1
|
Grinberg M, Vodeneev V. The role of signaling systems of plant in responding to key astrophysical factors: increased ionizing radiation, near-null magnetic field and microgravity. PLANTA 2025; 261:31. [PMID: 39797920 DOI: 10.1007/s00425-025-04610-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 01/06/2025] [Indexed: 01/13/2025]
Abstract
Plants will form the basis of artificial ecosystems in space exploration and the creation of bases on other planets. Astrophysical factors, such as ionizing radiation (IR), magnetic fields (MF) and gravity, can significantly affect the growth and development of plants beyond Earth. However, to date, the ways in which these factors influence plants remain largely unexplored. The review shows that, despite the lack of specialized receptors, plants are able to perceive changes in astrophysical factors. Potential mechanisms for perceiving changes in IR, MF and gravity levels are considered. The main pathway for inducing effects in plants is caused by primary physicochemical reactions and change in the levels of secondary messengers, including ROS and Ca2+. The presence of common components, including secondary messengers, in the chain of responses to astrophysical factors determines the complex nature of the response under their combined action. The analysis performed and the proposed hypothesis will help in planning space missions, as well as identifying the most important areas of research in space biology.
Collapse
Affiliation(s)
- Marina Grinberg
- Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia.
| | - Vladimir Vodeneev
- Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| |
Collapse
|
2
|
Kwon EJ, Lee H, Shin U, Kim ES, Myung K, Kim J, Park JH, Kim K, Lee Y, Oh CK, Kim YH. Ionizing radiation inhibits zebrafish embryo hatching through induction of tissue inhibitors of metalloproteinases (TIMPs) expression. FEBS J 2024; 291:5470-5485. [PMID: 39547957 DOI: 10.1111/febs.17318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 09/26/2024] [Accepted: 10/28/2024] [Indexed: 11/17/2024]
Abstract
Ionizing radiation (IR) has garnered growing attention because of its biological effects on aquatic organisms and humans. Here, we identify the most impacted organs and uncover the molecular mechanisms causing the changes in the context of vertebrate development using single-cell RNA sequencing. Alterations in cellular composition and biological functions were explored using transcriptomic profiling of zebrafish embryos exposed to 5 Gy. Single-cell RNA sequencing analyses unveiled notable shifts in the proportions of brain/central nervous system and hatching gland clusters. Although IR exposure led to increased expression of hatching enzymes, a significant but mild delay in hatching was observed following 5 Gy IR exposure. Gene Ontology analysis showed an increased expression of tissue inhibitors of metalloproteinases (TIMPs), known as matrix metalloproteinase inhibitors, which was confirmed via whole-mount in situ hybridization. Correlation analysis linked TIMPs to transcription factors cebpb and cebpd, which were significantly correlated post-IR exposure. Although no morphological changes were observed in some organs, including the brain, the study reveals substantial alterations in developing vertebrates. Notably, despite increased hatching enzymes, elevated TIMPs in the hatching gland suggest a regulatory mechanism impacting hatching activity. This research contributes to comprehending the ecological repercussions of IR exposure, emphasizing the importance of safety measures for aquatic ecosystems and overall environmental health.
Collapse
Affiliation(s)
- Eun Jung Kwon
- Medical Research Institute, Pusan National University, Yangsan, Korea
| | - Hansong Lee
- Medical Research Institute, Pusan National University, Yangsan, Korea
| | - Unbum Shin
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan, Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Korea
| | - Eun-Sun Kim
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan, Korea
| | - Kyungjae Myung
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan, Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Korea
| | - Jeongmo Kim
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Korea
| | - Jung-Hoon Park
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Korea
| | - Kihun Kim
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, Korea
- Department of Biomedical Informatics, School of Medicine, Pusan National University, Yangsan, Korea
| | - Yoonsung Lee
- Research Institute of Clinical Medicine, Kyung Hee University Hospital at Gangdong, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Chang-Kyu Oh
- Department of Biochemistry, School of Medicine, Pusan National University, Yangsan, Korea
- Institute for Future Earth, Pusan National University, Busan, Korea
| | - Yun Hak Kim
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, Korea
- Department of Biomedical Informatics, School of Medicine, Pusan National University, Yangsan, Korea
| |
Collapse
|
3
|
Jin JY, Yuan J, Qin X, Li Y, Yan H, Oleinick NL, Yao M, Pan Q, Kong FM(S, Machtay M. Derivation of a comprehensive semi-empirical proton RBE model from published experimental cell survival data collected in the PIDE database. Front Oncol 2024; 14:1415213. [PMID: 39664177 PMCID: PMC11631728 DOI: 10.3389/fonc.2024.1415213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 10/24/2024] [Indexed: 12/13/2024] Open
Abstract
We aimed to develop a comprehensive proton relative biological effectiveness (RBE) model based on accumulated cell survival data in the literature. Our approach includes four major components: (1) Eligible cell survival data with various linear energy transfers (LETs) in the Particle Irradiation Data Ensemble (PIDE) database (72 datasets in four cell lines); (2) a cell survival model based on Poisson equation, with α and β defined as the ability to generate and repair damage, respectively, to replace the classic linear-quadratic model for fitting the cell survival data; (3) hypothetical linear relations of α and β on LET, orα ( L E T ) α x = α α + b α ∗ L E T andβ ( L E T ) β x = α β - b β ∗ L E T ; and (4) a multi-curve fitting (MCF) approach to fit all cell survival data into the survival model and derive the aα , bα , aβ , and bβ values for each cell line. Dependences of these parameters on cell type were thus determined and finally a comprehensive RBE model was derived. MCF showed that (aα , bα , aβ , bβ ) = (1.09, 0.0010, 0.96, 0.033), (1.10, 0.0015, 1.03, 0.023), (1.12, 0.0025, 0.99, 0.0085), and (1.17, 0.0025, 0.99, 0.013) for the four cell lines, respectively. Thus, aα = 1.12 ± 0.04, bα = 0.0019 ± 0.0008, aβ = 0.99 ± 0.03, and bβ = 0.013 ∗ αx , and approximately α ∼ 1.12 ∗ α x and β = ( 0.99 - 0.013 ∗ α x ∗ L E T ) ∗ β x . Consequently, a relatively reliable and comprehensive RBE model with dependence on LET, αx , βx , and dose per fraction was finally derived for potential clinical application.
Collapse
Affiliation(s)
- Jian-Yue Jin
- School of Biomedical Engineering, Capital Medical University, Beijing, China
- Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Jiankui Yuan
- Seidman Cancer Center, University Hospitals, Cleveland Medical Center, Cleveland, OH, United States
| | - Xiaohang Qin
- School of Biomedical Engineering, Capital Medical University, Beijing, China
| | - Yinghui Li
- School of Biomedical Engineering, Capital Medical University, Beijing, China
| | - Huagang Yan
- School of Biomedical Engineering, Capital Medical University, Beijing, China
| | - Nancy L. Oleinick
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Min Yao
- Department of Radiation Oncology, Penn State University Cancer Institute, Hershey, PA, United States
| | - Quintin Pan
- Seidman Cancer Center, University Hospitals, Cleveland Medical Center, Cleveland, OH, United States
| | - Feng-Ming (Spring) Kong
- Department of Clinical Oncology, Hong Kong University Shenzhen Hospital, Shenzhen, China
- Department of Clinical Oncology, Queen Mary Hospital, Li Ka Shing Medical School, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Mitchell Machtay
- Department of Radiation Oncology, Penn State University Cancer Institute, Hershey, PA, United States
| |
Collapse
|
4
|
Cucinotta FA. Modeling Clustered DNA Damage by Ionizing Radiation Using Multinomial Damage Probabilities and Energy Imparted Spectra. Int J Mol Sci 2024; 25:12532. [PMID: 39684242 DOI: 10.3390/ijms252312532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/18/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Simple and complex clustered DNA damage represent the critical initial damage caused by radiation. In this paper, a multinomial probability model of clustered damage is developed with probabilities dependent on the energy imparted to DNA and surrounding water molecules. The model consists of four probabilities: (A) direct damage of sugar-phosphate moieties leading to SSB, (B) OH- radical formation with subsequent SSB and BD formation, (C) direct damage to DNA bases, and (D) energy imparted to histone proteins and other molecules in a volume not leading to SSB or BD. These probabilities are augmented by introducing probabilities for the relative location of SSB using a ≤10 bp criteria for a double-strand break (DSB) and for the possible success of a radical attack that leads to SSB or BD. Model predictions for electrons, 4He, and 12C ions are compared to the experimental data and show good agreement. Thus, the developed model allows an accurate and rapid computational method to predict simple and complex clustered DNA damage as a function of radiation quality and to explore the resulting challenges to DNA repair.
Collapse
Affiliation(s)
- Francis A Cucinotta
- Department of Health Physics and Diagnostic Sciences, University of Nevada Las Vegas, 4505 S. Maryland Parkway, Box 453037, Las Vegas, NV 89154, USA
| |
Collapse
|
5
|
Hammad M, Salma R, Balosso J, Rezvani M, Haghdoost S. Role of Oxidative Stress Signaling, Nrf2, on Survival and Stemness of Human Adipose-Derived Stem Cells Exposed to X-rays, Protons and Carbon Ions. Antioxidants (Basel) 2024; 13:1035. [PMID: 39334694 PMCID: PMC11429097 DOI: 10.3390/antiox13091035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/30/2024] Open
Abstract
Some cancers have a poor prognosis and often lead to local recurrence because they are resistant to available treatments, e.g., glioblastoma. Attempts have been made to increase the sensitivity of resistant tumors by targeting pathways involved in the resistance and combining it, for example, with radiotherapy (RT). We have previously reported that treating glioblastoma stem cells with an Nrf2 inhibitor increases their radiosensitivity. Unfortunately, the application of drugs can also affect normal cells. In the present study, we aim to investigate the role of the Nrf2 pathway in the survival and differentiation of normal human adipose-derived stem cells (ADSCs) exposed to radiation. We treated ADSCs with an Nrf2 inhibitor and then exposed them to X-rays, protons or carbon ions. All three radiation qualities are used to treat cancer. The survival and differentiation abilities of the surviving ADSCs were studied. We found that the enhancing effect of Nrf2 inhibition on cell survival levels was radiation-quality-dependent (X-rays > proton > carbon ions). Furthermore, our results indicate that Nrf2 inhibition reduces stem cell differentiation by 35% and 28% for adipogenesis and osteogenesis, respectively, using all applied radiation qualities. Interestingly, the results show that the cells that survive proton and carbon ion irradiations have an increased ability, compared with X-rays, to differentiate into osteogenesis and adipogenesis lineages. Therefore, we can conclude that the use of carbon ions or protons can affect the stemness of irradiated ADSCs at lower levels than X-rays and is thus more beneficial for long-time cancer survivors, such as pediatric patients.
Collapse
Affiliation(s)
- Mira Hammad
- Centre de Recherche sur les Ions, les Matériaux et la Photonique (CIMAP) UMR 6252, University of Caen Normandy, Cedex 04, F-14050 Caen, France
| | - Rima Salma
- Centre de Recherche sur les Ions, les Matériaux et la Photonique (CIMAP) UMR 6252, University of Caen Normandy, Cedex 04, F-14050 Caen, France
| | - Jacques Balosso
- Department of Radiation Oncology, Centre François Baclesse, F-14000 Caen, France
- Advanced Resource Center for HADrontherapy in Europe (ARCHADE), F-14000 Caen, France
| | - Mohi Rezvani
- Swiss Bioscience GmbH, Wagistrasse 27a, CH-8952 Schlieren, Switzerland
| | - Siamak Haghdoost
- Centre de Recherche sur les Ions, les Matériaux et la Photonique (CIMAP) UMR 6252, University of Caen Normandy, Cedex 04, F-14050 Caen, France
- Advanced Resource Center for HADrontherapy in Europe (ARCHADE), F-14000 Caen, France
- Le Laboratoire "Aliments, Bioprocédés, Toxicologie et Environnement (ABTE) UR 4651, ToxEMAC Team, University of Caen Normandy, Cedex 04, F-14050 Caen, France
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-10691 Stockholm, Sweden
| |
Collapse
|
6
|
Negoita RD, Ilisanu MA, Irimescu IN, Popescu RC, Tudor M, Mihailescu M, Scarlat EN, Pleava AM, Dinischiotu A, Savu D. Specific spectral sub-images for machine learning evaluation of optical differences between carbon ion and X ray radiation effects. Heliyon 2024; 10:e35249. [PMID: 39170121 PMCID: PMC11336423 DOI: 10.1016/j.heliyon.2024.e35249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 06/05/2024] [Accepted: 07/25/2024] [Indexed: 08/23/2024] Open
Abstract
Advances in radiotherapy, particularly the exploration of alternative radiation types such as carbon ions have updated our understanding of its effects and applicability on chondrosarcoma cells. Here we compare the optical effects produced by carbon ions (CI) and X-rays (XR) radiations on chondrosarcoma cells nuclei and set an automated method for evaluating the radiation-induced alterations without the need of chemical marking. Hyperspectral images (HSI) of SW1353 chondrosarcoma line carry detectable optical changes of the cells irradiated either with CI or XR compared to non-irradiated ones (REF). The differences between the spectral profiles of CI, XR and REF nuclei classes led to partitioning the HSIs into spectral sub-images. The changes are detected by support vector machine (SVM) classifiers whose performances are evaluated by the most used point metrics: sensitivity (SEN), accuracy (ACC), and precision (PREC), applied on spatial feature values. Specific interaction mechanisms by radiation type reveal distinct subintervals where HSIs changes are more prominent, and the classifiers perform at best. For CI the best classifiers are obtained for sub-images in the interval (424-436 nm), while for XR the best classifiers are obtained for sub-images in the interval (436-445 nm). The classifiers work better with texture features than roughness features in both cases. The classifier with the best SEN point metric in the testing phase is the most suitable to measure the irradiation efficiency irrespective of the radiation type. The altered nuclei are easier to discriminate when irradiated with CI than with XR. The study proves that SVM with optical data offers a rapid, automated, and label-free method for evaluating radiation-induced alterations in chondrosarcoma nuclei, thereby enabling effective analysis of extensive data.
