1
|
Sandhu S, Keyworth M, Karimi-Jashni S, Alomar D, Smith BJ, Kozbenko T, Doty S, Hocking R, Hamada N, Reynolds RJ, Scott RT, Costes SV, Beheshti A, Yauk C, Wilkins RC, Chauhan V. AOP Report: Development of an adverse outcome pathway for deposition of energy leading to bone loss. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2024; 65 Suppl 3:85-111. [PMID: 39387375 DOI: 10.1002/em.22631] [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: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 10/15/2024]
Abstract
Bone loss, commonly seen in osteoporosis, is a condition that entails a progressive decline of bone mineral density and microarchitecture, often seen in post-menopausal women. Bone loss has also been widely reported in astronauts exposed to a plethora of stressors and in patients with osteoporosis following radiotherapy for cancer. Studies on mechanisms are well documented but the causal connectivity of events to bone loss development remains incompletely understood. Herein, the adverse outcome pathway (AOP) framework was used to organize data and develop a qualitative AOP beginning from deposition of energy (the molecular initiating event) to bone loss (the adverse outcome). This qualitative AOP was developed in collaboration with bone loss research experts to aggregate relevant findings, supporting ongoing efforts to understand and mitigate human system risks associated with radiation exposures. A literature review was conducted to compile and evaluate the state of knowledge based on the modified Bradford Hill criteria. Following review of 2029 studies, an empirically supported AOP was developed, showing the progression to bone loss through many factors affecting the activities of bone-forming osteoblasts and bone-resorbing osteoclasts. The structural, functional, and quantitative basis of each proposed relationship was defined, for inference of causal changes between key events. Current knowledge and its gaps relating to dose-, time- and incidence-concordance across the key events were identified, as well as modulating factors that influence linkages. The new priorities for research informed by the AOP highlight areas for improvement to enable development of a quantitative AOP used to support risk assessment strategies for space travel or cancer radiotherapy.
Collapse
Affiliation(s)
- Snehpal Sandhu
- Consumer and Clinical Radiation Protection Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Mitchell Keyworth
- Consumer and Clinical Radiation Protection Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Syna Karimi-Jashni
- Consumer and Clinical Radiation Protection Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Dalya Alomar
- Consumer and Clinical Radiation Protection Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Benjamin J Smith
- Consumer and Clinical Radiation Protection Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Tatiana Kozbenko
- Consumer and Clinical Radiation Protection Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Stephen Doty
- Hospital for Special Surgery Research Institute, New York City, New York, USA
| | - Robyn Hocking
- Learning and Knowledge and Library Services, Health Canada, Ottawa, Ontario, Canada
| | - Nobuyuki Hamada
- Biology and Environmental Chemistry Division, Substantiable System Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), Chiba, Japan
| | | | - Ryan T Scott
- KBR, NASA Ames Research Center, Moffett Field, California, USA
| | - Sylvain V Costes
- NASA Ames Research Center, Space Biosciences Research Branch, Mountain View, California, USA
| | - Afshin Beheshti
- McGowan Institute for Regenerative Medicine - Center for Space Biomedicine, Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Carole Yauk
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Ruth C Wilkins
- Consumer and Clinical Radiation Protection Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Vinita Chauhan
- Consumer and Clinical Radiation Protection Bureau, Health Canada, Ottawa, Ontario, Canada
| |
Collapse
|
2
|
Park SH, Peters M, Aguayo C, Farris MK, Hughes RT, Moore J, Munley MT, Reno KE, Gardin J, Cline JM, Peters CM, Willey JS. Secretory factors released from high dose radiation-activated osteoclasts increase the expression level of pain-associated neuropeptides in sensory neuronal cultures. RESEARCH SQUARE 2024:rs.3.rs-4534694. [PMID: 39011106 PMCID: PMC11247937 DOI: 10.21203/rs.3.rs-4534694/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
Stereotactic Body Radiation Therapy for lung tumors near the chest wall often causes significant chest wall pain (CWP), negatively impacting patients' quality of life. The mechanisms behind SBRT-induced CWP remain unclear and may involve multiple factors. We investigated the potential crosstalk between radiation-activated osteoclasts and sensory neurons, focusing on osteoclast-derived factors in CWP. Using the murine pre-osteoclast cell line Raw264.7, we induced differentiation with RANKL, followed by 10Gy gamma-irradiation. Conditioned media from these irradiated osteoclasts was used to treat sensory neuronal cultures from mouse dorsal root ganglia. Neuronal cultures were also directly exposed to 10Gy radiation, with and without osteoclast co-culture. Analysis of osteoclast markers and pain-associated neuropeptides was conducted using RT-qPCR and histochemical staining. Osteoclast differentiation and activity were inhibited using Osteoprotegerin and risedronate. Results showed that high-dose radiation significantly increased osteoclast size, resorption pit size, and activity biomarkers. Neurons treated with CM from irradiated osteoclasts showed increased expression of pain-associated neuropeptides CGRP and Substance P, which was mitigated by osteoprotegerin and risedronate. This study suggests that high-dose radiation enhances osteoclast activity, upregulating pain-associated neuropeptides in sensory neurons, and that inhibitors like osteoprotegerin and risedronate may offer therapeutic strategies for managing radiation-induced pain.
Collapse
Affiliation(s)
- Sun H Park
- Wake Forest University School of Medicine
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Xiang L, Li F, Xiang Y, Zhang W, Shi D, Zhang X, Chen L, Ran Q, Li Z. CR6-Interacting Factor-1 Promotes Osteoclastogenesis Through the NF-κB Signaling Pathway after Irradiation. Radiat Res 2023; 200:489-502. [PMID: 37815199 DOI: 10.1667/rade-22-00066.1] [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: 04/11/2022] [Accepted: 09/11/2023] [Indexed: 10/11/2023]
Abstract
Radiation exposure arising from radiotherapy may induce rapid bone loss and an increase in the extent of bone resorption. Reactive oxygen species (ROS) caused by radiation exposure play a crucial role during the process of osteoclastogenesis. However, the pathological mechanisms underlying radiation-induced osteoclastogenesis have yet to be fully elucidated. CR6-interacting factor-1 (Crif1) as a multifunctional protein is involved in regulating multiple biological functions in cells. Here, we investigated the role of Crif1 in radiation-induced osteoclastogenesis and found that radiation exposure induced an increase in the expression level of Crif1 and enhanced osteoclastogenesis in osteoclast progenitors. Crif1 and NF-κB p65 co-localized in the cytoplasm after radiation exposure. Crif1 knockdown did not affect the phosphorylation and total protein levels of extracellular signal-regulated kinases (ERK), c-Jun amino (N)-terminal kinases (JNK), p38, and IκB-α before and after irradiation. However, Crif1 knockdown did lead to the reduced phosphorylation and nuclear translocation of NF-κB p65 after irradiation and resulted in a reduced level of osteoclastogenesis in RAW264.7 cells after irradiation. In vivo studies involving Lyz2Cre;Crif1fl/fl mice possessing the myeloid-specific deletion of Crif1 demonstrated the alleviation of bone loss after irradiation when compared with Crif1fl/fl mice. Our findings demonstrate that Crif1 mediated the phosphorylation and nuclear translocation of NF-κB p65 and promoted osteoclastogenesis via the NF-κB signaling pathway after radiation exposure. Thus, our analysis revealed a specific role for Crif1 in the mediation of radiation-induced bone loss and may provide new insight into potential therapeutic strategies for radiation-induced bone loss.
Collapse
Affiliation(s)
- Lixin Xiang
- Basic Research Innovation Center for Acute Radiation Syndrome, Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Fengjie Li
- Basic Research Innovation Center for Acute Radiation Syndrome, Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Yang Xiang
- Basic Research Innovation Center for Acute Radiation Syndrome, Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Weiwei Zhang
- Basic Research Innovation Center for Acute Radiation Syndrome, Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Dongling Shi
- Basic Research Innovation Center for Acute Radiation Syndrome, Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Xiaomei Zhang
- Basic Research Innovation Center for Acute Radiation Syndrome, Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Li Chen
- Basic Research Innovation Center for Acute Radiation Syndrome, Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Qian Ran
- Basic Research Innovation Center for Acute Radiation Syndrome, Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Zhongjun Li
- Basic Research Innovation Center for Acute Radiation Syndrome, Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, China
| |
Collapse
|
4
|
Wei F, Tuong ZK, Omer M, Ngo C, Asiatico J, Kinzel M, Pugazhendhi AS, Khaled AR, Ghosh R, Coathup M. A novel multifunctional radioprotective strategy using P7C3 as a countermeasure against ionizing radiation-induced bone loss. Bone Res 2023; 11:34. [PMID: 37385982 DOI: 10.1038/s41413-023-00273-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 05/16/2023] [Accepted: 05/28/2023] [Indexed: 07/01/2023] Open
Abstract
Radiotherapy is a critical component of cancer care but can cause osteoporosis and pathological insufficiency fractures in surrounding and otherwise healthy bone. Presently, no effective countermeasure exists, and ionizing radiation-induced bone damage continues to be a substantial source of pain and morbidity. The purpose of this study was to investigate a small molecule aminopropyl carbazole named P7C3 as a novel radioprotective strategy. Our studies revealed that P7C3 repressed ionizing radiation (IR)-induced osteoclastic activity, inhibited adipogenesis, and promoted osteoblastogenesis and mineral deposition in vitro. We also demonstrated that rodents exposed to clinically equivalent hypofractionated levels of IR in vivo develop weakened, osteoporotic bone. However, the administration of P7C3 significantly inhibited osteoclastic activity, lipid formation and bone marrow adiposity and mitigated tissue loss such that bone maintained its area, architecture, and mechanical strength. Our findings revealed significant enhancement of cellular macromolecule metabolic processes, myeloid cell differentiation, and the proteins LRP-4, TAGLN, ILK, and Tollip, with downregulation of GDF-3, SH2B1, and CD200. These proteins are key in favoring osteoblast over adipogenic progenitor differentiation, cell matrix interactions, and shape and motility, facilitating inflammatory resolution, and suppressing osteoclastogenesis, potentially via Wnt/β-catenin signaling. A concern was whether P7C3 afforded similar protection to cancer cells. Preliminarily, and remarkably, at the same protective P7C3 dose, a significant reduction in triple-negative breast cancer and osteosarcoma cell metabolic activity was found in vitro. Together, these results indicate that P7C3 is a previously undiscovered key regulator of adipo-osteogenic progenitor lineage commitment and may serve as a novel multifunctional therapeutic strategy, leaving IR an effective clinical tool while diminishing the risk of adverse post-IR complications. Our data uncover a new approach for the prevention of radiation-induced bone damage, and further work is needed to investigate its ability to selectively drive cancer cell death.
Collapse
Affiliation(s)
- Fei Wei
- Biionix Cluster, and Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Zewen Kelvin Tuong
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK
- Cellular Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Mahmoud Omer
- Biionix Cluster, and Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Christopher Ngo
- Biionix Cluster, and Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Jackson Asiatico
- Department of Mechanical and Aerospace Engineering, University of Central Florida, Orlando, FL, USA
| | - Michael Kinzel
- Department of Mechanical and Aerospace Engineering, University of Central Florida, Orlando, FL, USA
| | - Abinaya Sindu Pugazhendhi
- Biionix Cluster, and Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Annette R Khaled
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Ranajay Ghosh
- Department of Mechanical and Aerospace Engineering, University of Central Florida, Orlando, FL, USA
| | - Melanie Coathup
- Biionix Cluster, and Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, USA.
| |
Collapse
|
5
|
Bareke H, Ibáñez-Navarro A, Guerra-García P, González Pérez C, Rubio-Aparicio P, Plaza López de Sabando D, Sastre-Urgelles A, Ortiz-Cruz EJ, Pérez-Martínez A. Prospects and Advances in Adoptive Natural Killer Cell Therapy for Unmet Therapeutic Needs in Pediatric Bone Sarcomas. Int J Mol Sci 2023; 24:ijms24098324. [PMID: 37176035 PMCID: PMC10178897 DOI: 10.3390/ijms24098324] [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: 03/21/2023] [Revised: 04/25/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
Malignant bone tumors are aggressive tumors, with a high tendency to metastasize, that are observed most frequently in adolescents during rapid growth spurts. Pediatric patients with malignant bone sarcomas, Ewing sarcoma and osteosarcoma, who present with progressive disease have dire survival rates despite aggressive therapy. These therapies can have long-term effects on bone growth, such as decreased bone mineral density and reduced longitudinal growth. New therapeutic approaches are therefore urgently needed for targeting pediatric malignant bone tumors. Harnessing the power of the immune system against cancer has improved the survival rates dramatically in certain cancer types. Natural killer (NK) cells are a heterogeneous group of innate effector cells that possess numerous antitumor effects, such as cytolysis and cytokine production. Pediatric sarcoma cells have been shown to be especially susceptible to NK-cell-mediated killing. NK-cell adoptive therapy confers numerous advantages over T-cell adoptive therapy, including a good safety profile and a lack of major histocompatibility complex restriction. NK-cell immunotherapy has the potential to be a new therapy for pediatric malignant bone tumors. In this manuscript, we review the general characteristics of osteosarcoma and Ewing sarcoma, discuss the long-term effects of sarcoma treatment on bones, and the barriers to effective immunotherapy in bone sarcomas. We then present the laboratory and clinical studies on NK-cell immunotherapy for pediatric malignant bone tumors. We discuss the various donor sources and NK-cell types, the engineering of NK cells and combinatorial treatment approaches that are being studied to overcome the current challenges in adoptive NK-cell therapy, while suggesting approaches for future studies on NK-cell immunotherapy in pediatric bone tumors.
