1
|
Liu J, Ren W, Wang S, Yang J, Zhang H, Zeng Y, Yin D, Shang P. The effects and mechanisms of electromagnetic fields on bone remodeling: From clinical to laboratory. J Orthop Translat 2025; 52:14-26. [PMID: 40226335 PMCID: PMC11986540 DOI: 10.1016/j.jot.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 02/14/2025] [Accepted: 03/10/2025] [Indexed: 04/15/2025] Open
Abstract
Electromagnetic fields (EMFs) are physical fields generated by electrically charged objects, and play a vital role in the growth and development of living organisms. Bone is a highly dynamic structure that undergoes a constant remodeling process. From 1962 to 1977, Bassett discovered the piezoelectric effect in bone tissue and found that EMFs accelerated osteogenesis, promoted tibial fracture healing in dogs, and had positive effects in clinical trials. Since then, EMFs have been increasingly studied in bone remodeling disorders as a non-invasive physical therapy. This review summarizes clinical trials and laboratory studies on EMF interventions in bone remodeling disorders over the past few decades, outlining the effects of EMFs on various bone cells and their underlying molecular mechanisms. In addition, we propose issues in current studies and give an outlook on the research and application of EMFs as a non-invasive physical therapy. The translational potential of this article This article systematically reviews the research ranging from biological and physical mechanisms to medical applications of EMFs on bone remodeling and related diseases, identifies key challenges in future basic research, and proposes new strategies for developing novel medical equipment and advancing clinical applications in this field. These insights contribute to the advancement of non-invasive physical therapies in orthopedics.
Collapse
Affiliation(s)
- Junyu Liu
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, 518057, China
- School of Life Science, Northwestern Polytechnical University, Xi'an, 710072, China
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environment Biophysics, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Weihao Ren
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, 518057, China
- School of Life Science, Northwestern Polytechnical University, Xi'an, 710072, China
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environment Biophysics, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Shenghang Wang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases (TMBJ), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Department of Spine Surgery, People's Hospital of Longhua, Affiliated Hospital of Southern Medical University, Shenzhen, 518109, China
| | - Jiancheng Yang
- Department of Osteoporosis, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Hao Zhang
- Department of Spine Surgery, People's Hospital of Longhua, Affiliated Hospital of Southern Medical University, Shenzhen, 518109, China
| | - Yuhong Zeng
- Department of Osteoporosis, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, China
| | - Dachuan Yin
- School of Life Science, Northwestern Polytechnical University, Xi'an, 710072, China
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environment Biophysics, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Peng Shang
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, 518057, China
- School of Life Science, Northwestern Polytechnical University, Xi'an, 710072, China
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environment Biophysics, Northwestern Polytechnical University, Xi'an, 710072, China
| |
Collapse
|
2
|
Gerdesmeyer L, Tübel J, Obermeier A, Harrasser N, Glowalla C, von Eisenhart-Rothe R, Burgkart R. Extracorporeal Magnetotransduction Therapy as a New Form of Electromagnetic Wave Therapy: From Gene Upregulation to Accelerated Matrix Mineralization in Bone Healing. Biomedicines 2024; 12:2269. [PMID: 39457582 PMCID: PMC11505246 DOI: 10.3390/biomedicines12102269] [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: 09/11/2024] [Revised: 09/28/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Electromagnetic field therapy is gaining attention for its potential in treating bone disorders, with Extracorporeal Magnetotransduction Therapy (EMTT) emerging as an innovative approach. EMTT offers a higher oscillation frequency and magnetic field strength compared to traditional Pulsed Electromagnetic Field (PEMF) therapy, showing promise in enhancing fracture healing and non-union recovery. However, the mechanisms underlying these effects remain unclear. RESULTS This study demonstrates that EMTT significantly enhances osteoblast bone formation at multiple levels, from gene expression to extracellular matrix mineralization. Key osteoblastogenesis regulators, including SP7 and RUNX2, and bone-related genes such as COL1A1, ALPL, and BGLAP, were upregulated, with expression levels surpassing those of the control group by over sevenfold (p < 0.001). Enhanced collagen synthesis and mineralization were confirmed by von Kossa and Alizarin Red staining, indicating increased calcium and phosphate deposition. Additionally, calcium imaging revealed heightened calcium influx, suggesting a cellular mechanism for EMTT's osteogenic effects. Importantly, EMTT did not compromise cell viability, as confirmed by live/dead staining and WST-1 assays. CONCLUSION This study is the first to show that EMTT can enhance all phases of osteoblastogenesis and improve the production of critical mineralization components, offering potential clinical applications in accelerating fracture healing, treating osteonecrosis, and enhancing implant osseointegration.
Collapse
Affiliation(s)
- Lennart Gerdesmeyer
- Department of Orthopaedics and Sports Orthopaedics, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Jutta Tübel
- Department of Orthopaedics and Sports Orthopaedics, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Andreas Obermeier
- Department of Orthopaedics and Sports Orthopaedics, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Norbert Harrasser
- Department of Orthopaedics and Sports Orthopaedics, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
- ECOM Excellent Center of Medicine, Arabellastraße 17, 81925 Munich, Germany
| | - Claudio Glowalla
- Department of Orthopaedics and Sports Orthopaedics, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
- BG Unfallklinik Murnau, Professor-Küntscher-Straße 8, 82418 Murnau am Staffelsee, Germany
| | - Rüdiger von Eisenhart-Rothe
- Department of Orthopaedics and Sports Orthopaedics, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Rainer Burgkart
- Department of Orthopaedics and Sports Orthopaedics, Klinikum rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| |
Collapse
|
3
|
Liang Y, Yuan X, Dai X, Zhang G, Li C, Yang H, Zhang T, Qin J. The effects of simvastatin on the bone microstructure and mechanics of ovariectomized mice: a micro-CT and micro-finite element analysis study. BMC Musculoskelet Disord 2024; 25:748. [PMID: 39294613 PMCID: PMC11409800 DOI: 10.1186/s12891-024-07860-w] [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: 02/14/2024] [Accepted: 09/09/2024] [Indexed: 09/20/2024] Open
Abstract
BACKGROUND Osteoporosis is a major health concern for postmenopausal women, and the effect of simvastatin (Sim) on bone metabolism is controversial. This study aimed to investigate the effect of simvastatin on the bone microstructure and bone mechanical properties in ovariectomized (OVX) mice. METHODS 24 female C57BL/6J mice (8-week-old) were randomly allocated into three groups including the OVX + Sim group, the OVX group and the control group. At 8 weeks after operation, the L4 vertebral bones were dissected completely for micro-Computed Tomography (micro-CT) scanning and micro-finite element analysis (µFEA). The differences between three groups were compared using ANOVA with a LSD correction, and the relationship between bone microstructure and mechanical properties was analyzed using linear regression. RESULTS Bone volume fraction, trabecular number, connectivity density and trabecular tissue mineral density in the OVX + Sim group were significantly higher than those in the OVX group (P < 0.05). For the mechanical properties detected via µFEA, the OVX + Sim group had lower total deformation, equivalent elastic strain and equivalent stress compared to the OVX group (P < 0.05). In the three groups, the mechanical parameters were significantly correlated with bone volume fraction and trabecular bone mineral density. CONCLUSIONS The findings suggested that simvastatin had a potential role in the treatment of osteoporosis. The results of this study could guide future research on simvastatin and support the development of simvastatin-based treatments to improve bone health.
