1
|
Dai Y, Yu Y, Nie J, Gu K, Pei H. X-ray-downregulated nucleophosmin induces abnormal polarization by anchoring to G-actin. LIFE SCIENCES IN SPACE RESEARCH 2024; 40:81-88. [PMID: 38245352 DOI: 10.1016/j.lssr.2023.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/02/2023] [Accepted: 09/04/2023] [Indexed: 01/22/2024]
Abstract
Ionizing radiation poses significant risks to astronauts during deep space exploration. This study investigates the impact of radiation on nucleophosmin (NPM), a protein involved in DNA repair, cell cycle regulation, and proliferation. Using X-rays, a common space radiation, we found that radiation suppresses NPM expression. Knockdown of NPM increases DNA damage after irradiation, disrupts cell cycle distribution and enhances cellular radiosensitivity. Additionally, NPM interacts with globular actin (G-actin), affecting its translocation and centrosome binding during mitosis. These findings provide insights into the role of NPM in cellular processes in responding to radiation. This article enhances our comprehension of radiation-induced genomic instability and provides a foundational platform for prospective investigations within the realm of space radiation and its implications for cancer therapy.
Collapse
Affiliation(s)
- Yingchu Dai
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, 215123, China.
| | - Yongduo Yu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Jing Nie
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, 215123, China; Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Ke Gu
- Department of Radiotherapy and Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Hailong Pei
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, 215123, China; Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China.
| |
Collapse
|
2
|
Montesinos CA, Khalid R, Cristea O, Greenberger JS, Epperly MW, Lemon JA, Boreham DR, Popov D, Gorthi G, Ramkumar N, Jones JA. Space Radiation Protection Countermeasures in Microgravity and Planetary Exploration. Life (Basel) 2021; 11:life11080829. [PMID: 34440577 PMCID: PMC8398261 DOI: 10.3390/life11080829] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/03/2021] [Accepted: 08/06/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Space radiation is one of the principal environmental factors limiting the human tolerance for space travel, and therefore a primary risk in need of mitigation strategies to enable crewed exploration of the solar system. METHODS We summarize the current state of knowledge regarding potential means to reduce the biological effects of space radiation. New countermeasure strategies for exploration-class missions are proposed, based on recent advances in nutrition, pharmacologic, and immune science. RESULTS Radiation protection can be categorized into (1) exposure-limiting: shielding and mission duration; (2) countermeasures: radioprotectors, radiomodulators, radiomitigators, and immune-modulation, and; (3) treatment and supportive care for the effects of radiation. Vehicle and mission design can augment the overall exposure. Testing in terrestrial laboratories and earth-based exposure facilities, as well as on the International Space Station (ISS), has demonstrated that dietary and pharmacologic countermeasures can be safe and effective. Immune system modulators are less robustly tested but show promise. Therapies for radiation prodromal syndrome may include pharmacologic agents; and autologous marrow for acute radiation syndrome (ARS). CONCLUSIONS Current radiation protection technology is not yet optimized, but nevertheless offers substantial protection to crews based on Lunar or Mars design reference missions. With additional research and human testing, the space radiation risk can be further mitigated to allow for long-duration exploration of the solar system.
Collapse
Affiliation(s)
| | - Radina Khalid
- School of Engineering, Rice University, Houston, TX 77005, USA;
| | - Octav Cristea
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Joel S. Greenberger
- Department of Radiation Oncology, University of Pittsburg Medical Center, Pittsburgh, PA 15213, USA; (J.S.G.); (M.W.E.)
| | - Michael W. Epperly
- Department of Radiation Oncology, University of Pittsburg Medical Center, Pittsburgh, PA 15213, USA; (J.S.G.); (M.W.E.)
| | - Jennifer A. Lemon
- Medical Sciences Division, Northern Ontario School of Medicine, Sudbury, ON P3E 2C6, Canada; (J.A.L.); (D.R.B.)
| | - Douglas R. Boreham
- Medical Sciences Division, Northern Ontario School of Medicine, Sudbury, ON P3E 2C6, Canada; (J.A.L.); (D.R.B.)