Collapse
Affiliation(s)
- Halin Bareke
- Translational Research Group in Pediatric Oncology, Haematopoietic Transplantation and Cell Therapy, Hospital La Paz Institute for Health Research, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain
| | - Adrián Ibáñez-Navarro
- Translational Research Group in Pediatric Oncology, Haematopoietic Transplantation and Cell Therapy, Hospital La Paz Institute for Health Research, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain
| | - Pilar Guerra-García
- Department of Pediatric Hemato-Oncology, La Paz University Hospital, 28046 Madrid, Spain
| | - Carlos González Pérez
- Department of Pediatric Hemato-Oncology, La Paz University Hospital, 28046 Madrid, Spain
| | - Pedro Rubio-Aparicio
- Department of Pediatric Hemato-Oncology, La Paz University Hospital, 28046 Madrid, Spain
| | | | - Ana Sastre-Urgelles
- Department of Pediatric Hemato-Oncology, La Paz University Hospital, 28046 Madrid, Spain
| | - Eduardo José Ortiz-Cruz
- Department of Orthopedic Surgery and Traumatology, La Paz University Hospital, 28046 Madrid, Spain
| | - Antonio Pérez-Martínez
- Translational Research Group in Pediatric Oncology, Haematopoietic Transplantation and Cell Therapy, Hospital La Paz Institute for Health Research, IdiPAZ, La Paz University Hospital, 28046 Madrid, Spain
- Department of Pediatric Hemato-Oncology, La Paz University Hospital, 28046 Madrid, Spain
- School of Medicine, Autonomous University of Madrid, 28046 Madrid, Spain
| |
Collapse
|
6
|
Wei F, Neal CJ, Sakthivel TS, Fu Y, Omer M, Adhikary A, Ward S, Ta KM, Moxon S, Molinari M, Asiatico J, Kinzel M, Yarmolenko SN, San Cheong V, Orlovskaya N, Ghosh R, Seal S, Coathup M. A novel approach for the prevention of ionizing radiation-induced bone loss using a designer multifunctional cerium oxide nanozyme. Bioact Mater 2022; 21:547-565. [PMID: 36185749 PMCID: PMC9507991 DOI: 10.1016/j.bioactmat.2022.09.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/18/2022] Open
Abstract
The disability, mortality and costs due to ionizing radiation (IR)-induced osteoporotic bone fractures are substantial and no effective therapy exists. Ionizing radiation increases cellular oxidative damage, causing an imbalance in bone turnover that is primarily driven via heightened activity of the bone-resorbing osteoclast. We demonstrate that rats exposed to sublethal levels of IR develop fragile, osteoporotic bone. At reactive surface sites, cerium ions have the ability to easily undergo redox cycling: drastically adjusting their electronic configurations and versatile catalytic activities. These properties make cerium oxide nanomaterials fascinating. We show that an engineered artificial nanozyme composed of cerium oxide, and designed to possess a higher fraction of trivalent (Ce3+) surface sites, mitigates the IR-induced loss in bone area, bone architecture, and strength. These investigations also demonstrate that our nanozyme furnishes several mechanistic avenues of protection and selectively targets highly damaging reactive oxygen species, protecting the rats against IR-induced DNA damage, cellular senescence, and elevated osteoclastic activity in vitro and in vivo. Further, we reveal that our nanozyme is a previously unreported key regulator of osteoclast formation derived from macrophages while also directly targeting bone progenitor cells, favoring new bone formation despite its exposure to harmful levels of IR in vitro. These findings open a new approach for the specific prevention of IR-induced bone loss using synthesis-mediated designer multifunctional nanomaterials.
Collapse
Key Words
- ALP, Alkaline phosphatase
- BMSC, Bone marrow derived mesenchymal stem cells
- Bone resorption
- Bone strength
- CAT, Catalase
- COLI, Collagen type I
- CTSK, Cathepsin K
- CTX-1, Cross-linked C-telopeptide of type I collagen
- CeONPs, Cerium oxide nanoparticles
- Cerium oxide
- DFT, Density functional theory
- DNA, Deoxyribonucleic acid
- EPR, Electron paramagnetic resonance
- FDA, Food and Drug Administration
- GPX, Glutathione peroxidase
- Gy, Gray
- HIF1α, Hypoxia-inducible factor 1 alpha
- IL-1β, Interleukin 1 beta
- IL-6, Interleukin 6
- IR, Ionizing radiation
- Ionizing radiation
- MNGC, Multinucleated giant cell
- Nanozyme
- OCN, Osteocalcin
- Osteoporosis
- RANKL, Receptor activator of nuclear factor kappa-Β ligand
- ROS, Reactive oxygen species
- SAED, Selected area electron diffraction
- SOD, Superoxide dismutase
- TRAP, Tartrate-resistant acid phosphatase
- XPS, X-ray photoelectron spectroscopy
Collapse
Affiliation(s)
- Fei Wei
- Biionix Cluster, Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Craig J. Neal
- Advanced Materials Processing and Analysis Centre, Nanoscience Technology Center (NSTC), Materials Science and Engineering, College of Medicine, University of Central Florida, Orlando, FL, USA
| | | | - Yifei Fu
- Advanced Materials Processing and Analysis Centre, Nanoscience Technology Center (NSTC), Materials Science and Engineering, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Mahmoud Omer
- Biionix Cluster, Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Amitava Adhikary
- Department of Chemistry, Oakland University, Rochester, MI, MI, USA
| | - Samuel Ward
- Department of Chemistry, Oakland University, Rochester, MI, MI, USA
| | - Khoa Minh Ta
- School of Applied Sciences, Department of Chemical Sciences, University of Huddersfield, UK
| | - Samuel Moxon
- School of Applied Sciences, Department of Chemical Sciences, University of Huddersfield, UK
| | - Marco Molinari
- School of Applied Sciences, Department of Chemical Sciences, University of Huddersfield, UK
| | - Jackson Asiatico
- Department of Mechanical and Aerospace Engineering, University of Central Florida, Orlando, FL, USA
| | - Michael Kinzel
- Department of Mechanical and Aerospace Engineering, University of Central Florida, Orlando, FL, USA
| | - Sergey N. Yarmolenko
- Engineering Research Center for Revolutionizing Biomaterials, North Carolina A & T University, Greensboro, NC, USA
| | - Vee San Cheong
- Department of Automatic Control and Systems Engineering, Insigneo Institute for In Silico Medicine, University of Sheffield, Sheffield, S1 3JD, UK
| | - Nina Orlovskaya
- Department of Mechanical and Aerospace Engineering, University of Central Florida, Orlando, FL, USA
| | - Ranajay Ghosh
- Department of Mechanical and Aerospace Engineering, University of Central Florida, Orlando, FL, USA
| | - Sudipta Seal
- Biionix Cluster, Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, USA
- Advanced Materials Processing and Analysis Centre, Nanoscience Technology Center (NSTC), Materials Science and Engineering, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Melanie Coathup
- Biionix Cluster, Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, USA
- Corresponding author. Biionix Cluster, Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, USA.
| |
Collapse
|
7
|
Maier I, Ruegger PM, Deutschmann J, Helbich TH, Pietschmann P, Schiestl RH, Borneman J. Particle Radiation Side-Effects: Intestinal Microbiota Composition Shapes Interferon-γ-Induced Osteo-Immunogenicity. Radiat Res 2022; 197:184-192. [PMID: 35130347 DOI: 10.1667/rade-21-00065.1] [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: 03/31/2021] [Accepted: 11/09/2021] [Indexed: 07/25/2024]
Abstract
Microbiota can both negatively and positively impact radiation-induced bone loss. Our prior research showed that compared to mice with conventional gut microbiota (CM), mice with restricted gut microbiota (RM) reduced inflammatory tumor necrosis factor (TNF) in bone marrow, interleukin (IL)-17 in blood, and chemokine (C-C motif) ligand 20 (CCL20) in bone marrow under anti-IL-17 treatment. We showed that Muribaculum intestinale was more abundant in intestinal epithelial cells (IECs) from the small intestine of female RM mice and positively associated with augmented skeletal bone structure. Female C57BL/6J pun RM mice, which were injected with anti-IL-17 antibody one day before exposure to 1.5 Gy 28Si ions of 850 MeV/u, showed high trabecular numbers in tibiae at 6 weeks postirradiation. Irradiated CM mice were investigated for lower interferon-γ and IL-17 levels in the small intestine than RM mice. IL-17 blockage resulted in bacterial indicator phylotypes being different between both microbiota groups before and after irradiation. Analysis of the fecal bacteria were performed in relation to bone quality and body weight, showing reduced tibia cortical thickness in irradiated CM mice (-15%) vs. irradiated RM mice (-9.2%). Correlation analyses identified relationships among trabecular bone parameters (TRI-BV/TV, Tb.N, Tb.Th, Tb.Sp) and Bacteroides massiliensis, Muribaculum sp. and Prevotella denticola. Turicibacter sp. was found directly correlated with trabecular separation in anti-IL-17 treated mice, whereas an unidentified Bacteroidetes correlated with trabecular thickness in anti-IL-17 neutralized and radiation-exposed mice. We demonstrated radiation-induced osteolytic damage to correlate with bacterial indicator phylotypes of the intestinal microbiota composition, and these relationships were determined from the previously discovered dose-dependent particle radiation effects on cell proliferation in bone tissue. New translational approaches were designed to investigate dynamic changes of gut microbiota in correlation with conditions of treatment and disease as well as mechanisms of systemic side-effects in radiotherapy.
Collapse
Affiliation(s)
- Irene Maier
- Department of Environmental Health Sciences, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, California
| | - Paul M Ruegger
- Department of Microbiology and Plant Pathology, University of California, Riverside, Riverside, California
| | - Julia Deutschmann
- Department for Radiologic Technology, University of Applied Sciences Wiener Neustadt for Business and Engineering Ltd., Lower Austria, Austria
| | - Thomas H Helbich
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Peter Pietschmann
- Institute of Pathophysiology and Allergy Research, Medical University of Vienna, Waehringer Guertel 18-20, A-1090 Vienna, Austria
| | - Robert H Schiestl
- Departments of Pathology and Environmental Health Sciences, University of California, Los Angeles, Los Angeles, California
| | - James Borneman
- Department of Microbiology and Plant Pathology, University of California, Riverside, Riverside, California
| |
Collapse
|
8
|
Richardson KK, Ling W, Krager K, Fu Q, Byrum SD, Pathak R, Aykin-Burns N, Kim HN. Ionizing Radiation Activates Mitochondrial Function in Osteoclasts and Causes Bone Loss in Young Adult Male Mice. Int J Mol Sci 2022; 23:675. [PMID: 35054859 PMCID: PMC8775597 DOI: 10.3390/ijms23020675] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/03/2022] [Accepted: 01/07/2022] [Indexed: 12/13/2022] Open
Abstract
The damaging effects of ionizing radiation (IR) on bone mass are well-documented in mice and humans and are most likely due to increased osteoclast number and function. However, the mechanisms leading to inappropriate increases in osteoclastic bone resorption are only partially understood. Here, we show that exposure to multiple fractions of low-doses (10 fractions of 0.4 Gy total body irradiation [TBI]/week, i.e., fractionated exposure) and/or a single exposure to the same total dose of 4 Gy TBI causes a decrease in trabecular, but not cortical, bone mass in young adult male mice. This damaging effect was associated with highly activated bone resorption. Both osteoclast differentiation and maturation increased in cultures of bone marrow-derived macrophages from mice exposed to either fractionated or singular TBI. IR also increased the expression and enzymatic activity of mitochondrial deacetylase Sirtuin-3 (Sirt3)-an essential protein for osteoclast mitochondrial activity and bone resorption in the development of osteoporosis. Osteoclast progenitors lacking Sirt3 exposed to IR exhibited impaired resorptive activity. Taken together, targeting impairment of osteoclast mitochondrial activity could be a novel therapeutic strategy for IR-induced bone loss, and Sirt3 is likely a major mediator of this effect.