Collapse
Affiliation(s)
- Yanbo Liang
- Department of Radiology, The Second Affiliated Hospital of Shandong First Medical University, No.366 Taishan Street, Tai'an City, Shandong Province, 271000, China
| | - Xiaoqing Yuan
- Department of Radiology, The Second Affiliated Hospital of Shandong First Medical University, No.366 Taishan Street, Tai'an City, Shandong Province, 271000, China
- Chinese institutes for medical research, Capital Medical University, Beijing, 100050, China
| | - Xiaoxue Dai
- The First Affiliated Hospital of Shandong First Medical University, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, 271000, China
| | - Guohui Zhang
- Shandong First Medical University, Jinan, Shandong, 271000, China
| | - Changqin Li
- Department of Radiology, The Second Affiliated Hospital of Shandong First Medical University, No.366 Taishan Street, Tai'an City, Shandong Province, 271000, China
| | - Hui Yang
- Department of Radiology, The Second Affiliated Hospital of Shandong First Medical University, No.366 Taishan Street, Tai'an City, Shandong Province, 271000, China
| | - Tingting Zhang
- Department of Radiology, The Second Affiliated Hospital of Shandong First Medical University, No.366 Taishan Street, Tai'an City, Shandong Province, 271000, China
| | - Jian Qin
- Department of Radiology, The Second Affiliated Hospital of Shandong First Medical University, No.366 Taishan Street, Tai'an City, Shandong Province, 271000, China.
| |
Collapse
|
4
|
Gerdesmeyer L, Burgkart R, Saxena A. Clavicle fracture and triathlon performance: a case report. J Med Case Rep 2024; 18:197. [PMID: 38566165 PMCID: PMC10988895 DOI: 10.1186/s13256-024-04482-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 02/29/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Collarbone fracture is a common injury, particularly among athletes involved in contact sports and participating in endurance activities. Conventional treatment requires surgery and postoperative immobilization, resulting in an average return-to-sport timeframe of approximately 13 weeks. This case challenges the established treatment protocols, aiming to expedite recovery and enable a quicker resumption of high-intensity athletic activities. CASE PRESENTATION A 24-year-old Caucasian athlete completed a Half-Ironman Triathlon (70.3) merely three weeks post-collarbone fracture. Utilizing Extracorporeal Magneto-Transduction Therapy (EMTT) alongside surgical intervention, the patient achieved accelerated healing and remarkable performance outcomes without encountering any adverse effects. CONCLUSIONS The integration of EMTT into the treatment paradigm for bone fractures alters the traditional understanding of recovery timelines and rehabilitation strategies. This case highlights the potential benefits of electromagnetic wave therapy in expediting the healing process and enabling athletes to resume high-level sports activities at an earlier stage.
Collapse
Affiliation(s)
- Lennart Gerdesmeyer
- Department of Orthopaedics and Sports Orthopaedics, Klinikum Rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany.
| | - Rainer Burgkart
- Department of Orthopaedics and Sports Orthopaedics, Klinikum Rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675, Munich, Germany
| | - Amol Saxena
- PAMF-Sutter Department of Sports Medicine, 795 El Camino Real, Clark Building, Level 3, Palo Alto, CA, 94301, USA
| |
Collapse
|
5
|
Ozawa Y, Takegami Y, Osawa Y, Asamoto T, Tanaka S, Imagama S. Anti-sclerostin antibody therapy prevents post-ischemic osteonecrosis bone collapse via interleukin-6 association. Bone 2024; 181:117030. [PMID: 38309414 DOI: 10.1016/j.bone.2024.117030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/29/2024] [Accepted: 01/29/2024] [Indexed: 02/05/2024]
Abstract
Osteonecrosis of the femoral head (ONFH) is a debilitating condition characterized by subchondral bone necrosis, which frequently culminates in joint destruction. Although total hip arthroplasty is conventionally practiced to remediate ONFH, for patients under the age of 60, the outcomes can be suboptimal. Chronic inflammation, particularly that mediated by interleukin-6 (IL-6), has been conjectured to be a potential mechanism underlying the etiology of ONFH. This study aimed at exploring the interplay between IL-6, the canonical Wnt signaling pathway, and ONFH to provide insights for potential therapeutic interventions. Human ONFH specimens depicted an elevation in β-catenin expression in the transitional layer, while IL-6 levels were pronounced in the same region. Subsequently, mouse models of ischemic osteonecrosis were treated with an anti-sclerostin antibody to assess its effects on bone metabolism and cellular processes. Histological analysis revealed that the administration of anti-sclerostin antibodies effectuated early recovery from bone necrosis, reduced empty lacunae, and suppressed IL-6 expression. The treatment evidently initiated the activation of the Wnt/β-catenin signaling pathway, presenting a potential mechanism associated with IL-6-mediated inflammation. Furthermore, the antibody upregulated osteoblast formation, downregulated osteoclast formation, and increased bone volume. Micro-CT imaging demonstrated increased bone volume, prevented epiphyseal deformity, and improved compression strength. Therefore, this study yields significant findings, indicating the potency of anti-sclerostin antibodies in effectively modulating the Wnt/β-catenin pathway, associating with IL-6 expression, and preventing post-ONFH bone collapse. Additionally, this preclinical investigation in mouse models offers an avenue for prospective research on potential therapeutic interventions against human ONFH.