| | - Dmitri Popov
- Advanced Medical Technologies and Systems Inc., Richmond Hill, ON L4B 1N1, Canada;
| | | | - Nandita Ramkumar
- Center for Space Medicine, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Jeffrey A. Jones
- Center for Space Medicine, Department of Urology, Baylor College of Medicine, Houston, TX 77030, USA
- Correspondence:
| |
Collapse
|
3
|
Guo Z, Dai Y, Hu W, Zhang Y, Cao Z, Pei W, Liu N, Nie J, Wu A, Mao W, Chang L, Li B, Pei H, Hei TK, Zhou G. The long noncoding RNA CRYBG3 induces aneuploidy by interfering with spindle assembly checkpoint via direct binding with Bub3. Oncogene 2021; 40:1821-1835. [PMID: 33564066 PMCID: PMC7946627 DOI: 10.1038/s41388-020-01601-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 11/22/2020] [Accepted: 12/02/2020] [Indexed: 01/31/2023]
Abstract
Aneuploidy is a hallmark of genomic instability that leads to tumor initiation, progression, and metastasis. CDC20, Bub1, and Bub3 form the mitosis checkpoint complex (MCC) that binds the anaphase-promoting complex or cyclosome (APC/C), a crucial factor of the spindle assembly checkpoint (SAC), to ensure the bi-directional attachment and proper segregation of all sister chromosomes. However, just how MCC is regulated to ensure normal mitosis during cellular division remains unclear. In the present study, we demonstrated that LNC CRYBG3, an ionizing radiation-inducible long noncoding RNA, directly binds with Bub3 and interrupts its interaction with CDC20 to result in aneuploidy. The 261-317 (S3) residual of the LNC CRYBG3 sequence is critical for its interaction with Bub3 protein. Overexpression of LNC CRYBG3 leads to aneuploidy and promotes tumorigenesis and metastasis of lung cancer cells, implying that LNC CRYBG3 is a novel oncogene. These findings provide a novel mechanistic basis for the pathogenesis of NSCLC after exposure to ionizing radiation as well as a potential target for the diagnosis, treatment, and prognosis of an often fatal disease.
Collapse
Affiliation(s)
- Ziyang Guo
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Institute of Space Life Sciences, Medical College of Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
- Center for Radiological Research, College of Physician and Surgeons, Columbia University Medical Center, New York, NY, USA
| | - Yingchu Dai
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Institute of Space Life Sciences, Medical College of Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Wentao Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Institute of Space Life Sciences, Medical College of Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Yongsheng Zhang
- Department of Pathology, the Second Affiliated Hospital, Medical College of Soochow University, Suzhou, 215123, China
| | - Zhifei Cao
- Department of Pathology, the Second Affiliated Hospital, Medical College of Soochow University, Suzhou, 215123, China
| | - Weiwei Pei
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Institute of Space Life Sciences, Medical College of Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Ningang Liu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Institute of Space Life Sciences, Medical College of Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Jing Nie
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Institute of Space Life Sciences, Medical College of Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Anqing Wu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Institute of Space Life Sciences, Medical College of Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Weidong Mao
- Department of Pathology, the Second Affiliated Hospital, Medical College of Soochow University, Suzhou, 215123, China
| | - Lei Chang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Institute of Space Life Sciences, Medical College of Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Bingyan Li
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Institute of Space Life Sciences, Medical College of Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Hailong Pei
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Institute of Space Life Sciences, Medical College of Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China.
| | - Tom K Hei
- Center for Radiological Research, College of Physician and Surgeons, Columbia University Medical Center, New York, NY, USA.
| | - Guangming Zhou
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Institute of Space Life Sciences, Medical College of Soochow University, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China.