Collapse
Affiliation(s)
- Kimberly K. Richardson
- Center for Musculoskeletal Disease Research and Center for Osteoporosis and Metabolic Bone Diseases, Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.K.R.); (W.L.); (Q.F.)
| | - Wen Ling
- Center for Musculoskeletal Disease Research and Center for Osteoporosis and Metabolic Bone Diseases, Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.K.R.); (W.L.); (Q.F.)
| | - Kimberly Krager
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.K.); (R.P.); (N.A.-B.)
| | - Qiang Fu
- Center for Musculoskeletal Disease Research and Center for Osteoporosis and Metabolic Bone Diseases, Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.K.R.); (W.L.); (Q.F.)
| | - Stephanie D. Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Rupak Pathak
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.K.); (R.P.); (N.A.-B.)
| | - Nukhet Aykin-Burns
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.K.); (R.P.); (N.A.-B.)
| | - Ha-Neui Kim
- Center for Musculoskeletal Disease Research and Center for Osteoporosis and Metabolic Bone Diseases, Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (K.K.R.); (W.L.); (Q.F.)
| |
Collapse
|
9
|
Kumar K, Datta K, Fornace AJ, Suman S. Total body proton and heavy-ion irradiation causes cellular senescence and promotes pro-osteoclastogenic activity in mouse bone marrow. Heliyon 2022; 8:e08691. [PMID: 35028468 PMCID: PMC8741516 DOI: 10.1016/j.heliyon.2021.e08691] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/01/2021] [Accepted: 12/27/2021] [Indexed: 12/12/2022] Open
Abstract
Low-LET photon radiation-induced persistent alterations in bone marrow (BM) cells are well documented in total-body irradiated (TBI) rodents and also among radiotherapy patients. However, the late effects of protons and high-LET heavy-ion radiation on BM cells and its implications in osteoclastogenesis are not fully understood. Therefore, C57BL6/J female mice (8 weeks; n = 10/group) were irradiated to sham, and 1 Gy of the proton (0.22 keV/μm), or high-LET 56Fe-ions (148 keV/μm) and at 60 d post-exposure, mice were sacrificed and femur sections were obtained for histological, cellular and molecular analysis. Cell proliferation (PCNA), cell death (active caspase-3), senescence (p16), osteoclast (RANK), osteoblast (OPG), osteoblast progenitor (c-Kit), and osteoclastogenesis-associated secretory factors (like RANKL) were assessed using immunostaining. While no change in cell proliferation and apoptosis between control and irradiated groups was noted, the number of BM megakaryocytes was significantly reduced in irradiated mice at 60 d post-exposure. A remarkable increase in p16 positive cells indicated a persistent increase in cell senescence, whereas increased RANKL/OPG ratio, reductions in the number of osteoblast progenitor cells, and osteocalcin provided clear evidence that exposure to both proton and 56Fe-ions promotes pro-osteoclastogenic activity in BM. Among irradiated groups, 56Fe-induced alterations in the BM cellularity and osteoclastogenesis were significantly greater than the protons that demonstrated a radiation quality-dependent effect. This study has implications in understanding the role of IR-induced late changes in the BM cells and its involvement in bone degeneration among deep-space astronauts, and also in patients undergoing proton or heavy-ion radiotherapy.
Collapse
Affiliation(s)
- Kamendra Kumar
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Kamal Datta
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Albert J. Fornace
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Shubhankar Suman
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
- Corresponding author.
| |
Collapse
|
10
|
Padala SR, Kashyap B, Dekker H, Mikkonen JJW, Palander A, Bravenboer N, Kullaa AM. Irradiation affects the structural, cellular and molecular components of jawbones. Int J Radiat Biol 2021; 98:136-147. [PMID: 34855558 DOI: 10.1080/09553002.2022.2013568] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE Emerging evidence shows that changes in the bone and its microenvironment following radiotherapy are associated with either an inhibition or a state of low bone formation. Ionizing radiation is damaging to the jawbone as it increases the complication rate due to the development of hypovascular, hypocellular, and hypoxic tissue. This review summarizes and correlates the current knowledge on the effects of irradiation on the bone with an emphasis on jawbone, as these have been a less extensively studied area. CONCLUSIONS The stringent regulation of bone formation and bone resorption can be influenced by radiation, causing detrimental effects at structural, cellular, vascular, and molecular levels. It is also associated with a high risk of damage to surrounding healthy tissues and an increased risk of fracture. Technological advances and research on animal models as well as a few human bone tissue studies have provided novel insights into the ways in which bone can be affected by high, low and sublethal dose of radiation. The influence of radiation on bone metabolism, cellular properties, vascularity, collagen, and other factors like inflammation, reactive oxygen species are discussed.
Collapse
Affiliation(s)
- Sridhar Reddy Padala
- Institute of Dentistry, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Bina Kashyap
- Institute of Dentistry, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Hannah Dekker
- Amsterdam University Medical Centers, Academic Centre for Dentistry Amsterdam (ACTA), Department of Oral and Maxillofacial Surgery/Oral Pathology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Jopi J W Mikkonen
- Institute of Dentistry, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Anni Palander
- Institute of Dentistry, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Nathalie Bravenboer
- Amsterdam UMC, Department of Clinical Chemistry, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands.,Department of Internal Medicine, Division of Endocrinology and Center for Bone Quality, Leiden University Medical Center, Leiden, The Netherlands
| | - Arja M Kullaa
- Institute of Dentistry, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
11
|
Treatment of Radiation Bone Injury with Transplanted hUCB-MSCs via Wnt/ β-Catenin. Stem Cells Int 2021; 2021:5660927. [PMID: 34876908 PMCID: PMC8645406 DOI: 10.1155/2021/5660927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 11/16/2021] [Indexed: 11/19/2022] Open
Abstract
Radiation-induced bone injury (RIBI) is one of the complications after radiotherapy for malignant tumors. However, there are no effective measures for the treatment of RIBI in clinical practice, and the mechanism of RIBI is unclear. We use a single high-dose ionizing radiation (6Gy) to analyze the effect of radiotherapy on osteoblast function. Human umbilical cord blood mesenchymal stem cells (hUCB-MSCs) were cocultured with irradiated osteoblasts to examine their therapeutic effects and mechanisms on osteoblast injury. The hUCB-MSC transplantation mouse model is used to confirm the in vivo role of hUCB-MSC treatment in radiation bone injury. Western blot analysis, qRT-PCR, immunohistochemistry, and immunofluorescence staining were used to analyze gene expression and angiogenesis. The apoptosis and migration of osteoblasts were measured by Hoechst staining, scratch test, and transwell. The differentiation of osteoblasts was measured by ALP and Alizarin red staining and transmission electron microscopy. The bone-related parameters of mice were evaluated by micro-CT analysis. We found that radiation can damage the DNA of osteoblasts; induce apoptosis; reduce the differentiation, migration, and adhesion of osteoblasts, leading to lipogenesis of bone marrow mesenchymal stem cells (BMSCs) and reducing the source of osteoblasts; and increase the number of osteoclasts in bone tissue, while MSC treatment prevents these changes. Our results reveal the inhibitory effect of radiation on osteoblast function. hUCB-MSCs can be used as a therapeutic target for the development of new therapeutic strategies for radiotherapy of bone injury diseases.
Collapse
|
12
|
Kim HN, Richardson KK, Krager KJ, Ling W, Simmons P, Allen AR, Aykin-Burns N. Simulated Galactic Cosmic Rays Modify Mitochondrial Metabolism in Osteoclasts, Increase Osteoclastogenesis and Cause Trabecular Bone Loss in Mice. Int J Mol Sci 2021; 22:11711. [PMID: 34769141 PMCID: PMC8583929 DOI: 10.3390/ijms222111711] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 11/24/2022] Open
Abstract
Space is a high-stress environment. One major risk factor for the astronauts when they leave the Earth's magnetic field is exposure to ionizing radiation from galactic cosmic rays (GCR). Several adverse changes occur in mammalian anatomy and physiology in space, including bone loss. In this study, we assessed the effects of simplified GCR exposure on skeletal health in vivo. Three months following exposure to 0.5 Gy total body simulated GCR, blood, bone marrow and tissue were collected from 9 months old male mice. The key findings from our cell and tissue analysis are (1) GCR induced femoral trabecular bone loss in adult mice but had no effect on spinal trabecular bone. (2) GCR increased circulating osteoclast differentiation markers and osteoclast formation but did not alter new bone formation or osteoblast differentiation. (3) Steady-state levels of mitochondrial reactive oxygen species, mitochondrial and non-mitochondrial respiration were increased without any changes in mitochondrial mass in pre-osteoclasts after GCR exposure. (4) Alterations in substrate utilization following GCR exposure in pre-osteoclasts suggested a metabolic rewiring of mitochondria. Taken together, targeting radiation-mediated mitochondrial metabolic reprogramming of osteoclasts could be speculated as a viable therapeutic strategy for space travel induced bone loss.
Collapse
Affiliation(s)
- Ha-Neui Kim
- Center for Musculoskeletal Disease Research and Center for Osteoporosis and Metabolic Bone Diseases, Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Little Rock, AR 72205, USA; (K.K.R.); (W.L.)
| | - Kimberly K. Richardson
- Center for Musculoskeletal Disease Research and Center for Osteoporosis and Metabolic Bone Diseases, Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Little Rock, AR 72205, USA; (K.K.R.); (W.L.)
| | - Kimberly J. Krager
- Department of Pharmaceutical Sciences, Division of Radiation Health, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Little Rock, AR 72205, USA; (K.J.K.); (P.S.); (A.R.A.)
| | - Wen Ling
- Center for Musculoskeletal Disease Research and Center for Osteoporosis and Metabolic Bone Diseases, Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Little Rock, AR 72205, USA; (K.K.R.); (W.L.)
| | - Pilar Simmons
- Department of Pharmaceutical Sciences, Division of Radiation Health, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Little Rock, AR 72205, USA; (K.J.K.); (P.S.); (A.R.A.)
| | - Antino R. Allen
- Department of Pharmaceutical Sciences, Division of Radiation Health, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Little Rock, AR 72205, USA; (K.J.K.); (P.S.); (A.R.A.)
| | - Nukhet Aykin-Burns
- Department of Pharmaceutical Sciences, Division of Radiation Health, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Little Rock, AR 72205, USA; (K.J.K.); (P.S.); (A.R.A.)
| |
Collapse
|
13
|
Wang Y, Xu L, Wang J, Bai J, Zhai J, Zhu G. Radiation induces primary osteocyte senescence phenotype and affects osteoclastogenesis in vitro. Int J Mol Med 2021; 47:76. [PMID: 33693957 PMCID: PMC7949628 DOI: 10.3892/ijmm.2021.4909] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 02/10/2021] [Indexed: 02/06/2023] Open
Abstract
Irradiation-induced bone remodeling imbalances arise as a consequence of the dysregulation of bone formation and resorption. Due to the abundance of osteocytes, their long life and their dual-regulatory effects on both osteoblast and osteoclast function, they serve as critical coordinators of bone remolding. In the present study, femur and tibia-derived primary osteocytes were cultured and irradiated to observe the functional changes and the cellular senescence phenotype in vitro. Irradiation directly reduced cell viability, affected the crucial dendritic morphology and altered the expression of functional proteins, including upregulation of receptor activator of nuclear factor-κB ligand and sclerostin, and downregulation of osteoprotegerin. Irradiated osteocytes were shown to exhibit notable DNA damage, which resulted in the initiation of a typical cellular senescence phenotype. Furthermore, it was found that irradiation-induced prematurely senescent osteocytes stimulate molecular secretion, referred to as senescence-associated secretory phenotype (SASP), which may be involved in modulation of the bone microenvironment, including the promotion of osteoclastogenesis. Taken together, the results showed that irradiation triggered osteocyte senescence and the acquisition of an associated secretory phenotype. This further resulted in an imbalance of bone remodeling through senescent influence on proliferation, morphology and marker protein production, but also indirectly via a paracrine pathway through SASP secretion. The results of the present study may highlight the potential of SASP-targeted interventions for the management of radiation-induced bone loss.
Collapse
Affiliation(s)
- Yuyang Wang
- Department of Radiation Protection, Institute of Radiation Medicine, Fudan University, Shanghai 200032, P.R. China
| | - Linshan Xu
- Department of Radiation Protection, Institute of Radiation Medicine, Fudan University, Shanghai 200032, P.R. China
| | - Jianping Wang
- Department of Radiation Protection, Institute of Radiation Medicine, Fudan University, Shanghai 200032, P.R. China
| | - Jiangtao Bai
- Department of Radiation Protection, Institute of Radiation Medicine, Fudan University, Shanghai 200032, P.R. China
| | - Jianglong Zhai
- Department of Radiation Protection, Institute of Radiation Medicine, Fudan University, Shanghai 200032, P.R. China
| | - Guoying Zhu
- Department of Radiation Protection, Institute of Radiation Medicine, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
14
|
Little-Letsinger SE, Turner ND, Ford JR, Suva LJ, Bloomfield SA. Omega-3 fatty acid modulation of serum and osteocyte tumor necrosis factor-α in adult mice exposed to ionizing radiation. J Appl Physiol (1985) 2021; 130:627-639. [PMID: 33411639 DOI: 10.1152/japplphysiol.00848.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Chronic inflammation leads to bone loss and fragility. Proinflammatory cytokines such as tumor necrosis factor-alpha (TNF-α) consistently promote bone resorption. Dietary modulation of proinflammatory cytokines is an accepted therapeutic approach to treat chronic inflammation, including that induced by space-relevant radiation exposure. As such, these studies were designed to determine whether an anti-inflammatory diet, high in omega-3 fatty acids, could reduce radiation-mediated bone damage via reductions in the levels of inflammatory cytokines in osteocytes and serum. Lgr5-EGFP C57BL/6 mice were randomized to receive diets containing fish oil and pectin (FOP; high in omega-3 fatty acids) or corn oil and cellulose (COC; high in omega-6 fatty acids) and then acutely exposed to 0.5-Gy 56Fe or 2.0-Gy gamma-radiation. Mice fed the FOP diet exhibited consistent reductions in serum TNF-α in the 56Fe experiment but not the gamma-experiment. The percentage osteocytes (%Ot) positive for TNF-α increased in gamma-exposed COC, but not FOP, mice. Minimal changes in %Ot positive for sclerostin were observed. FOP mice exhibited modest improvements in several measures of cancellous microarchitecture and volumetric bone mineral density (BMD) postexposure to 56Fe and gamma-radiation. Reduced serum TNF-α in FOP mice exposed to 56Fe was associated with either neutral or modestly positive changes in bone structural integrity. Collectively, these data are generally consistent with previous findings that dietary intake of omega-3 fatty acids may effectively mitigate systemic inflammation after acute radiation exposure and facilitate maintenance of BMD during spaceflight in humans.NEW & NOTEWORTHY This is the first investigation, to our knowledge, to test the impact of a diet high in omega-3 fatty acids on multiple bone structural and biological outcomes following space-relevant radiation exposure. Novel in biological outcomes is the assessment of osteocyte responses to this stressor. These data also add to the growing evidence that low-dose exposures to even high-energy ion species like 56Fe may have neutral or even small positive impacts on bone.
