1
|
Chu T, Wasi M, Guerra RM, Song X, Wang S, Sims-Mourtada J, You L, Wang L. Skeletal response to Yoda1 and whole-body vibration in mice varied with animal age, bone compartment, treatment duration, and radiation exposure. Bone 2025:117525. [PMID: 40389188 DOI: 10.1016/j.bone.2025.117525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Revised: 05/13/2025] [Accepted: 05/13/2025] [Indexed: 05/21/2025]
Abstract
In this study, we investigated the skeletal effects of Yoda1, an agonist of the mechanosensitive Piezo1 channels, and whole-body vibration (WBV), alone and combined, in young mice (8-week-old) and in mature (31- to 36-week-old) mice after radiation exposure. Our goal was to determine whether the two mechanobiology-based interventions, known to induce anabolic response individually in young subjects, could promote the bone health of older subjects undergoing cancer treatments such as radiotherapy. Our hypothesis was that the combination of Yoda1 and WBV could improve young skeletons and protect mature skeletons after radiotherapy better than Yoda1 or WBV alone. Our in vivo experiments demonstrated (1) that Yoda1 (5 mg/kg body weight) alone or combined with WBV (0.3 g, 13 Hz, 30 min/day, 5 days/week, 4 weeks) enhanced bone growth similarly (~2 folds relative to nontreated controls) in young mice; (2) that mature mice were unresponsive to individual interventions but exhibited less polar moment of inertia loss (-56 %) in the tibiae receiving the combination of Yoda1 and WBV (15 min/day) but no radiation exposure; and (3) that the contralateral tibiae receiving fractionated radiation (2 × 8 Gy over three days) did not show different treatment responses in Week 4, while they responded to the combination therapy (increased cortical bone formation) in Week 2. Interestingly, pair comparisons of the irradiated and non-irradiated tibiae of the same animals revealed that radiation exposure resulted in decreased trabecular bone loss regardless of the treatments and increased the percentage of tibiae maintaining better cortical polar moment of inertia and cortical area in the groups receiving Yoda1 or the combination therapy. The complex skeletal responses to Yoda1 and/or WBV were compartment specific (cortical or trabecular bone) and dependent on animal age, radiation exposure, and treatment duration. This study partially supported our original hypothesis, while suggesting the need of finetuning the Yoda1 and WBV regimens and elucidating the underlying mechanisms in order to effectively treat age and radiation induced bone loss.
Collapse
Affiliation(s)
- Tiankuo Chu
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Murtaza Wasi
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Rosa M Guerra
- Department of Biomedical Engineering, University of Delaware, Newark, DE, USA
| | - Xin Song
- Department of Mechanical and Industrial Engineering, Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Shubo Wang
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | | | - Lidan You
- Department of Mechanical and Industrial Engineering, Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada; Department of Mechanical and Materials Engineering, Queens University, Ontario, Canada
| | - Liyun Wang
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA; Department of Biomedical Engineering, University of Delaware, Newark, DE, USA.
| |
Collapse
|
2
|
Carney TE, Biggs AE, Miller MA, Mann KA, Oest ME. Therapeutic radiation directly alters bone fatigue strength and microdamage accumulation. J Mech Behav Biomed Mater 2024; 160:106766. [PMID: 39378671 DOI: 10.1016/j.jmbbm.2024.106766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/28/2024] [Accepted: 10/01/2024] [Indexed: 10/10/2024]
Abstract
Radiotherapy (RTx) is an essential and efficacious oncologic treatment, however, post-RTx bone fragility fractures present a challenging clinical problem. Cancer survivors treated with RTx are at variable risk for these late-onset, complex fragility fractures. Little data exists regarding the effects of RTx on bone fatigue properties despite the likelihood of fatigue loading as a mechanism leading up to atraumatic fracture. In this study, femurs collected from adult male rats were irradiated ex vivo with a therapeutic dose of x-irradiation (20 Gy), and then fatigued using a three-point bend setup. Femurs positioned in an isotonic bath at room temperature were loaded to a range of prescribed initial strain levels (based on beam theory equations, prior to any fatigue damage) at 3 Hz in force control. The goals of this study were to determine the feasibility of assessing RTx-induced alterations in 1) femur fatigue strength, 2) structural microdamage (creep and stiffness), and 3) tissue damage (diffuse damage and/or linear microcracking). Mid-diaphyseal morphology and tissue mineral density were not different between the RTx and Sham groups (p ≥ 0.35). With increasing applied apparent strain, the number of cycles to failure was reduced for the RTx femurs when compared to the Sham femurs (treatment x εapp, p = 0.041). RTx femurs had a greater phase II (steady state) creep rate (p = 0.0462) compared to Sham femurs. For femurs that reached 500k cycles, the RTx group had greater diffuse damage area (p = 0.015) than the Sham. This study provides evidence that radiation at therapeutic doses can directly diminish bone fatigue properties. This loss of fatigue properties is associated with increased structural fatigue damage and diffuse microdamage, without alterations in morphology or tissue mineral density, indicating a reduction in bone quality.
Collapse
Affiliation(s)
- Tara E Carney
- Department of Orthopedic Surgery, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, 13210, USA; Department of Cell and Developmental Biology, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, 13210, USA.
| | - Amy E Biggs
- Department of Orthopedic Surgery, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, 13210, USA.
| | - Mark A Miller
- Department of Orthopedic Surgery, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, 13210, USA.
| | - Kenneth A Mann
- Department of Orthopedic Surgery, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, 13210, USA.
| | - Megan E Oest
- Department of Orthopedic Surgery, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, 13210, USA.
| |
Collapse
|
3
|
Stering JA, Biggs AE, Carney TE, Oest ME, Simone BA. Caloric Restriction Diet Attenuates Systemic Bone Fragility after Radiotherapy. Radiat Res 2024; 202:765-774. [PMID: 39307529 DOI: 10.1667/rade-23-00227.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 09/04/2024] [Indexed: 11/09/2024]
Abstract
Bone fragility is a well-documented long-term side effect of radiotherapy, which currently has no preventative treatments. In this study, we applied a caloric restriction (CR) diet to attenuate both local and systemic bone loss after irradiation (RTx) in an established female Balb/c mouse model (4 consecutive daily doses of 5 Gy to the right hindlimb only). CR mice were tapered down to a 30% reduced calorie diet (RTx/CR) one week before irradiation, while regular diet (RD) mice received food ad libitum (RTx/RD). Unirradiated (sham) mice received either a 30% CR diet (SH/CR) or received food ad libitum (SH/RD). Irradiated, contralateral, and unirradiated hindlimbs were evaluated at 2, 4, and 8 weeks postirradiation using micro-computed tomography (μCT) to assess bone morphology and 3-point bending to quantify femur strength. Histological analysis of irradiated and unirradiated tibiae was performed to examine general bone tissue cytology and serum biomarker analysis was performed using terminal blood draw samples. After treatment, femur strength and metaphyseal bone quantity was decreased in irradiated and contralateral femora of RTx/RD mice compared to SH/RD femurs; this finding is consistent with previous studies. RTx/CR mice had positive effects when compared to RTx/RD mice, including increased strength relative to body mass in both the irradiated and contralateral limb, increased trabecular bone mass, and decreased marrow adiposity. However, a number of adverse effects were also observed, including a significant decrease in body mass and decreased cortical bone. Overall, CR shows promise as a preventative treatment for postirradiated bone fragility, yet questions remain to be addressed in future studies. Ideal diet duration, impact to normal tissue, and mechanism of action must be explored to better understand the clinical implication of a CR diet.
Collapse
Affiliation(s)
- Jessica A Stering
- Department of Orthopedic Surgery, SUNY Upstate Medical University, Syracuse, New York
| | - Amy E Biggs
- Department of Orthopedic Surgery, SUNY Upstate Medical University, Syracuse, New York
| | - Tara E Carney
- Department of Orthopedic Surgery, SUNY Upstate Medical University, Syracuse, New York
| | - Megan E Oest
- Department of Orthopedic Surgery, SUNY Upstate Medical University, Syracuse, New York
| | - Brittany A Simone
- Department of Radiation Oncology, SUNY Upstate Medical University, Syracuse, New York
| |
Collapse
|
4
|
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
|
5
|
Gottsauner M, Sroka AM, Eichberger J, Schuderer J, Zeman F, Fiedler M, Maurer M, Einspieler I, Reichert TE, Ettl T. Progress of Ossification after Mandibular Reconstruction by Free Fibula Flap Depending on Different Timing of Radiotherapy: A Retrospective 3D Analysis by CT Scans. J Clin Med 2024; 13:4104. [PMID: 39064143 PMCID: PMC11278501 DOI: 10.3390/jcm13144104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Background: The aim of this study was to evaluate the difference between pre- and post-operative radiotherapy on the progress of ossification after free fibula flap reconstruction of the mandible using three-dimensional (3D) analysis. Methods: A total of 38 free fibula reconstructions of the mandible were evaluated retrospectively for ossification between bone segments by measuring Hounsfield Units (HU) in at least two postoperative computer tomography scans (average of 2.4 scans per patient; around the 5th, 12th, 16th, and 19th month postoperative). Three subgroups were created according to the time of irradiation: preoperative radiotherapy (preORT) (n = 11), postoperative radiotherapy (postORT) (n = 16), and patients without any radiation therapy (n = 11) as the control group (noRT). HU in eight regions of interest (ROI) and overlapping surfaces between segments per contact point, as well as influencing factors, were analyzed. Results: The fastest progress in gain of HU ossification with a difference of 0.30 HU/day was observed in noRT compared to preORT (p = 0.002). postORT was -0.24 HU/day slower than preORT (p = 0.005). Original and grafted bone showed a significantly slower HU uptake than between two graft segments with -84.18 HU/day (p < 0.001). Moreover, a larger initial overlapping surface between the segments in cm2 resulted in a higher rise of HU/day (p < 0.001). Conclusions: 3D analysis of post-reconstructive CT scans shows prolonged ossification of mandible reconstructions by free fibula after head and neck radiation. The effect is distinct in cases with post-operative adjuvant radiotherapy. The effects of radiotherapy on ossification may be minimized by a larger initial contact surface and improved operational techniques. Moreover, HU longitudinal measurements and 3D analysis offer new perspectives for clinical evaluation of successful bony healing.
Collapse
Affiliation(s)
- Maximilian Gottsauner
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Anne Marie Sroka
- Department of Prosthetic Dentistry, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Jonas Eichberger
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Johannes Schuderer
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Florian Zeman
- Center for Clinical Studies, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Mathias Fiedler
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Michael Maurer
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Ingo Einspieler
- Department of Radiology, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Torsten E. Reichert
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Tobias Ettl
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| |
Collapse
|
6
|
Iwasaki R, Yoshikawa R, Umeno R, Seki A, Matsukawa T, Takeno S, Yokoyama K, Mori T, Suzuki M, Ono K. The effects of BPA-BNCT on normal bone: determination of the CBE value in mice‡. JOURNAL OF RADIATION RESEARCH 2023; 64:795-803. [PMID: 37517393 PMCID: PMC10516729 DOI: 10.1093/jrr/rrad054] [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: 06/05/2023] [Revised: 07/03/2023] [Indexed: 08/01/2023]
Abstract
Boron neutron capture therapy (BNCT) with p-boronophenylalanine (BPA) is expected to have less effect on the decrease in normal bone strength than X-ray therapy. However, the compound biological effectiveness (CBE) value necessary to convert the boron neutron capture reaction (BNCR) dose into a bioequivalent X-ray dose has not been determined yet. The purpose of this study was to evaluate the influence of BNCT on normal bone in mice and to elucidate the CBE factor. We first searched the distribution of BPA in the normal bone of C3H/He mice and then measured the changes in bone strength after irradiation. The CBE value was determined when the decrease in bone strength was set as an index of the BNCT effect. The 10B concentrations in the tibia after subcutaneous injection of 125, 250 and 500 mg/kg BPA were measured by prompt gamma-ray spectroscopy and inductively coupled plasma (ICP)-atomic emission spectrometry. The 10B mapping in the tibia was examined by alpha-track autoradiography and laser ablation-ICP-mass spectrometry. The 10B concentration increased dose-dependently; moreover, the concentrations were maintained until 120 min after BPA administration. The administered 10B in the tibia was abundantly accumulated in the growth cartilage, trabecular bone and bone marrow. The bone strength was analyzed by a three-point bending test 12 weeks after irradiation. The bending strength of the tibia decreased dose-dependently after the irradiation of X-ray, neutron and BNCR. The CBE factor was obtained as 2.27 by comparing these dose-effect curves; the value determined in this study will enable an accurate dosimetry of normal bone.