| |
Collapse
|
4
|
Slaba TC, Plante I, Ponomarev A, Patel ZS, Hada M. Determination of Chromosome Aberrations in Human Fibroblasts Irradiated by Mixed Fields Generated with Shielding. Radiat Res 2020; 194:246-258. [PMID: 32942302 DOI: 10.1667/rr15366.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 05/14/2020] [Indexed: 11/03/2022]
Abstract
To better study biological effects of space radiation using ground-based facilities, the NASA Space Radiation Laboratory (NSRL) at the Brookhaven National Laboratory has been upgraded to rapidly switch ions and energies. This has allowed investigators to design irradiation protocols comprising a mixture of ions and energies more indicative of the galactic cosmic ray (GCR) environment. Despite these advancements, beam selection and delivery schemes should be optimized against facility and experimental constraints and validated to ensure such irradiations are a suitable representation of the space environment. Importantly, since experiments are time consuming and expensive, models capable of predicting biological outcomes over a range of irradiation conditions (single ion, sequential multi ion or mixed fields) are needed to support such efforts. In this work, human fibroblasts were placed behind 20 g/cm2 aluminum and 10.345 g/cm2 polyethylene and irradiated separately by 344 MeV hydrogen, 344 MeV/n helium, 450 MeV/n oxygen and 950 MeV/n iron ions at various doses. The fluorescence in situ hybridization (FISH) whole chromosome painting technique was then used to assess the cells for chromosome aberrations (CAs), notably simple exchanges. A multi-scale modeling approach was also developed to predict the formation of chromosome aberrations in these experiments. The Geant4 simulation toolkit was used to determine the spectra of particles and energies produced by interactions between the incident beams and shielding. The simulated mixed field generated by shielding was then transferred into the track structure code, RITRACKS (relativistic ion tracks), to generate three-dimensional (3D) voxelized dose maps at the nanometer scale. Finally, these voxel dose maps were input into the new damage and repair model, RITCARD (radiation-induced tracks, chromosome aberrations, repair and damage), to predict the formation of various CAs. The multi-scale model described herein is a significant advancement for the computational tools used to predict biological outcomes in cells exposed to highly complex, mixed ion fields related to the GCR environment. Results show that the simulation and experimental data are in good agreement for the complex radiation fields generated by all ions incident on shielding for most data points. The differences between model predictions and measurements are discussed. Although improvements are needed, the model extends current capabilities for evaluating beam selection and delivery schemes at the NSRL ground-based GCR simulator and for informing NASA risk projection models in the future.
Collapse
Affiliation(s)
- Tony C Slaba
- NASA Langley Research Center, Hampton, Virginia 23681
| | | | | | | | - Megumi Hada
- Prairie View A&M University, Prairie View, Texas 77446
| |
Collapse
|
5
|
Affiliation(s)
- T. Cahill
- Faculty of Medicine, Health and Life Sciences School of Biological Sciences Institute for Global Food Security (IGFS) Belfast UK
| | - G. Hardiman
- Faculty of Medicine, Health and Life Sciences School of Biological Sciences Institute for Global Food Security (IGFS) Belfast UK
- Department of Medicine Medical University of South Carolina Charleston SC USA
| |
Collapse
|
6
|
Guo Z, Pei H, Nie J, Hu W, Zhang J, Ding J, Pan S, Li B, Hei TK, Chen W, Zhou G. Anti-cancer effects of CQBTO, a chloroquine, and benzo(e)triazine oxide conjugate. Chem Biol Drug Des 2019; 93:874-882. [PMID: 30637976 DOI: 10.1111/cbdd.13477] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 12/11/2018] [Accepted: 12/16/2018] [Indexed: 12/13/2022]
Abstract
AIM Autophagy is a self-protective process, and it confers cancer cells resistance against radio-chemotherapeutics. To induce cancer cell death, a series of compounds of 3-((4-((7-chloroquinolin-4-yl)amino)butyl)amino)-7-substituted benzo[e][1,2,4]triazine 1-oxide or CQBTO containing two critical chemical groups were designed and synthesized. One compound, benzo[e][1,2,4]triazine 1-oxide, yielded free radicals to trigger autophagy, and the other one, chloroquine (CQ), was an inhibitor of autophagy. We hypothesized that the compounds could kill cancer cells effectively by inducing incomplete autophagy. METHODS In vitro cultured non-small cell lung carcinoma cells and primary lung tumors in mice in vivo were used to test the lethal effects of CQBTO on cancer cells and toxicity to normal tissues. Cell viability was examined using the CCK8 assay. Genomic instability was determined with the cytochalasin B-blocked micronucleus assay. Cell cycle distribution was analyzed by propidium iodide staining and flow cytometry. Western blotting and immunofluorescence were used to detect the induction and localization of LC3, a biomarker for autophagy. RESULTS Compared with CQ, three CQBTO compounds were lethal to lung cancer cells, and CQBTO-3 was the most effective. The LD50 for CQBTO-3 was 21 μΜ in A549 cells and 21.5 μΜ in Calu-1 cells, which was lower than that of CQBTO-2 or CQBTO-1. Induction of LC3 foci and an increase in the LC3II/LC3I ratio demonstrated the induction of autophagy by CQBTO-3 in A549 cells, whereas no obvious micronuclei or cell cycle arrest was observed. No detectable toxicity to normal mice was observed. CQBTO-3 improved the quality of mouse life, reduced the number and size of existing tumors, and suppressed tumor formation. CONCLUSION CQBTO-3 is a potential chemical compound for lung cancer treatment.