Collapse
Affiliation(s)
- Raluca D. Negoita
- Applied Sciences Doctoral School, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, Bucharest, 060042, Romania
| | - Mihaela A. Ilisanu
- Doctoral School of Automatic Control and Computers, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, Bucharest, 060042, Romania
- Holographic Imaging and Processing Laboratory, Physics Department, Faculty of Applied Sciences, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, Bucharest, 060042, Romania
| | - Ionela N. Irimescu
- Applied Sciences Doctoral School, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, Bucharest, 060042, Romania
- Tehnoplus Medical SRL, 1 Odobesti str, Bucharest, Romania
| | - Roxana C. Popescu
- Department of Life and Environmental Physics, Horia Hulubei National Institute for R&D in Physics and Nuclear Engineering, Reactorului 30, P.O. Box MG-6, 077125 Magurele, Romania
- Department of Bioengineering and Biotechnology, Faculty of Medical Engineering, National University of Science and Technology Politehnica Bucharest, G. Polizu Street, 1-7, 011061 Bucharest, Romania
| | - Mihaela Tudor
- Department of Life and Environmental Physics, Horia Hulubei National Institute for R&D in Physics and Nuclear Engineering, Reactorului 30, P.O. Box MG-6, 077125 Magurele, Romania
- Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania
| | - Mona Mihailescu
- Holographic Imaging and Processing Laboratory, Physics Department, Faculty of Applied Sciences, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, Bucharest, 060042, Romania
- Research Centre in Fundamental Sciences Applied in Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, Bucharest, 060042, Romania
| | - Eugen N. Scarlat
- Holographic Imaging and Processing Laboratory, Physics Department, Faculty of Applied Sciences, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, Bucharest, 060042, Romania
| | - Ana M. Pleava
- CAMPUS Research Centre, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, Bucharest, 060042, Romania
| | - Anca Dinischiotu
- Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania
| | - Diana Savu
- Department of Life and Environmental Physics, Horia Hulubei National Institute for R&D in Physics and Nuclear Engineering, Reactorului 30, P.O. Box MG-6, 077125 Magurele, Romania
| |
Collapse
|
7
|
Gao Y, Wen P, Shao C, Ye C, Chen Y, You J, Su Z. CDC20 Holds Novel Regulation Mechanism in RPA1 during Different Stages of DNA Damage to Induce Radio-Chemoresistance. Int J Mol Sci 2024; 25:8383. [PMID: 39125953 PMCID: PMC11312485 DOI: 10.3390/ijms25158383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/25/2024] [Accepted: 07/28/2024] [Indexed: 08/12/2024] Open
Abstract
Targeting CDC20 can enhance the radiosensitivity of tumor cells, but the function and mechanism of CDC20 on DNA damage repair response remains vague. To examine that issue, tumor cell lines, including KYSE200, KYSE450, and HCT116, were utilized to detect the expression, function, and underlying mechanism of CDC20 in radio-chemoresistance. Western blot and immunofluorescence staining were employed to confirm CDC20 expression and location, and radiation could upregulate the expression of CDC20 in the cell nucleus. The homologous recombination (HR) and non-homologous end joining (NHEJ) reporter gene systems were utilized to explore the impact of CDC20 on DNA damage repair, indicating that CDC20 could promote HR repair and radio/chemo-resistance. In the early stages of DNA damage, CDC20 stabilizes the RPA1 protein through protein-protein interactions, activating the ATR-mediated signaling cascade, thereby aiding in genomic repair. In the later stages, CDC20 assists in the subsequent steps of damage repair by the ubiquitin-mediated degradation of RPA1. CCK-8 and colony formation assay were used to detect the function of CDC20 in cell vitality and proliferation, and targeting CDC20 can exacerbate the increase in DNA damage levels caused by cisplatin or etoposide. A tumor xenograft model was conducted in BALB/c-nu/nu mice to confirm the function of CDC20 in vivo, confirming the in vitro results. In conclusion, this study provides further validation of the potential clinical significance of CDC20 as a strategy to overcome radio-chemoresistance via uncovering a novel role of CDC20 in regulating RPA1 during DNA damage repair.
Collapse
Affiliation(s)
- Yang Gao
- Department of Histology and Embryology, Shantou University Medical College, Shantou 515041, China; (Y.G.); (C.S.); (C.Y.); (Y.C.); (J.Y.)
| | - Pengbo Wen
- School of Medical Information and Engineering, Xuzhou Medical University, Xuzhou 221002, China;
| | - Chenran Shao
- Department of Histology and Embryology, Shantou University Medical College, Shantou 515041, China; (Y.G.); (C.S.); (C.Y.); (Y.C.); (J.Y.)
| | - Cheng Ye
- Department of Histology and Embryology, Shantou University Medical College, Shantou 515041, China; (Y.G.); (C.S.); (C.Y.); (Y.C.); (J.Y.)
| | - Yuji Chen
- Department of Histology and Embryology, Shantou University Medical College, Shantou 515041, China; (Y.G.); (C.S.); (C.Y.); (Y.C.); (J.Y.)
| | - Junyu You
- Department of Histology and Embryology, Shantou University Medical College, Shantou 515041, China; (Y.G.); (C.S.); (C.Y.); (Y.C.); (J.Y.)
| | - Zhongjing Su
- Department of Histology and Embryology, Shantou University Medical College, Shantou 515041, China; (Y.G.); (C.S.); (C.Y.); (Y.C.); (J.Y.)
| |
Collapse
|
8
|
Hirano SI, Takefuji Y. Molecular Hydrogen Protects against Various Tissue Injuries from Side Effects of Anticancer Drugs by Reducing Oxidative Stress and Inflammation. Biomedicines 2024; 12:1591. [PMID: 39062164 PMCID: PMC11274581 DOI: 10.3390/biomedicines12071591] [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: 05/08/2024] [Revised: 07/07/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
While drug therapy plays a crucial role in cancer treatment, many anticancer drugs, particularly cytotoxic and molecular-targeted drugs, cause severe side effects, which often limit the dosage of these drugs. Efforts have been made to alleviate these side effects by developing derivatives, analogues, and liposome formulations of existing anticancer drugs and by combining anticancer drugs with substances that reduce side effects. However, these approaches have not been sufficiently effective in reducing side effects. Molecular hydrogen (H2) has shown promise in this regard. It directly reduces reactive oxygen species, which have very strong oxidative capacity, and indirectly exerts antioxidant, anti-inflammatory, and anti-apoptotic effects by regulating gene expression. Its clinical application in various diseases has been expanded worldwide. Although H2 has been reported to reduce the side effects of anticancer drugs in animal studies and clinical trials, the underlying molecular mechanisms remain unclear. Our comprehensive literature review revealed that H2 protects against tissue injuries induced by cisplatin, oxaliplatin, doxorubicin, bleomycin, and gefitinib. The underlying mechanisms involve reductions in oxidative stress and inflammation. H2 itself exhibits anticancer activity. Therefore, the combination of H2 and anticancer drugs has the potential to reduce the side effects of anticancer drugs and enhance their anticancer activities. This is an exciting prospect for future cancer treatments.
Collapse
Affiliation(s)
- Shin-ichi Hirano
- Independent Researcher, 5-8-1-207 Honson, Chigasaki 253-0042, Japan
| | - Yoshiyasu Takefuji
- Keio University, 2-15-45 Mita, Minato-ku, Tokyo 108-8345, Japan;
- Faculty of Data Science, Musashino University, 3-3-3 Ariake, Koto-Ku, Tokyo 135-8181, Japan
| |
Collapse
|
9
|
Toprani SM, Scheibler C, Mordukhovich I, McNeely E, Nagel ZD. Cosmic Ionizing Radiation: A DNA Damaging Agent That May Underly Excess Cancer in Flight Crews. Int J Mol Sci 2024; 25:7670. [PMID: 39062911 PMCID: PMC11277465 DOI: 10.3390/ijms25147670] [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: 06/04/2024] [Revised: 06/20/2024] [Accepted: 06/30/2024] [Indexed: 07/28/2024] Open
Abstract
In the United States, the Federal Aviation Administration has officially classified flight crews (FC) consisting of commercial pilots, cabin crew, or flight attendants as "radiation workers" since 1994 due to the potential for cosmic ionizing radiation (CIR) exposure at cruising altitudes originating from solar activity and galactic sources. Several epidemiological studies have documented elevated incidence and mortality for several cancers in FC, but it has not yet been possible to establish whether this is attributable to CIR. CIR and its constituents are known to cause a myriad of DNA lesions, which can lead to carcinogenesis unless DNA repair mechanisms remove them. But critical knowledge gaps exist with regard to the dosimetry of CIR, the role of other genotoxic exposures among FC, and whether possible biological mechanisms underlying higher cancer rates observed in FC exist. This review summarizes our understanding of the role of DNA damage and repair responses relevant to exposure to CIR in FC. We aimed to stimulate new research directions and provide information that will be useful for guiding regulatory, public health, and medical decision-making to protect and mitigate the risks for those who travel by air.
Collapse
Affiliation(s)
- Sneh M. Toprani
- John B. Little Center for Radiation Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA;
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (C.S.); (I.M.); (E.M.)
| | - Christopher Scheibler
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (C.S.); (I.M.); (E.M.)
| | - Irina Mordukhovich
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (C.S.); (I.M.); (E.M.)
- Sustainability and Health Initiative (SHINE), Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA 02115, USA
| | - Eileen McNeely
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (C.S.); (I.M.); (E.M.)
- Sustainability and Health Initiative (SHINE), Department of Environmental Health, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA 02115, USA
| | - Zachary D. Nagel
- John B. Little Center for Radiation Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA;
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (C.S.); (I.M.); (E.M.)
| |
Collapse
|
10
|
Forenzo C, Larsen J. Bridging clinical radiotherapy and space radiation therapeutics through reactive oxygen species (ROS)-triggered delivery. Free Radic Biol Med 2024; 219:88-103. [PMID: 38631648 DOI: 10.1016/j.freeradbiomed.2024.04.219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/15/2024] [Accepted: 04/09/2024] [Indexed: 04/19/2024]
Abstract
This review explores the convergence of clinical radiotherapy and space radiation therapeutics, focusing on ionizing radiation (IR)-generated reactive oxygen species (ROS). IR, with high-energy particles, induces precise cellular damage, particularly in cancer treatments. The paper discusses parallels between clinical and space IR, highlighting unique characteristics of high-charge and energy particles in space and potential health risks for astronauts. Emphasizing the parallel occurrence of ROS generation in both clinical and space contexts, the review identifies ROS as a crucial factor with dual roles in cellular responses and potential disease initiation. The analysis covers ROS generation mechanisms, variations, and similarities in terrestrial and extraterrestrial environments leading to innovative ROS-responsive delivery systems adaptable for both clinical and space applications. The paper concludes by discussing applications of personalized ROS-triggered therapeutic approaches and discussing the challenges and prospects of implementing these strategies in clinical radiotherapy and extraterrestrial missions. Overall, it underscores the potential of ROS-targeted delivery for advancing therapeutic strategies in terrestrial clinical settings and space exploration, contributing to human health improvement on Earth and beyond.
Collapse
Affiliation(s)
- Chloe Forenzo
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, SC, 29631, USA
| | - Jessica Larsen
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, SC, 29631, USA; Department of Bioengineering, Clemson University, Clemson, SC, 29631, USA.
| |
Collapse
|
11
|
Chattaraj A, Selvam TP. Radiation-induced DNA damage by proton, helium and carbon ions in human fibroblast cell: Geant4-DNA and MCDS-based study. Biomed Phys Eng Express 2024; 10:045059. [PMID: 38870909 DOI: 10.1088/2057-1976/ad57ce] [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: 01/25/2024] [Accepted: 06/13/2024] [Indexed: 06/15/2024]
Abstract
Background. Radiation-induced DNA damages such as Single Strand Break (SSB), Double Strand Break (DSB) and Complex DSB (cDSB) are critical aspects of radiobiology with implications in radiotherapy and radiation protection applications.Materials and Methods. This study presents a thorough investigation into the effects of protons (0.1-100 MeV/u), helium ions (0.13-100 MeV/u) and carbon ions (0.5-480 MeV/u) on DNA of human fibroblast cells using Geant4-DNA track structure code coupled with DBSCAN algorithm and Monte Carlo Damage Simulations (MCDS) code. Geant4-DNA-based simulations consider 1μm × 1μm × 0.5μm water box as the target to calculate energy deposition on event-by-event basis and the three-dimensional coordinates of the interaction location, and then DBSCAN algorithm is used to calculate yields of SSB, DSB and cDSB in human fibroblast cell. The study investigated the influence of Linear Energy Transfer (LET) of protons, helium ions and carbon ions on the yields of DNA damages. Influence of cellular oxygenation on DNA damage patterns is investigated using MCDS code.Results. The study shows that DSB and SSB yields are influenced by the LET of the particles, with distinct trends observed for different particles. The cellular oxygenation is a key factor, with anoxic cells exhibiting reduced SSB and DSB yields, underscoring the intricate relationship between cellular oxygen levels and DNA damage. The study introduced DSB/SSB ratio as an informative metric for evaluating the severity of radiation-induced DNA damage, particularly in higher LET regions.Conclusions. The study highlights the importance of considering particle type, LET, and cellular oxygenation in assessing the biological effects of ionizing radiation.
Collapse
Affiliation(s)
- Arghya Chattaraj
- Radiological Physics and Advisory Division, Health, Safety and Environment Group, Bhabha Atomic Research Centre, Mumbai, 400085, India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai, 400094, India
| | - T Palani Selvam
- Radiological Physics and Advisory Division, Health, Safety and Environment Group, Bhabha Atomic Research Centre, Mumbai, 400085, India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai, 400094, India
| |
Collapse
|
12
|
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
|
13
|
Teh S, Bowland K, Halper-Stromberg E, Kotwal A, Bennett A, Skaist A, Tang J, Cai F, Macoretta A, Liang H, Kamiyama H, Wheelan S, Lin MT, Hruban R, Hung CF, Goldstein M, Scharpf R, Roberts N, Eshleman J. CRISPR-Cas9 for selective targeting of somatic mutations in pancreatic cancers. NAR Cancer 2024; 6:zcae028. [PMID: 38915758 PMCID: PMC11195629 DOI: 10.1093/narcan/zcae028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 05/23/2024] [Accepted: 06/03/2024] [Indexed: 06/26/2024] Open
Abstract
Somatic mutations are desirable targets for selective elimination of cancer, yet most are found within noncoding regions. We have adapted the CRISPR-Cas9 gene editing tool as a novel, cancer-specific killing strategy by targeting the subset of somatic mutations that create protospacer adjacent motifs (PAMs), which have evolutionally allowed bacterial cells to distinguish between self and non-self DNA for Cas9-induced double strand breaks. Whole genome sequencing (WGS) of paired tumor minus normal (T-N) samples from three pancreatic cancer patients (Panc480, Panc504, and Panc1002) showed an average of 417 somatic PAMs per tumor produced from single base substitutions. Further analyses of 591 paired T-N samples from The International Cancer Genome Consortium found medians of ∼455 somatic PAMs per tumor in pancreatic, ∼2800 in lung, and ∼3200 in esophageal cancer cohorts. Finally, we demonstrated 69-99% selective cell death of three targeted pancreatic cancer cell lines using 4-9 sgRNAs designed using the somatic PAM discovery approach. We also showed no off-target activity from these tumor-specific sgRNAs in either the patient's normal cells or an irrelevant cancer using WGS. This study demonstrates the potential of CRISPR-Cas9 as a novel and selective anti-cancer strategy, and supports the genetic targeting of adult cancers.
Collapse
Affiliation(s)
- Selina Shiqing K Teh
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kirsten Bowland
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eitan Halper-Stromberg
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Akhil Kotwal
- Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alexis Bennett
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alyza Skaist
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jacqueline Tang
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Fidel Cai
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Antonella Macoretta
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hong Liang
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Sarah Wheelan
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Scientific Review Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - Ming-Tseh Lin
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ralph H Hruban
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chien-Fu Hung
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael Goldstein
- Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert B Scharpf
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nicholas J Roberts
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - James R Eshleman
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
14
|
Zhang D, Li L, Li M, Cao X. Biological functions and clinic significance of SAF‑A (Review). Biomed Rep 2024; 20:88. [PMID: 38665420 PMCID: PMC11040223 DOI: 10.3892/br.2024.1776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
As one member of the heterogeneous ribonucleoprotein (hnRNP) family, scaffold attachment factor A (SAF-A) or hnRNP U, is an abundant nuclear protein. With RNA and DNA binding activities, SAF-A has multiple functions. The present review focused on the biological structure and different roles of SAF-A and SAF-A-related diseases. It was found that SAF-A maintains the higher-order chromatin organization via RNA and DNA, and regulates transcription at the initiation and elongation stages. In addition to regulating pre-mRNA splicing, mRNA transportation and stabilization, SAF-A participates in double-strand breaks and mitosis repair. Therefore, the aberrant expression and mutation of SAF-A results in tumors and impaired neurodevelopment. Moreover, SAF-A may play a role in the anti-virus system. In conclusion, due to its essential biological functions, SAF-A may be a valuable clinical prediction factor or therapeutic target. Since the role of SAF-A in tumors and viral infections may be controversial, more animal experiments and clinical assays are needed.
