX foci were analyzed by flow cytometry, conventional light microscopy and 3D superresolution microscopy. RESULTS DSBs were repaired delayed and with slower kinetics after carbon ions versus photons. Carbon ions caused stronger and longer-lasting cell cycle delays, predominantly in G2 phase, and a higher rate of apoptosis. Compared to photons, the effectiveness of carbon ions was less cell cycle-dependent. Homologous recombination (HR) appeared to be more important for DSB repair after carbon ions versus photons in phosphatase and tensin homolog (PTEN)-deficient U87 cells, as opposed to PTEN-proficient LN229 cells. CONCLUSION Carbon ions induced more severe DSB damage than photons, which was repaired less efficiently in both cell lines. Thus, carbon ion radiotherapy may help to overcome resistance mechanisms of glioblastoma associated with DNA repair for example in combination with repair pathway-specific drugs in the context of personalized radiotherapy.
Collapse
Affiliation(s)
- Ramon Lopez Perez
- CCU Molecular and Radiation Oncology, German Cancer Research Center and Department of Radiation Oncology, Heidelberg University Hospital, Germany.
| | - Nils H Nicolay
- CCU Molecular and Radiation Oncology, German Cancer Research Center and Department of Radiation Oncology, Heidelberg University Hospital, Germany; Department of Radiation Oncology, Freiburg University Medical Center, Germany
| | - Jörg-Christian Wolf
- CCU Molecular and Radiation Oncology, German Cancer Research Center and Department of Radiation Oncology, Heidelberg University Hospital, Germany
| | - Moritz Frister
- CCU Molecular and Radiation Oncology, German Cancer Research Center and Department of Radiation Oncology, Heidelberg University Hospital, Germany
| | - Peter Schmezer
- Epigenomics and Cancer Risk Factors, German Cancer Research Center, Heidelberg, Germany
| | - Klaus-Josef Weber
- CCU Molecular and Radiation Oncology, German Cancer Research Center and Department of Radiation Oncology, Heidelberg University Hospital, Germany
| | - Peter E Huber
- CCU Molecular and Radiation Oncology, German Cancer Research Center and Department of Radiation Oncology, Heidelberg University Hospital, Germany.
| |
Collapse
|
40
|
Recruitment of 53BP1 Proteins for DNA Repair and Persistence of Repair Clusters Differ for Cell Types as Detected by Single Molecule Localization Microscopy. Int J Mol Sci 2018; 19:ijms19123713. [PMID: 30469529 PMCID: PMC6321197 DOI: 10.3390/ijms19123713] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 11/19/2018] [Accepted: 11/20/2018] [Indexed: 01/30/2023] Open
Abstract
DNA double stranded breaks (DSBs) are the most serious type of lesions introduced into chromatin by ionizing radiation. During DSB repair, cells recruit different proteins to the damaged sites in a manner dependent on local chromatin structure, DSB location in the nucleus, and the repair pathway entered. 53BP1 is one of the important players participating in repair pathway decision of the cell. Although many molecular biology details have been investigated, the architecture of 53BP1 repair foci and its development during the post-irradiation time, especially the period of protein recruitment, remains to be elucidated. Super-resolution light microscopy is a powerful new tool to approach such studies in 3D-conserved cell nuclei. Recently, we demonstrated the applicability of single molecule localization microscopy (SMLM) as one of these highly resolving methods for analyses of dynamic repair protein distribution and repair focus internal nano-architecture in intact cell nuclei. In the present study, we focused our investigation on 53BP1 foci in differently radio-resistant cell types, moderately radio-resistant neonatal human dermal fibroblasts (NHDF) and highly radio-resistant U87 glioblastoma