Collapse
Affiliation(s)
| | - Nancy D Turner
- Nutrition and Food Sciences, Texas A&M University, College Station, Texas
| | - John R Ford
- Nuclear Engineering, Texas A&M University, College Station, Texas
| | - Larry J Suva
- Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Susan A Bloomfield
- Departments of Health and Kinesiology, Texas A&M University, College Station, Texas
| |
Collapse
|
15
|
Maier I, Liu J, Ruegger PM, Deutschmann J, Patsch JM, Helbich TH, Borneman J, Schiestl RH. Intestinal bacterial indicator phylotypes associate with impaired DNA double-stranded break sensors but augmented skeletal bone micro-structure. Carcinogenesis 2020; 41:483-489. [PMID: 31840161 DOI: 10.1093/carcin/bgz204] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 12/07/2019] [Accepted: 12/13/2019] [Indexed: 12/17/2022] Open
Abstract
Intestinal microbiota are considered a sensor for molecular pathways, which orchestrate energy balance, immune responses, and cell regeneration. We previously reported that microbiota restriction promoted higher levels of systemic radiation-induced genotoxicity, proliferative lymphocyte activation, and apoptotic polarization of metabolic pathways. Restricted intestinal microbiota (RM) that harbors increased abundance of Lactobacillus johnsonii (LBJ) has been investigated for bacterial communities that correlated radiation-induced genotoxicity. Indicator phylotypes were more abundant in RM mice and increased in prevalence after whole body irradiation in conventional microbiota (CM) mice, while none of the same ten most abundant phylotypes were different in abundance between CM mice before and after heavy ion irradiation. Muribaculum intestinale was detected highest in female small intestines in RM mice, which were lacking Ureaplasma felinum compared with males, and thus these bacteria could be contributing to the differential amounts of radiation-induced systemic genotoxicity between the CM and RM groups. Helicobacter rodentium and M.intestinale were found in colons in the radiation-resistant CM phenotype. While the expression of interferon-γ was elevated in the small intestine, and lower in blood in CM mice, high-linear energy transfer radiation reduced transforming growth factor-β with peripheral interleukin (IL)-17 in RM mice, particularly in females. We found that female RM mice showed improved micro-architectural bone structure and anti-inflammatory radiation response compared with CM mice at a delayed phase 6 weeks postexposure to particle radiation. However, microbiota restriction reduced inflammatory markers of tumor necrosis factor in marrow, when IL-17 was reduced by intraperitoneal injection of IL-17 neutralizing antibody.
Collapse
Affiliation(s)
- Irene Maier
- Department of Environmental Health Sciences, Fielding School of Public Health, University of California, Los Angeles 650 Charles E. Young Dr. South, Los Angeles, CA, USA
| | - Jared Liu
- Department of Environmental Health Sciences, Fielding School of Public Health, University of California, Los Angeles 650 Charles E. Young Dr. South, Los Angeles, CA, USA
| | - Paul M Ruegger
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA, USA
| | - Julia Deutschmann
- Department for Radiologic Technology, University of Applied Sciences Wiener Neustadt for Business and Engineering Ltd., Lower Austria, Austria
| | - Janina M Patsch
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Währinger Gürtel,Vienna, Austria
| | - Thomas H Helbich
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Währinger Gürtel,Vienna, Austria
| | - James Borneman
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA, USA
| | - Robert H Schiestl
- Department of Environmental Health Sciences, Fielding School of Public Health, University of California, Los Angeles 650 Charles E. Young Dr. South, Los Angeles, CA, USA.,Department of Pathology, University of California, Los Angeles, CA, USA
| |
Collapse
|
16
|
Li R, Yang W, Hu X, Zhou D, Huang K, Wang C, Li Y, Liu B. Effect of autophagy on irradiation‑induced damage in osteoblast‑like MC3T3‑E1 cells. Mol Med Rep 2020; 22:3473-3481. [PMID: 32945432 PMCID: PMC7453677 DOI: 10.3892/mmr.2020.11425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 07/16/2020] [Indexed: 12/05/2022] Open
Abstract
Autophagy is activated under radiation stress, which serves an important role in maintaining bone homeostasis. However, the underlying mechanisms of irradiation-induced autophagy in bone homeostasis is not well understood. The present study aimed to determine the effects of radiation-activated autophagy on pre-osteoblastic MC3T3-E1 cells. X-ray irradiation activated autophagy in a dose-dependent manner, with an increased fluorescence intensity of monodansylcadaverine staining, increased ratio of microtubule-associated protein 1 light chain 3β (LC3)-II/LC3-I, decreased p62 expression, and increased ATG5 and beclin-1 expression levels in MC3T3-E1 cells 72 h after irradiation compared with those in non-irradiated MC3T3-E1 cells. Irradiation reduced colony formation and mineralization in a dose-dependent manner in MC3T3-E1 cells at 2 and 3 weeks after irradiation, respectively. Decreased levels of alkaline phosphatase activity and runt-related transcription factor 2 expression were observed at 72 h post-irradiation. In addition, irradiation-induced apoptosis was accompanied by a decreased ratio of Bcl-2/BAX protein and increased the activity of caspase-3. By contrast, doxycycline (DOX)-inhibited autophagy attenuated the decreased colony formation and mineralization, and aggravated the increased cell apoptosis in irradiated MC3T3-E1 cells. Furthermore, the ratio of phosphorylated P38/P38 was observed to be higher following DOX treatment within 1 week of irradiation, which was reversed 2 weeks post-irradiation. In conclusion, DOX-inhibited autophagy aggravated X-ray irradiation-induced apoptosis at an early stage, but maintained cell proliferation and mineralization at a late stage in irradiated MC3T3-E1 cells.
Collapse
Affiliation(s)
- Rui Li
- School of Civil Engineering and Mechanics, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Wenke Yang
- School of Basic Medical Science, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Xurui Hu
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Dan Zhou
- Department of Cardiology, Gansu Provincial Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Ke Huang
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Chenwei Wang
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Yi Li
- School of Civil Engineering and Mechanics, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Bin Liu
- School/Hospital of Stomatology, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
17
|
The Influence of Radiation on Bone and Bone Cells-Differential Effects on Osteoclasts and Osteoblasts. Int J Mol Sci 2020; 21:ijms21176377. [PMID: 32887421 PMCID: PMC7504528 DOI: 10.3390/ijms21176377] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 02/06/2023] Open
Abstract
The bone is a complex organ that is dependent on a tight regulation between bone formation by osteoblasts (OBs) and bone resorption by osteoclasts (OCs). These processes can be influenced by environmental factors such as ionizing radiation (IR). In cancer therapy, IR is applied in high doses, leading to detrimental effects on bone, whereas radiation therapy with low doses of IR is applied for chronic degenerative and inflammatory diseases, with a positive impact especially on bone homeostasis. Moreover, the effects of IR are of particular interest in space travel, as astronauts suffer from bone loss due to space radiation and microgravity. This review summarizes the current state of knowledge on the effects of IR on bone with a special focus on the influence on OCs and OBs, as these cells are essential in bone remodeling. In addition, the influence of IR on the bone microenvironment is discussed. In summary, the effects of IR on bone and bone remodeling cells strongly depend on the applied radiation dose, as differential results are provided from in vivo as well as in vitro studies with varying doses of IR. Furthermore, the isolated effects of IR on a single cell type are difficult to determine, as the bone cells and bone microenvironment are building a tightly regulated network, influencing on one another. Therefore, future research is necessary in order to elucidate the influence of different bone cells on the overall radiation-induced effects on bone.
Collapse
|
18
|
Schreurs AS, Torres S, Truong T, Moyer EL, Kumar A, Tahimic CGT, Alwood JS, Globus RK. Skeletal tissue regulation by catalase overexpression in mitochondria. Am J Physiol Cell Physiol 2020; 319:C734-C745. [PMID: 32783660 DOI: 10.1152/ajpcell.00068.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Accumulation of oxidative damage from excess reactive oxygen species (ROS) may contribute to skeletal aging and mediate adverse responses to physiological challenges. Wild-type (WT) mice and transgenic mice (male, 16 wk of age) with human catalase targeted to the mitochondria (mCAT) were analyzed for skeletal responses to the remodeling stimuli of combined hind-limb unloading and exposure to ionizing radiation (137Cs, 2 Gy). Treatment for 2 wk caused lipid peroxidation in the bones WT but not mCAT mice, showing that transgene expression mitigated oxidative stress. Ex vivo osteoblast colony growth rate was 95% greater in mCAT than WT mice and correlated with catalase activity levels (P < 0.005, r = 0.67), although terminal osteoblast and osteoclast differentiation were unaffected. mCAT mice had lower cancellous bone volume and cortical size than WT mice. Ambulatory control mCAT animals also displayed reduced cancellous and cortical structural properties compared with control WT mice. In mCAT but not WT mice, treatment caused an unexpectedly rapid radial expansion (+8% cortical area, +22% moment of inertia), reminiscent of compensatory bone growth during advancing age. In contrast, treatment caused similar structural deficits in cancellous tissue of mCAT and WT mice. In sum, mitochondrial ROS signaling via H2O2 was important for the acquisition of adult bone structure and catalase overexpression failed to protect cancellous tissue from treatment. In contrast, catabolic stimuli caused radial expansion in mCAT not WT mice, suggesting that mitochondrial ROS in skeletal cells act to suppress tissue turnover in response to remodeling challenges.
Collapse
Affiliation(s)
- Ann-Sofie Schreurs
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, California.,Universities Space Research Association, Moffett Field, California
| | - Samantha Torres
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, California.,Blue Marble Space Institute of Science, Seattle, Washington
| | - Tiffany Truong
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, California.,Blue Marble Space Institute of Science, Seattle, Washington
| | - Eric L Moyer
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, California.,Blue Marble Space Institute of Science, Seattle, Washington
| | - Akhhilesh Kumar
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, California.,Universities Space Research Association, Moffett Field, California
| | - Candice G T Tahimic
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, California.,KBR, Moffett Field, California
| | - Joshua S Alwood
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, California
| | - Ruth K Globus
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, California
| |
Collapse
|
19
|
Tong L, Wang Y, Wang J, He F, Zhai J, Bai J, Zhu G. Radiation alters osteoclastogenesis by regulating the cytoskeleton and lytic enzymes in RAW 264.7 cells and mouse bone marrow-derived macrophages. Int J Radiat Biol 2020; 96:1296-1308. [PMID: 32687425 DOI: 10.1080/09553002.2020.1798542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE The aim of the present study was to investigate the duality of irradiation effect on osteoclastogenesis, particularly on the cytoskeleton and expression of lytic enzymes in osteoclast precursors. Therefore, the present study may serve as a useful reference for the prevention and treatment of radiation-induced bone loss in the clinic. MATERIALS AND METHODS Two typical osteoclast precursors, murine RAW 264.7 macrophage cells and mouse bone marrow-derived macrophages (BMMs), were exposed to radiation in the order of 0.25-8 Gy, and the effects on cell viability, TRAP activity and bone resorption were subsequently investigated. Furthermore, changes in the cytoskeleton, cell apoptosis, and expression of lytic enzymes in osteoclasts were examined to elucidate the molecular mechanism of the duality of irradiation on osteoclastogenesis. RESULTS Morphological changes and impaired viability were observed in RAW 264.7 cells and BMMs treated with 1-8 Gy irradiation with or without RANKL. However, the cell fusion tendency of osteoclasts was enhanced after 2 Gy irradiation, and an increased number of fused giant osteoclasts and enhanced F-actin ring formation were observed. Consistently, the bone resorption activity and the enzyme expression of TRAP, cathepsin K, matrix metalloproteinase 9, activator protein 1, and Caspase 9 were increased following irradiation with 2 Gy. Furthermore, intracellular ROS production and apoptosis of osteoclast precursors were increased. CONCLUSIONS Irradiation with 2 Gy inhibited the viability of osteoclast precursors, but increased osteoclastogenesis by enhancing cell fusion and increasing the secretion of lytic enzymes, which may be an important mechanism of radiation-induced bone loss.