Collapse
Affiliation(s)
- Daiquan Zhang
- Department of Traditional Chinese Medicine, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Li Li
- Immune Mechanism and Therapy of Major Diseases of Luzhou Key Laboratory, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Mengni Li
- Department of Pediatrics, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Xinmei Cao
- Immune Mechanism and Therapy of Major Diseases of Luzhou Key Laboratory, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
15
|
Rajpurohit YS, Sharma DK, Lal M, Soni I. A perspective on tumor radiation resistance following high-LET radiation treatment. J Cancer Res Clin Oncol 2024; 150:226. [PMID: 38696003 PMCID: PMC11065934 DOI: 10.1007/s00432-024-05757-8] [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: 02/24/2024] [Accepted: 04/22/2024] [Indexed: 05/05/2024]
Abstract
High-linear energy transfer (LET) radiation is a promising alternative to conventional low-LET radiation for therapeutic gain against cancer owing to its ability to induce complex and clustered DNA lesions. However, the development of radiation resistance poses a significant barrier. The potential molecular mechanisms that could confer resistance development are translesion synthesis (TLS), replication gap suppression (RGS) mechanisms, autophagy, epithelial-mesenchymal transition (EMT) activation, release of exosomes, and epigenetic changes. This article will discuss various types of complex clustered DNA damage, their repair mechanisms, mutagenic potential, and the development of radiation resistance strategies. Furthermore, it highlights the importance of careful consideration and patient selection when employing high-LET radiotherapy in clinical settings.
Collapse
Affiliation(s)
- Yogendra Singh Rajpurohit
- Molecular Biology Division, Bhabha Atomic Research Centre, 2-46-S, Modular Lab, A-Block, Mumbai, 400085, India.
- Homi Bhabha National Institute, DAE- Deemed University, Mumbai, 400094, India.
| | - Dhirendra Kumar Sharma
- Molecular Biology Division, Bhabha Atomic Research Centre, 2-46-S, Modular Lab, A-Block, Mumbai, 400085, India
| | - Mitu Lal
- Molecular Biology Division, Bhabha Atomic Research Centre, 2-46-S, Modular Lab, A-Block, Mumbai, 400085, India
| | - Ishu Soni
- Homi Bhabha National Institute, DAE- Deemed University, Mumbai, 400094, India
| |
Collapse
|
16
|
Bivehed E, Hellman B, Wenson L, Stenerlöw B, Söderberg O, Heldin J. Visualizing DNA single- and double-strand breaks in the Flash comet assay by DNA polymerase-assisted end-labelling. Nucleic Acids Res 2024; 52:e22. [PMID: 38261985 PMCID: PMC10899772 DOI: 10.1093/nar/gkae009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 12/14/2023] [Accepted: 01/02/2024] [Indexed: 01/25/2024] Open
Abstract
In the comet assay, tails are formed after single-cell gel electrophoresis if the cells have been exposed to genotoxic agents. These tails include a mixture of both DNA single-strand breaks (SSBs) and double-strand breaks (DSBs). However, these two types of strand breaks cannot be distinguished using comet assay protocols with conventional DNA stains. Since DSBs are more problematic for the cells, it would be useful if the SSBs and DSBs could be differentially identified in the same comet. In order to be able to distinguish between SSBs and DSBs, we designed a protocol for polymerase-assisted DNA damage analysis (PADDA) to be used in combination with the Flash comet protocol, or on fixed cells. By using DNA polymerase I to label SSBs and terminal deoxynucleotidyl transferase to label DSBs with fluorophore-labelled nucleotides. Herein, TK6-cells or HaCat cells were exposed to either hydrogen peroxide (H2O2), ionising radiation (X-rays) or DNA cutting enzymes, and then subjected to a comet protocol followed by PADDA. PADDA offers a wider detection range, unveiling previously undetected DNA strand breaks.
Collapse
Affiliation(s)
- Erik Bivehed
- Department of Pharmaceutical Biosciences, Uppsala University, Biomedical Centre, Uppsala SE-751 24, Sweden
| | - Björn Hellman
- Department of Pharmaceutical Biosciences, Uppsala University, Biomedical Centre, Uppsala SE-751 24, Sweden
| | - Leonie Wenson
- Department of Pharmaceutical Biosciences, Uppsala University, Biomedical Centre, Uppsala SE-751 24, Sweden
| | - Bo Stenerlöw
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, Uppsala SE-751 85, Sweden
| | - Ola Söderberg
- Department of Pharmaceutical Biosciences, Uppsala University, Biomedical Centre, Uppsala SE-751 24, Sweden
| | - Johan Heldin
- Department of Pharmaceutical Biosciences, Uppsala University, Biomedical Centre, Uppsala SE-751 24, Sweden
| |
Collapse
|
17
|
Ilicic K, Dollinger G, Dombrowsky A, Greubel C, Girst S, Sammer M, Siebenwirth C, Schmid E, Friedrich T, Kundrát P, Friedland W, Scholz M, Combs SE, Schmid TE, Reindl J. Enhanced RBE of Particle Radiation Depends on Beam Size in the Micrometer Range. Radiat Res 2024; 201:140-149. [PMID: 38214379 DOI: 10.1667/rade-23-00217.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/01/2023] [Indexed: 01/13/2024]
Abstract
High-linear energy transfer (LET) radiation, such as heavy ions is associated with a higher relative biological effectiveness (RBE) than low-LET radiation, such as photons. Irradiation with low- and high-LET particles differ in the interaction with the cellular matter and therefore in the spatial dose distribution. When a single high-LET particle interacts with matter, it results in doses of up to thousands of gray (Gy) locally concentrated around the ion trajectory, whereas the mean dose averaged over the target, such as a cell nucleus is only in the range of a Gy. DNA damage therefore accumulates in this small volume. In contrast, up to hundreds of low-LET particle hits are required to achieve the same mean dose, resulting in a quasi-homogeneous damage distribution throughout the cell nucleus. In this study, we investigated the dependence of RBE from different spatial dose depositions using different focused beam spot sizes of proton radiation with respect to the induction of chromosome aberrations and clonogenic cell survival. Human-hamster hybrid (AL) as well as Chinese hamster ovary cells (CHO-K1) were irradiated with focused low LET protons of 20 MeV (LET = 2.6 keV/µm) beam energy with a mean dose of 1.7 Gy in a quadratic matrix pattern with point spacing of 5.4 × 5.4 µm2 and 117 protons per matrix point at the ion microbeam SNAKE using different beam spot sizes between 0.8 µm and 2.8 µm (full width at half maximum). The dose-response curves of X-ray reference radiation were used to determine the RBE after a 1.7 Gy dose of radiation. The RBE for the induction of dicentric chromosomes and cell inactivation was increased after irradiation with the smallest beam spot diameter (0.8 µm for chromosome aberration experiments and 1.0 µm for cell survival experiments) compared to homogeneous proton radiation but was still below the RBE of a corresponding high LET single ion hit. By increasing the spot size to 1.6-1.8 µm, the RBE decreased but was still higher than for homogeneously distributed protons. By further increasing the spot size to 2.7-2.8 µm, the RBE was no longer different from the homogeneous radiation. Our experiments demonstrate that varying spot size of low-LET radiation gradually modifies the RBE. This underlines that a substantial fraction of enhanced RBE originates from inhomogeneous energy concentrations on the µm scale (mean intertrack distances of low-LET particles below 0.1 µm) and quantifies the link between such energy concentration and RBE. The missing fraction of RBE enhancement when comparing with high-LET ions is attributed to the high inner track energy deposition on the nanometer scale. The results are compared with model results of PARTRAC and LEM for chromosomal aberration and cell survival, respectively, which suggest mechanistic interpretations of the observed radiation effects.
Collapse
Affiliation(s)
- K Ilicic
- Institute of Radiation Medicine (IRM), Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Department of Radiation Oncology, School of Medicine, Technische Universität München, Germany
| | - G Dollinger
- Institute for Applied Physics and Measurement Technology, Universität der Bundeswehr München, Neubiberg, Germany
| | - A Dombrowsky
- Institute of Radiation Medicine (IRM), Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Department of Radiation Oncology, School of Medicine, Technische Universität München, Germany
| | - C Greubel
- Institute for Applied Physics and Measurement Technology, Universität der Bundeswehr München, Neubiberg, Germany
| | - S Girst
- Institute for Applied Physics and Measurement Technology, Universität der Bundeswehr München, Neubiberg, Germany
| | - M Sammer
- Institute for Applied Physics and Measurement Technology, Universität der Bundeswehr München, Neubiberg, Germany
| | - C Siebenwirth
- Department of Radiation Oncology, School of Medicine, Technische Universität München, Germany
- Institute for Applied Physics and Measurement Technology, Universität der Bundeswehr München, Neubiberg, Germany
| | - E Schmid
- Department for Anatomy and Cell Biology, Ludwig-Maximilians Universität München, Germany
| | - T Friedrich
- GSI Helmholtz Center for Heavy Ion Research, Department of Biophysics, Darmstadt, Germany
| | - P Kundrát
- GSI Helmholtz Center for Heavy Ion Research, Department of Biophysics, Darmstadt, Germany
| | - W Friedland
- Institute of Radiation Medicine (IRM), Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - M Scholz
- GSI Helmholtz Center for Heavy Ion Research, Department of Biophysics, Darmstadt, Germany
| | - S E Combs
- Institute of Radiation Medicine (IRM), Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Department of Radiation Oncology, School of Medicine, Technische Universität München, Germany
| | - T E Schmid
- Institute of Radiation Medicine (IRM), Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Department of Radiation Oncology, School of Medicine, Technische Universität München, Germany
| | - J Reindl
- Institute for Applied Physics and Measurement Technology, Universität der Bundeswehr München, Neubiberg, Germany
| |
Collapse
|
18
|
Khorasani M. Role of cGAS-STING in colorectal cancer: A new window for treatment strategies. Cytokine 2024; 173:156422. [PMID: 37948979 DOI: 10.1016/j.cyto.2023.156422] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 10/29/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023]
Abstract
Colorectal cancer (CRC) is a common and deadly form of cancer, leading to the need for new therapeutic targets and strategies for treatment. Recent studies have shown the cGAS-STING pathway to be a promising target for cancer therapy. The cGAS-STING pathway is a part of the innate immune system and serves to identify DNA damage and viral infection, promoting an immune response. Activation of this pathway leads to the production of immune mediators, such as type I interferons, that activate immune cells to attack cancer cells. Research has identified the cGAS-STING pathway as a frequently dysregulated component in CRC, promoting tumor growth and metastasis, or leading to chronic inflammation and tissue damage. The modulation of this pathway presents a potential therapeutic approach, either activating or inhibiting the pathway to enhance the immune response and prevent inflammation, respectively. Developing drugs that can modulate the cGAS-STING pathway offers promise for improving treatment outcomes for CRC patients. The present review explores recent research on the role of cGAS-STING in CRC and highlights the potential therapeutic benefits of targeting this pathway.
Collapse
Affiliation(s)
- Milad Khorasani
- Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur, Iran; Department of Biochemistry and Nutrition, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| |
Collapse
|
19
|
Helm A, Fournier C. High-LET charged particles: radiobiology and application for new approaches in radiotherapy. Strahlenther Onkol 2023; 199:1225-1241. [PMID: 37872399 PMCID: PMC10674019 DOI: 10.1007/s00066-023-02158-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 09/17/2023] [Indexed: 10/25/2023]
Abstract
The number of patients treated with charged-particle radiotherapy as well as the number of treatment centers is increasing worldwide, particularly regarding protons. However, high-linear energy transfer (LET) particles, mainly carbon ions, are of special interest for application in radiotherapy, as their special physical features result in high precision and hence lower toxicity, and at the same time in increased efficiency in cell inactivation in the target region, i.e., the tumor. The radiobiology of high-LET particles differs with respect to DNA damage repair, cytogenetic damage, and cell death type, and their increased LET can tackle cells' resistance to hypoxia. Recent developments and perspectives, e.g., the return of high-LET particle therapy to the US with a center planned at Mayo clinics, the application of carbon ion radiotherapy using cost-reducing cyclotrons and the application of helium is foreseen to increase the interest in this type of radiotherapy. However, further preclinical research is needed to better understand the differential radiobiological mechanisms as opposed to photon radiotherapy, which will help to guide future clinical studies for optimal exploitation of high-LET particle therapy, in particular related to new concepts and innovative approaches. Herein, we summarize the basics and recent progress in high-LET particle radiobiology with a focus on carbon ions and discuss the implications of current knowledge for charged-particle radiotherapy. We emphasize the potential of high-LET particles with respect to immunogenicity and especially their combination with immunotherapy.
Collapse
Affiliation(s)
- Alexander Helm
- Biophysics Department, GSI Helmholtz Center for Heavy Ion Research, Darmstadt, Germany
| | - Claudia Fournier
- Biophysics Department, GSI Helmholtz Center for Heavy Ion Research, Darmstadt, Germany.
| |
Collapse
|
20
|
Noonepalle SKR, Grindrod S, Aghdam N, Li X, Gracia-Hernandez M, Zevallos-Delgado C, Jung M, Villagra A, Dritschilo A. Radiotherapy-induced Immune Response Enhanced by Selective HDAC6 Inhibition. Mol Cancer Ther 2023; 22:1376-1389. [PMID: 37586844 PMCID: PMC10878032 DOI: 10.1158/1535-7163.mct-23-0215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 07/05/2023] [Accepted: 08/15/2023] [Indexed: 08/18/2023]
Abstract
Radiotherapy is a curative cancer treatment modality that imparts damage to cellular DNA, induces immunogenic cell death, and activates antitumor immunity. Despite the radiotherapy-induced direct antitumor effect seen within the treated volume, accumulating evidence indicates activation of innate antitumor immunity. Acute proinflammatory responses mediated by anticancer M1 macrophages are observed in the immediate aftermath following radiotherapy. However, after a few days, these M1 macrophages are converted to anti-inflammatory and pro-cancer M2 phenotype, leading to cancer resistance and underlying potential tumor relapse. Histone deacetylase 6 (HDAC6) plays a crucial role in regulating macrophage polarization and innate immune responses. Here, we report targeting HDAC6 function with a novel selective inhibitor (SP-2-225) as a potential therapeutic candidate for combination therapy with radiotherapy. This resulted in decreased tumor growth and enhanced M1/M2 ratio of infiltrating macrophages within tumors. These observations support the use of selective HDAC6 inhibitors to improve antitumor immune responses and prevent tumor relapse after radiotherapy.
Collapse
Affiliation(s)
- Satish Kumar R. Noonepalle
- Department of Oncology, Georgetown University Lombardi Comprehensive Cancer Center, Washington, District of Columbia
| | | | - Nima Aghdam
- Department of Radiation Medicine, Georgetown University Medical Center, Washington, District of Columbia
| | - Xintang Li
- Department of Oncology, Georgetown University Lombardi Comprehensive Cancer Center, Washington, District of Columbia
| | - Maria Gracia-Hernandez
- Department of Oncology, Georgetown University Lombardi Comprehensive Cancer Center, Washington, District of Columbia
| | - Christian Zevallos-Delgado
- Department of Oncology, Georgetown University Lombardi Comprehensive Cancer Center, Washington, District of Columbia
| | - Mira Jung
- Department of Radiation Medicine, Georgetown University Medical Center, Washington, District of Columbia
| | - Alejandro Villagra
- Department of Oncology, Georgetown University Lombardi Comprehensive Cancer Center, Washington, District of Columbia
| | - Anatoly Dritschilo
- Shuttle Pharmaceuticals, Inc., Rockville, Maryland
- Department of Radiation Medicine, Georgetown University Medical Center, Washington, District of Columbia
| |
Collapse
|
21
|
Nickoloff JA, Jaiswal AS, Sharma N, Williamson EA, Tran MT, Arris D, Yang M, Hromas R. Cellular Responses to Widespread DNA Replication Stress. Int J Mol Sci 2023; 24:16903. [PMID: 38069223 PMCID: PMC10707325 DOI: 10.3390/ijms242316903] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/22/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Replicative DNA polymerases are blocked by nearly all types of DNA damage. The resulting DNA replication stress threatens genome stability. DNA replication stress is also caused by depletion of nucleotide pools, DNA polymerase inhibitors, and DNA sequences or structures that are difficult to replicate. Replication stress triggers complex cellular responses that include cell cycle arrest, replication fork collapse to one-ended DNA double-strand breaks, induction of DNA repair, and programmed cell death after excessive damage. Replication stress caused by specific structures (e.g., G-rich sequences that form G-quadruplexes) is localized but occurs during the S phase of every cell division. This review focuses on cellular responses to widespread stress such as that caused by random DNA damage, DNA polymerase inhibition/nucleotide pool depletion, and R-loops. Another form of global replication stress is seen in cancer cells and is termed oncogenic stress, reflecting dysregulated replication origin firing and/or replication fork progression. Replication stress responses are often dysregulated in cancer cells, and this too contributes to ongoing genome instability that can drive cancer progression. Nucleases play critical roles in replication stress responses, including MUS81, EEPD1, Metnase, CtIP, MRE11, EXO1, DNA2-BLM, SLX1-SLX4, XPF-ERCC1-SLX4, Artemis, XPG, FEN1, and TATDN2. Several of these nucleases cleave branched DNA structures at stressed replication forks to promote repair and restart of these forks. We recently defined roles for EEPD1 in restarting stressed replication forks after oxidative DNA damage, and for TATDN2 in mitigating replication stress caused by R-loop accumulation in BRCA1-defective cells. We also discuss how insights into biological responses to genome-wide replication stress can inform novel cancer treatment strategies that exploit synthetic lethal relationships among replication stress response factors.