cells, exposed to high-LET 15N-ion radiation. At given time points up to 24 h post irradiation with doses of 1.3 Gy and 4.0 Gy, the coordinates and spatial distribution of fluorescently tagged 53BP1 molecules was quantitatively evaluated at the resolution of 10⁻20 nm. Clusters of these tags were determined as sub-units of repair foci according to SMLM parameters. The formation and relaxation of such clusters was studied. The higher dose generated sufficient numbers of DNA breaks to compare the post-irradiation dynamics of 53BP1 during DSB processing for the cell types studied. A perpendicular (90°) irradiation scheme was used with the 4.0 Gy dose to achieve better separation of a relatively high number of particle tracks typically crossing each nucleus. For analyses along ion-tracks, the dose was reduced to 1.3 Gy and applied in combination with a sharp angle irradiation (10° relative to the cell plane). The results reveal a higher ratio of 53BP1 proteins recruited into SMLM defined clusters in fibroblasts as compared to U87 cells. Moreover, the speed of foci and thus cluster formation and relaxation also differed for the cell types. In both NHDF and U87 cells, a certain number of the detected and functionally relevant clusters remained persistent even 24 h post irradiation; however, the number of these clusters again varied for the cell types. Altogether, our findings indicate that repair cluster formation as determined by SMLM and the relaxation (i.e., the remaining 53BP1 tags no longer fulfill the cluster definition) is cell type dependent and may be functionally explained and correlated to cell specific radio-sensitivity. The present study demonstrates that SMLM is a highly appropriate method for investigations of spatiotemporal protein organization in cell nuclei and how it influences the cell decision for a particular repair pathway at a given DSB site.
Collapse
|
41
|
Ochola DO, Sharif R, Bedford JS, Keefe TJ, Kato TA, Fallgren CM, Demant P, Costes SV, Weil MM. Persistence of Gamma-H2AX Foci in Bronchial Cells Correlates with Susceptibility to Radiation Associated Lung Cancer in Mice. Radiat Res 2018; 191:67-75. [PMID: 30398394 DOI: 10.1667/rr14979.1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The risk of developing radiation-induced lung cancer differs between different strains of mice, but the underlying cause of the strain differences is unknown. Strains of mice also differ in how quickly they repair radiation-induced DNA double-strand breaks (DSBs). We assayed mouse strains from the CcS/Dem recombinant congenic strain set for their efficacy in repairing DNA DSBs during protracted irradiation. We measured unrepaired γ-H2AX radiation-induced foci (RIF), which persisted after chronic 24-h gamma irradiation, as a surrogate marker for repair efficiency in bronchial epithelial cells for 17 of the CcS/Dem strains and the BALB/c founder strain. We observed a very strong correlation (R2 = 79.18%, P < 0.001) between the level of unrepaired RIF and radiogenic lung cancer incidence measured in the same strains. Interestingly, spontaneous levels of foci in nonirradiated mice also showed good correlation with lung cancer incidence when incidence data from male and female mice were combined. These results suggest that genetic differences in DNA repair capacity largely account for differing susceptibilities to radiation-induced lung cancer among CcS/Dem mouse strains, and that high levels of spontaneous DNA damage are also a relatively good marker of cancer predisposition. In a smaller pilot study, we found that the repair capacity measured in peripheral blood leucocytes also correlated well with radiogenic lung cancer susceptibility, raising the possibility that the assay could be used to detect radiogenic lung cancer susceptibility in humans.