Collapse
Affiliation(s)
- Ziyang Guo
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Institute of Space Life Sciences, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Medical College of Soochow University, Suzhou, China
| | - Hailong Pei
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Institute of Space Life Sciences, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Medical College of Soochow University, Suzhou, China
| | - Jing Nie
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Institute of Space Life Sciences, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Medical College of Soochow University, Suzhou, China
| | - Wentao Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Institute of Space Life Sciences, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Medical College of Soochow University, Suzhou, China
| | - Jian Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Institute of Space Life Sciences, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Medical College of Soochow University, Suzhou, China
| | - Jiahan Ding
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Institute of Space Life Sciences, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Medical College of Soochow University, Suzhou, China
| | - Shuxian Pan
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Institute of Space Life Sciences, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Medical College of Soochow University, Suzhou, China
| | - Bingyan Li
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Institute of Space Life Sciences, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Medical College of Soochow University, Suzhou, China
| | - Tom K Hei
- Center for Radiological Research, College of Physician and Surgeons, Columbia University, New York, New York
| | - Weiqiang Chen
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Guangming Zhou
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Institute of Space Life Sciences, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Medical College of Soochow University, Suzhou, China
| |
Collapse
|
7
|
Galactic Cosmic Radiation Induces Persistent Epigenome Alterations Relevant to Human Lung Cancer. Sci Rep 2018; 8:6709. [PMID: 29712937 PMCID: PMC5928241 DOI: 10.1038/s41598-018-24755-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 03/27/2018] [Indexed: 12/14/2022] Open
Abstract
Human deep space and planetary travel is limited by uncertainties regarding the health risks associated with exposure to galactic cosmic radiation (GCR), and in particular the high linear energy transfer (LET), heavy ion component. Here we assessed the impact of two high-LET ions 56Fe and 28Si, and low-LET X rays on genome-wide methylation patterns in human bronchial epithelial cells. We found that all three radiation types induced rapid and stable changes in DNA methylation but at distinct subsets of CpG sites affecting different chromatin compartments. The 56Fe ions induced mostly hypermethylation, and primarily affected sites in open chromatin regions including enhancers, promoters and the edges ("shores") of CpG islands. The 28Si ion-exposure had mixed effects, inducing both hyper and hypomethylation and affecting sites in more repressed heterochromatic environments, whereas X rays induced mostly hypomethylation, primarily at sites in gene bodies and intergenic regions. Significantly, the methylation status of 56Fe ion sensitive sites, but not those affected by X ray or 28Si ions, discriminated tumor from normal tissue for human lung adenocarcinomas and squamous cell carcinomas. Thus, high-LET radiation exposure leaves a lasting imprint on the epigenome, and affects sites relevant to human lung cancer. These methylation signatures may prove useful in monitoring the cumulative biological impact and associated cancer risks encountered by astronauts in deep space.