Collapse
Affiliation(s)
- Jac A. Nickoloff
- Department of Environmental and Radiological Health Sciences, Colorado State University, Ft. Collins, CO 80523, USA
| | - Aruna S. Jaiswal
- Department of Medicine and the Mays Cancer Center, The University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA; (A.S.J.); (M.T.T.); (R.H.)
| | - Neelam Sharma
- Department of Environmental and Radiological Health Sciences, Colorado State University, Ft. Collins, CO 80523, USA
| | - Elizabeth A. Williamson
- Department of Medicine and the Mays Cancer Center, The University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA; (A.S.J.); (M.T.T.); (R.H.)
| | - Manh T. Tran
- Department of Medicine and the Mays Cancer Center, The University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA; (A.S.J.); (M.T.T.); (R.H.)
| | - Dominic Arris
- Department of Medicine and the Mays Cancer Center, The University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA; (A.S.J.); (M.T.T.); (R.H.)
| | - Ming Yang
- Department of Medicine and the Mays Cancer Center, The University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA; (A.S.J.); (M.T.T.); (R.H.)
| | - Robert Hromas
- Department of Medicine and the Mays Cancer Center, The University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA; (A.S.J.); (M.T.T.); (R.H.)
| |
Collapse
|
22
|
van Barele M, Akdeniz D, Heemskerk-Gerritsen BAM, Andrieu N, Noguès C, van Asperen CJ, Wevers M, Ausems MGEM, de Bock GH, Dommering CJ, Gómez-García EB, van Leeuwen FE, Mooij TM, Easton DF, Antoniou AC, Evans DG, Izatt L, Tischkowitz M, Frost D, Brewer C, Olah E, Simard J, Singer CF, Thomassen M, Kast K, Rhiem K, Engel C, de la Hoya M, Foretová L, Jakubowska A, Jager A, Sattler MGA, Schmidt MK, Hooning MJ. Contralateral breast cancer risk in patients with breast cancer and a germline-BRCA1/2 pathogenic variant undergoing radiation. J Natl Cancer Inst 2023; 115:1318-1328. [PMID: 37369040 PMCID: PMC10637040 DOI: 10.1093/jnci/djad116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 03/06/2023] [Accepted: 05/27/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Radiation-induced secondary breast cancer (BC) may be a concern after radiation therapy (RT) for primary breast cancer (PBC), especially in young patients with germline (g)BRCA-associated BC who already have high contralateral BC (CBC) risk and potentially increased genetic susceptibility to radiation. We sought to investigate whether adjuvant RT for PBC increases the risk of CBC in patients with gBRCA1/2-associated BC. METHODS The gBRCA1/2 pathogenic variant carriers diagnosed with PBC were selected from the prospective International BRCA1/2 Carrier Cohort Study. We used multivariable Cox proportional hazards models to investigate the association between RT (yes vs no) and CBC risk. We further stratified for BRCA status and age at PBC diagnosis (<40 and >40 years). Statistical significance tests were 2-sided. RESULTS Of 3602 eligible patients, 2297 (64%) received adjuvant RT. Median follow-up was 9.6 years. The RT group had more patients with stage III PBC than the non-RT group (15% vs 3%, P < .001), received chemotherapy more often (81% vs 70%, P < .001), and received endocrine therapy more often (50% vs 35%, P < .001). The RT group had an increased CBC risk compared with the non-RT group (adjusted hazard ratio [HR] = 1.44; 95% confidence interval [CI] = 1.12 to 1.86). Statistical significance was observed in gBRCA2 (HR = 1.77; 95% CI = 1.13 to 2.77) but not in gBRCA1 pathogenic variant carriers (HR = 1.29; 95% CI = 0.93 to 1.77; P = .39 for interaction). In the combined gBRCA1/2 group, patients irradiated when they were younger than or older than 40 years of age at PBC diagnosis showed similar risks (HR = 1.38; 95% CI = 0.93 to 2.04 and HR = 1.56; 95% CI = 1.11 to 2.19, respectively). CONCLUSIONS RT regimens minimizing contralateral breast dose should be considered in gBRCA1/2 pathogenic variant carriers.
Collapse
Affiliation(s)
- Mark van Barele
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Delal Akdeniz
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | | | - Genepso
- Département d’Anticipation et de Suivi des Cancers, Oncogénétique Clinique, Institut Paoli-Calmettes, Marseille, France
| | - Nadine Andrieu
- INSERM, U900, Paris, France
- Institut Curie, Paris, France
- PSL Research University, Paris, France
- Mines ParisTech, Fontainebleau, France
| | - Catherine Noguès
- Département d’Anticipation et de Suivi des Cancers, Oncogénétique Clinique, Institut Paoli-Calmettes, Marseille, France
- Institut Paoli-Calmettes & Aix Marseille University, INSERM, IRD, SESSTIM, Marseille, France
| | - HEBON
- The Hereditary Breast and Ovarian Cancer Research Group Netherlands (HEBON), Coordinating Center, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Christi J van Asperen
- Department of Clinical Genetics, Leiden University Medical Centre, Leiden, the Netherlands
| | - Marijke Wevers
- Department for Clinical Genetics, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Margreet G E M Ausems
- Division of Laboratories, Pharmacy and Biomedical Genetics, Department of Genetics, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Geertruida H de Bock
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Charlotte J Dommering
- Department of Clinical Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | | | - Flora E van Leeuwen
- Division of Psychosocial Research and Epidemiology, Netherlands Cancer Institute–Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Thea M Mooij
- Division of Psychosocial Research and Epidemiology, Netherlands Cancer Institute–Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - EMBRACE
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Douglas F Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Antonis C Antoniou
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - D Gareth Evans
- The Prevent Breast Cancer Research Unit, The Nightingale Centre, Manchester University NHS Foundation Trust, Manchester, UK
- Genomic Medicine, Division of Evolution and Genomic Sciences, The University of Manchester, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
- Manchester Breast Centre, Oglesby Cancer Research Centre, The Christie, University of Manchester, Manchester, UK
| | - Louise Izatt
- Department of Clinical Genetics, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
| | - Marc Tischkowitz
- Department of Medical Genetics, National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
- Program in Cancer Genetics, Departments of Human Genetics and Oncology, McGill University, Montréal, QC, Canada
| | - Debra Frost
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Carole Brewer
- Department of Clinical Genetics, Royal Devon & Exeter Hospital, Exeter, UK
| | - Edit Olah
- Department of Molecular Genetics, National Institute of Oncology, Budapest, Hungary
| | - Jacques Simard
- Genomics Center, Centre Hospitalier Universitaire de Québec, Université Laval Research Center, Quebec City, QC, Canada
| | - Christian F Singer
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Mads Thomassen
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Centre for Personalized Response Monitoring in Oncology (PREMIO), Odense University Hospital, Odense, Denmark
| | - Karin Kast
- Center of Familial Breast and Ovarian Cancer and Center of Integrated Oncology, University Hospital Cologne, Cologne, Germany
| | - Kerstin Rhiem
- Center of Familial Breast and Ovarian Cancer and Center of Integrated Oncology, University Hospital Cologne, Cologne, Germany
| | - Christoph Engel
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Miguel de la Hoya
- Molecular Oncology Laboratory, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - Lenka Foretová
- Department of Cancer Epidemiology and Genetics, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Anna Jakubowska
- Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
- Independent Laboratory of Molecular Biology and Genetic Diagnostics, Pomeranian Medical University, Szczecin, Poland
| | - Agnes Jager
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Margriet G A Sattler
- Department of Radiotherapy, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Marjanka K Schmidt
- Division of Psychosocial Research and Epidemiology, Netherlands Cancer Institute–Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
- Division of Molecular Pathology, The Netherlands Cancer Institute–Antoni Van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Maartje J Hooning
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| |
Collapse
|
23
|
Hou Z, Xu Z, Wu M, Ma L, Sui L, Bian P, Wang T. Enhancement of Repeat-Mediated Deletion Rearrangement Induced by Particle Irradiation in a RecA-Dependent Manner in Escherichia coli. BIOLOGY 2023; 12:1406. [PMID: 37998005 PMCID: PMC10669199 DOI: 10.3390/biology12111406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 10/30/2023] [Accepted: 11/06/2023] [Indexed: 11/25/2023]
Abstract
Repeat-mediated deletion (RMD) rearrangement is a major source of genome instability and can be deleterious to the organism, whereby the intervening sequence between two repeats is deleted along with one of the repeats. RMD rearrangement is likely induced by DNA double-strand breaks (DSBs); however, it is unclear how the complexity of DSBs influences RMD rearrangement. Here, a transgenic Escherichia coli strain K12 MG1655 with a lacI repeat-controlled amp activation was used while taking advantage of particle irradiation, such as proton and carbon irradiation, to generate different complexities of DSBs. Our research confirmed the enhancement of RMD under proton and carbon irradiation and revealed a positive correlation between RMD enhancement and LET. In addition, RMD enhancement could be suppressed by an intermolecular homologous sequence, which was regulated by its composition and length. Meanwhile, RMD enhancement was significantly stimulated by exogenous λ-Red recombinase. Further results investigating its mechanisms showed that the enhancement of RMD, induced by particle irradiation, occurred in a RecA-dependent manner. Our finding has a significant impact on the understanding of RMD rearrangement and provides some clues for elucidating the repair process and possible outcomes of complex DNA damage.
Collapse
Affiliation(s)
- Zhiyang Hou
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China; (Z.H.); (Z.X.); (M.W.); (P.B.)
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- Science Island Branch, Graduate School of USTC, Hefei 230026, China
| | - Zelin Xu
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China; (Z.H.); (Z.X.); (M.W.); (P.B.)
| | - Mengying Wu
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China; (Z.H.); (Z.X.); (M.W.); (P.B.)
| | - Liqiu Ma
- Department of Nuclear Physics, China Institute of Atomic Energy, Beijing 102413, China;
- National Innovation Center of Radiation Application, Beijing 102413, China
| | - Li Sui
- Department of Nuclear Physics, China Institute of Atomic Energy, Beijing 102413, China;
- National Innovation Center of Radiation Application, Beijing 102413, China
| | - Po Bian
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China; (Z.H.); (Z.X.); (M.W.); (P.B.)
| | - Ting Wang
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China; (Z.H.); (Z.X.); (M.W.); (P.B.)
| |
Collapse
|
24
|
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
|
25
|
Carrasco-Hernandez J, Ramos-Méndez J, Padilla-Rodal E, Avila-Rodriguez MA. Cellular lethal damage of 64Cu incorporated in mammalian genome evaluated with Monte Carlo methods. Front Med (Lausanne) 2023; 10:1253746. [PMID: 37841004 PMCID: PMC10575761 DOI: 10.3389/fmed.2023.1253746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 09/13/2023] [Indexed: 10/17/2023] Open
Abstract
Purpose Targeted Radionuclide Therapy (TRT) with Auger Emitters (AE) is a technique that allows targeting specific sites on tumor cells using radionuclides. The toxicity of AE is critically dependent on its proximity to the DNA. The aim of this study is to quantify the DNA damage and radiotherapeutic potential of the promising AE radionuclide copper-64 (64Cu) incorporated into the DNA of mammalian cells using Monte Carlo track-structure simulations. Methods A mammalian cell nucleus model with a diameter of 9.3 μm available in TOPAS-nBio was used. The cellular nucleus consisted of double-helix DNA geometrical model of 2.3 nm diameter surrounded by a hydration shell with a thickness of 0.16 nm, organized in 46 chromosomes giving a total of 6.08 giga base-pairs (DNA density of 14.4 Mbp/μm3). The cellular nucleus was irradiated with monoenergetic electrons and radiation emissions from several radionuclides including 111In, 125I, 123I, and 99mTc in addition to 64Cu. For monoenergetic electrons, isotropic point sources randomly distributed within the nucleus were modeled. The radionuclides were incorporated in randomly chosen DNA base pairs at two positions near to the central axis of the double-helix DNA model at (1) 0.25 nm off the central axis and (2) at the periphery of the DNA (1.15 nm off the central axis). For all the radionuclides except for 99mTc, the complete physical decay process was explicitly simulated. For 99mTc only total electron spectrum from published data was used. The DNA Double Strand Breaks (DSB) yield per decay from direct and indirect actions were quantified. Results obtained for monoenergetic electrons and radionuclides 111In, 125I, 123I, and 99mTc were compared with measured and calculated data from the literature for verification purposes. The DSB yields per decay incorporated in DNA for 64Cu are first reported in this work. The therapeutic effect of 64Cu (activity that led 37% cell survival after two cell divisions) was determined in terms of the number of atoms incorporated into the nucleus that would lead to the same DSBs that 100 decays of 125I. Simulations were run until a 2% statistical uncertainty (1 standard deviation) was achieved. Results The behavior of DSBs as a function of the energy for monoenergetic electrons was consistent with published data, the DSBs increased with the energy until it reached a maximum value near 500 eV followed by a continuous decrement. For 64Cu, when incorporated in the genome at evaluated positions (1) and (2), the DSB were 0.171 ± 0.003 and 0.190 ± 0.003 DSB/decay, respectively. The number of initial atoms incorporated into the genome (per cell) for 64Cu that would cause a therapeutic effect was estimated as 3,107 ± 28, that corresponds to an initial activity of 47.1 ± 0.4 × 10-3 Bq. Conclusion Our results showed that TRT with 64Cu has comparable therapeutic effects in cells as that of TRT with radionuclides currently used in clinical practice.