Collapse
Affiliation(s)
- Donasian O Ochola
- a Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado
| | - Rabab Sharif
- a Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado
| | - Joel S Bedford
- a Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado
| | - Thomas J Keefe
- a Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado
| | - Takamitsu A Kato
- a Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado
| | - Christina M Fallgren
- a Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado
| | - Peter Demant
- b Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, New York
| | - Sylvain V Costes
- c Biosciences Division, NASA Ames Research Center, Mountain View, California
| | - Michael M Weil
- a Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado
| |
Collapse
|
42
|
Rassamegevanon T, Löck S, Baumann M, Krause M, von Neubeck C. Heterogeneity of γH2AX Foci Increases in Ex Vivo Biopsies Relative to In Vivo Tumors. Int J Mol Sci 2018; 19:E2616. [PMID: 30181446 PMCID: PMC6163410 DOI: 10.3390/ijms19092616] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 08/13/2018] [Accepted: 08/16/2018] [Indexed: 12/30/2022] Open
Abstract
The biomarker for DNA double stand breaks, gammaH2AX (γH2AX), holds a high potential as an intrinsic radiosensitivity predictor of tumors in clinical practice. Here, two published γH2AX foci datasets from in and ex vivo exposed human head and neck squamous cell carcinoma (hHNSCC) xenografts were statistically re-evaluated for the effect of the assay setting (in or ex vivo) on cellular geometry and the degree of heterogeneity in γH2AX foci. Significant differences between the nucleus areas of in- and ex vivo exposed samples were found. However, the number of foci increased linearly with nucleus area in irradiated samples of both settings. Moreover, irradiated tumor cells showed changes of nucleus area distributions towards larger areas compared to unexposed samples, implying cell cycle alteration after radiation exposure. The number of residual γH2AX foci showed a higher degree of intra-tumoral heterogeneity in the ex vivo exposed samples relative to the in vivo exposed samples. In the in vivo setting, the highest intra-tumoral heterogeneity was observed in initial γH2AX foci numbers (foci detected 30 min following irradiation). These results suggest that the tumor microenvironment and the culture condition considerably influence cellular adaptation and DNA damage repair.
Collapse
Affiliation(s)
- Treewut Rassamegevanon
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.
- OncoRay-National Center for Radiation Research in Oncology, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany.
| | - Steffen Löck
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.
- OncoRay-National Center for Radiation Research in Oncology, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany.
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Michael Baumann
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.
- OncoRay-National Center for Radiation Research in Oncology, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany.
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, 01328 Dresden, Germany.
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: Helmholtz Association/Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany.
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Mechthild Krause
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.
- OncoRay-National Center for Radiation Research in Oncology, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany.
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, 01328 Dresden, Germany.
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: Helmholtz Association/Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany.
| | - Cläre von Neubeck
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.
- OncoRay-National Center for Radiation Research in Oncology, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany.
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| |
Collapse
|
43
|
Deycmar S, Pruschy M. Combined Treatment Modalities for High-Energy Proton Irradiation: Exploiting Specific DNA Repair Dependencies. Int J Part Ther 2018; 5:133-139. [DOI: 10.14338/ijpt-18-00020.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 07/05/2018] [Indexed: 11/21/2022] Open
Affiliation(s)
- Simon Deycmar
- Department of Radiation Oncology, Laboratory for Applied Radiobiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Martin Pruschy
- Department of Radiation Oncology, Laboratory for Applied Radiobiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
44
|
Live Dynamics of 53BP1 Foci Following Simultaneous Induction of Clustered and Dispersed DNA Damage in U2OS Cells. Int J Mol Sci 2018; 19:ijms19020519. [PMID: 29419809 PMCID: PMC5855741 DOI: 10.3390/ijms19020519] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 01/18/2018] [Accepted: 01/30/2018] [Indexed: 12/19/2022] Open
Abstract
Cells react differently to clustered and dispersed DNA double strand breaks (DSB). Little is known about the initial reaction to simultaneous induction of DSBs with different complexities. Here, we used live cell microscopy to analyse the behaviour of 53BP1-GFP (green fluorescence protein) foci formation at DSBs induced in U2OS cells by alpha particles, X-rays or mixed beams over a 75 min period post irradiation. X-ray-induced foci rapidly increased and declined over the observation interval. After an initial increase, mixed beam-induced foci remained at a constant level over the observation interval, similarly as alpha-induced foci. The average areas of radiation-induced foci were similar for mixed beams and X-rays, being significantly smaller than those induced by alpha particles. Pixel intensities were highest for mixed beam-induced foci and showed the lowest level of variability over time as compared to foci induced by alphas and X-rays alone. Finally, mixed beam-exposed foci showed the lowest level of mobility as compared to alpha and X-ray exposure. The results suggest paralysation of chromatin around foci containing clustered DNA damage.