Collapse
Affiliation(s)
- Ryota Iwasaki
- Department of Veterinary Medicine, Gifu University, 1-1 Yanagido, Gifu-shi, Gifu 501-1193, Japan
| | - Ryutaro Yoshikawa
- Department of Veterinary Medicine, Gifu University, 1-1 Yanagido, Gifu-shi, Gifu 501-1193, Japan
| | - Ryo Umeno
- Department of Veterinary Medicine, Gifu University, 1-1 Yanagido, Gifu-shi, Gifu 501-1193, Japan
| | - Azusa Seki
- HAMRI Co. Ltd., 2638-2 Ozaki, Koga-shi, Ibaragi 306-0101, Japan
| | - Takehisa Matsukawa
- Department of Epidemiology and Environmental Health, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Department of Forensic Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Satoshi Takeno
- Department of Radiation Oncology, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi Takatsuki-shi, Osaka 569-8686, Japan
- Kansai BNCT Medical Center, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi Takatsuki-shi, Osaka 569-8686, Japan
| | - Kazuhito Yokoyama
- Department of Epidemiology and Environmental Health, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Department of Epidemiology and Social Medicine, International University of Health and Welfare, 4-1-26 Akasaka, Minato-ku, Tokyo 107-8402, Japan
| | - Takashi Mori
- Department of Veterinary Medicine, Gifu University, 1-1 Yanagido, Gifu-shi, Gifu 501-1193, Japan
| | - Minoru Suzuki
- Particle Radiation Oncology Research Center, Institute for Integrated Radiation and Nuclear Science, Kyoto University, 2-1010, Asashiro-nishi, Kumatori-cho, Sennan-gun, Osaka 590-0494, Japan
| | - Koji Ono
- Kansai BNCT Medical Center, Osaka Medical and Pharmaceutical University, 2-7 Daigaku-machi Takatsuki-shi, Osaka 569-8686, Japan
| |
Collapse
|
7
|
Babu B, Pawar S, Mittal A, Kolanthai E, Neal CJ, Coathup M, Seal S. Nanotechnology enabled radioprotectants to reduce space radiation-induced reactive oxidative species. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1896. [PMID: 37190884 DOI: 10.1002/wnan.1896] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/04/2023] [Accepted: 04/18/2023] [Indexed: 05/17/2023]
Abstract
Interest in space exploration has seen substantial growth following recent launch and operation of modern space technologies. In particular, the possibility of travel beyond low earth orbit is seeing sustained support. However, future deep space travel requires addressing health concerns for crews under continuous, longer-term exposure to adverse environmental conditions. Among these challenges, radiation-induced health issues are a major concern. Their potential to induce chronic illness is further potentiated by the microgravity environment. While investigations into the physiological effects of space radiation are still under investigation, studies on model ionizing radiation conditions, in earth and micro-gravity conditions, can provide needed insight into relevant processes. Substantial formation of high, sustained reactive oxygen species (ROS) evolution during radiation exposure is a clear threat to physiological health of space travelers, producing indirect damage to various cell structures and requiring therapeutic address. Radioprotection toward the skeletal system components is essential to astronaut health, due to the high radio-absorption cross-section of bone mineral and local hematopoiesis. Nanotechnology can potentially function as radioprotectant and radiomitigating agents toward ROS and direct radiation damage. Nanoparticle compositions such as gold, silver, platinum, carbon-based materials, silica, transition metal dichalcogenides, and ceria have all shown potential as viable radioprotectants to mitigate space radiation effects with nanoceria further showing the ability to protect genetic material from oxidative damage in several studies. As research into space radiation-induced health problems develops, this review intends to provide insights into the nanomaterial design to ameliorate pathological effects from ionizing radiation exposure. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Nanotechnology Approaches to Biology > Cells at the Nanoscale Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Balaashwin Babu
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida, USA
- Nanoscience Technology Center, University of Central Florida, Orlando, Florida, USA
| | - Shreya Pawar
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida, USA
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Agastya Mittal
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida, USA
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Elayaraja Kolanthai
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida, USA
| | - Craig J Neal
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida, USA
| | - Melanie Coathup
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida, USA
| | - Sudipta Seal
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida, USA
- College of Medicine, Nanoscience Technology Center, University of Central Florida, Orlando, Florida, USA
| |
Collapse
|
8
|
Emerzian SR, Wu T, Vaidya R, Tang SY, Abergel RJ, Keaveny TM. Relative Effects of Radiation-Induced Changes in Bone Mass, Structure, and Tissue Material on Vertebral Strength in a Rat Model. J Bone Miner Res 2023; 38:1032-1042. [PMID: 37191221 PMCID: PMC10524463 DOI: 10.1002/jbmr.4828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 04/06/2023] [Accepted: 05/07/2023] [Indexed: 05/17/2023]
Abstract
The observed increased risk of fracture after cancer radiation therapy is presumably due to a radiation-induced reduction in whole-bone strength. However, the mechanisms for impaired strength remain unclear, as the increased fracture risk is not fully explained by changes in bone mass. To provide insight, a small animal model was used to determine how much of this whole-bone weakening effect for the spine is attributable to changes in bone mass, structure, and material properties of the bone tissue and their relative effects. Further, because women have a greater risk of fracture after radiation therapy than men, we investigated if sex had a significant influence on bone's response to irradiation. Fractionated in vivo irradiation (10 × 3 Gy) or sham irradiation (0 Gy) was administered daily to the lumbar spine in twenty-seven 17-week-old Sprague-Dawley rats (n = 6-7/sex/group). Twelve weeks after final treatment, animals were euthanized, and lumbar vertebrae (L4 and L5 ) were isolated. Using a combination of biomechanical testing, micro-CT-based finite element analysis, and statistical regression analysis, we separated out the effect of mass, structural, and tissue material changes on vertebral strength. Compared with the sham group (mean ± SD strength = 420 ± 88 N), the mean strength of the irradiated group was lower by 28% (117 N/420 N, p < 0.0001). Overall, the response of treatment did not differ with sex. By combining results from both general linear regression and finite element analyses, we calculated that mean changes in bone mass, structure, and material properties of the bone tissue accounted for 56% (66 N/117 N), 20% (23 N/117 N), and 24% (28 N/117 N), respectively, of the overall change in strength. As such, these results provide insight into why an elevated clinical fracture risk for patients undergoing radiation therapy is not well explained by changes in bone mass alone. © 2023 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Shannon R. Emerzian
- Department of Mechanical Engineering, University of
California, Berkeley, California, USA
| | - Tongge Wu
- Department of Mechanical Engineering, University of
California, Berkeley, California, USA
| | - Rachana Vaidya
- Department of Orthopaedic Surgery, Washington University,
St. Louis, Missouri, USA
| | - Simon Y. Tang
- Department of Orthopaedic Surgery, Washington University,
St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington
University, St. Louis, Missouri, USA
- Department of Material Science & Mechanical
Engineering, Washington University, St. Louis, Missouri, USA
| | - Rebecca J. Abergel
- Department of Nuclear Engineering, University of
California, Berkeley, California, USA
| | - Tony M. Keaveny
- Department of Mechanical Engineering, University of
California, Berkeley, California, USA
- Department of Bioengineering, University of California,
Berkeley, California, USA
| |
Collapse
|
9
|
Sweeney-Ambros AR, Biggs AE, Zimmerman ND, Mann KA, Damron TA, Oest ME. Orchestrated delivery of PTH [1-34] followed by zoledronic acid prevents radiotherapy-induced bone loss but does not abrogate marrow damage. J Orthop Res 2022; 40:2843-2855. [PMID: 35266584 PMCID: PMC9463412 DOI: 10.1002/jor.25317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 01/14/2022] [Accepted: 03/01/2022] [Indexed: 02/04/2023]
Abstract
Postradiotherapy bone fragility fractures are a frequent late-onset complication in cancer survivors. There is a critical need to develop preventative interventions, and the use of Food and Drug Administration-approved drugs remains an attractive option. Prior data from our lab and others have shown that parathyroid hormone [1-34] mitigates radiotherapy-induced bone loss, but only for the duration of drug delivery. Utilizing a murine hindlimb radiotherapy model, we investigated whether orchestrated delivery of single-dose zoledronic acid could extend these anabolic benefits after cessation of parathyroid hormone delivery. We then explored the potential use of parathyroid hormone as a bone marrow radioprotectant. While the addition of zoledronic acid to parathyroid hormone increased irradiated bone mass, there was no increase in femur bending strength. In this model, the parathyroid hormone was not effective as a marrow radioprotectant, although this could be due to the short course of parathyroid hormone treatment. Marrow repopulation kinetics differed from those in total body irradiation, with hematopoietic stem cell repopulation occurring relatively early at four weeks postirradiation. Furthermore, we found radiation induced a loss of marrow stromal cells and an increase in inflammatory monocytes. Statement of Clinical Significance: Staged delivery of parathyroid hormone and zoledronic acid shows promise as an off-the-shelf intervention to mitigate post-radiotherapy bone damage in cancer patients, but parathyroid hormone is unlikely to function as a broad-spectrum marrow radioprotectant.
Collapse
Affiliation(s)
| | - Amy E Biggs
- Department of Orthopedic Surgery, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Nicholas D Zimmerman
- Department of Orthopedic Surgery, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Kenneth A Mann
- Department of Orthopedic Surgery, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Timothy A Damron
- Department of Orthopedic Surgery, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Megan E Oest
- Department of Orthopedic Surgery, SUNY Upstate Medical University, Syracuse, New York, USA
| |
Collapse
|
10
|
Reddy Padala S, Saikia D, Mikkonen JJW, Uurasjärvi E, Dekker H, Schulten EAJM, Bravenboer N, Koistinen A, Chauhan A, Singh SP, Kullaa AM. Irradiation Induced Biochemical Changes in Human Mandibular Bone: A Raman Spectroscopic Study. APPLIED SPECTROSCOPY 2022; 76:1165-1173. [PMID: 35684992 DOI: 10.1177/00037028221109244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Understanding the biochemical changes in irradiated human mandible after radiotherapy of cancer patients is critical for oral rehabilitation. The underlying mechanism for radiation-associated changes in the bone at the molecular level could lead to implant failure and osteoradionecrosis. The study aimed to assess the chemical composition and bone quality in irradiated human mandibular bone using Raman spectroscopy. A total of 33 bone biopsies from 16 control and 17 irradiated patients were included to quantify different biochemical parameters from the Raman spectra. The differences in bone mineral and matrix band intensities between control and irradiated groups were analyzed using unpaired Student's t-test with statistical significance at p < 0.05. Findings suggest that the intensity of the phosphate band is significantly decreased and the carbonate band is significantly increased in the irradiated group. Further, the mineral crystallinity and carbonate to phosphate ratio are increased. The mineral to matrix ratio is decreased in the irradiated group. Principal component analysis (PCA) based on the local radiation dose and biopsy time interval of irradiated samples did not show any specific classification between irradiation sub-groups. Irradiation disrupted the interaction and bonding between the organic matrix and hydroxyapatite minerals affecting the bone biochemical properties. However, the normal clinical appearance of irradiated bone would have been accompanied by underlying biochemical and microscopical changes which might result in radiation-induced delayed complications.