Collapse
Affiliation(s)
- Ling Tong
- Institute of Radiation Medicine, Fudan University, Shanghai, PR China.,Shanghai Municipal Center for Disease Control & Prevention, Shanghai, PR China
| | - Yuyang Wang
- Institute of Radiation Medicine, Fudan University, Shanghai, PR China
| | - Jianping Wang
- Institute of Radiation Medicine, Fudan University, Shanghai, PR China
| | - Feilong He
- Institute of Radiation Medicine, Fudan University, Shanghai, PR China.,Shanghai Municipal Center for Disease Control & Prevention, Shanghai, PR China
| | - Jianglong Zhai
- Institute of Radiation Medicine, Fudan University, Shanghai, PR China
| | - Jiangtao Bai
- Institute of Radiation Medicine, Fudan University, Shanghai, PR China
| | - Guoying Zhu
- Institute of Radiation Medicine, Fudan University, Shanghai, PR China
| |
Collapse
|
20
|
Steczina S, Tahimic CGT, Pendleton M, M'Saad O, Lowe M, Alwood JS, Halloran BP, Globus RK, Schreurs AS. Dietary countermeasure mitigates simulated spaceflight-induced osteopenia in mice. Sci Rep 2020; 10:6484. [PMID: 32300161 PMCID: PMC7162976 DOI: 10.1038/s41598-020-63404-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/26/2020] [Indexed: 02/06/2023] Open
Abstract
Spaceflight is a unique environment that includes at least two factors which can negatively impact skeletal health: microgravity and ionizing radiation. We have previously shown that a diet supplemented with dried plum powder (DP) prevented radiation-induced bone loss in mice. In this study, we investigated the capacity of the DP diet to prevent bone loss in mice following exposure to simulated spaceflight, combining microgravity (by hindlimb unloading) and radiation exposure. The DP diet was effective at preventing most decrements in bone micro-architectural and mechanical properties due to hindlimb unloading alone and simulated spaceflight. Furthermore, we show that the DP diet can protect osteoprogenitors from impairments resulting from simulated microgravity. Based on our findings, a dietary supplementation with DP could be an effective countermeasure against the skeletal deficits observed in astronauts during spaceflight.
Collapse
Affiliation(s)
- Sonette Steczina
- Blue Marble Space Institute of Science, Seattle, WA, 98154, USA.,Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA
| | - Candice G T Tahimic
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA.,KBR, Moffett Field, California, USA
| | - Megan Pendleton
- Department of Mechanical Engineering, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Ons M'Saad
- Space Life Sciences Training Program, NASA Ames Research Center, Moffett Field, CA, 94035, USA
| | - Moniece Lowe
- Blue Marble Space Institute of Science, Seattle, WA, 98154, USA.,Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA
| | - Joshua S Alwood
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA
| | - Bernard P Halloran
- Department of Medicine, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Ruth K Globus
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA
| | - Ann-Sofie Schreurs
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA. .,Universities Space Research Association, Moffett Field, CA, USA.
| |
Collapse
|
21
|
Xiang LX, Ran Q, Chen L, Xiang Y, Li FJ, Zhang XM, Xiao YN, Zou LY, Zhong JF, Li SC, Li ZJ. CR6-interacting factor-1 contributes to osteoclastogenesis by inducing receptor activator of nuclear factor κB ligand after radiation. World J Stem Cells 2020; 12:222-240. [PMID: 32266053 PMCID: PMC7118287 DOI: 10.4252/wjsc.v12.i3.222] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 02/29/2020] [Accepted: 03/15/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Radiation induces rapid bone loss and enhances bone resorption and adipogenesis, leading to an increased risk of bone fracture. There is still a lack of effective preventive or therapeutic method for irradiation-induced bone injury. Receptor activator of nuclear factor κB ligand (RANKL) provides the crucial signal to induce osteoclast differentiation and plays an important role in bone resorption. However, the mechanisms of radiation-induced osteoporosis are not fully understood.
AIM To investigate the role of CR6-interacting factor-1 (Crif1) in osteoclastogenesis after radiation and its possible mechanism.
METHODS C57BL/6 mice were exposed to Co-60 gamma rays and received 5 Gy of whole-body sublethal irradiation at a rate of 0.69 Gy/min. For in vitro study, mouse bone marrow mesenchymal stem/stromal cells (BM-MSCs) were irradiated with Co-60 at a single dose of 9 Gy. For osteoclast induction, monocyte-macrophage RAW264.7 cells were cocultured with mouse BM-MSCs for 7 d. ClusPro and InterProSurf were used to investigate the interaction interface in Crif1 and protein kinase cyclic adenosine monophosphate (cAMP)-activited catalytic subunit alpha complex. Virtual screening using 462608 compounds from the Life Chemicals database around His120 of Crif1 was carried out using the program Autodock_vina. A tetrazolium salt (WST-8) assay was carried out to study the toxicity of compounds to different cells, including human BM-MSCs, mouse BM-MSCs, and Vero cells.
RESULTS Crif1 expression increased in bone marrow cells after radiation in mice. Overexpression of Crif1 in mouse BM-MSCs and radiation exposure could increase RANKL secretion and promote osteoclastogenesis in vitro. Deletion of Crif1 in BM-MSCs could reduce both adipogenesis and RANKL expression, resulting in the inhibition of osteoclastogenesis. Deletion of Crif1 in RAW264.7 cells did not affect the receptor activator of nuclear factor κB expression or osteoclast differentiation. Following treatment with protein kinase A (PKA) agonist (forskolin) and inhibitor (H-89) in mouse BM-MSCs, Crif1 induced RANKL secretion via the cAMP/PKA pathway. Moreover, we identified the Crif1-protein kinase cyclic adenosine monophosphate-activited catalytic subunit alpha interaction interface by in silico studies and shortlisted interface inhibitors through virtual screening on Crif1. Five compounds dramatically suppressed RANKL secretion and adipogenesis by inhibiting the cAMP/PKA pathway.
CONCLUSION Crif1 promotes RANKL expression via the cAMP/PKA pathway, which induces osteoclastogenesis by binding to receptor activator of nuclear factor κB on monocytes-macrophages in the mouse model. These results suggest a role for Crif1 in modulating osteoclastogenesis and provide insights into potential therapeutic strategies targeting the balance between osteogenesis and adipogenesis for radiation-induced bone injury.
Collapse
Affiliation(s)
- Li-Xin Xiang
- Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Qian Ran
- Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, 400037, China
- Department of Otolaryngology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States
| | - Li Chen
- Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Yang Xiang
- Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, 400037, China
- Department of Otolaryngology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States
| | - Feng-Jie Li
- Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Xiao-Mei Zhang
- Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Yan-Ni Xiao
- Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Ling-Yun Zou
- Bioinformatics Center, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| | - Jiang F Zhong
- Department of Otolaryngology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States
| | - Shengwen Calvin Li
- CHOC Children’s Research Institute, Children’s Hospital of Orange County, University of California, Irvine, CA 92868, United States
| | - Zhong-Jun Li
- Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, 400037, China
| |
Collapse
|
22
|
Sullivan LK, Livingston EW, Lau AG, Rao-Dayton S, Bateman TA. A Mouse Model for Skeletal Structure and Function Changes Caused by Radiation Therapy and Estrogen Deficiency. Calcif Tissue Int 2020; 106:180-193. [PMID: 31583426 DOI: 10.1007/s00223-019-00617-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 09/18/2019] [Indexed: 12/23/2022]
Abstract
Radiation therapy and estrogen deficiency can damage healthy bone and lead to an increased fracture risk. The goal of this study is to develop a mouse model for radiation therapy using a fractionated biologically equivalent dose for cervical cancer treatment in both pre- and postmenopausal women. Thirty-two female C57BL/6 mice 13 weeks of age were divided into four groups: Sham + non-irradiated (SHAM + NR), Sham + irradiated (SHAM + IRR), ovariectomy + non-irradiated (OVX + NR) and ovariectomy + irradiated (OVX + IRR). The irradiated mice received a 6 Gy dose of X-rays to the hindlimbs at Day 2, Day 4 and Day 7 (18 Gy total). Tissues were collected at Day 35. DEXA, microCT analysis and FEA were used to quantify structural and functional changes at the proximal tibia, midshaft femur, proximal femur and L1 vertebra. There was a significant (p < 0.05) decline in proximal tibia trabecular BV/TV from (1) IRR compared to NR mice within Sham (- 46%) and OVX (- 41%); (2) OVX versus Sham within NR mice (- 36%) and IRR mice (- 30%). With homogenous material properties applied to the proximal tibia mesh using FEA, there was (1) an increase in whole bone (trabecular + cortical) structural stiffness from IRR compared to NR mice within Sham (+ 10%) and OVX (+ 15%); (2) a decrease in stiffness from OVX versus Sham within NR mice (- 18%) and IRR mice (- 14%). Fractionated irradiation and ovariectomy both had a negative effect on skeletal microarchitecture. Ovariectomy had a systemic effect, while skeletal radiation damage was largely specific to trabecular bone within the X-ray field.
Collapse
Affiliation(s)
- Lindsay K Sullivan
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, USA.
| | - Eric W Livingston
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, USA
| | - Anthony G Lau
- Department of Biomedical Engineering, The College of New Jersey, Ewing, USA
| | - Sheila Rao-Dayton
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, USA
| | - Ted A Bateman
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, USA
- Department of Radiation Oncology, University of North Carolina, Chapel Hill, USA
| |
Collapse
|
23
|
Zhai J, He F, Wang J, Chen J, Tong L, Zhu G. Influence of radiation exposure pattern on the bone injury and osteoclastogenesis in a rat model. Int J Mol Med 2019; 44:2265-2275. [PMID: 31638191 PMCID: PMC6844641 DOI: 10.3892/ijmm.2019.4369] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 09/20/2019] [Indexed: 12/24/2022] Open
Abstract
Radiotherapy, one of the clinical treatments of cancer, is accompanied by a high risk of damage to healthy tissues, such as bone loss and increased risk of fractures. The aim of the present study was to establish a rat model of local and systemic bone injury by focal irradiation, in order to study the etiological mechanism and intervention. The proximal metaphyseal region of the left hindlimb of male Sprague-Dawley rats were exposed to a single 2 Gy or three 8 Gy doses delivered on days 1, 3 and 5 using a small animal irradiator, the changes in bone volume and microarchitecture were evaluated, and the mineral apposition rate (MAR) was assessed. Furthermore, bone marrow-derived macrophages (BMMs) were isolated and induced to osteoclasts. It has been demonstrated that a single dose of 2 Gy may result in a significant loss of lumbar bone density at 3 days post-irradiation, however this is restored at 30 days post-irradiation. In the 3x8 Gy irradiation rat model, there was a rapid decrease in the aBMD of lumbar spine at 3 days and at 7 days post-irradiation, and the aBMD decline persisted even at 60 days post-irradiation. In addition, microCT analysis revealed a persistent decline in bone volume and damage in microarchitecture in the 3x8 Gy irradiation model, accompanied by a decrease in MAR, index of the decline in bone-forming ability. In the cellular mechanism, a single 2 Gy local irradiation mainly manifested as an enhancement of the BMMs osteoclastogenesis potential, which was different from the osteoclastogenesis inhibition after high-dose focal irradiation (3x8 Gy). In summary, the irradiation with simulated clinical focal fractionated radiotherapy exerts short- and long-term systemic injury on bone tissue, characterized by different osteoclastogenesis potential between the high dose mode and a single 2 Gy focal irradiation. Physicians must consider the irreversibility of bone damage in clinical radiotherapy.
Collapse
Affiliation(s)
- Jianglong Zhai
- Department of Radiation Protection, Institute of Radiation Medicine, Fudan University, Shanghai 200032, P.R. China
| | - Feilong He
- Department of Radiation Protection, Institute of Radiation Medicine, Fudan University, Shanghai 200032, P.R. China
| | - Jianping Wang
- Department of Radiation Protection, Institute of Radiation Medicine, Fudan University, Shanghai 200032, P.R. China
| | - Junxiang Chen
- Department of Radiation Protection, Institute of Radiation Medicine, Fudan University, Shanghai 200032, P.R. China
| | - Ling Tong
- Department of Radiation Protection, Institute of Radiation Medicine, Fudan University, Shanghai 200032, P.R. China
| | - Guoying Zhu
- Department of Radiation Protection, Institute of Radiation Medicine, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
24
|
Bokhari RS, Metzger CE, Black JM, Franklin KA, Boudreaux RD, Allen MR, Macias BR, Hogan HA, Braby LA, Bloomfield SA. Positive impact of low-dose, high-energy radiation on bone in partial- and/or full-weightbearing mice. NPJ Microgravity 2019; 5:13. [PMID: 31231675 PMCID: PMC6547738 DOI: 10.1038/s41526-019-0074-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 03/28/2019] [Indexed: 02/07/2023] Open
Abstract
Astronauts traveling beyond low Earth orbit will be exposed to galactic cosmic radiation (GCR); understanding how high energy ionizing radiation modifies the bone response to mechanical unloading is important to assuring crew health. To investigate this, we exposed 4-mo-old female Balb/cBYJ mice to an acute space-relevant dose of 0.5 Gy 56Fe or sham (n = ~8/group); 4 days later, half of the mice were also subjected to a ground-based analog for 1/6 g (partial weightbearing) (G/6) for 21 days. Microcomputed tomography (µ-CT) of the distal femur reveals that 56Fe exposure resulted in 65–78% greater volume and improved microarchitecture of cancellous bone after 21 d compared to sham controls. Radiation also leads to significant increases in three measures of energy absorption at the mid-shaft femur and an increase in stiffness of the L4 vertebra. No significant effects of radiation on bone formation indices are detected; however, G/6 leads to reduced % mineralizing surface on the inner mid-tibial bone surface. In separate groups allowed 21 days of weightbearing recovery from G/6 and/or 56Fe exposure, radiation-exposed mice still exhibit greater bone mass and improved microarchitecture vs. sham control. However, femoral bone energy absorption values are no longer higher in the 56Fe-exposed WB mice vs. sham controls. We provide evidence for persistent positive impacts of high-LET radiation exposure preceding a period of full or partial weightbearing on bone mass and microarchitecture in the distal femur and, for full weightbearing mice only and more transiently, cortical bone energy absorption values.