Collapse
Affiliation(s)
- Yuto Ozawa
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, 8 Showa-ku, Nagoya, Japan
| | - Yasuhiko Takegami
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, 8 Showa-ku, Nagoya, Japan.
| | - Yusuke Osawa
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, 8 Showa-ku, Nagoya, Japan
| | - Takamune Asamoto
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, 8 Showa-ku, Nagoya, Japan
| | - Shinya Tanaka
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, 8 Showa-ku, Nagoya, Japan
| | - Shiro Imagama
- Department of Orthopaedic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, 8 Showa-ku, Nagoya, Japan
| |
Collapse
|
6
|
Sun W, Xie Q, Yan JJ, Huang Y, Zhou Y, Xiao H, Wang CQ. Observation of the Short-term Efficacy of Technetium-99 Conjugated with Methylene Diphosphonate Combined Therapy in the Treatment of Postmenopausal Osteoporosis. Comb Chem High Throughput Screen 2024; 27:1930-1937. [PMID: 38357942 DOI: 10.2174/0113862073266467231228124942] [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: 07/24/2023] [Revised: 11/01/2023] [Accepted: 11/08/2023] [Indexed: 02/16/2024]
Abstract
OBJECTIVE To investigate the short-term efficacy and safety of Yunke (technetium-99 conjugated methylene diphosphonate) combined with pulsed electromagnetic field (PEMF) and Gukang capsule in the treatment of postmenopausal osteoporosis (PMOP). METHODS A total of 112 patients with PMOP who received treatment in the Department of Nuclear Medicine of the hospital from January 2019 to June 2020 were selected and randomly divided into 4 groups of 28 patients each. Group A received Yunke and PEMFs, group B received Gukang capsules and PEMFs, group C received Yunke and Gukang capsules and PEMFs, and group D received PEMFs. All groups were given adequate amounts of calcium and active vitamin D. Intervention 2 sessions of 3 months each. Outcome measures were bone mineral density (BMD) and pain improvement. RESULTS Compared with 1 course of treatment, the symptoms of bone pain were relieved more significantly after 2 courses of treatment in group A (50.0% vs. 64.3%), group B (46.4% vs. 64.3%), group C (78.6% vs. 92.9%) and group D (21.4% vs. 28.6%) (P < 0.05). After 2 courses of treatment, bone pain symptoms were less relieved in group A (96.4% vs. 64.3%), group B (96.4% vs. 64.3%), and group D (96.4% vs. 28.6%) compared with group C (P < 0.05). Compared with group C, BMD values of L4 vertebrae and femoral neck were significantly decreased in groups A, B, and D (P < 0.05). Compared with those before treatment, BMD of L4 vertebrae and femoral neck increased significantly in groups A, B, C, and D after 2 courses of treatment (P < 0.05). CONCLUSION Yunke combined therapy can effectively relieve the pain symptoms, increase BMD, and reduce the risk of fracture in patients with PMOP in a short period, which is an effective method for the treatment of PMOP.
Collapse
Affiliation(s)
- Wen Sun
- Department of Nuclear Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, 570102, China
| | - Quan Xie
- Department of Nuclear Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, 570102, China
| | - Juan-Juan Yan
- Department of Nuclear Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, 570102, China
| | - Ying Huang
- Department of Nuclear Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, 570102, China
| | - Ying Zhou
- Department of Nuclear Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, 570102, China
| | - Huan Xiao
- Department of Nuclear Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, 570102, China
| | - Chao-Qun Wang
- Department of Nuclear Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, China
| |
Collapse
|
7
|
Suzuki N, Kakikawa M, Oda Y, Kobayashi-Sun J, Yamada S, Kuroda K, Kobayashi I, Honda M, Matsubara H, Tabuchi Y, Shimizu N, Watanabe K, Hirayama J, Hattori A. Bone regeneration-enhancing effects of extremely low-frequency electromag- netic fields: Analysis using fish scales as a bone model. Biomed Res 2024; 45:187-195. [PMID: 39370297 DOI: 10.2220/biomedres.45.187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Electromagnetic fields (EMFs) noninvasively promote fracture healing, prevent osteoporosis, promote diaphyseal growth, enhance differentiation, and stimulate cell division. However, no good model systems for analyzing bone regeneration have been reported. In this study, we examined the in vivo regeneration of scales having osteoblasts and osteoclasts using a new magnetic field generator for exposing aquatic animals to EMFs at a sine-wave frequency of 60 Hz. Goldfish scales were put into a fish-breeding space with the developed magnetic field generator and exposed to extremely low-frequency electromagnetic fields (ELF-EMFs) of 60 Hz at an intensity of 1, 3, and 5 mT for 10 days while being regenerated the scales. After exposure, alkaline phosphatase (ALP) and tartrate-resistant acid phosphatase (TRAP) activities in the goldfish scales were measured as markers of osteoblasts and osteoclasts, respectively. As a result, both ALP and TRAP activities in regenerating scales exposed to 3 mT ELF-EMFs were higher than those in regenerating scales exposed to 1 and 5 mT ELF-EMFs. Exposure of scales to 3 mT ELF-EMFs significantly enhanced the scale regeneration rate. Exposure of rat calvaria to 3 mT ELF-EMFs also increased both ALP and TRAP activities like in goldfish scales. Thus, we concluded that 3 mT ELF-EMFs contribute to the medical treatment of bone diseases.