Collapse
|
8
|
Sasi SP, Yan X, Zuriaga-Herrero M, Gee H, Lee J, Mehrzad R, Song J, Onufrak J, Morgan J, Enderling H, Walsh K, Kishore R, Goukassian DA. Different Sequences of Fractionated Low-Dose Proton and Single Iron-Radiation-Induced Divergent Biological Responses in the Heart. Radiat Res 2017; 188:191-203. [PMID: 28613990 DOI: 10.1667/rr14667.1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Deep-space travel presents risks of exposure to ionizing radiation composed of a spectrum of low-fluence protons (1H) and high-charge and energy (HZE) iron nuclei (e.g., 56Fe). When exposed to galactic cosmic rays, each cell in the body may be traversed by 1H every 3-4 days and HZE nuclei every 3-4 months. The effects of low-dose sequential fractionated 1H or HZE on the heart are unknown. In this animal model of simulated ionizing radiation, middle-aged (8-9 months old) male C57BL/6NT mice were exposed to radiation as follows: group 1, nonirradiated controls; group 2, three fractionated doses of 17 cGy 1H every other day (1H × 3); group 3, three fractionated doses of 17 cGy 1H every other day followed by a single low dose of 15 cGy 56Fe two days after the final 1H dose (1H × 3 + 56Fe); and group 4, a single low dose of 15 cGy 56Fe followed (after 2 days) by three fractionated doses of 17 cGy 1H every other day (56Fe + 1H × 3). A subgroup of mice from each group underwent myocardial infarction (MI) surgery at 28 days postirradiation. Cardiac structure and function were assessed in all animals at days 7, 14 and 28 after MI surgery was performed. Compared to the control animals, the treatments that groups 2 and 3 received did not induce negative effects on cardiac function or structure. However, compared to all other groups, the animals in group 4, showed depressed left ventricular (LV) functions at 1 month with concomitant enhancement in cardiac fibrosis and induction of cardiac hypertrophy signaling at 3 months. In the irradiated and MI surgery groups compared to the control group, the treatments received by groups 2 and 4 did not induce negative effects at 1 month postirradiation and MI surgery. However, in group 3 after MI surgery, there was a 24% increase in mortality, significant decreases in LV function and a 35% increase in post-infarction size. These changes were associated with significant decreases in the angiogenic and cell survival signaling pathways. These data suggest that fractionated doses of radiation induces cellular and molecular changes that result in depressed heart functions both under basal conditions and particularly after myocardial infarction.
Collapse
Affiliation(s)
- Sharath P Sasi
- a Cardiovascular Research Center, GeneSys Research Institute, Boston, Massachusetts
| | - Xinhua Yan
- a Cardiovascular Research Center, GeneSys Research Institute, Boston, Massachusetts.,b Tufts University School of Medicine, Boston, Massachusetts
| | - Marian Zuriaga-Herrero
- f Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| | - Hannah Gee
- a Cardiovascular Research Center, GeneSys Research Institute, Boston, Massachusetts
| | - Juyong Lee
- c Calhoun Cardiology Center, University of Connecticut Health Center, Farmington, Connecticut
| | - Raman Mehrzad
- d Steward Carney Hospital, Dorchester, Massachusetts
| | - Jin Song
- a Cardiovascular Research Center, GeneSys Research Institute, Boston, Massachusetts
| | - Jillian Onufrak
- a Cardiovascular Research Center, GeneSys Research Institute, Boston, Massachusetts
| | - James Morgan
- b Tufts University School of Medicine, Boston, Massachusetts.,d Steward Carney Hospital, Dorchester, Massachusetts
| | - Heiko Enderling
- e Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Kenneth Walsh
- f Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| | - Raj Kishore
- 7 Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - David A Goukassian
- a Cardiovascular Research Center, GeneSys Research Institute, Boston, Massachusetts.,f Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts.,7 Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
9
|
Pei H, Zhang J, Nie J, Ding N, Hu W, Hua J, Hirayama R, Furusawa Y, Liu C, Li B, Hei TK, Zhou G. RAC2-P38 MAPK-dependent NADPH oxidase activity is associated with the resistance of quiescent cells to ionizing radiation. Cell Cycle 2017; 16:113-122. [PMID: 27936335 PMCID: PMC5270549 DOI: 10.1080/15384101.2016.1259039] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 11/01/2016] [Accepted: 11/04/2016] [Indexed: 01/13/2023] Open
Abstract
Our recent study showed that quiescent G0 cells are more resistant to ionizing radiation than G1 cells; however, the underlying mechanism for this increased radioresistance is unknown. Based on the relatively lower DNA damage induced in G0 cells, we hypothesize that these cells are exposed to less oxidative stress during exposure. As a catalytic subunit of NADPH oxidase, Ras-related C3 botulinum toxin substrate 2 (RAC2) may be involved in the cellular response to ionizing radiation. Here, we show that RAC2 was expressed at low levels in G0 cells but increased substantially in G1 cells. Relative to G1 cells, the total antioxidant capacity in G0 phase cells increased upon exposure to X-ray radiation, whereas the intracellular concentration of ROS and malondialdehyde increased only slightly. The induction of DNA single- and double-stranded breaks in G1 cells by X-ray radiation was inhibited by knockdown of RAC2. P38 MAPK interaction with RAC2 resulted in a decrease of functional RAC2. Increased phosphorylation of P38 MAPK in G0 cells also increased cellular radioresistance; however, excessive production of ROS caused P38 MAPK dephosphorylation. P38 MAPK, phosphorylated P38 MAPK, and RAC2 regulated in mutual feedback and negative feedback regulatory pathways, resulting in the radioresistance of G0 cells.