Collapse
Affiliation(s)
- Rihana S Bokhari
- 1Health and Kinesiology, Texas A&M University, College Station, TX USA
| | - Corinne E Metzger
- 1Health and Kinesiology, Texas A&M University, College Station, TX USA
| | - Jeremy M Black
- 2Mechanical Engineering, Texas A&M University, College Station, TX USA
| | | | - Ramon D Boudreaux
- 3Biomedical Engineering, Texas A&M University, College Station, TX USA
| | - Matthew R Allen
- 4Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, IN USA
| | - Brandon R Macias
- 1Health and Kinesiology, Texas A&M University, College Station, TX USA.,5KBRwyle, Cardiovascular and Vision Laboratory, NASA Johnson Space Center, Houston, TX USA
| | - Harry A Hogan
- 2Mechanical Engineering, Texas A&M University, College Station, TX USA.,3Biomedical Engineering, Texas A&M University, College Station, TX USA
| | - Leslie A Braby
- 6Nuclear Engineering, Texas A&M University, College Station, TX USA
| | | |
Collapse
|
25
|
Deyhle RT, Wong CP, Martin SA, McDougall MQ, Olson DA, Branscum AJ, Menn SA, Iwaniec UT, Hamby DM, Turner RT. Maintenance of Near Normal Bone Mass and Architecture in Lethally Irradiated Female Mice following Adoptive Transfer with as few as 750 Purified Hematopoietic Stem Cells. Radiat Res 2019; 191:413-427. [PMID: 30870097 DOI: 10.1667/rr15164.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Total-body irradiation (TBI) followed by transfer of bone marrow cells from donors is routinely performed in immunology research and can be used to manipulate differentiation and/or function of bone cells. However, exposure to high-dose radiation can result in irreversible osteopenia, and transfer of heterogeneous cell populations can complicate interpretation of results. The goal of this research was to establish an approach for reconstituting bone marrow using small numbers of purified donor-derived hematopoietic stem cells (HSCs) without negatively affecting bone metabolism. Gamma-irradiated (9 Gy) WBB6F1 mice were engrafted with bone marrow cells (5 × 106 cells) or purified HSCs (3,000 cells) obtained from GFP transgenic mice. In vivo analysis and in vitro differentiation assays performed two months later established that both methods were effective in reconstituting the hematopoietic compartment with donor-derived cells. We confirmed these findings by engrafting C57Bl/6 (B6) mice with bone marrow cells or purified HSCs from CD45.1 B6 congenic mice. We next performed adoptive transfer of purified HSCs (750 cells) into WBB6F1 and radiosensitive KitW/W-v mice and evaluated the skeleton two months later. Minimal differences were observed between controls and WBB6F1-engrafted mice that received fractionated doses of 2 × 5 Gy. Kitw/wv mice lost weight and became osteopenic after 2 × 5 Gy irradiations but these abnormalities were negligible after 5 Gy irradiation. Importantly, adoptive transfer of wild-type cells into Kitw/wv mice restored normal Kit expression in bone marrow. Together, these findings provide strong evidence for efficient engraftment with purified HSCs after lethal TBI with minimal collateral damage to bone. This approach will be useful for investigating mechanisms by which hematopoietic lineage cells regulate bone metabolism.
Collapse
Affiliation(s)
- Richard T Deyhle
- a Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331.,c Nuclear Science and Engineering, Oregon State University, Corvallis, Oregon 97331.,f Belgian Nuclear Research Centre (SCK•CEN), Boeretang 200, BE-2400 Mol, Belgium
| | - Carmen P Wong
- a Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331
| | - Stephen A Martin
- a Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331
| | - Melissa Q McDougall
- a Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331
| | - Dawn A Olson
- a Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331
| | - Adam J Branscum
- b Biostatistics Program, School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331
| | - Scott A Menn
- d Radiation Center, Oregon State University, Corvallis, Oregon 97331
| | - Urszula T Iwaniec
- a Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331.,e Center for Healthy Aging Research, Oregon State University, Corvallis, Oregon 97331
| | - David M Hamby
- c Nuclear Science and Engineering, Oregon State University, Corvallis, Oregon 97331
| | - Russell T Turner
- a Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon 97331.,e Center for Healthy Aging Research, Oregon State University, Corvallis, Oregon 97331
| |
Collapse
|
26
|
Zhang J, Zheng L, Wang Z, Pei H, Hu W, Nie J, Shang P, Li B, Hei TK, Zhou G. Lowering iron level protects against bone loss in focally irradiated and contralateral femurs through distinct mechanisms. Bone 2019; 120:50-60. [PMID: 30304704 DOI: 10.1016/j.bone.2018.10.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 09/21/2018] [Accepted: 10/06/2018] [Indexed: 12/20/2022]
Abstract
Radiation therapy leads to increased risk of late-onset fragility and bone fracture due to the loss of bone mass. On the other hand, iron overloading causes osteoporosis by enhancing bone resorption. It has been shown that total body irradiation increases iron level, but whether the systemic bone loss is related to the changes in iron level and hepcidin regulation following bone irradiation remains unknown. To investigate the potential link between them, we first created an animal model of radiation-induced systemic bone loss by targeting the mid-shaft femur with a single 2 Gy dose of X-rays. We found that mid-shaft femur focal irradiation led to structural deterioration in the distal region of the trabecular bone with increased osteoclasts surface and expressions of bone resorption markers in both irradiated and contralateral femurs relative to non-irradiated controls. Following irradiation, reduced hepcidin activity of the liver contributed to elevated iron levels in the serum and liver. By injecting hepcidin or deferoxamine (an iron chelator) to reduce iron level, deterioration of trabecular bone microarchitecture in irradiated mice was abrogated. The ability of iron chelation to inhibit radiation-induced osteoclast differentiation was observed in vitro as well. We further showed that ionizing radiation (IR) directly stimulated osteoclast differentiation and bone resorption in bone marrow cells isolated not from contralateral femurs but from directly irradiated femurs. These results suggest that increased iron levels after focal radiation is at least one of the main reasons for systemic bone loss. Furthermore, bone loss in directly irradiated bones is not only due to the elevated iron level, but also from increased osteoclast differentiation. In contrast, the bone loss in the contralateral femurs is mainly due to the elevated iron level induced by IR alone. These novel findings provide proof-of-principle evidence for the use of iron chelation or hepcidin as therapeutic treatments for IR-induced osteoporosis.
Collapse
Affiliation(s)
- Jian Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, China
| | - Lijun Zheng
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, China
| | - Ziyang Wang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, China
| | - Hailong Pei
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, China
| | - Wentao Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, China
| | - Jing Nie
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, China
| | - Peng Shang
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China; Research & Development Institute in Shenzhen, Northwestern Polytechnical University, Shenzhen, China
| | - Bingyan Li
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China; Department of Nutrition and Food Hygiene, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Tom K Hei
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, China; Center for Radiological Research, College of Physician and Surgeons, Columbia University, New York, USA.
| | - Guangming Zhou
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, China.
| |
Collapse
|
27
|
Oxidative Stress as Cause, Consequence, or Biomarker of Altered Female Reproduction and Development in the Space Environment. Int J Mol Sci 2018; 19:ijms19123729. [PMID: 30477143 PMCID: PMC6320872 DOI: 10.3390/ijms19123729] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/12/2018] [Accepted: 11/20/2018] [Indexed: 12/22/2022] Open
Abstract
Oxidative stress has been implicated in the pathophysiology of numerous terrestrial disease processes and associated with morbidity following spaceflight. Furthermore, oxidative stress has long been considered a causative agent in adverse reproductive outcomes. The purpose of this review is to summarize the pathogenesis of oxidative stress caused by cosmic radiation and microgravity, review the relationship between oxidative stress and reproductive outcomes in females, and explore what role spaceflight-induced oxidative damage may have on female reproductive and developmental outcomes.
Collapse
|
28
|
Zhang J, Qiu X, Xi K, Hu W, Pei H, Nie J, Wang Z, Ding J, Shang P, Li B, Zhou G. Therapeutic ionizing radiation induced bone loss: a review of in vivo and in vitro findings. Connect Tissue Res 2018; 59:509-522. [PMID: 29448860 DOI: 10.1080/03008207.2018.1439482] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Radiation therapy is one of the routine treatment modalities for cancer patients. Ionizing radiation (IR) can induce bone loss, and consequently increases the risk of fractures with delayed and nonunion of the bone in the cancer patients who receive radiotherapy. The orchestrated bone remodeling can be disrupted due to the affected behaviors of bone cells, including bone mesenchymal stem cells (BMSCs), osteoblasts and osteoclasts. BMSCs and osteoblasts are relatively radioresistant compared with osteoclasts and its progenitors. Owing to different radiosensitivities of bone cells, unbalanced bone remodeling caused by IR is closely associated with the dose absorbed. For doses less than 2 Gy, osteoclastogenesis and adipogenesis by BMSCs are enhanced, while there are limited effects on osteoblasts. High doses (>10 Gy) induce disrupted architecture of bone, which is usually related to decreased osteogenic potential. In this review, studies elucidating the biological effects of IR on bone cells (BMSCs, osteoblasts and osteoclasts) are summarized. Several potential preventions and therapies are also proposed.
Collapse
Affiliation(s)
- Jian Zhang
- a State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection , Soochow University , Suzhou , China.,b Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Suzhou , China
| | - Xinyu Qiu
- a State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection , Soochow University , Suzhou , China.,b Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Suzhou , China
| | - Kedi Xi
- a State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection , Soochow University , Suzhou , China.,b Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Suzhou , China
| | - Wentao Hu
- a State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection , Soochow University , Suzhou , China.,b Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Suzhou , China
| | - Hailong Pei
- a State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection , Soochow University , Suzhou , China.,b Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Suzhou , China
| | - Jing Nie
- a State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection , Soochow University , Suzhou , China.,b Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Suzhou , China
| | - Ziyang Wang
- a State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection , Soochow University , Suzhou , China.,b Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Suzhou , China
| | - Jiahan Ding
- a State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection , Soochow University , Suzhou , China.,b Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Suzhou , China
| | - Peng Shang
- a State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection , Soochow University , Suzhou , China.,b Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Suzhou , China.,c Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences , Northwestern Polytechnical University , Xi'an , China.,d Research & Development Institute in Shenzhen , Northwestern Polytechnical University, Fictitious College Garden , Shenzhen , China
| | - Bingyan Li
- a State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection , Soochow University , Suzhou , China
| | - Guangming Zhou
- a State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection , Soochow University , Suzhou , China.,b Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions , Suzhou , China
| |
Collapse
|
29
|
The Role of the Nuclear Factor κB Pathway in the Cellular Response to Low and High Linear Energy Transfer Radiation. Int J Mol Sci 2018; 19:ijms19082220. [PMID: 30061500 PMCID: PMC6121395 DOI: 10.3390/ijms19082220] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 07/24/2018] [Accepted: 07/24/2018] [Indexed: 12/19/2022] Open
Abstract
Astronauts are exposed to considerable doses of space radiation during long-term space missions. As complete shielding of the highly energetic particles is impracticable, the cellular response to space-relevant radiation qualities has to be understood in order to develop countermeasures and to reduce radiation risk uncertainties. The transcription factor Nuclear Factor κB (NF-κB) plays a fundamental role in the immune response and in the pathogenesis of many diseases. We have previously shown that heavy ions with a linear energy transfer (LET) of 100–300 keV/µm have a nine times higher potential to activate NF-κB compared to low-LET X-rays. Here, chemical inhibitor studies using human embryonic kidney cells (HEK) showed that the DNA damage sensor Ataxia telangiectasia mutated (ATM) and the proteasome were essential for NF-κB activation in response to X-rays and heavy ions. NF-κB’s role in cellular radiation response was determined by stable knock-down of the NF-κB subunit RelA. Transfection of a RelA short-hairpin RNA plasmid resulted in higher sensitivity towards X-rays, but not towards heavy ions. Reverse Transcriptase real-time quantitative PCR (RT-qPCR) showed that after exposure to X-rays and heavy ions, NF-κB predominantly upregulates genes involved in intercellular communication processes. This process is strictly NF-κB dependent as the response is completely absent in RelA knock-down cells. NF-κB’s role in the cellular radiation response depends on the radiation quality.