Collapse
|
45
|
Localization Microscopy Analyses of MRE11 Clusters in 3D-Conserved Cell Nuclei of Different Cell Lines. Cancers (Basel) 2018; 10:cancers10010025. [PMID: 29361783 PMCID: PMC5789375 DOI: 10.3390/cancers10010025] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 01/12/2018] [Accepted: 01/17/2018] [Indexed: 01/01/2023] Open
Abstract
In radiation biophysics, it is a subject of nowadays research to investigate DNA strand break repair in detail after damage induction by ionizing radiation. It is a subject of debate as to what makes up the cell’s decision to use a certain repair pathway and how the repair machinery recruited in repair foci is spatially and temporarily organized. Single-molecule localization microscopy (SMLM) allows super-resolution analysis by precise localization of single fluorescent molecule tags, resulting in nuclear structure analysis with a spatial resolution in the 10 nm regime. Here, we used SMLM to study MRE11 foci. MRE11 is one of three proteins involved in the MRN-complex (MRE11-RAD50-NBS1 complex), a prominent DNA strand resection and broken end bridging component involved in homologous recombination repair (HRR) and alternative non-homologous end joining (a-NHEJ). We analyzed the spatial arrangements of antibody-labelled MRE11 proteins in the nuclei of a breast cancer and a skin fibroblast cell line along a time-course of repair (up to 48 h) after irradiation with a dose of 2 Gy. Different kinetics for cluster formation and relaxation were determined. Changes in the internal nano-scaled structure of the clusters were quantified and compared between the two cell types. The results indicate a cell type-dependent DNA damage response concerning MRE11 recruitment and cluster formation. The MRE11 data were compared to H2AX phosphorylation detected by γH2AX molecule distribution. These data suggested modulations of MRE11 signal frequencies that were not directly correlated to DNA damage induction. The application of SMLM in radiation biophysics offers new possibilities to investigate spatial foci organization after DNA damaging and during subsequent repair.
Collapse
|
46
|
Jezkova L, Zadneprianetc M, Kulikova E, Smirnova E, Bulanova T, Depes D, Falkova I, Boreyko A, Krasavin E, Davidkova M, Kozubek S, Valentova O, Falk M. Particles with similar LET values generate DNA breaks of different complexity and reparability: a high-resolution microscopy analysis of γH2AX/53BP1 foci. NANOSCALE 2018; 10:1162-1179. [PMID: 29271466 DOI: 10.1039/c7nr06829h] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Biological effects of high-LET (linear energy transfer) radiation have received increasing attention, particularly in the context of more efficient radiotherapy and space exploration. Efficient cell killing by high-LET radiation depends on the physical ability of accelerated particles to generate complex DNA damage, which is largely mediated by LET. However, the characteristics of DNA damage and repair upon exposure to different particles with similar LET parameters remain unexplored. We employed high-resolution confocal microscopy to examine phosphorylated histone H2AX (γH2AX)/p53-binding protein 1 (53BP1) focus streaks at the microscale level, focusing on the complexity, spatiotemporal behaviour and repair of DNA double-strand breaks generated by boron and neon ions accelerated at similar LET values (∼135 keV μm-1) and low energies (8 and 47 MeV per n, respectively). Cells were irradiated using sharp-angle geometry and were spatially (3D) fixed to maximize the resolution of these analyses. Both high-LET radiation types generated highly complex γH2AX/53BP1 focus clusters with a larger size, increased irregularity and slower elimination than low-LET γ-rays. Surprisingly, neon ions produced even more complex γH2AX/53BP1 focus clusters than boron ions, consistent with DSB repair kinetics. Although the exposure of cells to γ-rays and boron ions eliminated a vast majority of foci (94% and 74%, respectively) within 24 h, 45% of the foci persisted in cells irradiated with neon. Our calculations suggest that the complexity of DSB damage critically depends on (increases with) the particle track core diameter. Thus, different particles with similar LET and energy may generate different types of DNA damage, which should be considered in future research.