Collapse
Affiliation(s)
| | - Dimple Saikia
- Department of Bio-Sciences and Bio-Engineering, 477529Indian Institute of Technology, Dharwad, India
| | - Jopi J W Mikkonen
- Institute of Dentistry, University of Eastern Finland, Kuopio, Finland
- SIB Labs, University of Eastern Finland, Kuopio, Finland
| | | | - Hannah Dekker
- Department of Oral and Maxillofacial Surgery/Oral Pathology, Amsterdam UMC and Academic Centre for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam, De Boelelaan, The Netherlands
| | - Engelbert A J M Schulten
- Department of Oral and Maxillofacial Surgery/Oral Pathology, Amsterdam UMC and Academic Centre for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam, De Boelelaan, The Netherlands
| | - Nathalie Bravenboer
- Department of Clinical Chemistry, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan, The Netherlands
- Department of Internal Medicine, Division of Endocrinology and Center for Bone Quality, 4501Leiden University Medical Center, Leiden, The Netherlands
| | - Arto Koistinen
- SIB Labs, University of Eastern Finland, Kuopio, Finland
| | - Amrita Chauhan
- Department of Bio-Sciences and Bio-Engineering, 477529Indian Institute of Technology, Dharwad, India
| | - Surya P Singh
- Department of Bio-Sciences and Bio-Engineering, 477529Indian Institute of Technology, Dharwad, India
| | - Arja M Kullaa
- Institute of Dentistry, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
11
|
Influence of Radiotherapy on Ossification of Vascularized Osseous Reconstruction of the Jaw: A Radiological Retrospective Cohort Study Based on Panoramic Radiographs. J Clin Med 2022; 11:jcm11175041. [PMID: 36078969 PMCID: PMC9456693 DOI: 10.3390/jcm11175041] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/11/2022] [Accepted: 08/20/2022] [Indexed: 11/16/2022] Open
Abstract
Background: The aim of this study was to evaluate the impact of irradiation and time of irradiation on the ossification of jaws reconstructed with free bone grafts. Methods: In total, 100 reconstructions of the jaw were retrospectively evaluated for ossification between bone segments by two raters based on postoperative panoramic radiographs (immediate postOP, approximately 6, 12 and 24 months follow-up). Three subgroups were divided according to the time of irradiation: preoperative radiation therapy (n = 41), postoperative radiation therapy (n = 26) and patients without any radiation therapy (n = 33) as the control group. Ossification time and influencing factors were documented. Results: The fastest ossification with a median of 304 ± 37 days was observed (p < 0.001) in the nonirradiated control group. No significant difference (p = 0.087) in ossification was found between the pre- (447 ± 136 days) and postoperative (510 ± 112 days) radiation groups. Ossification between two graft segments (336 ± 38 days) showed significantly (p < 0.001) faster ossification than between the original and grafted bone (448 ± 85 days). Moreover, closer initial contact between the segments resulted in faster ossification (p < 0.001). When analyzing cofactors, tobacco consumption was the only negative factor aggravating ossification (p = 0.006). Conclusion: Head and neck radiation corresponded with the impaired and prolonged ossification of jaw reconstructions with free bone grafts. There was no difference in ossification if radiotherapy was performed before or after reconstructive surgery. A close bony contact was particularly important for ossification between the original and grafted bone.
Collapse
|
12
|
Dekker H, Schulten EA, Lichters I, van Ruijven L, van Essen HW, Blom GJ, Bloemena E, ten Bruggenkate CM, Kullaa AM, Bravenboer N. Osteocyte Apoptosis, Bone Marrow Adiposity, and Fibrosis in the Irradiated Human Mandible. Adv Radiat Oncol 2022; 7:100951. [PMID: 35662809 PMCID: PMC9156996 DOI: 10.1016/j.adro.2022.100951] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 03/21/2022] [Indexed: 11/30/2022] Open
Abstract
Purpose To assess the effect of radiation therapy on osteocyte apoptosis, osteocyte death, and bone marrow adipocytes in the human mandible and its contribution to the pathophysiology of radiation damage to the mandibular bone. Methods and Materials Mandibular cancellous bone biopsies were taken from irradiated patients and nonirradiated controls. Immunohistochemical detection of cleaved caspase-3 was performed to visualize apoptotic osteocytes. The number of apoptotic osteocytes per bone area and per total amount of osteocytes, osteocytes per bone area, and empty lacunae per bone area were counted manually. The percentage fibrotic tissue and adipose tissue per bone marrow area, the percentage bone marrow of total area, and the mean adipocyte diameter (μm) was determined digitally from adjacent Goldner stained sections. Results Biopsies of 15 irradiated patients (12 men and 3 women) and 7 nonirradiated controls (5 men and 2 women) were assessed. In the study group a significant increase was seen in the number of empty lacunae, the percentage of adipose tissue of bone marrow area, and the adipocyte diameter. There was no significant difference in bone marrow fibrosis nor apoptotic osteocytes between the irradiated group and the controls. Conclusions Irradiation alone does not seem to induce excessive bone marrow fibrosis. The damage to bone mesenchymal stem cells leads to increased marrow adipogenesis and decreased osteoblastogenic potential. Early osteocyte death resulting in avital persisting bone matrix with severely impaired regenerative potential may contribute to the vulnerability of irradiated bone to infection and necrosis.
Collapse
Affiliation(s)
- Hannah Dekker
- Department of Oral and Maxillofacial Surgery/Oral Pathology, Amsterdam University Medical Centers and Academic Centre for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Engelbert A.J.M. Schulten
- Department of Oral and Maxillofacial Surgery/Oral Pathology, Amsterdam University Medical Centers and Academic Centre for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Inez Lichters
- Department of Oral and Maxillofacial Surgery/Oral Pathology, Amsterdam University Medical Centers and Academic Centre for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Leo van Ruijven
- Department of Functional Anatomy, Academic Centre for Dentistry Amsterdam (ACTA), Amsterdam, The Netherlands
| | - Huib W. van Essen
- Department of Clinical Chemistry, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Gerrit-Jan Blom
- Department of Radiotherapy, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Elisabeth Bloemena
- Department of Oral and Maxillofacial Surgery/Oral Pathology, Amsterdam University Medical Centers and Academic Centre for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Chris M. ten Bruggenkate
- Department of Oral and Maxillofacial Surgery/Oral Pathology, Amsterdam University Medical Centers and Academic Centre for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Oral and Maxillofacial Surgery, Alrijne Hospital, Leiderdorp, The Netherlands
| | - Arja M. Kullaa
- Institute of Dentistry, Kuopio Campus, University of Eastern Finland, Kuopio, Finland
- Educational Dental Clinic, Kuopio University Hospital, Kuopio, Finland
| | - Nathalie Bravenboer
- Department of Clinical Chemistry, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
13
|
Systemic Literature Review on Multilevel Analysis of Radiation Effects on Bone Microarchitecture. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9890633. [PMID: 35782085 PMCID: PMC9249517 DOI: 10.1155/2022/9890633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 04/14/2022] [Accepted: 04/27/2022] [Indexed: 11/19/2022]
Abstract
Introduction Modern radiation therapy has become an effective method to treat and monitor tumour growth in cancer patients. It has proved to be a successful way to minimise mortality rates. However, the adverse effects of radiation have been historical evidence in the clinical environment involving diminishing the quality and density of bone and causing fragility fracture to the bone in the long run. This systematic review was aimed at identifying and evaluating the effects of irradiation on morphology and mechanical properties of murine model bone in previous publications. Methods A systematic literature review was undertaken following the Preferred Reporting Items for Systemic Reviews and Meta-analysis (PRISMA) guidelines. A comprehensive literature search was performed using Scopus, Web of Science, and Science Direct databases (English only studies published between 2015 and 2020). The selected studies were evaluated according to three criteria: (1) criteria for study sample selection; (2) criteria for methodological procedures; and (3) criteria for detection and evaluation. Results The initial search strategy identified 1408 related studies, 8 of were included based on inclusion and exclusion criteria. This review revealed an association between bone destruction and the magnitude of time and dose postirradiation. We agreed that the effect of radiation on bone morphology and strength primarily is a later stage event but noticeable in both low (1 Gy) and high dose (30 Gy) radiation. Trabecular and cortical bone microstructures were significantly altered at irradiation and contralateral sites. Besides, the mechanical strength was significantly impacted in both shorter and longer periods. Conclusion Overall, the radiotherapy altered bone microstructures and substantially decreases bone mechanical properties. The alteration was related to quantity and the activity of the osteoblast and osteoclast. Early detection of those most at risk for radiation-induced bone alterations could lead to better prophylactic intervention decisions.
Collapse
|
14
|
Eckert D, Rapp F, Tsedeke AT, Kraft D, Wente I, Molendowska J, Basheer S, Langhans M, Meckel T, Friedrich T, Donaubauer AJ, Becker I, Frey B, Fournier C. Modulation of Differentiation and Bone Resorbing Activity of Human (Pre-) Osteoclasts After X-Ray Exposure. Front Immunol 2022; 13:817281. [PMID: 35603191 PMCID: PMC9116137 DOI: 10.3389/fimmu.2022.817281] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 04/08/2022] [Indexed: 11/13/2022] Open
Abstract
Low-dose radiotherapy (LD-RT) is a local treatment option for patients with chronic degenerative and inflammatory diseases, in particular musculoskeletal diseases. Despite reported analgesic and anti-inflammatory effects, cellular and molecular mechanisms related to osteoimmunological effects are still elusive. Here we test the hypothesis that X-irradiation inhibits the differentiation of precursor osteoclasts into mature osteoclasts (mOC) and their bone resorbing activity. Circulating monocytes from healthy donors were isolated and irradiated after attachment with single or fractionated X-ray doses, comparable to an LD-RT treatment scheme. Then monocytes underwent ex vivo differentiation into OC during cultivation up to 21 days, under conditions mimicking the physiological microenvironment of OC on bone. After irradiation, apoptotic frequencies were low, but the total number of OC precursors and mOC decreased up to the end of the cultivation period. On top, we observed an impairment of terminal differentiation, i.e. a smaller fraction of mOC, reduced resorbing activity on bone, and release of collagen fragments. We further analyzed the effect of X-irradiation on multinucleation, resulting from the fusion of precursor OC, which occurs late during OC differentiation. At 21 days after exposure, the observation of smaller cellular areas and a reduced number of nuclei per mOC suggest an impaired fusion of OC precursors to form mOC. Before, at 14 days, the nuclear translocation of Nuclear Factor Of Activated T Cells 1 (NFATc1), a master regulator of osteoclast differentiation and fusion, was decreased. In first results, obtained in the frame of a longitudinal LD-RT study, we previously reported a pain-relieving effect in patients. However, in a subgroup of patients suffering from Calcaneodynia or Achillodynia, we did not observe a consistent decrease of established blood markers for resorption and formation of bone, or modified T cell subtypes involved in regulating these processes. To assess the relevance of changes in bone metabolism for other diseases treated with LD-RT will be subject of further studies. Taken together, we observed that in vitro X-irradiation of monocytes results in an inhibition of the differentiation into bone-resorbing OC and a concomitant reduction of resorbing activity. The detected reduced NFATc1 signaling could be one underlying mechanism.