Collapse
|
30
|
Huang B, Zhang Q, Yuan Y, Xin N, He K, Huang Y, Tang H, Gong P. Sema3a inhibits the differentiation of Raw264.7 cells to osteoclasts under 2Gy radiation by reducing inflammation. PLoS One 2018; 13:e0200000. [PMID: 29975739 PMCID: PMC6033434 DOI: 10.1371/journal.pone.0200000] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Accepted: 06/18/2018] [Indexed: 02/05/2023] Open
Abstract
Astronauts and cancer patients receive different types of radiation, and radiation decreases bone strength and leads to radiation-induced osteoporosis. This effect is attributed to the activation of osteoclasts. Our aim was to study the effect of Sema3a on the differentiation of the murine macrophage cell line Raw264.7 into osteoclasts upon irradiation. Raw264.7 cells were divided into four groups: A, receiving no radiation; B, receiving no radiation + 50ngng/ml Sema3a; C, receiving 2Gy radiation; and D, receiving 2Gy radiation +50ngng/ml Sema3a. After treatment, cells were subjected to a proliferation assay, migration assay, live and apoptosis assay, and an ROS assay, along with analyses of bone resorption activity, TRAP staining and RT-PCR to assess the effect of Sema3a on Raw264.7 cells under 2Gy radiation. Sema3a inhibited the proliferation of Raw264.7 cells and showed statistical significance at a concentration of 100ngng/ml (P<0.05). Under 2Gy radiation, cell migration was reduced (P<0.05). In addition, 2Gy radiation resulted in more apoptotic cells, a higher level of ROS, larger bone resorption lacunae and more Trap-positive cells (p<0.05), and radiation increased CSTK, NFAT, TRAP-5b, Rankl/OPG, IL-1, IL-6, TNFa and P53 gene expression (P<0.05). Sema3a had an inhibitory effect on the differentiation of Raw264.7 cells and the migration and activity of osteoclasts upon irradiation but did not affect ROS. Sema3a also decreased the expression of CSTK, NFAT, TRAP-5b, Rankl/OPG, IL-1, IL-6 and TNFa on the 3rd and 7th days after irradiation (p<0.05), whereas P53 expression was increased (P<0.05). Sema3a reduced the inflammation induced by radiation and negatively regulated osteoclast differentiation. Sema3a promoted Raw264.7 cell apoptosis after irradiation, indicating that Sema3a could be a potential therapeutic target for radiation-induced osteoporosis.
Collapse
Affiliation(s)
- Bo Huang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China
- Dental Implant Center, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China
| | - Qin Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China
- Dental Implant Center, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China
| | - Ying Yuan
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China
- Dental Implant Center, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China
| | - Na Xin
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China
- Dental Implant Center, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China
| | - Kun He
- Dental Implant Center, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China
| | - Yan Huang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China
- OMFS-IMPATH Research Group, Oral Imaging Center, Department of Imaging and Pathology, Biomedical Sciences Group, University of Leuven, Leuven, Belgium
| | - Hua Tang
- Dental Implant Center, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China
- * E-mail: (HT); (PG)
| | - Ping Gong
- Dental Implant Center, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China
- * E-mail: (HT); (PG)
| |
Collapse
|
31
|
Farris M, McTyre ER, Okoukoni C, Dugan G, Johnson BJ, Blackstock AW, Munley MT, Bourland JD, Cline JM, Willey JS. Cortical Thinning and Structural Bone Changes in Non-Human Primates after Single-Fraction Whole-Chest Irradiation. Radiat Res 2018; 190:63-71. [PMID: 29738279 PMCID: PMC6036641 DOI: 10.1667/rr15007.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Stereotactic body radiation therapy (SBRT) is associated with an increased risk of vertebral compression fracture. While bone is typically considered radiation resistant, fractures frequently occur within the first year of SBRT. The goal of this work was to determine if rapid deterioration of bone occurs in vertebrae after irradiation. Sixteen male rhesus macaque non-human primates (NHPs) were analyzed after whole-chest irradiation to a midplane dose of 10 Gy. Ages at the time of exposure varied from 45-134 months. Computed tomography (CT) scans were taken 2 months prior to irradiation and 2, 4, 6 and 8 months postirradiation for all animals. Bone mineral density (BMD) and cortical thickness were calculated longitudinally for thoracic (T) 9, lumbar (L) 2 and L4 vertebral bodies; gross morphology and histopathology were assessed per vertebra. Greater mortality (related to pulmonary toxicity) was noted in NHPs <50 months at time of exposure versus NHPs >50 months ( P = 0.03). Animals older than 50 months at time of exposure lost cortical thickness in T9 by 2 months postirradiation ( P = 0.0009), which persisted to 8 months. In contrast, no loss of cortical thickness was observed in vertebrae out-of-field (L2 and L4). Loss of BMD was observed by 4 months postirradiation for T9, and 6 months postirradiation for L2 and L4 ( P < 0.01). For NHPs younger than 50 months at time of exposure, both cortical thickness and BMD decreased in T9, L2 and L4 by 2 months postirradiation ( P < 0.05). Regions that exhibited the greatest degree of cortical thinning as determined from CT scans also exhibited increased porosity histologically. Rapid loss of cortical thickness was observed after high-dose chest irradiation in NHPs. Younger age at time of exposure was associated with increased pneumonitis-related mortality, as well as greater loss of both BMD and cortical thickness at both in- and out-of-field vertebrae. Older NHPs exhibited rapid loss of BMD and cortical thickness from in-field vertebrae, but only loss of BMD in out-of-field vertebrae. Bone is sensitive to high-dose radiation, and rapid loss of bone structure and density increases the risk of fractures.
Collapse
Affiliation(s)
| | | | | | - Greg Dugan
- c Pathology/Section on Comparative Medicine
| | - Brendan J Johnson
- e Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
| | | | - Michael T Munley
- Departments of a Radiation Oncology
- b Biomedical Engineering
- d Physics, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - J Daniel Bourland
- Departments of a Radiation Oncology
- d Physics, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | | | | |
Collapse
|
32
|
Chishti AA, Baumstark-Khan C, Koch K, Kolanus W, Feles S, Konda B, Azhar A, Spitta LF, Henschenmacher B, Diegeler S, Schmitz C, Hellweg CE. Linear Energy Transfer Modulates Radiation-Induced NF-kappa B Activation and Expression of its Downstream Target Genes. Radiat Res 2018; 189:354-370. [DOI: 10.1667/rr14905.1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Arif Ali Chishti
- German Aerospace Centre (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Linder Höhe, D-51147 Köln, Germany
| | - Christa Baumstark-Khan
- German Aerospace Centre (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Linder Höhe, D-51147 Köln, Germany
| | - Kristina Koch
- German Aerospace Centre (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Linder Höhe, D-51147 Köln, Germany
| | - Waldemar Kolanus
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Karlrobert-Kreiten-Straße 13, 53115 Bonn, Germany
| | - Sebastian Feles
- German Aerospace Centre (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Linder Höhe, D-51147 Köln, Germany
| | - Bikash Konda
- German Aerospace Centre (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Linder Höhe, D-51147 Köln, Germany
| | - Abid Azhar
- The Karachi Institute of Biotechnology and Genetic Engineering, University of Karachi, Karachi-75270, Pakistan
| | - Luis F. Spitta
- German Aerospace Centre (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Linder Höhe, D-51147 Köln, Germany
| | - Bernd Henschenmacher
- German Aerospace Centre (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Linder Höhe, D-51147 Köln, Germany
| | - Sebastian Diegeler
- German Aerospace Centre (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Linder Höhe, D-51147 Köln, Germany
| | - Claudia Schmitz
- German Aerospace Centre (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Linder Höhe, D-51147 Köln, Germany
| | - Christine E. Hellweg
- German Aerospace Centre (DLR), Institute of Aerospace Medicine, Radiation Biology Department, Linder Höhe, D-51147 Köln, Germany
| |
Collapse
|
33
|
Ethiraj P, Link JR, Sinkway JM, Brown GD, Parler WA, Reddy SV. Microgravity modulation of syncytin-A expression enhance osteoclast formation. J Cell Biochem 2018; 119:5696-5703. [PMID: 29388695 DOI: 10.1002/jcb.26750] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 01/25/2018] [Indexed: 12/25/2022]
Abstract
Microgravity (μXg) experienced by astronauts during space flights causes accelerated bone loss. However, the molecular basis of μXg induced bone loss in space is unclear. Osteoclast (OCL) is the primary bone-resorbing cell. We previously demonstrated that simulated μXg promotes OCL formation. In this study, we identified that μXg induces syncytin-A expression in RAW264.7 preosteoclast cells without RANKL stimulation. We further tested the effect of osteotropic factors such as CXCL5 and 1,25(OH)2 D3 to regulate the syncytin-A expression in preosteoclast cells subjected to μXg compared to ground based (Xg) cultures. CXCL5 (25 ng/mL) and 1,25(OH)2 D3 (10 ng/mL) increased syncytin-A expression under Xg conditions. However, μXg alone upregulates syncytin-A expression compared to Xg control preosteoclast cells. Confocal microscopy using Lyso-Tracker identified syncytin-A expression co-localized with lysosomes in preosteoclast cells. Acridine orange staining showed RANKL elevated autophagy activity in these cells. Further, siRNA suppression of syncytin-A significantly inhibits autophagy activity in RAW264.7 cells. In addition, knockdown of syncytin-A expression inhibits μXg increased OCL formation in mouse bone marrow cultures. Thus, our findings suggest that targeting syncytin-A expression may be an effective countermeasure to control bone loss under microgravity conditions.
Collapse
Affiliation(s)
- Purushoth Ethiraj
- Darby Children's Research Institute, Department of Pediatrics/Endocrinology, Medical University of South Carolina, Charleston, South Carolina
| | - Jessica R Link
- Darby Children's Research Institute, Department of Pediatrics/Endocrinology, Medical University of South Carolina, Charleston, South Carolina
| | - James M Sinkway
- Darby Children's Research Institute, Department of Pediatrics/Endocrinology, Medical University of South Carolina, Charleston, South Carolina
| | - Gabriella D Brown
- Darby Children's Research Institute, Department of Pediatrics/Endocrinology, Medical University of South Carolina, Charleston, South Carolina
| | - William A Parler
- Darby Children's Research Institute, Department of Pediatrics/Endocrinology, Medical University of South Carolina, Charleston, South Carolina
| | - Sakamuri V Reddy
- Darby Children's Research Institute, Department of Pediatrics/Endocrinology, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
34
|
|
35
|
Zhang J, Wang Z, Wu A, Nie J, Pei H, Hu W, Wang B, Shang P, Li B, Zhou G. Differences in responses to X-ray exposure between osteoclast and osteoblast cells. JOURNAL OF RADIATION RESEARCH 2017; 58:791-802. [PMID: 28541506 PMCID: PMC5710662 DOI: 10.1093/jrr/rrx026] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Indexed: 05/07/2023]
Abstract
Radiation-induced bone loss is a potential health concern for cancer patients undergoing radiotherapy. Enhanced bone resorption by osteoclasts and decreased bone formation by osteoblasts were thought to be the main reasons. In this study, we showed that both pre-differentiating and differentiating osteoclasts were relatively sensitive to X-rays compared with osteoblasts. X-rays decreased cell viability to a greater degree in RAW264.7 cells and in differentiating cells than than in osteoblastic MC3T3-E1 cells. X-rays at up to 8 Gy had little effects on osteoblast mineralization. In contrast, X-rays at 1 Gy induced enhanced osteoclastogenesis by enhanced cell fusion, but had no effects on bone resorption. A higher dose of X-rays at 8 Gy, however, had an inhibitory effect on bone resorption. In addition, actin ring formation was disrupted by 8 Gy of X-rays and reorganized into clusters. An increased activity of Caspase 3 was found after X-ray exposure. Actin disorganization and increased apoptosis may be the potential effects of X-rays at high doses, by inhibiting osteoclast differentiation. Taken together, our data indicate high radiosensitivity of osteoclasts. X-ray irradiation at relatively low doses can activate osteoclastogenesis, but not osteogenic differentiation. The radiosensitive osteoclasts are the potentially responsive cells for X-ray-induced bone loss.
Collapse
Affiliation(s)
- Jian Zhang
- School of Radiation Medicine and Protection, Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, 199 Renai Road, Suzhou 215123, China
| | - Ziyang Wang
- School of Radiation Medicine and Protection, Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, 199 Renai Road, Suzhou 215123, China
| | - Anqing Wu
- School of Radiation Medicine and Protection, Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, 199 Renai Road, Suzhou 215123, China
| | - Jing Nie
- School of Radiation Medicine and Protection, Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, 199 Renai Road, Suzhou 215123, China
| | - Hailong Pei
- School of Radiation Medicine and Protection, Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, 199 Renai Road, Suzhou 215123, China
| | - Wentao Hu
- School of Radiation Medicine and Protection, Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, 199 Renai Road, Suzhou 215123, China
| | - Bing Wang
- Department of Radiation Effects Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Anagawa 4-9-1, Inage-ku, Chiba 263-555, Japan
| | - Peng Shang
- School of Radiation Medicine and Protection, Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, 199 Renai Road, Suzhou 215123, China
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, 127 Youyi Road, Xi'an 710072, China
| | - Bingyan Li
- Department of Nutrition and Food Hygiene, School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China
| | - Guangming Zhou
- School of Radiation Medicine and Protection, Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, 199 Renai Road, Suzhou 215123, China
- Corresponding author. School of Radiation Medicine and Protection, Medical College of Soochow University, 199 Renai Road, Suzhou 215123, China. Tel: +86-512-6588-4829; Fax: +86-512-6588-4830;
| |
Collapse
|
36
|
Tahimic CGT, Globus RK. Redox Signaling and Its Impact on Skeletal and Vascular Responses to Spaceflight. Int J Mol Sci 2017; 18:ijms18102153. [PMID: 29035346 PMCID: PMC5666834 DOI: 10.3390/ijms18102153] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 09/30/2017] [Accepted: 10/10/2017] [Indexed: 12/16/2022] Open
Abstract
Spaceflight entails exposure to numerous environmental challenges with the potential to contribute to both musculoskeletal and vascular dysfunction. The purpose of this review is to describe current understanding of microgravity and radiation impacts on the mammalian skeleton and associated vasculature at the level of the whole organism. Recent experiments from spaceflight and ground-based models have provided fresh insights into how these environmental stresses influence mechanisms that are related to redox signaling, oxidative stress, and tissue dysfunction. Emerging mechanistic knowledge on cellular defenses to radiation and other environmental stressors, including microgravity, are useful for both screening and developing interventions against spaceflight-induced deficits in bone and vascular function.