Collapse
Affiliation(s)
- Nobuo Suzuki
- Noto Marine Laboratory, Institute of Nature and Environmental Technology, Kanazawa University, Noto-cho, Japan
| | - Makiko Kakikawa
- Faculty of Biological Science and Technology, Institute of Science and Engineering, Kanazawa University, Kanazawa, Japan
| | - Yuta Oda
- Faculty of Biological Science and Technology, Institute of Science and Engineering, Kanazawa University, Kanazawa, Japan
| | - Jingjing Kobayashi-Sun
- Faculty of Biological Science and Technology, Institute of Science and Engineering, Kanazawa University, Kanazawa, Japan
- Department of Clinical Engineering, Faculty of Health Sciences, Komatsu University, Komatsu, Japan
- Department of Food Science, Faculty of Bioresources and Environmental Sciences, Ishikawa Prefectural University, Nonoichi, Japan
| | - Sotoshi Yamada
- Noto Marine Laboratory, Institute of Nature and Environmental Technology, Kanazawa University, Noto-cho, Japan
| | - Kouhei Kuroda
- Noto Marine Laboratory, Institute of Nature and Environmental Technology, Kanazawa University, Noto-cho, Japan
| | - Isao Kobayashi
- Faculty of Biological Science and Technology, Institute of Science and Engineering, Kanazawa University, Kanazawa, Japan
| | - Masato Honda
- Botanical Garden, Institute of Nature and Environmental Technology, Kanazawa University, Kanazawa, Japan
| | - Hajime Matsubara
- Noto Center for Fisheries Science and Technology, Kanazawa University, Noto-cho, Japan
| | - Yoshiaki Tabuchi
- Life Science Research Center, University of Toyama, Toyama, Japan
| | - Nobuaki Shimizu
- Noto Marine Laboratory, Institute of Nature and Environmental Technology, Kanazawa University, Noto-cho, Japan
| | - Kazuki Watanabe
- Department of Clinical Engineering, Faculty of Health Sciences, Komatsu University, Komatsu, Japan
- Department of Immunobiology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Jun Hirayama
- Department of Clinical Engineering, Faculty of Health Sciences, Komatsu University, Komatsu, Japan
- Faculty of Education, Bunkyo University, Koshigaya, Japan
| | - Atsuhiko Hattori
- Department of Sport and Wellness, College of Sport and Wellness, Rikkyo University, Saitama, Japan
| |
Collapse
|
8
|
Guan Z, Xuanqi Z, Zhu J, Yuan W, Jia J, Zhang C, Sun T, Leng H, Jiang C, Xu Y, Song C. Estrogen deficiency induces bone loss through the gut microbiota. Pharmacol Res 2023; 196:106930. [PMID: 37722518 DOI: 10.1016/j.phrs.2023.106930] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/20/2023]
Abstract
Postmenopausal osteoporosis is a common bone metabolic disease, and gut microbiota (GM) imbalance plays an important role in the development of metabolic bone disease. Here, we show that ovariectomized mice had high levels of lipopolysaccharide in serum and gut microbiota dysbiosis through increases in luminal Firmicutes:Bacteroidetes ratio. We depleted the GM through antibiotic treatment and observed improvements in bone mass, bone microstructure, and bone strength in ovariectomized mice. Conversely, transplantation of GM adapted to ovariectomy induced bone loss. However, GM depletion reversed ovariectomy-induced gene expression in the tibia and increased periosteal bone formation. Furthermore, bioinformatics analysis revealed that the G-protein-coupled bile acid receptor (TGR5) and systemic inflammatory factors play key roles in bone metabolism. Silencing TGR5 expression through small interfering RNA (siRNA) in the local tibia and knockout of TGR5 attenuated the effects of GM depletion in ovariectomized mice, confirming these findings. Thus, this study highlights the critical role of the GM in inducing bone loss in ovariectomized mice and suggests that targeting TGR5 within the GM may have therapeutic potential for postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Zhiyuan Guan
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
| | - Zheng Xuanqi
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
| | - Junxiong Zhu
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
| | - Wanqiong Yuan
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
| | - Jialin Jia
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
| | - Chenggui Zhang
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
| | - Tiantong Sun
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
| | - Huijie Leng
- Beijing Key Laboratory of Spinal Diseases, Beijing, China
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, and the Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Yingsheng Xu
- Department of Neurology, Peking University Third Hospital, Beijing, China
| | - Chunli Song
- Department of Orthopedics, Peking University Third Hospital, Beijing, China; Beijing Key Laboratory of Spinal Diseases, Beijing, China.
| |
Collapse
|
9
|
Zhai M, Zhang C, Cui J, Liu J, Li Y, Xie K, Luo E, Tang C. Electromagnetic fields ameliorate hepatic lipid accumulation and oxidative stress: potential role of CaMKKβ/AMPK/SREBP-1c and Nrf2 pathways. Biomed Eng Online 2023; 22:51. [PMID: 37217972 DOI: 10.1186/s12938-023-01114-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 05/13/2023] [Indexed: 05/24/2023] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease worldwide, and is related to disturbed lipid metabolism and redox homeostasis. However, a definitive drug treatment has not been approved for this disease. Studies have found that electromagnetic fields (EMF) can ameliorate hepatic steatosis and oxidative stress. Nevertheless, the mechanism remains unclear. METHODS NAFLD models were established by feeding mice a high-fat diet. Simultaneously, EMF exposure is performed. The effects of the EMF on hepatic lipid deposition and oxidative stress were investigated. Additionally, the AMPK and Nrf2 pathways were analysed to confirm whether they were activated by the EMF. RESULTS Exposure to EMF decreased the body weight, liver weight and serum triglyceride (TG) levels and restrained the excessive hepatic lipid accumulation caused by feeding the HFD. The EMF boosted CaMKKβ protein expression, activated AMPK phosphorylation and suppressed mature SREBP-1c protein expression. Meanwhile, the activity of GSH-Px was enhanced following an increase in nuclear Nrf2 protein expression by PEMF. However, no change was observed in the activities of SOD and CAT. Consequently, EMF reduced hepatic reactive oxygen species (ROS) and MDA levels, which means that EMF relieved liver damage caused by oxidative stress in HFD-fed mice. CONCLUSIONS EMF may activate the CaMKKβ/AMPK/SREBP-1c and Nrf2 pathways to control hepatic lipid deposition and oxidative stress. This investigation indicates that EMF may be a novel therapeutic method for NAFLD.
Collapse
Affiliation(s)
- Mingming Zhai
- Department of Biomedical Engineering, Fourth Military Medical University, No. 169 Changle West Road, Xi'an, 710032, China
- Shaanxi Provincial Key Laboratory of Bioelectromagnetic Detection and Intelligent Perception, Xi'an, China
| | - Chenxu Zhang
- Department of Biomedical Engineering, Fourth Military Medical University, No. 169 Changle West Road, Xi'an, 710032, China
| | - Jinxiu Cui
- Department of Biomedical Engineering, Fourth Military Medical University, No. 169 Changle West Road, Xi'an, 710032, China
| | - Juan Liu
- Department of Biomedical Engineering, Fourth Military Medical University, No. 169 Changle West Road, Xi'an, 710032, China
| | - Yuanzhe Li
- Department of Biomedical Engineering, Fourth Military Medical University, No. 169 Changle West Road, Xi'an, 710032, China
| | - Kangning Xie
- Department of Biomedical Engineering, Fourth Military Medical University, No. 169 Changle West Road, Xi'an, 710032, China
| | - Erping Luo
- Department of Biomedical Engineering, Fourth Military Medical University, No. 169 Changle West Road, Xi'an, 710032, China.
| | - Chi Tang
- Department of Biomedical Engineering, Fourth Military Medical University, No. 169 Changle West Road, Xi'an, 710032, China.