Collapse
Affiliation(s)
- Jhonatan Carrasco-Hernandez
- Departamento de Estructura de la Materia, Instituto de Ciencias Nucleares, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - José Ramos-Méndez
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, United States
| | - Elizabeth Padilla-Rodal
- Departamento de Estructura de la Materia, Instituto de Ciencias Nucleares, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Miguel A. Avila-Rodriguez
- Unidad Radiofarmacia-Ciclotrón, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
26
|
Buglewicz DJ, Su C, Banks AB, Stenger-Smith J, Elmegerhi S, Hirakawa H, Fujimori A, Kato TA. Metal Ions Modify In Vitro DNA Damage Yields with High-LET Radiation. TOXICS 2023; 11:773. [PMID: 37755783 PMCID: PMC10537317 DOI: 10.3390/toxics11090773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 09/04/2023] [Accepted: 09/07/2023] [Indexed: 09/28/2023]
Abstract
Cu2+ and Co2+ are metals known to increase DNA damage in the presence of hydrogen peroxide through a Fenton-type reaction. We hypothesized that these metals could increase DNA damage following irradiations of increasing LET values as hydrogen peroxide is a product of the radiolysis of water. The reaction mixtures contain either double- or single-stranded DNA in solution with Cu2+ or Co2+ and were irradiated either with X-ray, carbon-ion or iron-ion beams, or they were treated with hydrogen peroxide or bleomycin at increasing radiation dosages or chemical concentrations. DNA damage was then assessed via gel electrophoresis followed with a band intensity analysis. DNA damage was the greatest when DNA in the solution with either metal was treated with only hydrogen peroxide followed by the DNA damage of DNA in the solution with either metal post irradiation of low-LET (X-Ray) or high-LET (carbon-ion and iron-ion), respectively, and demonstrated the least damage after treatment with bleomycin. Cu2+ portrayed greater DNA damage than Co2+ following all experimental conditions. The metals' effect caused more DNA damage and was observed to be LET-dependent for single-strand break formation but inversely dependent for double-strand break formation. These results suggest that Cu2+ is more efficient than Co2+ at inducing both DNA single-strand and double-strand breaks following all irradiations and chemical treatments.
Collapse
Affiliation(s)
- Dylan J. Buglewicz
- National Institute of Radiological Sciences, National Institutes of Quantum Science and Technology, Chiba 263-8555, Japan; (D.J.B.); (H.H.); (A.F.)
- Department of Environmental & Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA; (C.S.); (A.B.B.); (J.S.-S.); (S.E.)
| | - Cathy Su
- Department of Environmental & Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA; (C.S.); (A.B.B.); (J.S.-S.); (S.E.)
| | - Austin B. Banks
- Department of Environmental & Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA; (C.S.); (A.B.B.); (J.S.-S.); (S.E.)
| | - Jazmine Stenger-Smith
- Department of Environmental & Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA; (C.S.); (A.B.B.); (J.S.-S.); (S.E.)
| | - Suad Elmegerhi
- Department of Environmental & Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA; (C.S.); (A.B.B.); (J.S.-S.); (S.E.)
| | - Hirokazu Hirakawa
- National Institute of Radiological Sciences, National Institutes of Quantum Science and Technology, Chiba 263-8555, Japan; (D.J.B.); (H.H.); (A.F.)
| | - Akira Fujimori
- National Institute of Radiological Sciences, National Institutes of Quantum Science and Technology, Chiba 263-8555, Japan; (D.J.B.); (H.H.); (A.F.)
| | - Takamitsu A. Kato
- Department of Environmental & Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA; (C.S.); (A.B.B.); (J.S.-S.); (S.E.)
| |
Collapse
|
27
|
Handwerk L, Schreier HK, Kraft D, Shreder K, Hemmersbach R, Hauslage J, Bonig H, Wiesmüller L, Fournier C, Rall-Scharpf M. Simulating Space Conditions Evokes Different DNA Damage Responses in Immature and Mature Cells of the Human Hematopoietic System. Int J Mol Sci 2023; 24:13761. [PMID: 37762064 PMCID: PMC10531023 DOI: 10.3390/ijms241813761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
The impact of space radiation and microgravity on DNA damage responses has been discussed controversially, largely due to the variety of model systems engaged. Here, we performed side-by-side analyses of human hematopoietic stem/progenitor cells (HSPC) and peripheral blood lymphocytes (PBL) cultivated in a 2D clinostat to simulate microgravity before, during and after photon and particle irradiation. We demonstrate that simulated microgravity (SMG) accelerates the early phase of non-homologous end joining (NHEJ)-mediated repair of simple, X-ray-induced DNA double-strand breaks (DSBs) in PBL, while repair kinetics in HSPC remained unaltered. Repair acceleration was lost with increasing LET of ion exposures, which increases the complexity of DSBs, precluding NHEJ and requiring end resection for successful repair. Such cell-type specific effect of SMG on DSB repair was dependent on the NF-кB pathway pre-activated in PBL but not HSPC. Already under unperturbed growth conditions HSPC and PBL suffered from SMG-induced replication stress associated with accumulation of single-stranded DNA and DSBs, respectively. We conclude that in PBL, SMG-induced DSBs promote repair of radiation-induced damage in an adaptive-like response. HSPC feature SMG-induced single-stranded DNA and FANCD2 foci, i.e., markers of persistent replication stress and senescence that may contribute to a premature decline of the immune system in space.
Collapse
Affiliation(s)
- Leonie Handwerk
- Department of Biophysics, GSI Helmholtz Center for Heavy Ion Research, 64291 Darmstadt, Germany
| | | | - Daniela Kraft
- Department of Biophysics, GSI Helmholtz Center for Heavy Ion Research, 64291 Darmstadt, Germany
| | - Kateryna Shreder
- Department of Biophysics, GSI Helmholtz Center for Heavy Ion Research, 64291 Darmstadt, Germany
| | - Ruth Hemmersbach
- Department of Gravitational Biology, German Aerospace Center, Institute of Aerospace Medicine, 51147 Cologne, Germany; (R.H.)
| | - Jens Hauslage
- Department of Gravitational Biology, German Aerospace Center, Institute of Aerospace Medicine, 51147 Cologne, Germany; (R.H.)
| | - Halvard Bonig
- Institute for Transfusion Medicine and Immunohematology, Johann Wolfgang Goethe-University Hospital, and German Red Cross Blood Service, Baden-Wuerttemberg–Hessen, 60528 Frankfurt, Germany
| | - Lisa Wiesmüller
- Department of Obstetrics and Gynecology, Ulm University, 89075 Ulm, Germany
| | - Claudia Fournier
- Department of Biophysics, GSI Helmholtz Center for Heavy Ion Research, 64291 Darmstadt, Germany
| | | |
Collapse
|
28
|
Hardwick SW, Stavridi AK, Chirgadze DY, De Oliveira TM, Charbonnier JB, Ropars V, Meek K, Blundell TL, Chaplin AK. Cryo-EM structure of a DNA-PK trimer: higher order oligomerisation in NHEJ. Structure 2023; 31:895-902.e3. [PMID: 37311458 DOI: 10.1016/j.str.2023.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/11/2023] [Accepted: 05/18/2023] [Indexed: 06/15/2023]
Abstract
The ability of humans to maintain the integrity of the genome is imperative for cellular survival. DNA double-strand breaks (DSBs) are considered the most critical type of DNA lesion, which can ultimately lead to diseases including cancer. Non-homologous end joining (NHEJ) is one of two core mechanisms utilized to repair DSBs. DNA-PK is a key component in this process and has recently been shown to form alternate long-range synaptic dimers. This has led to the proposal that these complexes can be formed before transitioning to a short-range synaptic complex. Here we present cryo-EM data representing an NHEJ supercomplex consisting of a trimer of DNA-PK in complex with XLF, XRCC4, and DNA Ligase IV. This trimer represents a complex of both long-range synaptic dimers. We discuss the potential role of the trimeric structure, and possible higher order oligomers, as structural intermediates in the NHEJ mechanism, or as functional DNA repair centers.
Collapse
Affiliation(s)
- Steven W Hardwick
- Cryo-EM Facility, Department of Biochemistry, University of Cambridge, Sanger Building, Tennis Court Road, CB2 1GA Cambridge, UK
| | - Antonia Kefala Stavridi
- Department of Biochemistry, University of Cambridge, Sanger Building, Tennis Court Road, CB2 1GA Cambridge, UK
| | - Dimitri Y Chirgadze
- Cryo-EM Facility, Department of Biochemistry, University of Cambridge, Sanger Building, Tennis Court Road, CB2 1GA Cambridge, UK
| | | | - Jean-Baptiste Charbonnier
- Institute for Integrative Biology of the Cell (I2BC), Institute Joliot, CEA, CNRS, Université Paris-Saclay, 91198 Gif-sur-Yvette cedex, France
| | - Virginie Ropars
- Institute for Integrative Biology of the Cell (I2BC), Institute Joliot, CEA, CNRS, Université Paris-Saclay, 91198 Gif-sur-Yvette cedex, France
| | - Katheryn Meek
- College of Veterinary Medicine, Department of Microbiology & Molecular Genetics, Department of Pathobiology & Diagnostic Investigation, Michigan State University, East Lansing, MI 48824, USA
| | - Tom L Blundell
- Department of Biochemistry, University of Cambridge, Sanger Building, Tennis Court Road, CB2 1GA Cambridge, UK
| | - Amanda K Chaplin
- Leicester Institute for Structural and Chemical Biology, Department of Molecular and Cell Biology, University of Leicester, Leicester, UK.
| |
Collapse
|
29
|
Marshall SK, Kaewpradit N, Mudmarn T, Buathong J, Sriwirote P. Evaluation of Single Dose and Fractionated Dose of I-131 Radiolabeled Nanoparticles for Triple-Negative Breast Cancer Treatment. Biomedicines 2023; 11:2169. [PMID: 37626666 PMCID: PMC10452573 DOI: 10.3390/biomedicines11082169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/27/2023] [Accepted: 07/29/2023] [Indexed: 08/27/2023] Open
Abstract
Combination chemotherapy is still the standard clinical care for triple-negative breast cancer (TNBC). However, sodium iodide symporter (NIS) uptake by TNBC has opened the potential of NIS as a molecular target for radioiodine theranostic treatments. Radiolabeled poly(lactic-co-glycolic) acid nanocarrier (NINP) was developed for NIS targeted delivery of I-131 to MDA-MB-231 cells to overcome I-131 low uptake in cancer cells and rapid clearance. The NINP diameter of 237 nm has good particle size uniformity and excellent particle stability. Radiochemical purity, radioactive stability, and radiolabeling yield of NINPs over 72 h were >95%. Cytotoxicity confirmed fractionated NINPs over 72 h to be more effective in cell death than single-dose NINP and both single and fractionated Na131I. Cellular uptake in a three-dimensional spheroid confirmed that NINP fractionated-dose achieved ~4.8-fold-higher mean fluorescent intensity than Na131I and ~2.7-fold greater reduction in cell viability compared to single-dose. The NINP fractionated-dose initiated greater cellular DNA damage to cells than single-dose NINP, resulting in inhibition of cell cycle progression, resulting in cell cycle progression being inhibited by cyclin-dependent kinases, which play a vital role in the control of MDA-MB-231 cell cycle. NINPs are biocompatible with blood, and were found to have no negative impact on red blood cells.
Collapse
Affiliation(s)
- Suphalak Khamruang Marshall
- Department of Radiology, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
- Molecular Imaging and Cyclotron Center, Department of Radiology, Division of Nuclear Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Nutnicha Kaewpradit
- Department of Radiology, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Tavadee Mudmarn
- Department of Radiology, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Jirassaya Buathong
- Department of Radiology, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Palmuk Sriwirote
- Department of Radiology, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| |
Collapse
|
30
|
Li Z, Wang Y. Short Double-Stranded DNA (≤40-bp) Affects Repair Pathway Choice. Int J Mol Sci 2023; 24:11836. [PMID: 37511594 PMCID: PMC10380458 DOI: 10.3390/ijms241411836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
To repair ionizing radiation (IR)-induced double strand breaks (DSBs), mammalian cells primarily use canonical non-homologous end-joining (cNHEJ), the homologous recombination (HR) pathway, and the alternative non-homologous end-joining (aEJ) as a backup. These pathways function either compensatively or competitively. High linear energy transfer (LET) compared to low-LET IR kills more cells at the same doses by inhibiting only cNHEJ, but not HR or aEJ. The mechanism remains unclear. The activation of each repair pathway requires the binding of different proteins to DNA fragments of varying lengths. We previously observed an increased generation of small DNA fragments (≤40 bp) in cells following high-LET IR compared to low-LET IR, suggesting that short DNA fragments were one of the major factors interfering with cNHEJ. To provide direct evidence, here we compare the efficiencies of cNHEJ, HR, or aEJ in repairing DSBs containing 30- or 60-bp fragments in vitro and in cells. We show that only cNHEJ but not HR or a-EJ was inefficient for repairing DSBs with 30-bp fragments compared to 60-bp ones, which strongly supports our hypothesis. These results not only enhance our understanding of the DSB repair pathway choice but also hold potential benefits for protection against high-LET IR-induced damage or improving high-LET radiotherapy.
Collapse
Affiliation(s)
- Zhentian Li
- Department of Radiation Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ya Wang
- Department of Radiation Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
31
|
Teh SSK, Halper-Stromberg E, Morsberger L, Bennett A, Bowland K, Skaist A, Cai F, Liang H, Hruban RH, Roberts NJ, Scharpf RB, Zou YS, Eshleman JR. Mechanism of delayed cell death following simultaneous CRISPR-Cas9 targeting in pancreatic cancers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.03.535384. [PMID: 37066222 PMCID: PMC10103988 DOI: 10.1101/2023.04.03.535384] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
When we transduced pancreatic cancers with sgRNAs that targeted 2-16 target sites in the human genome, we found that increasing the number of CRISPR-Cas9 target sites produced greater cytotoxicity, with >99% growth inhibition observed by targeting only 12 sites. However, cell death was delayed by 2-3 weeks after sgRNA transduction, in contrast to the repair of double strand DNA breaks (DSBs) that happened within 3 days after transduction. To explain this discrepancy, we used both cytogenetics and whole genome sequencing to interrogate the genome. We first detected chromatid and chromosome breaks, followed by radial formations, dicentric, ring chromosomes, and other chromosomal aberrations that peaked at 14 days after transduction. Structural variants (SVs) were detected at sites that were directly targeted by CRISPR-Cas9, including SVs generated from two sites that were targeted, but the vast majority of SVs (89.4%) were detected elsewhere in the genome that arose later than those directly targeted. Cells also underwent polyploidization that peaked at day 10 as detected by XY FISH assay, and ultimately died via apoptosis. Overall, we found that the simultaneous DSBs induced by CRISPR-Cas9 in pancreatic cancers caused chromosomal instability and polyploidization that ultimately led to delayed cell death.
Collapse
Affiliation(s)
- Selina Shiqing K. Teh
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eitan Halper-Stromberg
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Laura Morsberger
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alexis Bennett
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kirsten Bowland
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alyza Skaist
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Fidel Cai
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hong Liang
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ralph H. Hruban
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nicholas J. Roberts
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert B. Scharpf
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ying S. Zou
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - James R. Eshleman
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
32
|
Yi G, Sung Y, Kim C, Ra JS, Hirakawa H, Kato T, Fujimori A, Kim H, Takata KI. DNA polymerase θ-mediated repair of high LET radiation-induced complex DNA double-strand breaks. Nucleic Acids Res 2023; 51:2257-2269. [PMID: 36805268 PMCID: PMC10018357 DOI: 10.1093/nar/gkad076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 01/21/2023] [Accepted: 01/25/2023] [Indexed: 02/22/2023] Open
Abstract
DNA polymerase θ (POLQ) is a unique DNA polymerase that is able to perform microhomology-mediated end-joining as well as translesion synthesis (TLS) across an abasic (AP) site and thymine glycol (Tg). However, the biological significance of the TLS activity is currently unknown. Herein we provide evidence that the TLS activity of POLQ plays a critical role in repairing complex DNA double-strand breaks (DSBs) induced by high linear energy transfer (LET) radiation. Radiotherapy with high LET radiation such as carbon ions leads to more deleterious biological effects than corresponding doses of low LET radiation such as X-rays. High LET-induced DSBs are considered to be complex, carrying additional DNA damage such as AP site and Tg in close proximity to the DSB sites. However, it is not clearly understood how complex DSBs are processed in mammalian cells. We demonstrated that genetic disruption of POLQ results in an increase of chromatid breaks and enhanced cellular sensitivity following treatment with high LET radiation. At the biochemical level, POLQ was able to bypass an AP site and Tg during end-joining and was able to anneal two single-stranded DNA tails when DNA lesions were located outside the microhomology. This study offers evidence that POLQ is directly involved in the repair of complex DSBs.