Collapse
Affiliation(s)
- Candice G T Tahimic
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA.
- KBRWyle, Moffett Field, CA 94035, USA.
| | - Ruth K Globus
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA.
| |
Collapse
|
37
|
|
38
|
Alwood JS, Tran LH, Schreurs AS, Shirazi-Fard Y, Kumar A, Hilton D, Tahimic CGT, Globus RK. Dose- and Ion-Dependent Effects in the Oxidative Stress Response to Space-Like Radiation Exposure in the Skeletal System. Int J Mol Sci 2017; 18:ijms18102117. [PMID: 28994728 PMCID: PMC5666799 DOI: 10.3390/ijms18102117] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 09/29/2017] [Accepted: 09/30/2017] [Indexed: 12/12/2022] Open
Abstract
Space radiation may pose a risk to skeletal health during subsequent aging. Irradiation acutely stimulates bone remodeling in mice, although the long-term influence of space radiation on bone-forming potential (osteoblastogenesis) and possible adaptive mechanisms are not well understood. We hypothesized that ionizing radiation impairs osteoblastogenesis in an ion-type specific manner, with low doses capable of modulating expression of redox-related genes. 16-weeks old, male, C57BL6/J mice were exposed to low linear-energy-transfer (LET) protons (150 MeV/n) or high-LET 56Fe ions (600 MeV/n) using either low (5 or 10 cGy) or high (50 or 200 cGy) doses at NASA's Space Radiation Lab. Five weeks or one year after irradiation, tissues were harvested and analyzed by microcomputed tomography for cancellous microarchitecture and cortical geometry. Marrow-derived, adherent cells were grown under osteoblastogenic culture conditions. Cell lysates were analyzed by RT-PCR during the proliferative or mineralizing phase of growth, and differentiation was analyzed by imaging mineralized nodules. As expected, a high dose (200 cGy), but not lower doses, of either 56Fe or protons caused a loss of cancellous bone volume/total volume. Marrow cells produced mineralized nodules ex vivo regardless of radiation type or dose; 56Fe (200 cGy) inhibited osteoblastogenesis by more than 90% (5 weeks and 1 year post-IR). After 5 weeks, irradiation (protons or 56Fe) caused few changes in gene expression levels during osteoblastogenesis, although a high dose 56Fe (200 cGy) increased Catalase and Gadd45. The addition of exogenous superoxide dismutase (SOD) protected marrow-derived osteoprogenitors from the damaging effects of exposure to low-LET (137Cs γ) when irradiated in vitro, but had limited protective effects on high-LET 56Fe-exposed cells. In sum, either protons or 56Fe at a relatively high dose (200 cGy) caused persistent bone loss, whereas only high-LET 56Fe increased redox-related gene expression, albeit to a limited extent, and inhibited osteoblastogenesis. Doses below 50 cGy did not elicit widespread responses in any parameter measured. We conclude that high-LET irradiation at 200 cGy impaired osteoblastogenesis and regulated steady-state gene expression of select redox-related genes during osteoblastogenesis, which may contribute to persistent bone loss.
Collapse
Affiliation(s)
- Joshua S Alwood
- Bone and Signaling Laboratory, Space BioSciences Division, NASA Ames Research Center, Mail-Stop 236-7, Moffett Field, CA 94035, USA.
| | - Luan H Tran
- Bone and Signaling Laboratory, Space BioSciences Division, NASA Ames Research Center, Mail-Stop 236-7, Moffett Field, CA 94035, USA.
| | - Ann-Sofie Schreurs
- Bone and Signaling Laboratory, Space BioSciences Division, NASA Ames Research Center, Mail-Stop 236-7, Moffett Field, CA 94035, USA.
| | - Yasaman Shirazi-Fard
- Bone and Signaling Laboratory, Space BioSciences Division, NASA Ames Research Center, Mail-Stop 236-7, Moffett Field, CA 94035, USA.
| | - Akhilesh Kumar
- Bone and Signaling Laboratory, Space BioSciences Division, NASA Ames Research Center, Mail-Stop 236-7, Moffett Field, CA 94035, USA.
| | - Diane Hilton
- Bone and Signaling Laboratory, Space BioSciences Division, NASA Ames Research Center, Mail-Stop 236-7, Moffett Field, CA 94035, USA.
| | - Candice G T Tahimic
- Bone and Signaling Laboratory, Space BioSciences Division, NASA Ames Research Center, Mail-Stop 236-7, Moffett Field, CA 94035, USA.
- Wyle Laboratories, Mail-Stop 236-7, Moffett Field, CA 94035, USA.
| | - Ruth K Globus
- Bone and Signaling Laboratory, Space BioSciences Division, NASA Ames Research Center, Mail-Stop 236-7, Moffett Field, CA 94035, USA.
| |
Collapse
|
39
|
Osteoclast profile of medication-related osteonecrosis of the jaw secondary to bisphosphonate therapy: a comparison with osteoradionecrosis and osteomyelitis. J Transl Med 2017; 15:128. [PMID: 28587628 PMCID: PMC5461625 DOI: 10.1186/s12967-017-1230-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 05/30/2017] [Indexed: 12/25/2022] Open
Abstract
Background The medication-related osteonecrosis of the jaw secondary to bisphosphonate therapy [MRONJ (BP)] is characterized by non-healing exposed bone in the maxillofacial region. The pathogenesis of MRONJ (BP) is not fully understood. Giant, hypernucleated, inactive osteoclasts were found in MRONJ (BP) tissues, which indicated that accelerated cell–cell fusion might play a role. Dendritic cell-specific transmembrane protein (DC-STAMP) is associated with the cell–cell fusion of osteoclasts and precursor cells. Tartrate-resistant acid phosphatase (TRAP) is essential for osteoclastic bone resorption. The cell–cell fusion, as part of the osteoclastogenesis, and the resorptive activity can determine the morphology of osteoclasts. This study analyzed jaw bone from patients with MRONJ (BP), osteomyelitis (OM) and osteoradionecrosis (ORN) because a comparison with the osteoclast profiles of OM and ORN is essential for characterizing the osteoclast profile of MRONJ (BP). Methods Formalin-fixed routine jaw bone specimens from 70 patients [MRONJ (BP) n = 30; OM: n = 15, ORN: n = 15, control: n = 10] were analyzed retrospectively for osteoclast quantity, morphology and the expression of TRAP and DC-STAMP. The specimens were processed for hematoxylin and eosin staining (H&E), histochemistry (TRAP) and immunohistochemistry (anti-DC-STAMP) and were analyzed via virtual microscopy. Results The quantity, diameter and nuclearity of osteoclasts were significantly higher in MRONJ (BP) specimens than in OM, ORN and control specimens. Giant, hypernucleated osteoclasts were detected in MRONJ (BP) specimens only. Osteoclastic TRAP expression was lower in MRONJ (BP) and ORN specimens than in OM and control specimens. The DC-STAMP expression of osteoclasts and mononuclear cells was significantly higher in MRONJ (BP) and ORN specimens than in OM and control specimens. Conclusions This study indicates that the osteoclast profile of MRONJ (BP) is characterized by osteoclast inactivation and a high cell–cell fusion rate; however, the presence of giant, hypernucleated osteoclasts cannot be attributed to increased DC-STAMP-triggered cell–cell fusion alone. The incidental characterization of the osteoclast profiles of OM and ORN revealed differences that might facilitate the histopathological differentiation of these diseases from MRONJ (BP), which is essential because their therapies are somewhat different.
Collapse
|
40
|
Afshar SF, Zawaski JA, Inoue T, Rendon DA, Zieske AW, Punia JN, Sabek OM, Gaber MW. Investigating the Abscopal Effects of Radioablation on Shielded Bone Marrow in Rodent Models Using Multimodality Imaging. Radiat Res 2017; 188:56-65. [PMID: 28475423 DOI: 10.1667/rr14692.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The abscopal effect is the response to radiation at sites that are distant from the irradiated site of an organism, and it is thought to play a role in bone marrow (BM) recovery by initiating responses in the unirradiated bone marrow. Understanding the mechanism of this effect has applications in treating BM failure (BMF) and BM transplantation (BMT), and improving survival of nuclear disaster victims. Here, we investigated the use of multimodality imaging as a translational tool to longitudinally assess bone marrow recovery. We used positron emission tomography/computed tomography (PET/CT), magnetic resonance imaging (MRI) and optical imaging to quantify bone marrow activity, vascular response and marrow repopulation in fully and partially irradiated rodent models. We further measured the effects of radiation on serum cytokine levels, hematopoietic cell counts and histology. PET/CT imaging revealed a radiation-induced increase in proliferation in the shielded bone marrow (SBM) compared to exposed bone marrow (EBM) and sham controls. T2-weighted MRI showed radiation-induced hemorrhaging in the EBM and unirradiated SBM. In the EBM and SBM groups, we found alterations in serum cytokine and hormone levels and in hematopoietic cell population proportions, and histological evidence of osteoblast activation at the bone marrow interface. Importantly, we generated a BMT mouse model using fluorescent-labeled bone marrow donor cells and performed fluorescent imaging to reveal the migration of bone marrow cells from shielded to radioablated sites. Our study validates the use of multimodality imaging to monitor bone marrow recovery and provides evidence for the abscopal response in promoting bone marrow recovery after irradiation.
Collapse
Affiliation(s)
- Solmaz F Afshar
- a Department of Surgery, Houston Methodist Hospital Research Institute, Houston, Texas
| | - Janice A Zawaski
- b Hematology-Oncology Section, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Taeko Inoue
- b Hematology-Oncology Section, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - David A Rendon
- b Hematology-Oncology Section, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Arthur W Zieske
- d Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas
| | - Jyotinder N Punia
- c Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
| | - Omaima M Sabek
- a Department of Surgery, Houston Methodist Hospital Research Institute, Houston, Texas
| | - M Waleed Gaber
- b Hematology-Oncology Section, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
41
|
Poort LJ, Ludlage JH, Lie N, Böckmann RA, Odekerken JC, Hoebers FJ, Kessler PA. The histological and histomorphometric changes in the mandible after radiotherapy: An animal model. J Craniomaxillofac Surg 2017; 45:716-721. [DOI: 10.1016/j.jcms.2017.02.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 01/15/2017] [Accepted: 02/13/2017] [Indexed: 02/07/2023] Open
|
42
|
Schreurs AS, Shirazi-Fard Y, Shahnazari M, Alwood JS, Truong TA, Tahimic CGT, Limoli CL, Turner ND, Halloran B, Globus RK. Dried plum diet protects from bone loss caused by ionizing radiation. Sci Rep 2016; 6:21343. [PMID: 26867002 PMCID: PMC4750446 DOI: 10.1038/srep21343] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 01/21/2016] [Indexed: 12/21/2022] Open
Abstract
Bone loss caused by ionizing radiation is a potential health concern for radiotherapy patients, radiation workers and astronauts. In animal studies, exposure to ionizing radiation increases oxidative damage in skeletal tissues, and results in an imbalance in bone remodeling initiated by increased bone-resorbing osteoclasts. Therefore, we evaluated various candidate interventions with antioxidant or anti-inflammatory activities (antioxidant cocktail, dihydrolipoic acid, ibuprofen, dried plum) both for their ability to blunt the expression of resorption-related genes in marrow cells after irradiation with either gamma rays (photons, 2 Gy) or simulated space radiation (protons and heavy ions, 1 Gy) and to prevent bone loss. Dried plum was most effective in reducing the expression of genes related to bone resorption (Nfe2l2, Rankl, Mcp1, Opg, TNF-α) and also preventing later cancellous bone decrements caused by irradiation with either photons or heavy ions. Thus, dietary supplementation with DP may prevent the skeletal effects of radiation exposures either in space or on Earth.
Collapse
Affiliation(s)
- A-S Schreurs
- Bone and Signaling Laboratory, Space Biosciences Division, NASA Ames Research Center
| | - Y Shirazi-Fard
- Bone and Signaling Laboratory, Space Biosciences Division, NASA Ames Research Center
| | - M Shahnazari
- Bone and Signaling Laboratory, Space Biosciences Division, NASA Ames Research Center
| | - J S Alwood
- Bone and Signaling Laboratory, Space Biosciences Division, NASA Ames Research Center
| | - T A Truong
- Bone and Signaling Laboratory, Space Biosciences Division, NASA Ames Research Center
| | - C G T Tahimic
- Bone and Signaling Laboratory, Space Biosciences Division, NASA Ames Research Center
| | - C L Limoli
- Department of Radiation Oncology, University of California Irvine
| | - N D Turner
- Department of Nutrition and Food Science, Texas A&M University
| | - B Halloran
- Department of Medicine, Division of Endocrinology, University of California San Francisco
| | - R K Globus
- Bone and Signaling Laboratory, Space Biosciences Division, NASA Ames Research Center
| |
Collapse
|