Collapse
Affiliation(s)
- Denise Eckert
- Department of Biophysics, Gesellschaft für Schwerionenforschung (GSI) Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Felicitas Rapp
- Department of Biophysics, Gesellschaft für Schwerionenforschung (GSI) Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Ayele Taddese Tsedeke
- Department of Biophysics, Gesellschaft für Schwerionenforschung (GSI) Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Daniela Kraft
- Department of Biophysics, Gesellschaft für Schwerionenforschung (GSI) Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Isabell Wente
- Department of Biophysics, Gesellschaft für Schwerionenforschung (GSI) Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Jessica Molendowska
- Department of Biophysics, Gesellschaft für Schwerionenforschung (GSI) Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Sidra Basheer
- Department of Biophysics, Gesellschaft für Schwerionenforschung (GSI) Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Markus Langhans
- Department of Macromolecular and Paper Chemistry and Membrane Dynamics, Technical University Darmstadt, Darmstadt, Germany
| | - Tobias Meckel
- Department of Macromolecular and Paper Chemistry and Membrane Dynamics, Technical University Darmstadt, Darmstadt, Germany
| | - Thomas Friedrich
- Department of Biophysics, Gesellschaft für Schwerionenforschung (GSI) Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Anna-Jasmina Donaubauer
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Ina Becker
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Benjamin Frey
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Claudia Fournier
- Department of Biophysics, Gesellschaft für Schwerionenforschung (GSI) Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| |
Collapse
|
15
|
Chandra A, Lagnado AB, Farr JN, Doolittle M, Tchkonia T, Kirkland JL, LeBrasseur NK, Robbins PD, Niedernhofer LJ, Ikeno Y, Passos JF, Monroe DG, Pignolo RJ, Khosla S. Targeted clearance of p21- but not p16-positive senescent cells prevents radiation-induced osteoporosis and increased marrow adiposity. Aging Cell 2022; 21:e13602. [PMID: 35363946 PMCID: PMC9124310 DOI: 10.1111/acel.13602] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/09/2022] [Accepted: 03/20/2022] [Indexed: 12/14/2022] Open
Abstract
Cellular senescence, which is a major cause of tissue dysfunction with aging and multiple other conditions, is known to be triggered by p16Ink4a or p21Cip1 , but the relative contributions of each pathway toward inducing senescence are unclear. Here, we directly addressed this issue by first developing and validating a p21-ATTAC mouse with the p21Cip1 promoter driving a "suicide" transgene encoding an inducible caspase-8 which, upon induction, selectively kills p21Cip1 -expressing senescent cells. Next, we used the p21-ATTAC mouse and the established p16-INK-ATTAC mouse to directly compare the contributions of p21Cip1 versus p16Ink4a in driving cellular senescence in a condition where a tissue phenotype (bone loss and increased marrow adiposity) is clearly driven by cellular senescence-specifically, radiation-induced osteoporosis. Using RNA in situ hybridization, we confirmed the reduction in radiation-induced p21Cip1 - or p16Ink4a -driven transcripts following senescent cell clearance in both models. However, only clearance of p21Cip1 +, but not p16Ink4a +, senescent cells prevented both radiation-induced osteoporosis and increased marrow adiposity. Reduction in senescent cells with dysfunctional telomeres following clearance of p21Cip1 +, but not p16Ink4a +, senescent cells also reduced several of the radiation-induced pro-inflammatory senescence-associated secretory phenotype factors. Thus, by directly comparing senescent cell clearance using two parallel genetic models, we demonstrate that radiation-induced osteoporosis is driven predominantly by p21Cip1 - rather than p16Ink4a -mediated cellular senescence. Further, this approach can be used to dissect the contributions of these pathways in other senescence-associated conditions, including aging across tissues.
Collapse
Affiliation(s)
- Abhishek Chandra
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
| | - Anthony B. Lagnado
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
| | - Joshua N. Farr
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
| | - Madison Doolittle
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
| | - Tamara Tchkonia
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
| | - James L. Kirkland
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
| | - Nathan K. LeBrasseur
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
- Department of Physical Medicine and RehabilitationMayo ClinicRochesterMinnesotaUSA
| | - Paul D. Robbins
- Institute on the Biology of Aging and MetabolismDepartment of Biochemistry, Molecular Biology and BiophysicsUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Laura J. Niedernhofer
- Institute on the Biology of Aging and MetabolismDepartment of Biochemistry, Molecular Biology and BiophysicsUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Yuji Ikeno
- Department of Pathology and Laboratory MedicineUniversity of Texas Health Science CenterSan AntonioTexasUSA
| | - João F. Passos
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
| | - David G. Monroe
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
| | - Robert J. Pignolo
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
| | - Sundeep Khosla
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
- Division of EndocrinologyMayo ClinicRochesterMinnesotaUSA
| |
Collapse
|
16
|
Sahbani K, Cardozo CP, Bauman WA, Tawfeek HA. Inhibition of TGF-β Signaling Attenuates Disuse-induced Trabecular Bone Loss After Spinal Cord Injury in Male Mice. Endocrinology 2022; 163:bqab230. [PMID: 34791098 DOI: 10.1210/endocr/bqab230] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Indexed: 11/19/2022]
Abstract
Bone loss is one of the most common complications of immobilization after spinal cord injury (SCI). Whether transforming growth factor (TGF)-β signaling plays a role in SCI-induced disuse bone loss has not been determined. Thus, 16-week-old male mice underwent sham or spinal cord contusion injury to cause complete hindlimb paralysis. Five days later, 10 mg/kg/day control (IgG) or anti-TGF-β1,2,3 neutralizing antibody (1D11) was administered twice weekly for 4 weeks. Femurs were examined by micro-computed tomography (micro-CT) scanning and histology. Bone marrow (BM) supernatants were analyzed by enzyme-linked immunosorbent assay for levels of procollagen type 1 intact N-terminal propeptide (P1NP), tartrate-resistant acid phosphatase (TRAcP-5b), receptor activator of nuclear factor-kappa B ligand (RANKL), osteoprotegerin (OPG), and prostaglandin E2 (PGE2). Distal femoral micro-CT analysis showed that SCI-1D11 mice had significantly (P < .05) attenuated loss of trabecular fractional bone volume (123% SCI-1D11 vs 69% SCI-IgG), thickness (98% vs 81%), and connectivity (112% vs 69%) and improved the structure model index (2.1 vs 2.7). Histomorphometry analysis revealed that osteoclast numbers were lower in the SCI-IgG mice than in sham-IgG control. Biochemically, SCI-IgG mice had higher levels of P1NP and PGE2 but similar TRAcP-5b and RANKL/OPG ratio to the sham-IgG group. The SCI-1D11 group exhibited higher levels of P1NP but similar TRAcP-5b, RANKL/OPG ratio, and PGE2 to the sham-1D11 group. Furthermore, 1D11 treatment prevented SCI-induced hyperphosphorylation of tau protein in osteocytes, an event that destabilizes the cytoskeleton. Together, inhibition of TGF-β signaling after SCI protects trabecular bone integrity, likely by balancing bone remodeling, inhibiting PGE2 elevation, and preserving the osteocyte cytoskeleton.
Collapse
Affiliation(s)
- Karim Sahbani
- National Center for the Medical Consequences of Spinal Cord Injury, James J Peters Veterans Affairs Medical Center, Bronx, NY 10468, USA
- Bronx Veterans Medical Research Foundation, Bronx, NY 10468, USA
| | - Christopher P Cardozo
- National Center for the Medical Consequences of Spinal Cord Injury, James J Peters Veterans Affairs Medical Center, Bronx, NY 10468, USA
- Bronx Veterans Medical Research Foundation, Bronx, NY 10468, USA
- Department of Medicine, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Rehabilitation Medicine and Human Performance, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mount Sinai Institute for Systems Biomedicine, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - William A Bauman
- National Center for the Medical Consequences of Spinal Cord Injury, James J Peters Veterans Affairs Medical Center, Bronx, NY 10468, USA
- Bronx Veterans Medical Research Foundation, Bronx, NY 10468, USA
- Department of Medicine, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Rehabilitation Medicine and Human Performance, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mount Sinai Institute for Systems Biomedicine, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Hesham A Tawfeek
- National Center for the Medical Consequences of Spinal Cord Injury, James J Peters Veterans Affairs Medical Center, Bronx, NY 10468, USA
- Bronx Veterans Medical Research Foundation, Bronx, NY 10468, USA
- Department of Medicine, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
17
|
Pendleton MM, Emerzian SR, Sadoughi S, Li A, Liu JW, Tang SY, O'Connell GD, Sibonga JD, Alwood JS, Keaveny TM. Relations Between Bone Quantity, Microarchitecture, and Collagen Cross-links on Mechanics Following In Vivo Irradiation in Mice. JBMR Plus 2021; 5:e10545. [PMID: 34761148 PMCID: PMC8567491 DOI: 10.1002/jbm4.10545] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 08/20/2021] [Indexed: 01/22/2023] Open
Abstract
Humans are exposed to ionizing radiation via spaceflight or cancer radiotherapy, and exposure from radiotherapy is known to increase risk of skeletal fractures. Although irradiation can reduce trabecular bone mass, alter trabecular microarchitecture, and increase collagen cross‐linking, the relative contributions of these effects to any loss of mechanical integrity remain unclear. To provide insight, while addressing both the monotonic strength and cyclic‐loading fatigue life, we conducted total‐body, acute, gamma‐irradiation experiments on skeletally mature (17‐week‐old) C57BL/6J male mice (n = 84). Mice were administered doses of either 0 Gy (sham), 1 Gy (motivated by cumulative exposures from a Mars mission), or 5 Gy (motivated by clinical therapy regimens) with retrieval of the lumbar vertebrae at either a short‐term (11‐day) or long‐term (12‐week) time point after exposure. Micro‐computed tomography was used to assess trabecular and cortical quantity and architecture, biochemical composition assays were used to assess collagen quality, and mechanical testing was performed to evaluate vertebral compressive strength and fatigue life. At 11 days post‐exposure, 5 Gy irradiation significantly reduced trabecular mass (p < 0.001), altered microarchitecture (eg, connectivity density p < 0.001), and increased collagen cross‐links (p < 0.001). Despite these changes, vertebral strength (p = 0.745) and fatigue life (p = 0.332) remained unaltered. At 12 weeks after 5 Gy exposure, the trends in trabecular bone persisted; in addition, regardless of irradiation, cortical thickness (p < 0.01) and fatigue life (p < 0.01) decreased. These results demonstrate that the highly significant effects of 5 Gy total‐body irradiation on the trabecular bone morphology and collagen cross‐links did not translate into detectable effects on vertebral mechanics. The only mechanical deficits observed were associated with aging. Together, these vertebral results suggest that for spaceflight, irradiation alone will likely not alter failure properties, and for radiotherapy, more investigations that include post‐exposure time as a positive control and testing of both failure modalities are needed to determine the cause of increased fracture risk. © 2021 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research. This article has been contributed to by US Government employees and their work is in the public domain in the USA.