Collapse
Affiliation(s)
- Geunil Yi
- Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, Republic of Korea
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Yubin Sung
- Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, Republic of Korea
| | - Chanwoo Kim
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Jae Sun Ra
- Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, Republic of Korea
| | - Hirokazu Hirakawa
- Department of Charged Particle Therapy Research, Institute for Quantum Medical Science, Chiba 263-8555, Japan
| | - Takamitsu A Kato
- Department of Environmental & Radiological Health Sciences, Colorado State University, Colorado 80523, USA
| | - Akira Fujimori
- Department of Charged Particle Therapy Research, Institute for Quantum Medical Science, Chiba 263-8555, Japan
| | - Hajin Kim
- Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, Republic of Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Kei-ichi Takata
- Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, Republic of Korea
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| |
Collapse
|
33
|
Georgieva M, Vassileva V. Stress Management in Plants: Examining Provisional and Unique Dose-Dependent Responses. Int J Mol Sci 2023; 24:ijms24065105. [PMID: 36982199 PMCID: PMC10049000 DOI: 10.3390/ijms24065105] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/05/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023] Open
Abstract
The purpose of this review is to critically evaluate the effects of different stress factors on higher plants, with particular attention given to the typical and unique dose-dependent responses that are essential for plant growth and development. Specifically, this review highlights the impact of stress on genome instability, including DNA damage and the molecular, physiological, and biochemical mechanisms that generate these effects. We provide an overview of the current understanding of predictable and unique dose-dependent trends in plant survival when exposed to low or high doses of stress. Understanding both the negative and positive impacts of stress responses, including genome instability, can provide insights into how plants react to different levels of stress, yielding more accurate predictions of their behavior in the natural environment. Applying the acquired knowledge can lead to improved crop productivity and potential development of more resilient plant varieties, ensuring a sustainable food source for the rapidly growing global population.
Collapse
|
34
|
Mare SD, Nishri Y, Shai A, Efrati M, Deutsch L, Den RB, Kelson I, Keisari Y, Domankevich V. Diffusing Alpha-Emitters Radiation Therapy Promotes a Proimmunogenic Tumor Microenvironment and Synergizes With Programmed Cell Death Protein 1 Blockade. Int J Radiat Oncol Biol Phys 2023; 115:707-718. [PMID: 36031029 DOI: 10.1016/j.ijrobp.2022.08.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/15/2022] [Accepted: 08/18/2022] [Indexed: 02/04/2023]
Abstract
PURPOSE Diffusing alpha-emitters Radiation Therapy (DaRT) releases alpha-emitting atoms into the tumor microenvironment. The treatment effectively ablates human and mice xenografts and shows 100% response rates in skin or head and neck squamous cell carcinoma patients. DaRT induces specific and systemic antitumor immune activation and synergizes with immune stimulation and modulation in mice. Here, the transcriptional profile activated by DaRT, and its potential to enhance responsiveness to immune checkpoint inhibition by programmed cell death protein 1 (PD-1) blockade were studied. METHODS AND MATERIALS Squamous cell carcinoma tumor- bearing BALB/C mice were treated with DaRT or inert seeds in combination with anti-PD-1 (aPD-1) or IgG control antibody. Sixteen days after seed insertion, tumors and spleens were subjected to immunophenotyping and immunohistochemical staining. Combination of DaRT and aPD-1 was tested for efficacy. Gene expression analysis was performed on mRNA extracted from tumors 7 days after DaRT or inert insertion using Nanostring PanCancer-IO-360 panel, and tumors and spleens were subjected to flow cytometry analysis. RESULTS DaRT in combination with aPD-1 delayed tumor development, induced CD3 and CD8 lymphocytes infiltration more efficiently than either monotherapy. The combined treatment reduced splenic polymorphonuclear myeloid derived suppressor cells more than aPD-1 therapy or control. Granzyme B release in the tumor was increased only in the combinational treatment and was correlated with T-lymphocyte infiltration. Gene expression and gene set enrichment analysis of mRNA levels 7 days after DaRT insertion indicated that DaRT upregulated apoptosis, p53 signaling, G1/S-related arrest, interferon signaling and myeloid related transcription, while downregulating DNA repair, cell proliferation, and notch-related transcription. Flow cytometry showed that DaRT increased dendritic cells activation and led to changes in MDSCs distribution. CONCLUSIONS DaRT promotes a "hot" tumor microenvironment and changes in immune suppression that lead to a potentiation of aPD-1 blockade induced effector T cell function and improved treatment efficacy. This study provides rationale for investigating DaRT and aPD-1 combination in patients with squamous cell carcinoma.
Collapse
Affiliation(s)
- Sara Del Mare
- Translational Research Laboratory, Alpha Tau Medical Ltd., Jerusalem, Israel
| | - Yossi Nishri
- Translational Research Laboratory, Alpha Tau Medical Ltd., Jerusalem, Israel
| | - Amit Shai
- Translational Research Laboratory, Alpha Tau Medical Ltd., Jerusalem, Israel
| | - Margalit Efrati
- Translational Research Laboratory, Alpha Tau Medical Ltd., Jerusalem, Israel
| | - Lisa Deutsch
- BioStats Statistical Consulting Ltd., Maccabim, Israel
| | - Robert B Den
- Translational Research Laboratory, Alpha Tau Medical Ltd., Jerusalem, Israel; Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Itzhak Kelson
- Sackler Faculty of Exact Sciences, School of Physics and Astronomy, Tel Aviv University, Tel Aviv, Israel
| | - Yona Keisari
- Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Vered Domankevich
- Translational Research Laboratory, Alpha Tau Medical Ltd., Jerusalem, Israel.
| |
Collapse
|
35
|
Hsu CF, Liu HM, Peir JJ, Liao JW, Chen KS, Chen YW, Chuang YJ, Chou FI. Therapeutic Efficacy and Radiobiological Effects of Boric-Acid-Mediated BNCT in an Osteosarcoma-Bearing SD Rat Model. Life (Basel) 2023; 13:life13020514. [PMID: 36836871 PMCID: PMC9964381 DOI: 10.3390/life13020514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/02/2023] [Accepted: 02/10/2023] [Indexed: 02/15/2023] Open
Abstract
BACKGROUND Osteosarcoma (OS) is the most common primary malignancy of the bone and is notoriously resistant to radiation therapy. High-dose cytotoxic chemotherapy and surgical resection have improved the survival rate and prognosis of patients with OS. Nonetheless, treatment challenges remain when the tumor cannot be removed by surgery. Boron neutron capture therapy (BNCT) provides high linear energy transfer (LET) radiation, and its internal targeted characteristics make BNCT a novel therapy for removing OS and reducing radiation damage to adjacent healthy tissues. METHODS In this study, a UMR-106-grafted OS rat model was developed, and boric acid (BA) was used as the boron drug for BNCT. The pharmacokinetics of BA, following intravenous injection, were evaluated to determine the optimal time window for neutron irradiation. OS-bearing rats were irradiated by an epithermal neutron beam at Tsing Hua Open-Pool Reactor (THOR). The therapeutic efficacy of and tissue response after BNCT were evaluated by radiographic and histopathological observations. RESULTS OS-bearing rats were irradiated by neutrons in the first hour following the intravenous injection of BA. The prescription-absorbed doses in the tumor regions were 5.8 and 11.0 Gy. BNCT reduced the body weight of the tumor-bearing rats, but they recovered after a few days. The BA-mediated BNCT effectively controlled the orthotopic OS tumor, reduced osteolysis, and induced bone healing. Autoradiography and histological analysis confirmed that the BA retention region is consistent with the calcification region in OS tissue. CONCLUSION BA is specifically retained in OS, and the BA-mediated BNCT can significantly reduce the tumor burden and osteolysis in OS-bearing rats.
Collapse
Affiliation(s)
- Chen-Fang Hsu
- Institute of Nuclear Engineering and Science, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Hong-Ming Liu
- Nuclear Science and Technology Development Center, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Jinn-Jer Peir
- Nuclear Science and Technology Development Center, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Jiunn-Wang Liao
- Graduate Institute of Veterinary Pathobiology, College of Veterinary Medicine, National Chung Hsing University, Taichung 40227, Taiwan
| | - Kuan-Sheng Chen
- Veterinary Medical Teaching Hospital, College of Veterinary Medicine, National Chung Hsing University, Taichung 40227, Taiwan
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung Hsing University, Taichung 40227, Taiwan
| | - Yi-Wei Chen
- Department of Oncology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Yung-Jen Chuang
- School of Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Fong-In Chou
- Institute of Nuclear Engineering and Science, National Tsing Hua University, Hsinchu 30013, Taiwan
- Nuclear Science and Technology Development Center, National Tsing Hua University, Hsinchu 30013, Taiwan
- Correspondence: ; Tel.: +886-3-5742884
| |
Collapse
|
36
|
Chitsike L, Bertucci A, Vazquez M, Lee S, Unternaehrer JJ, Duerksen-Hughes PJ. GA-OH enhances the cytotoxicity of photon and proton radiation in HPV + HNSCC cells. Front Oncol 2023; 13:1070485. [PMID: 36845698 PMCID: PMC9950506 DOI: 10.3389/fonc.2023.1070485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 01/23/2023] [Indexed: 02/12/2023] Open
Abstract
Introduction Treatment-related toxicity following either chemo- or radiotherapy can create significant clinical challenges for HNSCC cancer patients, particularly those with HPV-associated oropharyngeal squamous cell carcinoma. Identifying and characterizing targeted therapy agents that enhance the efficacy of radiation is a reasonable approach for developing de-escalated radiation regimens that result in less radiation-induced sequelae. We evaluated the ability of our recently discovered, novel HPV E6 inhibitor (GA-OH) to radio-sensitize HPV+ and HPV- HNSCC cell lines to photon and proton radiation. Methods Radiosensitivity to either photon or proton beams was assessed using various assays such as colony formation assay, DNA damage markers, cell cycle and apoptosis, western blotting, and primary cells. Calculations for radiosensitivity indices and relative biological effectiveness (RBE) were based on the linear quadratic model. Results Our results showed that radiation derived from both X-ray photons and protons is effective in inhibiting colony formation in HNSCC cells, and that GA-OH potentiated radiosensitivity of the cells. This effect was stronger in HPV+ cells as compared to their HPV- counterparts. We also found that GA-OH was more effective than cetuximab but less effective than cisplatin (CDDP) in enhancing radiosensitivity of HSNCC cells. Further tests indicated that the effects of GA-OH on the response to radiation may be mediated through cell cycle arrest, particularly in HPV+ cell lines. Importantly, the results also showed that GA-OH increases the apoptotic induction of radiation as measured by several apoptotic markers, even though radiation alone had little effect on apoptosis. Conclusion The enhanced combinatorial cytotoxicity found in this study indicates the strong potential of E6 inhibition as a strategy to sensitize cells to radiation. Future research is warranted to further characterize the interaction of GA-OH derivatives and other E6-specific inhibitors with radiation, as well as its potential to improve the safety and effectiveness of radiation treatment for patients with oropharyngeal cancer.
Collapse
Affiliation(s)
- Lennox Chitsike
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Antonella Bertucci
- Department of Radiation Medicine, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Marcelo Vazquez
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States
- Department of Radiation Medicine, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Steve Lee
- Department of Otolaryngology & Head/Neck Surgery, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Juli J. Unternaehrer
- Division of Biochemistry, Department of Basic Sciences, Loma Linda University, Loma Linda, CA, United States
| | | |
Collapse
|
37
|
Lohberger B, Glänzer D, Eck N, Stasny K, Falkner A, Leithner A, Georg D. The ATR Inhibitor VE-821 Enhances the Radiosensitivity and Suppresses DNA Repair Mechanisms of Human Chondrosarcoma Cells. Int J Mol Sci 2023; 24:2315. [PMID: 36768638 PMCID: PMC9917087 DOI: 10.3390/ijms24032315] [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: 12/13/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 01/27/2023] Open
Abstract
To overcome the resistance to radiotherapy in chondrosarcomas, the prevention of efficient DNA repair with an additional treatment was explored for particle beams as well as reference X-ray irradiation. The combined treatment with DNA repair inhibitors-with a focus on ATRi VE-821-and proton or carbon ions irradiation was investigated regarding cell viability, proliferation, cell cycle distribution, MAPK phosphorylation, and the expression of key DNA repair genes in two human chondrosarcoma cell lines. Pre-treatment with the PARPis Olaparib or Veliparib, the ATMi Ku-55933, and the ATRi VE-821 resulted in a dose-dependent reduction in viability, whereas VE-821 has the most efficient response. Quantification of γH2AX phosphorylation and protein expression of the DNA repair pathways showed a reduced regenerative capacity after irradiation. Furthermore, combined treatment with VE-821 and particle irradiation increased MAPK phosphorylation and the expression of apoptosis markers. At the gene expression and at the protein expression/phosphorylation level, we were able to demonstrate the preservation of DNA damage after combined treatment. The present data showed that the combined treatment with ATMi VE-821 increases the radiosensitivity of human chondrosarcoma cells in vitro and significantly suppresses efficient DNA repair mechanisms, thus improving the efficiency of radiotherapy.
Collapse
Affiliation(s)
- Birgit Lohberger
- Department of Orthopedics and Trauma, Medical University of Graz, 8036 Graz, Austria
| | - Dietmar Glänzer
- Department of Orthopedics and Trauma, Medical University of Graz, 8036 Graz, Austria
| | - Nicole Eck
- Department of Orthopedics and Trauma, Medical University of Graz, 8036 Graz, Austria
| | | | - Anna Falkner
- MedAustron Ion Therapy Center, 2700 Wiener Neustadt, Austria
| | - Andreas Leithner
- Department of Orthopedics and Trauma, Medical University of Graz, 8036 Graz, Austria
| | - Dietmar Georg
- MedAustron Ion Therapy Center, 2700 Wiener Neustadt, Austria
- Department of Radiation Oncology, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
38
|
The 'stealth-bomber' paradigm for deciphering the tumour response to carbon-ion irradiation. Br J Cancer 2023; 128:1429-1438. [PMID: 36639527 PMCID: PMC10070470 DOI: 10.1038/s41416-022-02117-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/01/2022] [Accepted: 12/08/2022] [Indexed: 01/14/2023] Open
Abstract
Numerous studies have demonstrated the higher biological efficacy of carbon-ion irradiation (C-ions) and their ballistic precision compared with photons. At the nanometre scale, the reactive oxygen species (ROS) produced by radiation and responsible for the indirect effects are differentially distributed according to the type of radiation. Photon irradiation induces a homogeneous ROS distribution, whereas ROS remain condensed in clusters in the C-ions tracks. Based on this linear energy transfer-dependent differential nanometric ROS distribution, we propose that the higher biological efficacy and specificities of the molecular response to C-ions rely on a 'stealth-bomber' effect. When biological targets are on the trajectories of the particles, the clustered radicals in the tracks are responsible for a 'bomber' effect. Furthermore, the low proportion of ROS outside the tracks is not able to trigger the cellular mechanisms of defence and proliferation. The ability of C-ions to deceive the cellular defence of the cancer cells is then categorised as a 'stealth' effect. This review aims to classify the biological arguments supporting the paradigm of the 'stealth-bomber' as responsible for the biological superiority of C-ions compared with photons. It also explains how and why C-ions will always be more efficient for treating patients with radioresistant cancers than conventional radiotherapy.
Collapse
|
39
|
Helm A, Totis C, Durante M, Fournier C. Are charged particles a good match for combination with immunotherapy? Current knowledge and perspectives. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 376:1-36. [PMID: 36997266 DOI: 10.1016/bs.ircmb.2023.01.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Charged particle radiotherapy, mainly using protons and carbon ions, provides physical characteristics allowing for a volume conformal irradiation and a reduction of the integral dose to normal tissue. Carbon ion therapy additionally features an increased biological effectiveness resulting in peculiar molecular effects. Immunotherapy, mostly performed with immune checkpoint inhibitors, is nowadays considered a pillar in cancer therapy. Based on the advantageous features of charged particle radiotherapy, we review pre-clinical evidence revealing a strong potential of its combination with immunotherapy. We argue that the combination therapy deserves further investigation with the aim of translation in clinics, where a few studies have been set up already.