- Shaanxi Provincial Key Laboratory of Bioelectromagnetic Detection and Intelligent Perception, Xi'an, China.
| |
Collapse
|
10
|
Zhang H, Yang Y, Yang E, Tian Z, Huang Y, Zhang Z, Bao M, Liao D, Ge J, Wang C, Li X, Luo P. Pulsed Electromagnetic Fields Protect Against Brain Ischemia by Modulating the Astrocytic Cholinergic Anti-inflammatory Pathway. Cell Mol Neurobiol 2023; 43:1301-1317. [PMID: 35831547 PMCID: PMC11414443 DOI: 10.1007/s10571-022-01251-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 06/27/2022] [Indexed: 01/11/2023]
Abstract
Neuroinflammation is one of the most important pathological processes following brain ischemia. Pulsed electromagnetic fields (PEMFs) protect against brain ischemia, but their role in regulating neuroinflammation remains unclear. In the present study, we investigated the biological effects of PEMF exposure on brain ischemia-induced neuroinflammation through the astrocytic cholinergic anti-inflammatory pathway. PEMF exposure reduced the activation of astrocytes and neuroinflammation following brain ischemia by directly modulating astrocytic injury and inflammatory cytokine release. Inhibition of nicotinic acetylcholine receptor alpha 7 subunit (α7nAChR) by a specific antagonist reversed the regulatory effects of PEMF on astrocytes. Furthermore, negative regulation of signal transducer and activator of transcription 3 (STAT3) by α7nAChR was found to be an important downstream mechanism through which PEMF regulates astrocyte-related neuroinflammation. PEMF suppressed STAT3 phosphorylation and nuclear translocation by activating α7nAChR. These results demonstrate that PEMF exerts anti-inflammatory effects in the context of brain ischemia by modulating astrocytic α7nAChR/STAT3 signaling.
Collapse
Affiliation(s)
- Haofuzi Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, 12769 Changle Xi Road, Xi'an, 710032, China
| | - Yuefan Yang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, 12769 Changle Xi Road, Xi'an, 710032, China
- Department of Biomedical Engineering, Fourth Military Medical University, Xi'an, China
| | - Erwan Yang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, 12769 Changle Xi Road, Xi'an, 710032, China
| | - Zhicheng Tian
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, 12769 Changle Xi Road, Xi'an, 710032, China
| | - Yutao Huang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, 12769 Changle Xi Road, Xi'an, 710032, China
| | - Zhuoyuan Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, 12769 Changle Xi Road, Xi'an, 710032, China
- School of Life Science, Northwest University, Xi'an, China
| | - Mingdong Bao
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, 12769 Changle Xi Road, Xi'an, 710032, China
| | - Dan Liao
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, 12769 Changle Xi Road, Xi'an, 710032, China
| | - Junmiao Ge
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, 12769 Changle Xi Road, Xi'an, 710032, China
| | - Chao Wang
- School of Electrical Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Xin Li
- Department of Anesthesiology, Xijing Hospital, Fourth Military Medical University, 127 Changle Xi Road, Xi'an, 710032, China.
| | - Peng Luo
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, 12769 Changle Xi Road, Xi'an, 710032, China.
| |
Collapse
|
11
|
Luo L, Guan Z, Jin X, Guan Z, Jiang Y. Identification of kukoamine a as an anti-osteoporosis drug target using network pharmacology and experiment verification. Mol Med 2023; 29:36. [PMID: 36941586 PMCID: PMC10029210 DOI: 10.1186/s10020-023-00625-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 02/16/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND Osteoporosis (OP) is a major and growing public health problem characterized by decreased bone mineral density and destroyed bone microarchitecture. Previous studies found that Lycium Chinense Mill (LC) has a potent role in inhibiting bone loss. Kukoamine A (KuA), a bioactive compound extract from LC was responsible for the anti-osteoporosis effect. This study aimed to investigate the anti-osteoporosis effect of KuA isolated from LC in treating OP and its potential molecular mechanism. METHOD In this study, network pharmacology and molecular docking were investigated firstly to find the active ingredients of LC such as KuA, and the target genes of OP by the TCMSP platform. The LC-OP-potential Target gene network was constructed by the STRING database and network maps were built by Cytoscape software. And then, the anti-osteoporotic effect of KuA in OVX-induced osteoporosis mice and MC3T3-E1 cell lines were investigated and the potential molecular mechanism including inflammation level, cell apoptosis, and oxidative stress was analyzed by dual-energy X-ray absorptiometry (DXA), micro-CT, ELISA, RT-PCR, and Western Blotting. RESULT A total of 22 active compounds were screened, and we found KuA was identified as the highest active ingredient. Glycogen Phosphorylase (PYGM) was the target gene associated with a maximum number of active ingredients of LC and regulated KuA. In vivo, KuA treatment significantly increased the bone mineral density and improve bone microarchitecture for example increased BV/TV, Tb.N and Tb.Th but reduced Tb.Sp in tibia and lumber 4. Furthermore, KuA increased mRNA expression of osteoblastic differentiation-related genes in OVX mice and protects against OVX-induced cell apoptosis, oxidative stress level and inflammation level. In vitro, KuA significantly improves osteogenic differentiation and mineralization in cells experiment. In addition, KuA also attenuated inflammation levels, cell apoptosis, and oxidative stress level. CONCLUSION The results suggest that KuA could protect against the development of OP in osteoblast cells and ovariectomized OP model mice and these found to provide a better understanding of the pharmacological activities of KuA again bone loss.