Collapse
Affiliation(s)
- Lucie Jezkova
- Joint Institute for Nuclear Research, Dubna, Russia and University of Chemistry and Technology Prague, Prague, Czech Republic
- University of Chemistry and Technology Prague, Prague, Czech Republic
| | - Mariia Zadneprianetc
- Joint Institute for Nuclear Research, Dubna, Russia and Dubna State University, Dubna, Russia
- Dubna State University, Dubna, Russia
| | - Elena Kulikova
- Joint Institute for Nuclear Research, Dubna, Russia and Dubna State University, Dubna, Russia
- Dubna State University, Dubna, Russia
| | | | - Tatiana Bulanova
- Joint Institute for Nuclear Research, Dubna, Russia and Dubna State University, Dubna, Russia
- Dubna State University, Dubna, Russia
| | - Daniel Depes
- Czech Academy of Sciences, Institute of Biophysics, Brno, Czech Republic.
| | - Iva Falkova
- Czech Academy of Sciences, Institute of Biophysics, Brno, Czech Republic.
| | - Alla Boreyko
- Joint Institute for Nuclear Research, Dubna, Russia and Dubna State University, Dubna, Russia
- Dubna State University, Dubna, Russia
| | - Evgeny Krasavin
- Joint Institute for Nuclear Research, Dubna, Russia and Dubna State University, Dubna, Russia
- Dubna State University, Dubna, Russia
| | - Marie Davidkova
- Czech Academy of Sciences, Nuclear Physics Institute, Prague, Czech Republic
| | - Stanislav Kozubek
- Czech Academy of Sciences, Institute of Biophysics, Brno, Czech Republic.
| | - Olga Valentova
- University of Chemistry and Technology Prague, Prague, Czech Republic
| | - Martin Falk
- Czech Academy of Sciences, Institute of Biophysics, Brno, Czech Republic.
| |
Collapse
|
47
|
McDonald JS, McDonald RJ, Ekins JB, Tin AS, Costes S, Hudson TM, Schroeder DJ, Kallmes K, Kaufmann SH, Young PM, Lu A, Kadirvel R, Kallmes DF. Gadolinium-enhanced cardiac MR exams of human subjects are associated with significant increases in the DNA repair marker 53BP1, but not the damage marker γH2AX. PLoS One 2018; 13:e0190890. [PMID: 29309426 PMCID: PMC5757995 DOI: 10.1371/journal.pone.0190890] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 12/21/2017] [Indexed: 12/13/2022] Open
Abstract
Magnetic resonance imaging is considered low risk, yet recent studies have raised a concern of potential damage to DNA in peripheral blood leukocytes. This prospective Institutional Review Board-approved study examined potential double-strand DNA damage by analyzing changes in the DNA damage and repair markers γH2AX and 53BP1 in patients who underwent a 1.5 T gadolinium-enhanced cardiac magnetic resonance (MR) exam. Sixty patients were enrolled (median age 55 years, 39 males). Patients with history of malignancy or who were receiving chemotherapy, radiation therapy, or steroids were excluded. MR sequence data were recorded and blood samples obtained immediately before and after MR exposure. An automated immunofluorescence assay quantified γH2AX or 53BP1 foci number in isolated peripheral blood mononuclear cells. Changes in foci number were analyzed using the Wilcoxon signed-rank test. Clinical and MR procedural characteristics were compared between patients who had a >10% increase in γH2AX or 53BP1 foci numbers and patients who did not. The number of γH2AX foci did not significantly change following cardiac MR (median foci per cell pre-MR = 0.11, post-MR = 0.11, p = .90), but the number of 53BP1 foci significantly increased following MR (median foci per cell pre-MR = 0.46, post-MR = 0.54, p = .0140). Clinical and MR characteristics did not differ significantly between patients who had at least a 10% increase in foci per cell and those who did not. We conclude that MR exposure leads to a small (median 25%) increase in 53BP1 foci, however the clinical relevance of this increase is unknown and may be attributable to normal variation instead of MR exposure.