Collapse
Affiliation(s)
- A Helm
- Biophysics Department, GSI, Darmstadt, Germany
| | - C Totis
- Biophysics Department, GSI, Darmstadt, Germany
| | - M Durante
- Biophysics Department, GSI, Darmstadt, Germany.
| | - C Fournier
- Biophysics Department, GSI, Darmstadt, Germany
| |
Collapse
|
40
|
Usman M, Beilerli A, Sufianov A, Kudryashov V, Ilyasova T, Balaev P, Danilov A, Lu H, Gareev I. Investigations into the impact of non-coding RNA on the sensitivity of gastric cancer to radiotherapy. Front Physiol 2023; 14:1149821. [PMID: 36909247 PMCID: PMC9998927 DOI: 10.3389/fphys.2023.1149821] [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: 01/23/2023] [Accepted: 02/16/2023] [Indexed: 02/26/2023] Open
Abstract
Non-coding RNAs (ncRNAs) are a newly discovered functional RNA different from messenger RNA, which can participate in regulating the occurrence and development of tumors. More and more research results show that ncRNAs can participate in the regulation of gastric cancer (GC) radiotherapy response, and its mechanism may be related to its effect on DNA damage repair, gastric cancer cell stemness, cell apoptosis, activation of epidermal growth factor receptor signaling pathway, etc. This article summarizes the relevant mechanisms of ncRNAs regulating the response to radiotherapy in gastric cancer, which will be directly important for the introduction of ncRNAs particularly microRNAs (miRNAs), long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) into clinical medicine as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Muhammad Usman
- Department of Medical Imaging, Central Hospital Affiliated to Chongqing University of Technology, Chongqing, China
| | - Aferin Beilerli
- Department of Obstetrics and Gynecology, Tyumen State Medical University, Tyumen, Russia
| | - Albert Sufianov
- Department of Neurosurgery, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia.,Department of Internal Diseases, Bashkir State Medical University, Ufa, Russia
| | - Valentin Kudryashov
- Gastric Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Tatiana Ilyasova
- Department of Internal Diseases, Bashkir State Medical University, Ufa, Russia
| | - Pavel Balaev
- Department of Oncology and Radiology, Ural State Medical University, Yekaterinburg, Russia
| | - Andrei Danilov
- Department of Clinical Pharmacology, Smolensk State Medical University, Smolensk, Russia
| | - Hong Lu
- Department of Medical Imaging, Central Hospital Affiliated to Chongqing University of Technology, Chongqing, China
| | - Ilgiz Gareev
- Educational and Scientific Institute of Neurosurgery, Рeoples' Friendship University of Russia (RUDN University), Moscow, Russia
| |
Collapse
|
41
|
Liu J, Hormuth DA, Yang J, Yankeelov TE. A data assimilation framework to predict the response of glioma cells to radiation. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2023; 20:318-336. [PMID: 36650768 PMCID: PMC11165419 DOI: 10.3934/mbe.2023015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
We incorporate a practical data assimilation methodology into our previously established experimental-computational framework to predict the heterogeneous response of glioma cells receiving fractionated radiation treatment. Replicates of 9L and C6 glioma cells grown in 96-well plates were irradiated with six different fractionation schemes and imaged via time-resolved microscopy to yield 360- and 286-time courses for the 9L and C6 lines, respectively. These data were used to calibrate a biology-based mathematical model and then make predictions within two different scenarios. For Scenario 1, 70% of the time courses are fit to the model and the resulting parameter values are averaged. These average values, along with the initial cell number, initialize the model to predict the temporal evolution for each test time course (10% of the data). In Scenario 2, the predictions for the test cases are made with model parameters initially assigned from the training data, but then updated with new measurements every 24 hours via four versions of a data assimilation framework. We then compare the predictions made from Scenario 1 and the best version of Scenario 2 to the experimentally measured microscopy measurements using the concordance correlation coefficient (CCC). Across all fractionation schemes, Scenario 1 achieved a CCC value (mean ± standard deviation) of 0.845 ± 0.185 and 0.726 ± 0.195 for the 9L and C6 cell lines, respectively. For the best data assimilation version from Scenario 2 (validated with the last 20% of the data), the CCC values significantly increased to 0.954 ± 0.056 (p = 0.002) and 0.901 ± 0.061 (p = 8.9e-5) for the 9L and C6 cell lines, respectively. Thus, we have developed a data assimilation approach that incorporates an experimental-computational system to accurately predict the in vitro response of glioma cells to fractionated radiation therapy.
Collapse
Affiliation(s)
- Junyan Liu
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX 78712, USA
| | - David A. Hormuth
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, 201 E. 24 Street POB 4.102 Stop C0200, Austin, TX 78712, USA
- Livestrong Cancer Institutes, The University of Texas at Austin, 1601 Trinity St. Bldg. B Mail Stop Z1100, TX 78712, USA
| | - Jianchen Yang
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX 78712, USA
| | - Thomas E. Yankeelov
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton Street Stop C0800, Austin, TX 78712, USA
- Department of Diagnostic Medicine, The University of Texas at Austin, 1601 Trinity St Bldg. B Stop Z0300, Austin, TX 78712, USA
- Department of Oncology, The University of Texas at Austin, 1601 Trinity St Bldg. B, Austin, TX 78712, USA
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, 201 E. 24 Street POB 4.102 Stop C0200, Austin, TX 78712, USA
- Livestrong Cancer Institutes, The University of Texas at Austin, 1601 Trinity St. Bldg. B Mail Stop Z1100, TX 78712, USA
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, PO Box 301402, Houston, TX, 77230-1402, USA
| |
Collapse
|
42
|
Huang CY, Lai ZY, Hsu TJ, Chou FI, Liu HM, Chuang YJ. Boron Neutron Capture Therapy Eliminates Radioresistant Liver Cancer Cells by Targeting DNA Damage and Repair Responses. J Hepatocell Carcinoma 2022; 9:1385-1401. [PMID: 36600987 PMCID: PMC9807134 DOI: 10.2147/jhc.s383959] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 12/04/2022] [Indexed: 12/29/2022] Open
Abstract
Introduction For advanced hepatocellular carcinoma (HCC), resistance to conservative treatments remains a challenge. In previous studies, the therapeutic effectiveness and DNA damage responses of boric acid-mediated boron neutron capture therapy (BA-BNCT) in HCC have been demonstrated in animal models and HCC cell line. On the other hand, numerous studies have shown that high linear energy transfer (LET) radiation can overcome tumor resistance. Since BNCT yields a mixture of high and low LET radiation, we aimed to explore whether and how BA-BNCT could eliminate radioresistant HCC cells. Methods Radioresistant human HCC (HepG2-R) cells were established from HepG2 cells via intermittent irradiation. HepG2 and HepG2-R cells were then irradiated with either γ-ray or neutron radiation of BA-BNCT. Colony formation assays were used to assess cell survival and the relative biological effectiveness (RBE). The expression of phosphorylated H2AX (γH2AX) was also examined by immunocytochemistry and Western blot assays to evaluate the extent of DNA double-strand breaks (DSBs). Finally, the expression levels of DNA damage response-associated proteins were determined, followed by cell cycle analysis and caspase-3 activity analysis. Results Our data demonstrated that under the same dose by γ-ray, BNCT effectively eliminated radioresistant HCC by increasing the number of DNA DSBs (p < 0.05) and impeding their repair (p < 0.05), which verified the high RBE of BNCT. We also found that BNCT resulted in delayed homologous recombination (HR) and inhibited the nonhomologous end-joining (NHEJ) pathway during DNA repair. Markedly, BNCT increased cell arrest (p < 0.05) in the G2/M phase by altering G2 checkpoint signaling and increased PUMA-mediated apoptosis (p < 0.05). Conclusion Our data suggest that DNA damage and repair responses could affect the anticancer efficiency of BNCT in radioresistant HepG2-R cells, which highlights the potential of BNCT as a viable treatment option for recurrent HCC.
Collapse
Affiliation(s)
- Chu-Yu Huang
- School of Medicine, National Tsing Hua University, Hsinchu, Taiwan,Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan,Nuclear Science and Technology Development Center, National Tsing Hua University, Hsinchu, Taiwan
| | - Zih-Yin Lai
- School of Medicine, National Tsing Hua University, Hsinchu, Taiwan,Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan,Nuclear Science and Technology Development Center, National Tsing Hua University, Hsinchu, Taiwan
| | - Tzu-Jung Hsu
- School of Medicine, National Tsing Hua University, Hsinchu, Taiwan,Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan,Nuclear Science and Technology Development Center, National Tsing Hua University, Hsinchu, Taiwan
| | - Fong-In Chou
- Nuclear Science and Technology Development Center, National Tsing Hua University, Hsinchu, Taiwan
| | - Hong-Ming Liu
- Nuclear Science and Technology Development Center, National Tsing Hua University, Hsinchu, Taiwan
| | - Yung-Jen Chuang
- School of Medicine, National Tsing Hua University, Hsinchu, Taiwan,Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan,Nuclear Science and Technology Development Center, National Tsing Hua University, Hsinchu, Taiwan,Correspondence: Yung-Jen Chuang, School of Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan, Tel +886-3-5742764, Fax +886-3-5715934, Email
| |
Collapse
|
43
|
Du TQ, Liu R, Zhang Q, Luo H, Chen Y, Tan M, Wang Q, Wu X, Liu Z, Sun S, Yang K, Tian J, Wang X. Does particle radiation have superior radiobiological advantages for prostate cancer cells? A systematic review of in vitro studies. Eur J Med Res 2022; 27:306. [PMID: 36572945 PMCID: PMC9793637 DOI: 10.1186/s40001-022-00942-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 12/07/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Charged particle beams from protons to carbon ions provide many significant physical benefits in radiation therapy. However, preclinical studies of charged particle therapy for prostate cancer are extremely limited. The aim of this study was to comprehensively investigate the biological effects of charged particles on prostate cancer from the perspective of in vitro studies. METHODS We conducted a systematic review by searching EMBASE (OVID), Medline (OVID), and Web of Science databases to identify the publications assessing the radiobiological effects of charged particle irradiation on prostate cancer cells. The data of relative biological effectiveness (RBE), surviving fraction (SF), standard enhancement ratio (SER) and oxygen enhancement ratio (OER) were extracted. RESULTS We found 12 studies met the eligible criteria. The relative biological effectiveness values of proton and carbon ion irradiation ranged from 0.94 to 1.52, and 1.67 to 3.7, respectively. Surviving fraction of 2 Gy were 0.17 ± 0.12, 0.55 ± 0.20 and 0.53 ± 0.16 in carbon ion, proton, and photon irradiation, respectively. PNKP inhibitor and gold nanoparticles were favorable sensitizing agents, while it was presented poorer performance in GANT61. The oxygen enhancement ratio values of photon and carbon ion irradiation were 2.32 ± 0.04, and 1.77 ± 0.13, respectively. Charged particle irradiation induced more G0-/G1- or G2-/M-phase arrest, more expression of γ-H2AX, more apoptosis, and lower motility and/or migration ability than photon irradiation. CONCLUSIONS Both carbon ion and proton irradiation have advantages over photon irradiation in radiobiological effects on prostate cancer cell lines. Carbon ion irradiation seems to have further advantages over proton irradiation.
Collapse
Affiliation(s)
- Tian-Qi Du
- grid.9227.e0000000119573309Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Rd, Lanzhou, 730000 Gansu People’s Republic of China ,grid.32566.340000 0000 8571 0482The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu People’s Republic of China
| | - Ruifeng Liu
- grid.9227.e0000000119573309Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Rd, Lanzhou, 730000 Gansu People’s Republic of China ,grid.410726.60000 0004 1797 8419Graduate School, University of Chinese Academy of Sciences, Beijing, People’s Republic of China ,Heavy Ion Therapy Center, Lanzhou Heavy Ion Hospital, Lanzhou, Gansu People’s Republic of China
| | - Qiuning Zhang
- grid.9227.e0000000119573309Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Rd, Lanzhou, 730000 Gansu People’s Republic of China ,grid.410726.60000 0004 1797 8419Graduate School, University of Chinese Academy of Sciences, Beijing, People’s Republic of China ,Heavy Ion Therapy Center, Lanzhou Heavy Ion Hospital, Lanzhou, Gansu People’s Republic of China
| | - Hongtao Luo
- grid.9227.e0000000119573309Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Rd, Lanzhou, 730000 Gansu People’s Republic of China ,grid.410726.60000 0004 1797 8419Graduate School, University of Chinese Academy of Sciences, Beijing, People’s Republic of China ,Heavy Ion Therapy Center, Lanzhou Heavy Ion Hospital, Lanzhou, Gansu People’s Republic of China
| | - Yanliang Chen
- grid.9227.e0000000119573309Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Rd, Lanzhou, 730000 Gansu People’s Republic of China ,grid.32566.340000 0000 8571 0482The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu People’s Republic of China
| | - Mingyu Tan
- grid.9227.e0000000119573309Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Rd, Lanzhou, 730000 Gansu People’s Republic of China ,grid.32566.340000 0000 8571 0482The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu People’s Republic of China
| | - Qian Wang
- grid.9227.e0000000119573309Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Rd, Lanzhou, 730000 Gansu People’s Republic of China ,grid.32566.340000 0000 8571 0482The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu People’s Republic of China
| | - Xun Wu
- grid.9227.e0000000119573309Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Rd, Lanzhou, 730000 Gansu People’s Republic of China ,grid.32566.340000 0000 8571 0482The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu People’s Republic of China
| | - Zhiqiang Liu
- grid.9227.e0000000119573309Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Rd, Lanzhou, 730000 Gansu People’s Republic of China ,grid.410726.60000 0004 1797 8419Graduate School, University of Chinese Academy of Sciences, Beijing, People’s Republic of China ,Heavy Ion Therapy Center, Lanzhou Heavy Ion Hospital, Lanzhou, Gansu People’s Republic of China
| | - Shilong Sun
- grid.9227.e0000000119573309Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Rd, Lanzhou, 730000 Gansu People’s Republic of China ,grid.410726.60000 0004 1797 8419Graduate School, University of Chinese Academy of Sciences, Beijing, People’s Republic of China ,Heavy Ion Therapy Center, Lanzhou Heavy Ion Hospital, Lanzhou, Gansu People’s Republic of China
| | - Kehu Yang
- grid.32566.340000 0000 8571 0482Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu People’s Republic of China
| | - Jinhui Tian
- grid.32566.340000 0000 8571 0482Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu People’s Republic of China
| | - Xiaohu Wang
- grid.9227.e0000000119573309Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Rd, Lanzhou, 730000 Gansu People’s Republic of China ,grid.32566.340000 0000 8571 0482The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu People’s Republic of China ,grid.410726.60000 0004 1797 8419Graduate School, University of Chinese Academy of Sciences, Beijing, People’s Republic of China ,Heavy Ion Therapy Center, Lanzhou Heavy Ion Hospital, Lanzhou, Gansu People’s Republic of China
| |
Collapse
|
44
|
Putt KS, Du Y, Fu H, Zhang ZY. High-throughput screening strategies for space-based radiation countermeasure discovery. LIFE SCIENCES IN SPACE RESEARCH 2022; 35:88-104. [PMID: 36336374 DOI: 10.1016/j.lssr.2022.07.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 06/13/2022] [Accepted: 07/19/2022] [Indexed: 06/16/2023]
Abstract
As humanity begins to venture further into space, approaches to better protect astronauts from the hazards found in space need to be developed. One particular hazard of concern is the complex radiation that is ever present in deep space. Currently, it is unlikely enough spacecraft shielding could be launched that would provide adequate protection to astronauts during long-duration missions such as a journey to Mars and back. In an effort to identify other means of protection, prophylactic radioprotective drugs have been proposed as a potential means to reduce the biological damage caused by this radiation. Unfortunately, few radioprotectors have been approved by the FDA for usage and for those that have been developed, they protect normal cells/tissues from acute, high levels of radiation exposure such as that from oncology radiation treatments. To date, essentially no radioprotectors have been developed that specifically counteract the effects of chronic low-dose rate space radiation. This review highlights how high-throughput screening (HTS) methodologies could be implemented to identify such a radioprotective agent. Several potential target, pathway, and phenotypic assays are discussed along with potential challenges towards screening for radioprotectors. Utilizing HTS strategies such as the ones proposed here have the potential to identify new chemical scaffolds that can be developed into efficacious radioprotectors that are specifically designed to protect astronauts during deep space journeys. The overarching goal of this review is to elicit broader interest in applying drug discovery techniques, specifically HTS towards the identification of radiation countermeasures designed to be efficacious towards the biological insults likely to be encountered by astronauts on long duration voyages.