Collapse
Affiliation(s)
- Megan M Pendleton
- Department of Mechanical Engineering University of California Berkeley CA USA
| | - Shannon R Emerzian
- Department of Mechanical Engineering University of California Berkeley CA USA
| | - Saghi Sadoughi
- Department of Mechanical Engineering University of California Berkeley CA USA
| | - Alfred Li
- Endocrine Research Unit University of California and Veteran Affairs Medical Center San Francisco CA USA
| | - Jennifer W Liu
- Department of Orthopaedic Surgery Washington University St. Louis MO USA
| | - Simon Y Tang
- Department of Orthopaedic Surgery Washington University St. Louis MO USA.,Department of Biomedical Engineering Washington University St. Louis MO USA.,Department of Mechanical Engineering and Materials Science Washington University St. Louis MO USA
| | - Grace D O'Connell
- Department of Mechanical Engineering University of California Berkeley CA USA.,Department of Orthopaedic Surgery University of California San Francisco CA USA
| | - Jean D Sibonga
- Biomedical Research and Environmental Sciences Division NASA Johnson Space Center Houston TX USA
| | - Joshua S Alwood
- Space Biosciences Division NASA Ames Research Center Moffett Field CA USA
| | - Tony M Keaveny
- Department of Mechanical Engineering University of California Berkeley CA USA.,Department of Bioengineering University of California Berkeley CA USA
| |
Collapse
|
18
|
Sweeney-Ambros AR, Nappi AN, Oest ME. In Vitro Radiosensitivity of Murine Marrow Stromal Cells Varies Across Donor Strains. Radiat Res 2021; 195:590-595. [PMID: 33826738 DOI: 10.1667/rade-20-00020.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 03/11/2021] [Indexed: 11/03/2022]
Abstract
Mouse models are widely used in the study of musculoskeletal radiobiology both in vivo and in vitro. Two of the most commonly used mouse strains are C57BL/6 and BALB/c. However, little is known about their equivalence in response to ionizing radiation. In this study we compare the responses of marrow stromal cells derived from both of these strains to X rays in vitro at passages 0 and 2. Colony-forming efficiency was significantly higher in BALB/c marrow stromal cells at passage 0. Radiation-induced decreases in colony-forming unit (CFU) formation at passage 0 were comparable across both strains at 0-2 Gy, but BALB/c stromal cells were more radiosensitive than C57BL/6 stromal cells at 3-7 Gy. Osteogenic differentiation at passage 2 was not affected by radiation for either strain. This work demonstrates that commonly used inbred mouse strains differ in their early-passage marrow stromal cell responses to X rays, including self-renewal and differentiation potential. This variability is an important point to consider when selecting an animal model for in vivo or in vitro study.
Collapse
Affiliation(s)
| | - Alexander N Nappi
- Department of Orthopedic Surgery, SUNY Upstate Medical University, Syracuse, New York
| | - Megan E Oest
- Department of Orthopedic Surgery, SUNY Upstate Medical University, Syracuse, New York
| |
Collapse
|
19
|
Bartlow CM, Mann KA, Damron TA, Oest ME. Altered mechanical behavior of demineralized bone following therapeutic radiation. J Orthop Res 2021; 39:750-760. [PMID: 32965711 PMCID: PMC8212945 DOI: 10.1002/jor.24868] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 07/15/2020] [Accepted: 09/21/2020] [Indexed: 02/04/2023]
Abstract
Post-radiotherapy (RTx) bone fragility fractures are a late-onset complication occurring in bone within or underlying the radiation field. These fractures are difficult to predict, as patients do not present with local osteopenia. Using a murine hindlimb RTx model, we previously documented decreased mineralized bone strength and fracture toughness, but alterations in material properties of the organic bone matrix are largely unknown. In this study, 4 days of fractionated hindlimb irradiation (4 × 5 Gy) or Sham irradiation was administered in a mouse model (BALB/cJ, end points: 0, 4, 8, and 12 weeks, n = 15/group/end point). Following demineralization, the viscoelastic stress relaxation, and monotonic tensile mechanical properties of tibiae were determined. Irradiated tibiae demonstrated an immediate (day after last radiation fraction) and sustained (4, 8, 12 weeks) increase in stress relaxation compared to the Sham group, with a 4.4% decrease in equilibrium stress (p < .017). While tensile strength was not different between groups, irradiated tibiae had a lower elastic modulus (-5%, p = .027) and energy to failure (-12.2%, p = .012) with monotonic loading. Gel electrophoresis showed that therapeutic irradiation (4 × 5 Gy) does not result in collagen fragmentation, while irradiation at a common sterilization dose (25 kGy) extensively fragmented collagen. These results suggest that altered collagen mechanical behavior has a role in postirradiation bone fragility, but this can occur without detectable collagen fragmentation. Statement of Clinical Significance: Therapeutic irradiation alters bone organic matrix mechanics and which contribute to diminished fatigue strength, but this does not occur via collagen fragmentation.
Collapse
Affiliation(s)
- Christopher M. Bartlow
- Department of Orthopedic Surgery State University of New York Upstate Medical University Syracuse New York USA
| | - Kenneth A. Mann
- Department of Orthopedic Surgery State University of New York Upstate Medical University Syracuse New York USA
| | - Timothy A. Damron
- Department of Orthopedic Surgery State University of New York Upstate Medical University Syracuse New York USA
| | - Megan E. Oest
- Department of Orthopedic Surgery State University of New York Upstate Medical University Syracuse New York USA
| |
Collapse
|
20
|
Postradiation Fractures after Combined Modality Treatment in Extremity Soft Tissue Sarcomas. Sarcoma 2021; 2021:8877567. [PMID: 33790687 PMCID: PMC7984930 DOI: 10.1155/2021/8877567] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 01/20/2021] [Accepted: 02/27/2021] [Indexed: 12/26/2022] Open
Abstract
Soft tissue sarcoma (STS) of the extremities is typically treated with limb-sparing surgery and radiation therapy; with this treatment approach, high local control rates can be achieved. However, postradiation bone fractures, fractures occurring in the prior radiation field with minimal or no trauma, are a serious late complication that occurs in 2–22% of patients who receive surgery and radiation for STS. Multiple risk factors for sustaining a postradiation fracture exist, including high radiation dose, female sex, periosteal stripping, older age, femur location, and chemotherapy administration. The treatment of these pathological fractures can be difficult, with complications including delayed union, nonunion, and infection posing particular challenges. Here, we review the mechanisms, risk factors, and treatment challenges associated with postradiation fractures in STS patients.
Collapse
|
21
|
Damron TA. CORR Insights®: Radiation Disrupts Protective Function of the Spinal Meninges in a Mouse Model of Tumor-induced Spinal Cord Compression. Clin Orthop Relat Res 2021; 479:177-179. [PMID: 33165040 PMCID: PMC7899582 DOI: 10.1097/corr.0000000000001544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 10/01/2020] [Indexed: 01/31/2023]
Affiliation(s)
- Timothy A Damron
- T. A. Damron, Institute for Human Performance, Musculoskeletal Science Research Center, Syracuse, NY, USA
| |
Collapse
|
22
|
Duan J, Yang Y, Zhang E, Wang H. Co-Cr-Mo-Cu alloys for clinical implants with osteogenic effect by increasing bone induction, formation and development in a rabbit model. BURNS & TRAUMA 2020; 8:tkaa036. [PMID: 33376752 PMCID: PMC7750714 DOI: 10.1093/burnst/tkaa036] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 02/12/2020] [Accepted: 08/17/2020] [Indexed: 12/12/2022]
Abstract
Background Co-Cr-Mo alloy has been widely used in clinical implants because of its excellent mechanical and anti-corrosion properties, but there is an urgent need to address its disadvantages, such as implant-related infections and implant loosening. We synthesized Co-Cr-Mo-Cu (Co-Cu) alloys with different Cu contents to modify implant performance to be suitable as a bone-compatible implant material. Methods Microstructure, phase content and mechanical properties of the Co-Cr-Mo alloy were characterized. Histological and immunohistochemical analyses were performed after implantation in rabbits. The experimental alloy was implanted on the lateral side of the lower tibial condyle and the tibial nodule. Results Phase content and mechanical properties revealed that the crystallographic structure and wear resistance were changed. Experimental implantation results demonstrated that osteogenic capability was markedly enhanced, ascribed to the excellent antibacterial and osseointegration capacities of Cu phases, and with the release of Cu ions. In particular, Co-Cu alloy containing 2 wt% Cu exhibited the best osteogenic performance among all samples. Conclusions The results indicated that osteogenic performance of the Co-Cr-Mo alloy could be enhanced by adding Cu. In particular, the Co-2Cu alloy exhibited the best properties according to both immunohistochemical and histological analyses. Our study not only provides deep insight into the osteogenic effect of Cu but presents a new Co-Cu alloy for clinical implants.
Collapse
Affiliation(s)
- Jingzhu Duan
- Department of Orthopaedic, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, China
| | - Yang Yang
- Department of Ophthalmology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, China
| | - Erlin Zhang
- Key Laboratory for Anisotropy and Texture of Materials, Education Ministry of China, School of Materials Science and Engineering, Northeastern University, No. 3-11 Wenhua Road, Heping District, Shenyang 110819, China
| | - Huan Wang
- Department of Orthopaedic, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, China
| |
Collapse
|
23
|
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
|
24
|
Chandra A, Lagnado AB, Farr JN, Monroe DG, Park S, Hachfeld C, Tchkonia T, Kirkland JL, Khosla S, Passos JF, Pignolo RJ. Targeted Reduction of Senescent Cell Burden Alleviates Focal Radiotherapy-Related Bone Loss. J Bone Miner Res 2020; 35:1119-1131. [PMID: 32023351 PMCID: PMC7357625 DOI: 10.1002/jbmr.3978] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 01/18/2020] [Accepted: 01/29/2020] [Indexed: 12/11/2022]
Abstract
Clinical radiotherapy treats life-threatening cancers, but the radiation often affects neighboring normal tissues including bone. Acute effects of ionizing radiation include oxidative stress, DNA damage, and cellular apoptosis. We show in this study that a large proportion of bone marrow cells, osteoblasts, and matrix-embedded osteocytes recover from these insults only to attain a senescent profile. Bone analyses of senescence-associated genes, senescence-associated beta-galactosidase (SA-β-gal) activity, and presence of telomere dysfunction-induced foci (TIF) at 1, 7, 14, 21, and 42 days post-focal radiation treatment (FRT) in C57BL/6 male mice confirmed the development of senescent cells and the senescence-associated secretory phenotype (SASP). Accumulation of senescent cells and SASP markers were correlated with a significant reduction in bone architecture at 42 days post-FRT. To test if senolytic drugs, which clear senescent cells, alleviate FRT-related bone damage, we administered the senolytic agents, dasatinib (D), quercetin (Q), fisetin (F), and a cocktail of D and Q (D+Q). We found moderate alleviation of radiation-induced bone damage with D and Q as stand-alone compounds, but no such improvement was seen with F. However, the senolytic cocktail of D+Q reduced senescent cell burden as assessed by TIF+ osteoblasts and osteocytes, markers of senescence (p16 Ink4a and p21), and key SASP factors, resulting in significant recovery in the bone architecture of radiated femurs. In summary, this study provides proof of concept that senescent cells play a role in radiotherapy-associated bone damage, and that reduction in senescent cell burden by senolytic agents is a potential therapeutic option for alleviating radiotherapy-related bone deterioration. © 2020 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Abhishek Chandra
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Rochester, MN, USA.,Department of Medicine, Division of Geriatric Medicine and Gerontology, Mayo Clinic College of Medicine, Rochester, MN, USA.,Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Anthony B Lagnado
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Rochester, MN, USA.,Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Joshua N Farr
- Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, MN, USA.,Division of Endocrinology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - David G Monroe
- Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, MN, USA.,Division of Endocrinology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Sean Park
- Department of Radiation Oncology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Christine Hachfeld
- Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - James L Kirkland
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Rochester, MN, USA.,Department of Medicine, Division of Geriatric Medicine and Gerontology, Mayo Clinic College of Medicine, Rochester, MN, USA.,Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Sundeep Khosla
- Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, MN, USA.,Division of Endocrinology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - João F Passos
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Rochester, MN, USA.,Department of Medicine, Division of Geriatric Medicine and Gerontology, Mayo Clinic College of Medicine, Rochester, MN, USA.,Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Robert J Pignolo
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Rochester, MN, USA.,Department of Medicine, Division of Geriatric Medicine and Gerontology, Mayo Clinic College of Medicine, Rochester, MN, USA.,Robert and Arlene Kogod Center on Aging, Mayo Clinic College of Medicine, Rochester, MN, USA.,Division of Endocrinology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| |
Collapse
|
25
|
Mandair GS, Oest ME, Mann KA, Morris MD, Damron TA, Kohn DH. Radiation-induced changes to bone composition extend beyond periosteal bone. Bone Rep 2020; 12:100262. [PMID: 32258252 PMCID: PMC7125315 DOI: 10.1016/j.bonr.2020.100262] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 03/25/2020] [Accepted: 03/26/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Cancer patients receiving radiotherapy for soft tissue sarcomas are often at risk of post-irradiation (post-RTx) bone fragility fractures, but our understanding of factors controlling radiation-induced bone injury is limited. Previous studies have evaluated post-RTx changes to cortical bone composition in the periosteum of irradiated tibiae, but have not evaluated effects of irradiation in deeper tissues, such as endosteal or mid-cortical bone, and whether there are differential spatial effects of irradiation. In this study, we hypothesize that post-RTx changes to cortical bone composition are greater in endosteal compared to mid-cortical or periosteal bone. METHODS A pre-clinical mouse model of limited field hindlimb irradiation was used to evaluate spatial and temporal post-RTx changes to the metaphyseal cortex of irradiated tibiae. Irradiation was delivered unilaterally to the hindlimbs of 12-wk old female BALB/cJ mice as 4 consecutive daily doses of 5 Gy each. RTx and non-RTx tibiae were obtained at 0, 2, 4, 8, and 12 wks post-RTx (n = 9 mice/group/time). Raman spectroscopy was used to evaluate spatial and temporal post-RTx changes to cortical bone composition in age-matched RTx and non-RTx groups. RESULTS Significant early spatial differences in mineral/matrix and collagen crosslink ratios were found between endosteal and periosteal or mid-cortical bone at 2-wks post-RTx. Although spatial differences were transient, mineral/matrix ratios significantly decreased and collagen crosslink ratios significantly increased with post-RTx time throughout the entire tibial metaphyseal cortex. CONCLUSIONS Irradiation negatively impacts the composition of cortical bone in a spatially-dependent manner starting as early as 2-wks post-RTx. Long-term progressive post-RTx changes across all cortical bone sites may eventually contribute to the increased risk of post-RTx bone fragility fractures.