Collapse
Affiliation(s)
- Jennifer S. McDonald
- Department of Radiology, College of Medicine, Mayo Clinic, Rochester, MN, United States of America
- * E-mail:
| | - Robert J. McDonald
- Department of Radiology, College of Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - Jacob B. Ekins
- Department of Radiology, College of Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - Anthony S. Tin
- Exogen Biotechnology Inc., Berkeley, CA, United States of America
| | - Sylvain Costes
- Exogen Biotechnology Inc., Berkeley, CA, United States of America
| | - Tamara M. Hudson
- Department of Radiology, College of Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - Dana J. Schroeder
- Department of Radiology, College of Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - Kevin Kallmes
- Department of Radiology, College of Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - Scott H. Kaufmann
- Department of Molecular Pharmacology and Experimental Therapeutics, College of Medicine, Mayo Clinic, Rochester, MN, United States of America
- Department of Oncology, College of Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - Philip M. Young
- Department of Radiology, College of Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - Aiming Lu
- Department of Radiology, College of Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - Ramanathan Kadirvel
- Department of Radiology, College of Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - David F. Kallmes
- Department of Radiology, College of Medicine, Mayo Clinic, Rochester, MN, United States of America
- Department of Neuroscience, College of Medicine, Mayo Clinic, Rochester, MN, United States of America
| |
Collapse
|
48
|
Patient-Specific Screening Using High-Grade Glioma Explants to Determine Potential Radiosensitization by a TGF-β Small Molecule Inhibitor. Neoplasia 2017; 18:795-805. [PMID: 27978994 PMCID: PMC5156509 DOI: 10.1016/j.neo.2016.08.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 08/26/2016] [Accepted: 08/29/2016] [Indexed: 12/26/2022] Open
Abstract
High-grade glioma (HGG), a deadly primary brain malignancy, manifests radioresistance mediated by cell-intrinsic and microenvironmental mechanisms. High levels of the cytokine transforming growth factor-β (TGF-β) in HGG promote radioresistance by enforcing an effective DNA damage response and supporting glioma stem cell self-renewal. Our analysis of HGG TCGA data and immunohistochemical staining of phosphorylated Smad2, which is the main transducer of canonical TGF-β signaling, indicated variable levels of TGF-β pathway activation across HGG tumors. These data suggest that evaluating the putative benefit of inhibiting TGF-β during radiotherapy requires personalized screening. Thus, we used explant cultures of seven HGG specimens as a rapid, patient-specific ex vivo platform to test the hypothesis that LY364947, a small molecule inhibitor of the TGF-β type I receptor, acts as a radiosensitizer in HGG. Immunofluorescence detection and image analysis of γ-H2AX foci, a marker of cellular recognition of radiation-induced DNA damage, and Sox2, a stem cell marker that increases post-radiation, indicated that LY364947 blocked these radiation responses in five of seven specimens. Collectively, our findings suggest that TGF-β signaling increases radioresistance in most, but not all, HGGs. We propose that short-term culture of HGG explants provides a flexible and rapid platform for screening context-dependent efficacy of radiosensitizing agents in patient-specific fashion. This time- and cost-effective approach could be used to personalize treatment plans in HGG patients.