Collapse
Affiliation(s)
- Karson S Putt
- Institute for Drug Discovery, Purdue University, West Lafayette IN 47907 USA
| | - Yuhong Du
- Department of Pharmacology and Chemical Biology and Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Haian Fu
- Department of Pharmacology and Chemical Biology and Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Zhong-Yin Zhang
- Institute for Drug Discovery, Purdue University, West Lafayette IN 47907 USA; Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette IN 47907 USA.
| |
Collapse
|
45
|
Baiocco G, Bartzsch S, Conte V, Friedrich T, Jakob B, Tartas A, Villagrasa C, Prise KM. A matter of space: how the spatial heterogeneity in energy deposition determines the biological outcome of radiation exposure. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2022; 61:545-559. [PMID: 36220965 PMCID: PMC9630194 DOI: 10.1007/s00411-022-00989-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 08/03/2022] [Indexed: 05/10/2023]
Abstract
The outcome of the exposure of living organisms to ionizing radiation is determined by the distribution of the associated energy deposition at different spatial scales. Radiation proceeds through ionizations and excitations of hit molecules with an ~ nm spacing. Approaches such as nanodosimetry/microdosimetry and Monte Carlo track-structure simulations have been successfully adopted to investigate radiation quality effects: they allow to explore correlations between the spatial clustering of such energy depositions at the scales of DNA or chromosome domains and their biological consequences at the cellular level. Physical features alone, however, are not enough to assess the entity and complexity of radiation-induced DNA damage: this latter is the result of an interplay between radiation track structure and the spatial architecture of chromatin, and further depends on the chromatin dynamic response, affecting the activation and efficiency of the repair machinery. The heterogeneity of radiation energy depositions at the single-cell level affects the trade-off between cell inactivation and induction of viable mutations and hence influences radiation-induced carcinogenesis. In radiation therapy, where the goal is cancer cell inactivation, the delivery of a homogenous dose to the tumour has been the traditional approach in clinical practice. However, evidence is accumulating that introducing heterogeneity with spatially fractionated beams (mini- and microbeam therapy) can lead to significant advantages, particularly in sparing normal tissues. Such findings cannot be explained in merely physical terms, and their interpretation requires considering the scales at play in the underlying biological mechanisms, suggesting a systemic response to radiation.
Collapse
Affiliation(s)
- Giorgio Baiocco
- Radiation Biophysics and Radiobiology Group, Physics Department, University of Pavia, Pavia, Italy.
| | - Stefan Bartzsch
- Institute for Radiation Medicine, Helmholtz Centre Munich, Munich, Germany
- Department of Radiation Oncology, Technical University of Munich, Munich, Germany
| | - Valeria Conte
- Istituto Nazionale Di Fisica Nucleare INFN, Laboratori Nazionali Di Legnaro, Legnaro, Italy
| | - Thomas Friedrich
- Department of Biophysics, GSI Helmholtz Centre for Heavy Ion Research, Darmstadt, Germany
| | - Burkhard Jakob
- Department of Biophysics, GSI Helmholtz Centre for Heavy Ion Research, Darmstadt, Germany
| | - Adrianna Tartas
- Biomedical Physics Division, Institute of Experimental Physics, University of Warsaw, Warsaw, Poland
| | - Carmen Villagrasa
- IRSN, Institut de Radioprotection et de Sûreté Nucléaire, Fontenay aux Roses, France
| | - Kevin M Prise
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| |
Collapse
|
46
|
Naumenko NV, Petruseva IO, Lavrik OI. Bulky Adducts in Clustered DNA Lesions: Causes of Resistance to the NER System. Acta Naturae 2022; 14:38-49. [PMID: 36694906 PMCID: PMC9844087 DOI: 10.32607/actanaturae.11741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 10/18/2022] [Indexed: 01/22/2023] Open
Abstract
The nucleotide excision repair (NER) system removes a wide range of bulky DNA lesions that cause significant distortions of the regular double helix structure. These lesions, mainly bulky covalent DNA adducts, are induced by ultraviolet and ionizing radiation or the interaction between exogenous/endogenous chemically active substances and nitrogenous DNA bases. As the number of DNA lesions increases, e.g., due to intensive chemotherapy and combination therapy of various diseases or DNA repair impairment, clustered lesions containing bulky adducts may occur. Clustered lesions are two or more lesions located within one or two turns of the DNA helix. Despite the fact that repair of single DNA lesions by the NER system in eukaryotic cells has been studied quite thoroughly, the repair mechanism of these lesions in clusters remains obscure. Identification of the structural features of the DNA regions containing irreparable clustered lesions is of considerable interest, in particular due to a relationship between the efficiency of some antitumor drugs and the activity of cellular repair systems. In this review, we analyzed data on the induction of clustered lesions containing bulky adducts, the potential biological significance of these lesions, and methods for quantification of DNA lesions and considered the causes for the inhibition of NER-catalyzed excision of clustered bulky lesions.
Collapse
Affiliation(s)
- N. V. Naumenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090 Russia
| | - I. O. Petruseva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090 Russia
| | - O. I. Lavrik
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, 630090 Russia
| |
Collapse
|
47
|
Bright SJ, Flint DB, Martinus DKJ, Turner BX, Manandhar M, Ben Kacem M, McFadden CH, Yap TA, Shaitelman SF, Sawakuchi GO. Targeted Inhibition of DNA-PKcs, ATM, ATR, PARP, and Rad51 Modulate Response to X Rays and Protons. Radiat Res 2022; 198:336-346. [PMID: 35939823 PMCID: PMC9648665 DOI: 10.1667/rade-22-00040.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 07/05/2022] [Indexed: 11/03/2022]
Abstract
Small molecule inhibitors are currently in preclinical and clinical development for the treatment of selected cancers, particularly those with existing genetic alterations in DNA repair and DNA damage response (DDR) pathways. Keen interest has also been expressed in combining such agents with other targeted antitumor strategies such as radiotherapy. Radiotherapy exerts its cytotoxic effects primarily through DNA damage-induced cell death; therefore, inhibiting DNA repair and the DDR should lead to additive and/or synergistic radiosensitizing effects. In this study we screened the response to X-ray or proton radiation in cell lines treated with DDR inhibitors (DDRis) targeting ATM, ATR, DNA-PKcs, Rad51, and PARP, with survival metrics established using clonogenic assays. We observed that DDRis generate significant radiosensitization in cancer and primary cells derived from normal tissue. Existing genetic defects in cancer cells appear to be an important consideration when determining the optimal inhibitor to use for synergistic combination with radiation. We also show that while greater radiosensitization can be achieved with protons (9.9 keV/µm) combined with DDRis, the relative biological effectiveness is unchanged or in some cases reduced. Our results indicate that while targeting the DDR can significantly radiosensitize cancer cells to such combinations, normal cells may also be equally or more severely affected, depending on the DDRi used. These data highlight the importance of identifying genetic defects as predictive biomarkers of response for combination treatment.
Collapse
Affiliation(s)
- Scott J. Bright
- Department of Radiation Physics, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David B. Flint
- Department of Radiation Physics, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David K. J. Martinus
- Department of Radiation Physics, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| | - Broderick X. Turner
- Department of Radiation Physics, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| | - Mandira Manandhar
- Department of Radiation Physics, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mariam Ben Kacem
- Department of Radiation Physics, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Conor H. McFadden
- Department of Radiation Physics, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Timothy A. Yap
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Division of Cancer Medicine; Khalifa Institute for Personalized Cancer Therapy; Department of Thoracic/Head and Neck Medical Oncology; and The Institute for Applied Cancer Science. The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Simona F. Shaitelman
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gabriel O. Sawakuchi
- Department of Radiation Physics, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| |
Collapse
|
48
|
Lohberger B, Barna S, Glänzer D, Eck N, Kerschbaum-Gruber S, Stasny K, Leithner A, Georg D. Cellular and Molecular Biological Alterations after Photon, Proton, and Carbon Ions Irradiation in Human Chondrosarcoma Cells Linked with High-Quality Physics Data. Int J Mol Sci 2022; 23:11464. [PMID: 36232764 PMCID: PMC9569755 DOI: 10.3390/ijms231911464] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/08/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022] Open
Abstract
Chondrosarcomas are particularly difficult to treat due to their resistance to chemotherapy and radiotherapy. However, particle therapy can enhance local control and patient survival rates. To improve our understanding of the basic cellular radiation response, as a function of dose and linear energy transfer (LET), we developed a novel water phantom-based setup for cell culture experiments and characterized it dosimetrically. In a direct comparison, human chondrosarcoma cell lines were analyzed with regard to their viability, cell proliferation, cell cycle, and DNA repair behavior after irradiation with X-ray, proton, and carbon ions. Our results clearly showed that cell viability and proliferation were inhibited according to the increasing ionization density, i.e., LET, of the irradiation modes. Furthermore, a prominent G2/M arrest was shown. Gene expression profiling proved the upregulation of the senescence genes CDKN1A (p21), CDKN2A (p16NK4a), BMI1, and FOXO4 after particle irradiation. Both proton or C-ion irradiation caused a positive regulation of the repair genes ATM, NBN, ATXR, and XPC, and a highly significant increase in XRCC1/2/3, ERCC1, XPC, and PCNA expression, with C-ions appearing to activate DNA repair mechanisms more effectively. The link between the physical data and the cellular responses is an important contribution to the improvement of the treatment system.
Collapse
Affiliation(s)
- Birgit Lohberger
- Department of Orthopaedics and Trauma, Medical University of Graz, 8036 Graz, Austria
| | - Sandra Barna
- Department of Radiation Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | - Dietmar Glänzer
- Department of Orthopaedics and Trauma, Medical University of Graz, 8036 Graz, Austria
| | - Nicole Eck
- Department of Orthopaedics and Trauma, Medical University of Graz, 8036 Graz, Austria
| | | | | | - Andreas Leithner
- Department of Orthopaedics and Trauma, Medical University of Graz, 8036 Graz, Austria
| | - Dietmar Georg
- Department of Radiation Oncology, Medical University of Vienna, 1090 Vienna, Austria
- MedAustron Ion Therapy Center, 2700 Wiener Neustadt, Austria
| |
Collapse
|
49
|
Averbeck NB, Barent C, Jakob B, Syzonenko T, Durante M, Taucher-Scholz G. The Ubiquitin Ligase RNF138 Cooperates with CtIP to Stimulate Resection of Complex DNA Double-Strand Breaks in Human G1-Phase Cells. Cells 2022; 11:cells11162561. [PMID: 36010636 PMCID: PMC9406464 DOI: 10.3390/cells11162561] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/04/2022] [Accepted: 08/05/2022] [Indexed: 11/30/2022] Open
Abstract
DNA double-strand breaks (DSBs) represent the molecular origin of ionizing-radiation inflicted biological effects. An increase in the ionization density causes more complex, clustered DSBs that can be processed by resection also in G1 phase, where repair of resected DSBs is considered erroneous and may contribute to the increased biological effectiveness of heavy ions in radiotherapy. To investigate the resection regulation of complex DSBs, we exposed G1 cells depleted for different candidate factors to heavy ions or α-particle radiation. Immunofluorescence microscopy was used to monitor the resection marker RPA, the DSB marker γH2AX and the cell-cycle markers CENP-F and geminin. The Fucci system allowed to select G1 cells, cell survival was measured by clonogenic assay. We show that in G1 phase the ubiquitin ligase RNF138 functions in resection regulation. RNF138 ubiquitinates the resection factor CtIP in a radiation-dependent manner to allow its DSB recruitment in G1 cells. At complex DSBs, RNF138′s participation becomes more relevant, consistent with the observation that also resection is more frequent at these DSBs. Furthermore, deficiency of RNF138 affects both DSB repair and cell survival upon induction of complex DSBs. We conclude that RNF138 is a regulator of resection that is influenced by DSB complexity and can affect the quality of DSB repair in G1 cells.
Collapse
Affiliation(s)
- Nicole B. Averbeck
- Department of Biophysics, GSI Helmholtzzentrum für Schwerionenforschung GmbH, Planckstr. 1, 64291 Darmstadt, Germany
- Correspondence:
| | - Carina Barent
- Department of Biophysics, GSI Helmholtzzentrum für Schwerionenforschung GmbH, Planckstr. 1, 64291 Darmstadt, Germany
| | - Burkhard Jakob
- Department of Biophysics, GSI Helmholtzzentrum für Schwerionenforschung GmbH, Planckstr. 1, 64291 Darmstadt, Germany
- Department of Biology, Technische Universität Darmstadt, Schnittspahnstr. 11, 64287 Darmstadt, Germany
| | - Tatyana Syzonenko
- Department of Biophysics, GSI Helmholtzzentrum für Schwerionenforschung GmbH, Planckstr. 1, 64291 Darmstadt, Germany
| | - Marco Durante
- Department of Biophysics, GSI Helmholtzzentrum für Schwerionenforschung GmbH, Planckstr. 1, 64291 Darmstadt, Germany
- Department of Condensed Matter Physics, Technische Universität Darmstadt, Hochschulstr. 6–8, 64289 Darmstadt, Germany
| | - Gisela Taucher-Scholz
- Department of Biophysics, GSI Helmholtzzentrum für Schwerionenforschung GmbH, Planckstr. 1, 64291 Darmstadt, Germany
- Department of Biology, Technische Universität Darmstadt, Schnittspahnstr. 11, 64287 Darmstadt, Germany
| |
Collapse
|
50
|
Frame CM, Chen Y, Gagnon J, Yuan Y, Ma T, Dritschilo A, Pang D. Proton induced DNA double strand breaks at the Bragg peak: Evidence of enhanced LET effect. Front Oncol 2022; 12:930393. [PMID: 35992825 PMCID: PMC9388940 DOI: 10.3389/fonc.2022.930393] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
PurposeTo investigate DNA double strand breaks (DSBs) induced by therapeutic proton beams in plateau and Bragg peak to demonstrate DSB induction due to the higher LET in the Bragg peak.Materials and MethodspUC19 plasmid DNA samples were irradiated to doses of 1000 and 3000 Gy on a Mevion S250i proton system with a monoenergetic, 110 MeV, proton beam at depths of 2 and 9.4 cm, corresponding to a position on the plateau and distal Bragg peak of the beam, respectively. The irradiated DNA samples were imaged by atomic force microscopy for visualization of individual DNA molecules, either broken or intact, and quantification of the DNA fragment length distributions for each of the irradiated samples. Percentage of the broken DNA and average number of DSBs per DNA molecule were obtained.ResultsCompared to irradiation effects in the plateau region, DNA irradiated at the Bragg peak sustained more breakage at the same dose, yielding more short DNA fragments and higher numbers of DSB per DNA molecule.ConclusionThe higher LET of proton beams at the Bragg peak results in more densely distributed DNA DSBs, which supports an underlying mechanism for the increased cell killing by protons at the Bragg peak.
Collapse
|