Collapse
Affiliation(s)
| | - Megan E. Oest
- Department of Orthopedic Surgery, Upstate Medical University, Syracuse, NY, USA
| | - Kenneth A. Mann
- Department of Orthopedic Surgery, Upstate Medical University, Syracuse, NY, USA
| | | | - Timothy A. Damron
- Department of Orthopedic Surgery, Upstate Medical University, Syracuse, NY, USA
| | - David H. Kohn
- School of Dentistry, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, USA
| |
Collapse
|
26
|
Farris MK, Helis CA, Hughes RT, LeCompte MC, Borg AM, Nieto K, Munley MT, Willey JS. Bench to Bedside: Animal Models of Radiation Induced Musculoskeletal Toxicity. Cancers (Basel) 2020; 12:cancers12020427. [PMID: 32059447 PMCID: PMC7073177 DOI: 10.3390/cancers12020427] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/04/2020] [Accepted: 02/05/2020] [Indexed: 12/29/2022] Open
Abstract
Ionizing radiation is a critical aspect of current cancer therapy. While classically mature bone was thought to be relatively radio-resistant, more recent data have shown this to not be the case. Radiation therapy (RT)-induced bone loss leading to fracture is a source of substantial morbidity. The mechanisms of RT likely involve multiple pathways, including changes in angiogenesis and bone vasculature, osteoblast damage/suppression, and increased osteoclast activity. The majority of bone loss appears to occur rapidly after exposure to ionizing RT, with significant changes in cortical thickness being detectable on computed tomography (CT) within three to four months. Additionally, there is a dose–response relationship. Cortical thinning is especially notable in areas of bone that receive >40 gray (Gy). Methods to mitigate toxicity due to RT-induced bone loss is an area of active investigation. There is an accruing clinical trial investigating the use of risderonate, a bisphosphonate, to prevent rib bone loss in patients undergoing lung stereotactic body radiation therapy (SBRT). Additionally, several other promising therapeutic/preventative approaches are being explored in preclinical studies, including parathyroid hormone (PTH), amifostine, and mechanical loading of irradiated bones.
Collapse
|
27
|
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: 0.6] [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
|
28
|
Effects of ionizing radiation on woven bone: influence on the osteocyte lacunar network, collagen maturation, and microarchitecture. Clin Oral Investig 2019; 24:2763-2771. [PMID: 31732880 DOI: 10.1007/s00784-019-03138-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 10/24/2019] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Evaluate the effects of ionizing radiation on microarchitecture, the osteocyte lacunar network, and collagen maturity in a bone repair site. MATERIALS AND METHODS Bone defects were created on tibias of 20 New Zealand rabbits. After 2 weeks, the animals were randomly divided into (n = 10) NoIr (nonirradiated group) and Ir (irradiated group). In the Ir, the animals received single-dose irradiation of 30 Gy on the tibia and were euthanized after 2 weeks. Bone microarchitecture parameters were analyzed by using micro-CT, and the osteocyte lacunar network, bone matrix, and collagen maturation by histomorphometric analysis. The data were analyzed using unpaired Student's t test (α = 0.05). RESULTS Trabecular thickness in Ir was lower than that in NoIr (P = 0.028). No difference was found for bone volume fraction and bone area. Lacunae filled with osteocytes were more numerous (P < 0.0001) in NoIr (2.6 ± 0.6) than in Ir (1.97 ± 0.53). Empty lacunae were more prevalent (P < 0.003) in Ir (0.14 ± 0.10) than in NoIr (0.1 ± 0.1). The mean osteocyte lacunae size was higher (P < 0.01) in Ir (15.4 ± 4.41) than in NoIr (12.7 ± 3.7). Picrosirius red analysis showed more (P < 0.05) mature collagen in NoIr (29.0 ± 5.3) than in Ir (23.4 ± 4.5). Immature collagen quantification revealed no difference between groups. CONCLUSIONS Ionizing radiation compromised bone formation and an impairment in bone repair in irradiated woven bone was observed. CLINICAL RELEVANCE Before radiotherapy, patients usually need surgical intervention, which may be better performed, if clinicians understand the repair process in irradiated bone, using novel approaches for treating these individuals.
Collapse
|
29
|
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: 1.8] [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
|
30
|
Chandra A, Park SS, Pignolo RJ. Potential role of senescence in radiation-induced damage of the aged skeleton. Bone 2019; 120:423-431. [PMID: 30543989 DOI: 10.1016/j.bone.2018.12.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 12/04/2018] [Accepted: 12/08/2018] [Indexed: 12/21/2022]
Abstract
Human aging-related changes are exacerbated in cases of disease and cancer, and conversely aging is a catalyst for the occurrence of disease and multimorbidity. For example, old age is the most significant risk factor for cancer and among people who suffer from cancer, >60% are above the age of 65. Oxidative stress and DNA damage, leading to genomic instability and telomere dysfunction, are prevalent in aging and radiation-induced damage and are major cellular events that lead to senescence. Human exposures from nuclear fallout, cosmic radiation and clinical radiotherapy (RT) are some common sources of irradiation that affect bone tissue. RT has been used to treat malignant tumors for over a century, but the effects of radiation damage on tumor-adjacent normal tissue has largely been overlooked. There is an increase in the percent survivorship among patients post-RT, and it is in older survivors where the deleterious synergy between aging and radiation exposure conspires to promote tissue deterioration and dysfunction which then negatively impacts their quality of life. Thus, an aging skeleton is already pre-disposed to architectural deterioration, which is further worsened by radiation-induced bone damage. Effects of senescence and the senescence associated secretory phenotype (SASP) have been implicated in age-associated bone loss, but their roles in radiation-associated bone damage are still elusive. RT is used in treatment for a variety of cancers and in different anatomical locations, the sequelae of which include long-term morbidity and lifelong discomfort. Therefore, consideration of the growing evidence that implicates the role of senescence in radiation-induced bone damage argues in favor of exploiting current senotherapeutic approaches as a possible prevention or treatment.
Collapse
Affiliation(s)
- Abhishek Chandra
- Department of Medicine, Mayo Clinic School of Medicine, Mayo Clinic, Rochester, MN, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic School of Medicine, Mayo Clinic, Rochester, MN, USA.
| | - Sean S Park
- Department of Radiation Oncology, Mayo Clinic School of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Robert J Pignolo
- Department of Medicine, Mayo Clinic School of Medicine, Mayo Clinic, Rochester, MN, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic School of Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
31
|
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: 4.0] [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
|
32
|
Soares PBF, Soares CJ, Limirio PHJO, de Jesus RNR, Dechichi P, Spin-Neto R, Zanetta-Barbosa D. Effect of ionizing radiation after-therapy interval on bone: histomorphometric and biomechanical characteristics. Clin Oral Investig 2018; 23:2785-2793. [PMID: 30368663 DOI: 10.1007/s00784-018-2724-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 10/18/2018] [Indexed: 01/06/2023]
Abstract
OBJECTIVES This study aimed to evaluate the effects of radiotherapy on biomechanical, histomorphometric, and microstructural characteristics of bone, in diverse periods, compared with intact bone tissue. MATERIALS AND METHODS Eighteen adult male New Zealand rabbits were treated with a single radiation dose of 30 Gy. The animals were randomly divided into six groups: NoIr, control group, no radiation, and five irradiated groups sacrificed after 24 h (Ir24h), 7 (Ir7d), 14 (Ir14d), 21 (Ir21d), and 28 (Ir28d) days. After these periods, the animals were sacrificed and their tibias (n = 6) evaluated using three-point bending test to calculate the ultimate force, work to failure, and bone stiffness. Dynamic indentation test was used to quantify Vickers hardness and elasticity modulus of bone tissue. Micro-CT was used to analyze the cortical volume (CtV), cortical thickness (CtTh), and porosity (Ct.Po). Histomorphometric assessment was based on the lacunarity of bone tissue. Data were analyzed using one-way ANOVA and Kruskal-Wallis tests followed by Tukey, Dunnet, and Dunn's post-tests (P < 0.05). RESULTS The ultimate force, work to failure, stiffness, elastic modulus, and Vickers hardness values of irradiated bone were significantly lower that non-irradiated bone. Irradiated bone showed significantly lower CtTh and CtV values and higher CtPo than non-irradiated bone. No significant difference was found for lacunarity between non-irradiated bone and irradiated bone. CONCLUSIONS Ionizing radiation decreases normal anisotropy on microarchitecture of cortical bone, and increases bone fragility compared with non-irradiated bone. Further, these changes were seen after longer periods (e.g., 14 and 21 days), and not immediately after radiation therapy. CLINICAL RELEVANCE The radiotherapy reduces bone mechanical properties and the normal structure of organic and inorganic bone matrix. For studying the protocols to protect the radiotherapy effect using rabbit model, the use of the sacrificing period between 14 and 21 days is recommended.