Collapse
Affiliation(s)
- Hailong Pei
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China
- Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, China
- Institute of Space Life Sciences, Soochow University, Suzhou, China
| | - Jian Zhang
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China
- Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, China
- Institute of Space Life Sciences, Soochow University, Suzhou, China
| | - Jing Nie
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China
- Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, China
- Institute of Space Life Sciences, Soochow University, Suzhou, China
| | - Nan Ding
- Department of Space Radiobiology, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Wentao Hu
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China
- Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, China
- Institute of Space Life Sciences, Soochow University, Suzhou, China
| | - Junrui Hua
- Department of Space Radiobiology, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Ryoichi Hirayama
- Research Center for Charged Particle Therapy, National Institutes for Quanturn and Radiological Science and Technology, Chiba, Japan
| | - Yoshiya Furusawa
- Research Center for Charged Particle Therapy, National Institutes for Quanturn and Radiological Science and Technology, Chiba, Japan
| | - Cuihua Liu
- Research Center for Charged Particle Therapy, National Institutes for Quanturn and Radiological Science and Technology, Chiba, Japan
| | - Bingyan Li
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China
- Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, China
| | - Tom K. Hei
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China
- Center for Radiological Research, College of Physician and Surgeons, Columbia University, New York, NY, USA
| | - Guangming Zhou
- School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China
- Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou, China
- Institute of Space Life Sciences, Soochow University, Suzhou, China
| |
Collapse
|
10
|
Wu X, Ding N, Hu W, He J, Xu S, Pei H, Hua J, Zhou G, Wang J. Down-regulation of BTG1 by miR-454-3p enhances cellular radiosensitivity in renal carcinoma cells. Radiat Oncol 2014; 9:179. [PMID: 25115181 PMCID: PMC4252025 DOI: 10.1186/1748-717x-9-179] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 08/08/2014] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND B cell translocation gene 1 (BTG1) has long been recognized as a tumor suppressor gene. Recent reports demonstrated that BTG1 plays an important role in progression of cell cycle and is involved in cellular response to stressors. However, the microRNAs mediated regulatory mechanism of BTG1 expression has not been reported so far. MicroRNAs can effectively influence tumor radiosensitivity by preventing cell cycle progression, resulting in enhancement of the cytotoxicity of radiotherapy efficacy. This study aimed to demonstrating the effects of microRNAs on the BTG1 expression and cellular radiosensitivity. METHODS The human renal carcinoma 786-O cells were treated with 5 Gy of X-rays. Expressions of BTG1 gene and miR-454-3p, which was predicted to target BTG1 by software algorithm, were analyzed by quantitative polymerase chain reaction. Protein expressions were assessed by Western blot. Luciferase assays were used to quantify the interaction between BTG1 3'-untranslated region (3'-UTR) and miR-454-3p. The radiosensitivity was quantified by the assay of cell viability, colony formation and caspase-3 activity. RESULTS The expression of the BTG1 gene in 786-O cells was significantly elevated after treatments with X-ray irradiation, DMSO, or serum starvation. The up-regulation of BTG1 after irradiation reduced cellular radiosensitivity as demonstrated by the enhanced cell viability and colony formation, as well as the repressed caspase-3 activity. In comparison, knock down of BTG1 by siRNA led to significantly enhanced cellular radiosensitivity. It was found that miR-454-3p can regulate the expression of BTG1 through a direct interaction with the 3'-UTR of BTG1 mRNA. Decreasing of its expression level correlates well with BTG1 up-regulation during X-ray irradiation. Particularly, we observed that over-expression of miR-454-3p by transfection inhibited the BTG1 expression and enhanced the radiosensitivity. In addition, cell cycle analysis showed that over-expression of miR-454-3p shifted the cell cycle arrest from G2/M phase to S phase. CONCLUSIONS Our results indicate that BTG1 is a direct target of miR-454-3p. Down-regulation of BTG1 by miR-454-3p renders tumor cells sensitive to radiation. These results may shed light on the potential application in tumor radiotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jufang Wang
- Department of Space Radiobiology, Key Laboratory of Heavy Ion Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 509 Nanchang Road, Lanzhou 730000, China.
| |
Collapse
|