Collapse
Affiliation(s)
- Liying Luo
- Department of Ophthalmology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zhiyuan Guan
- Department of Orthopedics, The Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - Xiao Jin
- Department of Rheumatology and Immunology, The First People's Hospital of Xuzhou, Xuzhou, Jiangsu, 221002, People's Republic of China.
| | - Zhiqiang Guan
- Department of Dermatology, Xuzhou Municipal Hospital Affiliated With Xuzhou Medical University, Xuzhou, Jiangsu, 221002, People's Republic of China.
| | - Yanyun Jiang
- Department of Ophthalmology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
12
|
Popović T, Matavulj M, Nežić L, Radulović TN, Škrbić R. Pulsed electromagnetic field attenuates bone fragility in estrogen-deficient osteoporosis in rats. Technol Health Care 2023:THC220642. [PMID: 36641696 DOI: 10.3233/thc-220642] [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: 01/12/2023]
Abstract
BACKGROUND The pulsed electromagnetic fields (PEMFs) seem effective in increasing bone mineral density and promoting osteogenesis and bone healing. OBJECTIVE To examine the effect of two different modalities of PEMFs therapy in comparison with the recommended pharmacological treatment on experimental osteoporosis in rats. METHODS The experimental model of estrogen-deficient osteoporosis induced by ovariectomy was used in this study. The animals were exposed to PEMFs of various frequencies (40 Hz and 25 Hzk), intensities (10 mT and 36.4 μT), lengths of exposure, and the effects were compared with the standard treatment with pamidronate, vitamin D, and calcium supplementation. RESULTS The application of PEMF40Hz, significantly reduced the osteoporotic bone loss in female rats that were confirmed with biochemical, biomechanical, and histological analyses. These effects were more pronounced than in osteoporotic animals treated with pamidronate, vitamin D, and calcium supplementation. On the contrary, the exposure to PEMF25Hz did not show restorative effects but led to further progression of osteoporosis. CONCLUSION The exposure to PEMF40Hz, significantly restored osteoporosis and attenuated bone fragility in comparison to the rats exposed to PEMF25Hz or those treated with pamidronate, vitamin D, and calcium supplementation.
Collapse
Affiliation(s)
- Tamara Popović
- Institute for Physical Medicine and Rehabilitation "Dr. Miroslav Zotović", Banja Luka, Bosnia and Herzegovina
| | - Milica Matavulj
- Department of Histology and Embryology, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | - Lana Nežić
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Tatjana Nožica Radulović
- Institute for Physical Medicine and Rehabilitation "Dr. Miroslav Zotović", Banja Luka, Bosnia and Herzegovina
| | - Ranko Škrbić
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| |
Collapse
|
13
|
Tian S, Gao J, Gong H, Zhang X, Wang S. Effects of whole-body vibration at different periods on lumbar vertebrae in female rats. Med Eng Phys 2022; 110:103918. [PMID: 36564133 DOI: 10.1016/j.medengphy.2022.103918] [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: 03/08/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022]
Abstract
The current study aimed to investigate the effects of whole-body vibration (WBV) before and after ovariectomy on lumbar vertebrae, and to observe whether the positive effects of WBV before and after ovariectomy on lumbar vertebrae in rats could be maintained after vibration stopped. Three-month-old female rats were divided into four groups (n = 45/group): control (CON), ovariectomy (OVA), WBV before ovariectomy (WBV-BO), and WBV after ovariectomy (WBV-AO) groups. For 1-8 weeks, WBV-BO group was subjected to vertical WBV. At the 9th week, the rats in WBV-BO, WBV-AO, and OVA groups were ovariectomized. During 11-18 weeks, WBV-AO group was subjected to vibration. For 19-26 weeks, no intervention was done for rats. The lumbar vertebrae were examined by Micro-CT, compressive test, creep test, and microindentation test. At the 8th week, the displacement of the L1-L2 annulus fibrosus in WBV-BO group was 18% smaller compared with CON group (p<0.05). At the 18th week, the elastic modulus of the L5 vertebral body in WBV-BO and WBV-AO groups was 53% and 57% higher than that in CON group, respectively (p<0.05); the displacement of the L1-L2 annulus fibrosus in WBV-BO group was 25% smaller than those in the other groups (p<0.05). At the 26th week, there was no significant difference in the displacement of the L1-L2 annulus fibrosus between WBV-BO group and other groups (p>0.05); the elastic modulus of the L5 vertebral body had no significant difference between WBV-AO group and CON group (p>0.05). Our results demonstrated that WBV before ovariectomy effectively prevented disc degeneration with significant effects up to 8 weeks after ovariectomy. The vertebral mechanical properties could be significantly improved by WBV after ovariectomy, but the residual effect did not maintain after WBV stopped.
Collapse
Affiliation(s)
- Sujing Tian
- Department of Engineering Mechanics, Jilin University, Changchun, Jilin, 130025, China
| | - Jiazi Gao
- Department of Engineering Mechanics, Jilin University, Changchun, Jilin, 130025, China
| | - He Gong
- Department of Engineering Mechanics, Jilin University, Changchun, Jilin, 130025, China.