Collapse
Affiliation(s)
- Priscilla Barbosa Ferreira Soares
- Department of Periodontology and Implantology, School of Dentistry, Federal University of Uberlândia, Avenida Pará s/n°, Campus Umuarama, Bloco 4L, Bairro Umuarama, Uberlândia, Minas Gerais, 38400-902, Brazil.
| | - Carlos José Soares
- Department of Operative Dentistry and Dental Materials, School of Dentistry, Federal University of Uberlândia, Avenida Pará s/n°, Campus Umuarama, Bloco 4L, Bairro Umuarama, Uberlândia, Minas Gerais, 38400-902, Brazil
| | - Pedro Henrique Justino Oliveira Limirio
- Department of Periodontology and Implantology, School of Dentistry, Federal University of Uberlândia, Avenida Pará s/n°, Campus Umuarama, Bloco 4L, Bairro Umuarama, Uberlândia, Minas Gerais, 38400-902, Brazil
| | - Rainde Naiara Rezende de Jesus
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Federal University of Uberlândia, Avenida Pará s/n°, Campus Umuarama, Bloco 4T, Bairro Umuarama, Uberlândia, Minas Gerais, 38400-902, Brazil
| | - Paula Dechichi
- Institute of Biomedical Sciences, Federal University of Uberlândia, Avenida Pará 1720, Campus Umuarama, Bloco 2B, Bairro Umuarama, Uberlândia, Minas Gerais, 38400-902, Brazil
| | - Rubens Spin-Neto
- Department of Dentistry and Oral Health, Department of Oral Radiology, Aarhus University, Vennelyst Boulevard 9, Building 1613, 130, 8000, Aarhus C, Denmark
| | - Darceny Zanetta-Barbosa
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Federal University of Uberlândia, Avenida Pará s/n°, Campus Umuarama, Bloco 4T, Bairro Umuarama, Uberlândia, Minas Gerais, 38400-902, Brazil
| |
Collapse
|
33
|
Bartlow CM, Mann KA, Damron TA, Oest ME. Limited field radiation therapy results in decreased bone fracture toughness in a murine model. PLoS One 2018; 13:e0204928. [PMID: 30281657 PMCID: PMC6169919 DOI: 10.1371/journal.pone.0204928] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 09/17/2018] [Indexed: 11/19/2022] Open
Abstract
Fragility fractures are a well-known complication following oncologic radiotherapy, and it is suspected that radiation-induced embrittlement of bone within the treatment field may contribute to fracture risk. To explore this phenomenon, a mouse model (BALB/cJ) of fractionated, limited field, bilateral hindlimb irradiation (4x5 Gy) was used. The effects of radiation on femoral (cortical) bone fracture toughness, morphology, and biochemistry-including advanced glycation end products (AGEs)-were quantified and compared to Sham group samples prior to irradiation and at 0, 4, 8, and 12 weeks post-irradiation. Additionally, alterations to bone fracture toughness mediated directly by radiation (independent of cellular mechanisms) were determined using devitalized mouse cadaver femurs. Finally, the contribution of AGEs to reduced fracture toughness was examined by artificially ribosylating mouse femurs ex vivo. These data demonstrate that in vivo irradiation results in an immediate (-42% at 0 weeks, p < 0.001) and sustained (-28% at 12 weeks, p < 0.001) decrease in fracture toughness with small changes in morphology (-5% in cortical area at 12 weeks), and minimal changes in bone composition (tissue mineral density, mineral:matrix ratio, and AGE content). Irradiation of devitalized femurs also reduced fracture toughness (-29%, p < 0.001), but to a lesser extent than was seen in vivo. While artificial ribosylation decreased fracture toughness with time, the extent of glycation needed to induce this effect exceeded the AGE accumulation that occurred in vivo. Overall, hindlimb irradiation induced a substantial and sustained decrease in bone fracture toughness. Approximately half of this decrease in fracture toughness is due to direct radiation damage, independent of cellular remodeling. Collagen glycation in vivo was not substantially altered, suggesting other matrix changes may contribute to post-radiotherapy bone embrittlement.
Collapse
Affiliation(s)
- Christopher M. Bartlow
- Department of Orthopedic Surgery, State University of New York Upstate Medical University, Syracuse, New York, United States of America
| | - Kenneth A. Mann
- Department of Orthopedic Surgery, State University of New York Upstate Medical University, Syracuse, New York, United States of America
| | - Timothy A. Damron
- Department of Orthopedic Surgery, State University of New York Upstate Medical University, Syracuse, New York, United States of America
| | - Megan E. Oest
- Department of Orthopedic Surgery, State University of New York Upstate Medical University, Syracuse, New York, United States of America
| |
Collapse
|
34
|
Camacho A, Jerez S. Bone metastasis treatment modeling via optimal control. J Math Biol 2018; 78:497-526. [DOI: 10.1007/s00285-018-1281-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 07/29/2018] [Indexed: 12/14/2022]
|
35
|
Abstract
BACKGROUND Effects of high-dose radiation using protons and photons on bone are relatively unexplored, but high rates of insufficiency fractures are reported, and the causes of this are incompletely understood. Imaging studies with pre- and postradiation scans can help one understand the effect of radiation on bone. QUESTIONS/PURPOSES The purpose of this study was to assess the effects of high-dose radiation on the trabecular density of bone in the sacrum using CT-derived Hounsfield units (HU). METHODS Between 2009 and 2015, we treated 57 patients (older then 18 years) with sacral chordoma. Fourteen (25%) of them were treated with radiation only. The general indication for this approach is inoperability resulting from tumor size. Forty-two (74%) patients were treated with transverse sacral resections and high-dose radiotherapy (using either protons or photons or a combination) before surgery and after surgery. During this time period, our indication for this approach generally was symptomatic sacral chordoma in which resection would prevent further growth and reasonable sacrifice of nerve roots was possible. Of those patients, 21 (50%) had CT scans both before and after radiation treatment. We used HU as a surrogate for bone density. CT uses HU to derive information on tissue and bone quantity. A recent study presented reference HU values for normal (mean 133 ± 38 HU), osteoporotic (101 ± 25 HU), and osteopenic bone (79 ± 32 HU). To adjust for scanning protocol-induced changes in HU, we calculated the ratio between bone inside and outside the radiation field rather than using absolute values. To assess the effect of radiation, we tested whether there was a difference in ratio (sacrum/L1) before and after radiation. A control measurement was performed (L2/L1) and also tested for a difference before and after radiation. Statistical analyses were performed using the paired t-test. RESULTS The effects of radiation appeared confined to the intended field, because the bone density outside the treated field was not observed to decrease. The ratio of HU (a surrogate for bone density) in L2 relative to L1 did not change after radiotherapy (preradiation mean: 0.979 ± 0.009, postradiation mean: 0.980 ± 0.009, mean difference outside the radiation field: -0.001, 95% confidence interval [CI], -0.009 to 0.007, p = 0.799). The ratio of HU within the radiation field relative to L1 decreased after radiotherapy (preradiation mean: 0.895 ± 0.050, postradiation mean: 0.658 ± 0.050, mean difference inside the radiation field: 0.237, 95% CI, 0.187-0.287, p < 0.001), suggesting the bone density stayed the same outside the radiation field but decreased inside the radiation field. CONCLUSIONS Trabecular bone density decreased after high-dose radiation therapy in a small group of patients with sacral chordoma. High-dose radiation is increasingly gaining acceptance for treating sacral malignancies; further long-term prospective studies using calibrated CT scanners and preferably bone biopsies are needed. LEVEL OF EVIDENCE Level IV, therapeutic study.
Collapse
|
36
|
Oest ME, Policastro CG, Mann KA, Zimmerman ND, Damron TA. Longitudinal Effects of Single Hindlimb Radiation Therapy on Bone Strength and Morphology at Local and Contralateral Sites. J Bone Miner Res 2018; 33:99-112. [PMID: 28902435 PMCID: PMC5776033 DOI: 10.1002/jbmr.3289] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 08/30/2017] [Accepted: 09/12/2017] [Indexed: 02/06/2023]
Abstract
Radiation therapy (RTx) is associated with increased risk for late-onset fragility fractures in bone tissue underlying the radiation field. Bone tissue outside the RTx field is often selected as a "normal" comparator tissue in clinical assessment of fragility fracture risk, but the robustness of this comparison is limited by an incomplete understanding of the systemic effects of local radiotherapy. In this study, a mouse model of limited field irradiation was used to quantify longitudinal changes in local (irradiated) and systemic (non-irradiated) femurs with respect to bone density, morphology, and strength. BALB/cJ mice aged 12 weeks underwent unilateral hindlimb irradiation (4 × 5 Gy) or a sham procedure. Femurs were collected at endpoints of 4 days before treatment and at 0, 1, 2, 4, 8, 12, and 26 weeks post-treatment. Irradiated (RTx), Contralateral (non-RTx), and Sham (non-RTx) femurs were imaged by micro-computed tomography and mechanically tested in three-point bending. In both the RTx and Contralateral non-RTx groups, the longer-term (12- to 26-week) outcomes included trabecular resorption, loss of diaphyseal cortical bone, and decreased bending strength. Contralateral femurs generally followed an intermediate response compared with RTx femurs. Change also varied by anatomic compartment; post-RTx loss of trabecular bone was more profound in the metaphyseal than the epiphyseal compartment, and cortical bone thickness decreased at the mid-diaphysis but increased at the metaphysis. These data demonstrate that changes in bone quantity, density, and architecture occur both locally and systemically after limited field irradiation and vary by anatomic compartment. Furthermore, the severity and persistence of systemic bone damage after limited field irradiation suggest selection of control tissues for assessment of fracture risk or changes in bone density after radiotherapy may be challenging. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Megan E Oest
- Department of Orthopedic Surgery, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Connor G Policastro
- Department of Orthopedic Surgery, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Kenneth A Mann
- Department of Orthopedic Surgery, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Nicholas D Zimmerman
- Department of Orthopedic Surgery, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Timothy A Damron
- Department of Orthopedic Surgery, SUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
37
|
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: 2.6] [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
|
38
|
|
39
|
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: 2.1] [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
|
40
|
Govey PM, Zhang Y, Donahue HJ. Mechanical Loading Attenuates Radiation-Induced Bone Loss in Bone Marrow Transplanted Mice. PLoS One 2016; 11:e0167673. [PMID: 27936104 PMCID: PMC5147933 DOI: 10.1371/journal.pone.0167673] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 11/20/2016] [Indexed: 12/16/2022] Open
Abstract
Exposure of bone to ionizing radiation, as occurs during radiotherapy for some localized malignancies and blood or bone marrow cancers, as well as during space travel, incites dose-dependent bone morbidity and increased fracture risk. Rapid trabecular and endosteal bone loss reflects acutely increased osteoclastic resorption as well as decreased bone formation due to depletion of osteoprogenitors. Because of this dysregulation of bone turnover, bone’s capacity to respond to a mechanical loading stimulus in the aftermath of irradiation is unknown. We employed a mouse model of total body irradiation and bone marrow transplantation simulating treatment of hematologic cancers, hypothesizing that compression loading would attenuate bone loss. Furthermore, we hypothesized that loading would upregulate donor cell presence in loaded tibias due to increased engraftment and proliferation. We lethally irradiated 16 female C57Bl/6J mice at age 16 wks with 10.75 Gy, then IV-injected 20 million GFP(+) total bone marrow cells. That same day, we initiated 3 wks compression loading (1200 cycles 5x/wk, 10 N) in the right tibia of 10 of these mice while 6 mice were irradiated, non-mechanically-loaded controls. As anticipated, before-and-after microCT scans demonstrated loss of trabecular bone (-48.2% Tb.BV/TV) and cortical thickness (-8.3%) at 3 wks following irradiation. However, loaded bones lost 31% less Tb.BV/TV and 8% less cortical thickness (both p<0.001). Loaded bones also had significant increases in trabecular thickness and tissue mineral densities from baseline. Mechanical loading did not affect donor cell engraftment. Importantly, these results demonstrate that both cortical and trabecular bone exposed to high-dose therapeutic radiation remain capable of an anabolic response to mechanical loading. These findings inform our management of bone health in cases of radiation exposure.