Collapse
|
49
|
Si J, Zhou R, Song J, Gan L, Zhou X, Di C, Liu Y, Mao A, Zhao Q, Wang Y, Zhang H. Toxic effects of 56Fe ion radiation on the zebrafish (Danio rerio) embryonic development. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2017; 186:87-95. [PMID: 28267650 DOI: 10.1016/j.aquatox.2017.02.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 02/20/2017] [Accepted: 02/27/2017] [Indexed: 06/06/2023]
Abstract
All living organisms and ecosystems are permanently exposed to ionizing radiation. Of all the types of ionizing radiation, heavy ions such as 56Fe have the potential to cause the most severe biological effects. We therefore examined the effects and potential mechanisms of iron ion irradiation on the induction of developmental toxicity and apoptosis in zebrafish embryos. Zebrafish embryos at 4h post-fertilization (hpf) were divided into five groups: a control group; and four groups irradiated with 0.5, 1, 2, and 4Gy radiation, respectively. Mortality and teratogenesis were significantly increased, and spontaneous movement, heart rate, and swimming distance were decreased in the irradiated groups, accompanied by increased apoptosis. mRNA levels of genes involved in the apoptotic pathway, including p53, bax, bcl-2, and caspase-3, were significantly affected by radiation exposure. Moreover, protein expression levels of P53 and Bcl-2 changed in accordance with the corresponding mRNA expression levels. In addition, we detected the protein expression levels of γ-H2AX, which is a biomarker for radiation-induced DNA double-strand breaks, and found that γ-H2AX protein levels were significantly increased in the irradiated groups. Overall, the results of this study improve our understanding of the mechanisms of iron ion radiation-induced developmental toxicity and apoptosis, potentially involving the induction of DNA damage and mitochondrial dysfunction. The findings of this study may aid future impact assessment of environmental radioactivity in fish.
Collapse
Affiliation(s)
- Jing Si
- Department of Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China
| | - Rong Zhou
- Department of Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China
| | - Jing'e Song
- Hospital of Stomatology, Lanzhou University, Lanzhou 730000, China
| | - Lu Gan
- Department of Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China
| | - Xin Zhou
- Department of Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China
| | - Cuixia Di
- Department of Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China
| | - Yang Liu
- Department of Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China
| | - Aihong Mao
- Department of Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China
| | - Qiuyue Zhao
- Department of Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China
| | - Yupei Wang
- Department of Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China
| | - Hong Zhang
- Department of Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China; Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China; Gansu Wuwei Institute of Medical Sciences, Wuwei 733000, China.
| |
Collapse
|
50
|
Herskind C, Ma L, Liu Q, Zhang B, Schneider F, Veldwijk MR, Wenz F. Biology of high single doses of IORT: RBE, 5 R's, and other biological aspects. Radiat Oncol 2017; 12:24. [PMID: 28107823 PMCID: PMC5251326 DOI: 10.1186/s13014-016-0750-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 12/21/2016] [Indexed: 01/14/2023] Open
Abstract
Intraoperative radiotherapy differs from conventional, fractionated radiotherapy in several aspects that may influence its biological effect. The radiation quality influences the relative biologic effectiveness (RBE), and the role of the five R’s of radiotherapy (reassortment, repair, reoxygenation, repopulation, radiosensitivity) is different. Furthermore, putative special biological effects and the small volume receiving a high single dose may be important. The present review focuses on RBE, repair, and repopulation, and gives an overview of the other factors that potentially contribute to the efficacy. The increased RBE should be taken into account for low-energy X-rays while evidence of RBE < 1 for high-energy electrons at higher doses is presented. Various evidence supports a hypothesis that saturation of the primary DNA double-strand break (DSB) repair mechanisms leads to increasing use of an error-prone backup repair system leading to genomic instability that may contribute to inactivate tumour cells at high single doses. Furthermore, the elimination of repopulation of residual tumour cells in the tumour bed implies that some patients are likely to have very few residual tumour cells which may be cured even by low doses to the tumour bed. The highly localised dose distribution of IORT has the potential to inactivate tumour cells while sparing normal tissue by minimising the volume exposed to high doses. Whether special effects of high single doses also contribute to the efficacy will require further experimental and clinical studies.
Collapse
Affiliation(s)
- Carsten Herskind
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
| | - Lin Ma
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.,Present Address: Department of Radiation Oncology, University of California, San Francisco, CA, USA
| | - Qi Liu
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.,Present Address: Department of Radiation Oncology, University of California, San Francisco, CA, USA
| | - Bo Zhang
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.,Present Address: Department of Oncology at No. 2 Hospital Anhui Medical University, and School of Life Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Frank Schneider
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Marlon R Veldwijk
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Frederik Wenz
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| |
Collapse
|