| | - Xiang Zhang
- Department of Engineering Mechanics, Jilin University, Changchun, Jilin, 130025, China
| | - Shuai Wang
- Department of Engineering Mechanics, Jilin University, Changchun, Jilin, 130025, China
| |
Collapse
|
14
|
Zhou J, Wang J, Qu M, Huang X, Yin L, Liao Y, Huang F, Ning P, Zhong P, Zeng Y. Effect of the Pulsed Electromagnetic Field Treatment in a Rat Model of Senile Osteoporosis In Vivo. Bioelectromagnetics 2022; 43:438-447. [DOI: 10.1002/bem.22423] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 10/30/2022] [Indexed: 11/21/2022]
Affiliation(s)
- Jun Zhou
- Department of Rehabilitation, Rehabilitation Medicine Center, Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School University of South China Hengyang China
| | - Jinling Wang
- Department of Rehabilitation, Rehabilitation Medicine Center, Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School University of South China Hengyang China
| | - Mengjian Qu
- Department of Rehabilitation, Rehabilitation Medicine Center, Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School University of South China Hengyang China
| | - Xiarong Huang
- Department of Rehabilitation, Rehabilitation Medicine Center, Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School University of South China Hengyang China
| | - Linwei Yin
- Department of Rehabilitation, Rehabilitation Medicine Center, Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School University of South China Hengyang China
| | - Yang Liao
- Department of Rehabilitation, Rehabilitation Medicine Center, Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School University of South China Hengyang China
| | - Fujin Huang
- Department of Rehabilitation, Rehabilitation Medicine Center, Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School University of South China Hengyang China
| | - Pengyun Ning
- Department of Rehabilitation, Rehabilitation Medicine Center, Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School University of South China Hengyang China
| | - Peirui Zhong
- Department of Rehabilitation, Rehabilitation Medicine Center, Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School University of South China Hengyang China
| | - Yahua Zeng
- Department of Rehabilitation, Rehabilitation Medicine Center, Rehabilitation Laboratory, The First Affiliated Hospital, Hengyang Medical School University of South China Hengyang China
| |
Collapse
|
15
|
A novel ceRNA regulatory network involving the long noncoding NEAT1, miRNA-466f-3p and its mRNA target in osteoblast autophagy and osteoporosis. J Mol Med (Berl) 2022; 100:1629-1646. [PMID: 36169673 DOI: 10.1007/s00109-022-02255-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 08/10/2022] [Accepted: 08/31/2022] [Indexed: 12/14/2022]
Abstract
Osteoporosis (OP) is a systemic metabolic disorder characterized by a reduction in bone tissue volume. LncRNAs have been reported to act as regulators of several human diseases. Specifically, lncRNA nuclear paraspeckle assembly transcript 1 (NEAT1) is involved in proliferation, differentiation and apoptosis in osteoclasts and bone marrow mesenchymal stem cells and regulates the occurrence and development of OP. However, the relationship between NEAT1 and osteoblast autophagy and its mechanism are still unclear. Western blotting of LC3 and P62 was used to evaluate the effect of fluid shear stress (FSS) on autophagy in MC3T3-E1 osteoblasts. Total transcriptome sequencing and bioinformatics analyses were performed on osteoblasts loaded with and without FSS. qPCR was performed to examine the expression of NEAT1 in OP bone tissues and osteoblasts. The RNA-FISH was performed to study the localization of lncRNA NEAT1 and miR-466f-3p in MC3T3-E1 osteoblasts. In vitro, western blotting, transmission electron microscopy (TEM), immunofluorescence (IF) staining and qPCR were performed to verify the biological functions of NEAT1, miR-466f-3p and HK2. Subsequently, we conducted bioinformatics analysis and dual luciferase reporter assays to identify the relationships among NEAT1, miR-466f-3p and HK2. Additionally, rescue assays were conducted on osteoblasts to clarify the regulatory network of the NEAT1/miR-466f-3p/HK2 signalling pathway. In vivo, the OVX mouse model was used to investigate the effects of si-NEAT1 on autophagy in OP mice. The distal femur and serum were collected for further micro-CT analysis, blood biochemistry, and haematoxylin-eosin and Alizarin red staining (ARS). Immunohistochemistry (IHC) was performed to assess the protein expression of LC3 and HK2. NEAT1 expression was upregulated in OP tissues and osteoblast lines exposed to FSS. Knockdown of NEAT1 inhibited autophagy in vitro and in vivo. Further studies demonstrated that NEAT1 positively regulated HK2 expression via its competing endogenous RNA effects on miR-466f-3p. Moreover, we found the NEAT1/miR-466f-3p/HK2 axis regulated autophagy in osteoblasts. Knocking down NEAT1 inhibited autophagy in osteoblasts via the miR-466f-3p/HK2 signalling pathway, which may provide new ideas for novel molecular therapeutic targets of postmenopausal OP. KEY MESSAGES: • Fluid shear stress (FSS) can promote autophagy of osteoblast and performed transcriptome sequencing. • NEAT1 is overexpressed in osteoporosis and could regulate osteoblast cells autophagy. • Knockdown of lncRNA NEAT1 inhibited osteoblast cells autophagy by sponging miRNA-466f-3p and targeting HK2 in osteoporosis.
Collapse
|
16
|
Liu X, Gao X, Tong J, Yu L, Xu M, Zhang J. Improvement of Osteoporosis in Rats With Hind-Limb Unloading Treated With Pulsed Electromagnetic Field and Whole-Body Vibration. Phys Ther 2022; 102:6652209. [PMID: 35906872 DOI: 10.1093/ptj/pzac097] [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: 04/30/2021] [Revised: 01/22/2022] [Accepted: 04/24/2022] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Physical factors have been used to address disuse osteoporosis, but their effects and mechanism remain unclear. The purpose of this study was to determine the effects of pulsed electromagnetic field (PEMF) and whole-body vibration (WBV) on disuse osteoporosis to increase knowledge about treating osteoporosis. METHODS A disuse osteoporosis rat model was developed by hind-limb unloading (HU) for 6 weeks. Forty 4-month-old female Sprague-Dawley rats were divided into 5 groups and given the following interventions: HU, HU treated with PEMF (HUP), HU treated with WBV (HUW), HU treated with both PEMF and WBV (HUPW), and no intervention (controls). After 8 weeks of intervention, measurements were taken. RESULTS HU induced a decrease in bone mineral density (BMD), whereas HUP, HUW, and HUPW increased it. Moreover, the bone resorption markers tartrate-resistant acid phosphatase (TRAP) and C-terminal peptide of type 1 collagen in the HU group significantly increased, whereas the osteogenesis markers osteocalcin and N-terminal propeptide of type 1 procollagen significantly decreased. The markers osteocalcin and N-terminal propeptide of type 1 procollagen significantly increased, but TRAP and C-terminal peptide of type 1 collagen significantly decreased in the HUPW, HUP, and HUW groups compared with the HU group. In particular, HUPW effectively increased osteocalcin and decreased TRAP compared with HUP and WBV. Microcomputed tomography analysis of the femur indicated that HUPW improved trabecular number, bone volume over total volume, bone surface over bone volume, trabecular separation, and the structure model index compared with HUP and that it improved bone surface over bone volume, trabecular separation, and structure model index compared with HUW. The HUPW group showed a significant increase in maximum load compared with the HUW group and a significant increase in elastic modulus compared with the HUP group. CONCLUSION PEMF, WBV, and their combination all attenuated bone resorption and enhanced osteogenesis. WBV and the combination of treatments have great potential to improve osteogenesis compared with PEMF. In addition, HUPW significantly attenuated bone resorption compared with HUW and HUP. IMPACT The results of this study indicated that HUPW could effectively improve disuse osteoporosis compared with HUP, given that trabecular number and bone volume over total volume are associated with disuse osteoporosis. Moreover, BMD recovered well with HUP, HUW, and HUPW but the bone structure-especially mechanical performance-did not, indicating that osteoporosis should be evaluated with BMD and mechanical performance, not with BMD in isolation.