Collapse
Affiliation(s)
- Peter M. Govey
- Division of Musculoskeletal Sciences, Department of Orthopaedics and Rehabilitation, Penn State College of Medicine, Hershey, PA, United States of America
- Department of Biomedical Engineering, Penn State College of Engineering, University Park, PA, United States of America
| | - Yue Zhang
- Division of Musculoskeletal Sciences, Department of Orthopaedics and Rehabilitation, Penn State College of Medicine, Hershey, PA, United States of America
- Department of Biomedical Engineering, Virginia Commonwealth College of Engineering, Richmond, VA, United States of America
| | - Henry J. Donahue
- Division of Musculoskeletal Sciences, Department of Orthopaedics and Rehabilitation, Penn State College of Medicine, Hershey, PA, United States of America
- Department of Biomedical Engineering, Penn State College of Engineering, University Park, PA, United States of America
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, United States of America
- Department of Biomedical Engineering, Virginia Commonwealth College of Engineering, Richmond, VA, United States of America
- * E-mail:
| |
Collapse
|
41
|
Guo C, Li C, Yang K, Kang H, Xu X, Xu X, Deng L. Increased EZH2 and decreased osteoblastogenesis during local irradiation-induced bone loss in rats. Sci Rep 2016; 6:31318. [PMID: 27499068 PMCID: PMC4976370 DOI: 10.1038/srep31318] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 07/05/2016] [Indexed: 01/06/2023] Open
Abstract
Radiation therapy is commonly used to treat cancer patients but exhibits adverse effects, including insufficiency fractures and bone loss. Epigenetic regulation plays an important role in osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). Here, we reported local bone changes after single-dose exposure to 137CS irradiation in rats. Femur bone mineral density (BMD) and trabecular bone volume in the tibia were significantly decreased at 12 weeks after irradiation. Micro-CT results showed that tBMD, Tb.h and Tb.N were also significantly reduced at 12 weeks after irradiation exposure. ALP-positive OB.S/BS was decreased by 42.3% at 2 weeks after irradiation and was decreased by 50.8% at 12 weeks after exposure. In contrast to the decreased expression of Runx2 and BMP2, we found EZH2 expression was significantly increased at 2 weeks after single-dose 137CS irradiation in BMSCs. Together, our results demonstrated that single-dose 137CS irradiation induces BMD loss and the deterioration of bone microarchitecture in the rat skeleton. Furthermore, EZH2 expression increased and osteoblastogenesis decreased after irradiation. The underlying mechanisms warrant further investigation.
Collapse
Affiliation(s)
- Changjun Guo
- Shanghai Key Laboratory for the Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopedics, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine. Address: No. 197, Rui Jin Er Road, Shanghai 200025 China
| | - Changwei Li
- Shanghai Key Laboratory for the Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopedics, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine. Address: No. 197, Rui Jin Er Road, Shanghai 200025 China
| | - Kai Yang
- Shanghai Key Laboratory for the Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopedics, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine. Address: No. 197, Rui Jin Er Road, Shanghai 200025 China
| | - Hui Kang
- Shanghai Key Laboratory for the Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopedics, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine. Address: No. 197, Rui Jin Er Road, Shanghai 200025 China
| | - Xiaoya Xu
- Department of Bone Metabolism, Institute of Radiation Medicine, Fudan University, Shanghai 200032, China. Address: No. 2094, Xietu Road, Shanghai 200032 China
| | - Xiangyang Xu
- Department of Orthopedics, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine. Address: No. 197, Rui Jin Er Road, Shanghai 200025 China
| | - Lianfu Deng
- Shanghai Key Laboratory for the Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopedics, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine. Address: No. 197, Rui Jin Er Road, Shanghai 200025 China
| |
Collapse
|
42
|
Oest ME, Mann KA, Zimmerman ND, Damron TA. Parathyroid Hormone (1-34) Transiently Protects Against Radiation-Induced Bone Fragility. Calcif Tissue Int 2016; 98:619-30. [PMID: 26847434 PMCID: PMC4860360 DOI: 10.1007/s00223-016-0111-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 01/18/2016] [Indexed: 01/05/2023]
Abstract
Radiation therapy for soft tissue sarcoma or tumor metastases is frequently associated with damage to the underlying bone. Using a mouse model of limited field hindlimb irradiation, we assessed the ability of parathyroid hormone (1-34) fragment (PTH) delivery to prevent radiation-associated bone damage, including loss of mechanical strength, trabecular architecture, cortical bone volume, and mineral density. Female BALB/cJ mice received four consecutive doses of 5 Gy to a single hindlimb, accompanied by daily injections of either PTH or saline (vehicle) for 8 weeks, and were followed for 26 weeks. Treatment with PTH maintained the mechanical strength of irradiated femurs in axial compression for the first eight weeks of the study, and the apparent strength of irradiated femurs in PTH-treated mice was greater than that of naïve bones during this time. PTH similarly protected against radiation-accelerated resorption of trabecular bone and transient decrease in mid-diaphyseal cortical bone volume, although this benefit was maintained only for the duration of PTH delivery. Overall, PTH conferred protection against radiation-induced fragility and morphologic changes by increasing the quantity of bone, but only during the period of administration. Following cessation of PTH delivery, bone strength and trabecular volume fraction rapidly decreased. These data suggest that PTH does not negate the longer-term potential for osteoclastic bone resorption, and therefore, finite-duration treatment with PTH alone may not be sufficient to prevent late onset radiotherapy-induced bone fragility.
Collapse
Affiliation(s)
- Megan E Oest
- Department of Orthopedic Surgery, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, 13210, USA.
| | - Kenneth A Mann
- Department of Orthopedic Surgery, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, 13210, USA
| | - Nicholas D Zimmerman
- Department of Orthopedic Surgery, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, 13210, USA
| | - Timothy A Damron
- Department of Orthopedic Surgery, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, 13210, USA
| |
Collapse
|
43
|
Okoukoni C, Lynch SK, McTyre ER, Randolph DM, Weaver AA, Blackstock AW, Lally BE, Munley MT, Willey JS. A cortical thickness and radiation dose mapping approach identifies early thinning of ribs after stereotactic body radiation therapy. Radiother Oncol 2016; 119:449-53. [DOI: 10.1016/j.radonc.2016.03.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 03/16/2016] [Accepted: 03/18/2016] [Indexed: 01/08/2023]
|
44
|
Oest ME, Gong B, Esmonde-White K, Mann KA, Zimmerman ND, Damron TA, Morris MD. Parathyroid hormone attenuates radiation-induced increases in collagen crosslink ratio at periosteal surfaces of mouse tibia. Bone 2016; 86:91-97. [PMID: 26960578 PMCID: PMC4833661 DOI: 10.1016/j.bone.2016.03.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 02/06/2016] [Accepted: 03/03/2016] [Indexed: 12/13/2022]
Abstract
As part of our ongoing efforts to understand underlying mechanisms contributing to radiation-associated bone fragility and to identify possible treatments, we evaluated the longitudinal effects of parathyroid hormone (PTH) treatment on bone quality in a murine model of limited field irradiation. We hypothesized PTH would mitigate radiation-induced changes in the chemical composition and structure of bone, as measured by microscope-based Raman spectroscopy. We further hypothesized that collagen crosslinking would be especially responsive to PTH treatment. Raman spectroscopy was performed on retrieved tibiae (6-7/group/time point) to quantify metrics associated with bone quality, including: mineral-to-matrix ratio, carbonate-to-phosphate ratio, mineral crystallinity, collagen crosslink (trivalent:divalent) ratio, and the mineral and matrix depolarization ratios. Irradiation disrupted the molecular structure and orientation of bone collagen, as evidenced by a higher collagen crosslink ratio and lower matrix depolarization ratio (vs. non-irradiated control bones), persisting until 12weeks post-irradiation. Radiation transiently affected the mineral phase, as evidenced by increased mineral crystallinity and mineral-to-matrix ratio at 4weeks compared to controls. Radiation decreased bone mineral depolarization ratios through 12weeks, indicating increased mineral alignment. PTH treatment partially attenuated radiation-induced increases in collagen crosslink ratio, but did not restore collagen or mineral alignment. These post-radiation matrix changes are consistent with our previous studies of radiation damage to bone, and suggest that the initial radiation damage to bone matrix has extensive effects on the quality of tissue deposited thereafter. In addition to maintaining bone quality, preventing initial radiation damage to the bone matrix (i.e. crosslink ratio, matrix orientation) may be critical to preventing late-onset fragility fractures.
Collapse
Affiliation(s)
- Megan E. Oest
- Department of Orthopedic Surgery, Upstate Medical University, Syracuse, NY 13210, USA
- Corresponding author: Megan E. Oest, Department of Orthopedic Surgery, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, 13210, USA, Tel: +1 315-464-9950; Fax: +1 315-464-36638,
| | - Bo Gong
- Department of Chemistry; University of Michigan, Ann Arbor, MI 48109, USA
| | - Karen Esmonde-White
- Department of Internal Medicine-Rheumatology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Kenneth A. Mann
- Department of Orthopedic Surgery, Upstate Medical University, Syracuse, NY 13210, USA
| | - Nicholas D. Zimmerman
- Department of Orthopedic Surgery, Upstate Medical University, Syracuse, NY 13210, USA
| | - Timothy A. Damron
- Department of Orthopedic Surgery, Upstate Medical University, Syracuse, NY 13210, USA
| | - Michael D. Morris
- Department of Chemistry; University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
45
|
Goyden J, Tawara K, Hedeen D, Willey JS, Thom Oxford J, Jorcyk CL. The Effect of OSM on MC3T3-E1 Osteoblastic Cells in Simulated Microgravity with Radiation. PLoS One 2015; 10:e0127230. [PMID: 26030441 PMCID: PMC4452373 DOI: 10.1371/journal.pone.0127230] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 04/12/2015] [Indexed: 12/20/2022] Open
Abstract
Bone deterioration is a challenge in long-term spaceflight with significant connections to patients experiencing disuse bone loss. Prolonged unloading and radiation exposure, defining characteristics of space travel, have both been associated with changes in inflammatory signaling via IL-6 class cytokines in bone. While there is also evidence for perturbed IL-6 class signaling in spaceflight, there has been scant examination of the connections between microgravity, radiation, and inflammatory stimuli in bone. Our lab and others have shown that the IL-6 class cytokine oncostatin M (OSM) is an important regulator of bone remodeling. We hypothesize that simulated microgravity alters osteoblast OSM signaling, contributing to the decoupling of osteolysis and osteogenesis in bone homeostasis. To test this hypothesis, we induced OSM signaling in murine MC3T3-E1 pre-osteoblast cells cultured in modeled microgravity using a rotating wall vessel bioreactor with and without exposure to radiation typical of a solar particle event. We measured effects on inflammatory signaling, osteoblast activity, and mineralization. Results indicated time dependent interactions among all conditions in the regulation of IL-6 production. Furthermore, OSM induced the transcription of OSM receptor ß, IL 6 receptor α subunits, collagen α1(I), osteocalcin, sclerostin, RANKL, and osteoprotegerin. Measurements of osteoid mineralization suggest that the spatial organization of the osteoblast environment is an important consideration in understanding bone formation. Taken together, these results support a role for altered OSM signaling in the mechanism of microgravity-induced bone loss.
Collapse
Affiliation(s)
- Jake Goyden
- Department of Biological Sciences, Boise State University, 1910 University Drive, Boise, Idaho 83725, United States of America
| | - Ken Tawara
- Department of Biological Sciences, Boise State University, 1910 University Drive, Boise, Idaho 83725, United States of America
| | - Danielle Hedeen
- Department of Biological Sciences, Boise State University, 1910 University Drive, Boise, Idaho 83725, United States of America
| | - Jeffrey S. Willey
- Department of Radiation Oncology, and the Comprehensive Cancer Center, Wake Forest School of Medicine, 1 Medical Center Blvd, Winston-Salem, North Carolina, 27157, United States of America
| | - Julia Thom Oxford
- Department of Biological Sciences, Boise State University, 1910 University Drive, Boise, Idaho 83725, United States of America
- Biomolecular Research Center, Boise State University 1910 University Drive, Boise, Idaho 83725, United States of America
| | - Cheryl L. Jorcyk
- Department of Biological Sciences, Boise State University, 1910 University Drive, Boise, Idaho 83725, United States of America
- Biomolecular Research Center, Boise State University 1910 University Drive, Boise, Idaho 83725, United States of America
- * E-mail:
| |
Collapse
|