Collapse
Affiliation(s)
- Xifang Liu
- Key Laboratory of Biomedical Information Engineering of Education Ministry, Institute of Health and Rehabilitation Science, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China.,Honghui Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Xiaohang Gao
- Key Laboratory of Biomedical Information Engineering of Education Ministry, Institute of Health and Rehabilitation Science, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Jie Tong
- Key Laboratory of Biomedical Information Engineering of Education Ministry, Institute of Health and Rehabilitation Science, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Liyin Yu
- Key Laboratory of Biomedical Information Engineering of Education Ministry, Institute of Health and Rehabilitation Science, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Minglong Xu
- State Key Laboratory for Strength and Vibration of Mechanical Structures, School of Aerospace, Xi'an Jiaotong University, Xi'an, China
| | - Jianbao Zhang
- Key Laboratory of Biomedical Information Engineering of Education Ministry, Institute of Health and Rehabilitation Science, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
17
|
Ganse B, Orth M, Roland M, Diebels S, Motzki P, Seelecke S, Kirsch SM, Welsch F, Andres A, Wickert K, Braun BJ, Pohlemann T. Concepts and clinical aspects of active implants for the treatment of bone fractures. Acta Biomater 2022; 146:1-9. [PMID: 35537678 DOI: 10.1016/j.actbio.2022.05.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/24/2022] [Accepted: 05/02/2022] [Indexed: 12/17/2022]
Abstract
Nonunion is a complication of long bone fractures that leads to disability, morbidity and high costs. Early detection is difficult and treatment through external stimulation and revision surgery is often a lengthy process. Therefore, alternative diagnostic and therapeutic options are currently being explored, including the use of external and internal sensors. Apart from monitoring fracture stiffness and displacement directly at the fracture site, it would be desirable if an implant could also vary its stiffness and apply an intervention to promote healing, if needed. This could be achieved either by a predetermined protocol, by remote control, or even by processing data and triggering the intervention itself (self-regulated 'intelligent' or 'smart' implant). So-called active or smart materials like shape memory alloys (SMA) have opened up opportunities to build active implants. For example, implants could stimulate fracture healing by active shortening and lengthening via SMA actuator wires; by emitting pulses, waves, or electromagnetic fields. However, it remains undefined which modes of application, forces, frequencies, force directions, time durations and periods, or other stimuli such implants should ideally deliver for the best result. The present paper reviews the literature on active implants and interventions for nonunion, discusses possible mechanisms of active implants and points out where further research and development are needed to build an active implant that applies the most ideal intervention. STATEMENT OF SIGNIFICANCE: Early detection of delays during fracture healing and timely intervention are difficult due to limitations of the current diagnostic strategies. New diagnostic options are under evaluation, including the use of external and internal sensors. In addition, it would be desirable if an implant could actively facilitate healing ('Intelligent' or 'smart' implant). Implants could stimulate fracture healing via active shortening and lengthening; by emitting pulses, waves, or electromagnetic fields. No such implants exist to date, but new composite materials and alloys have opened up opportunities to build such active implants, and several groups across the globe are currently working on their development. The present paper is the first review on this topic to date.
Collapse
|
18
|
Pulsed Electro-Magnetic Field (PEMF) Effect on Bone Healing in Animal Models: A Review of Its Efficacy Related to Different Type of Damage. BIOLOGY 2022; 11:biology11030402. [PMID: 35336776 PMCID: PMC8945722 DOI: 10.3390/biology11030402] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/18/2022] [Accepted: 03/03/2022] [Indexed: 11/17/2022]
Abstract
Simple Summary Pulsed electromagnetic fields (PEMFs) are a type of biophysical stimulation that has been shown to be effective in improving bone regeneration and preventing bone loss. Their use dates back to the 1970s, but a gold standard treatment protocol has not yet been defined. PEMF efficacy relies on the generation of biopotentials, which activate several molecular pathways. There is currently no clear understanding of the effects on bone healing and, in addition, there are several animal models relevant to this issue. Therefore, drawing guidelines and conclusions from the analysis of the studies is difficult. In vivo investigations on PEMF stimulation are reviewed in this paper, focusing on molecular and morphological improvements in bone. Currently, there is little knowledge about the biological mechanism of PEMF and its effect on bone healing. This is due to the variability of crucial characteristics of electro-magnetic fields, such as amplitude and exposure frequency, which may influence the type of biological response. Furthermore, a different responsiveness of cells involved in the bone healing process is documented. Heterogeneous setting parameters and different outcome measures are considered in various animal models. Therefore, achieving comparable results is difficult. Abstract Biophysical energies are a versatile tool to stimulate tissues by generating biopotentials. In particular, pulsed electromagnetic field (PEMF) stimulation has intrigued researchers since the 1970s. To date, many investigations have been carried out in vivo, but a gold standard treatment protocol has not yet been defined. The main obstacles are represented by the complex setting of PEMF characteristics, the variety of animal models (including direct and indirect bone damage) and the lack of a complete understanding of the molecular pathways involved. In the present review the main studies about PEMF stimulation in animal models with bone impairment were reviewed. PEMF signal characteristics were investigated, as well as their effect on molecular pathways and osseous morphological features. We believe that this review might be a useful starting point for a prospective study in a clinical setting. Consistent evidence from the literature suggests a potential beneficial role of PEMF in clinical practice. Nevertheless, the wide variability of selected parameters (frequency, duration, and amplitude) and the heterogeneity of applied protocols make it difficult to draw certain conclusions about PEMF effectiveness in clinical implementation to promote bone healing. Deepening the knowledge regarding the most consistent results reported in literature to date, we believe that this review may be a useful starting point to propose standardized experimental guidelines. This might provide a solid base for further controlled trials, to investigate PEMF efficacy in bone damage conditions during routine clinical practice.
Collapse
|
19
|
Wang Q, Zhou J, Wang X, Xu Y, Liang Z, Gu X, He C. Coupling induction of osteogenesis and type H vessels by pulsed electromagnetic fields in ovariectomy-induced osteoporosis in mice. Bone 2022; 154:116211. [PMID: 34560308 DOI: 10.1016/j.bone.2021.116211] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 09/13/2021] [Accepted: 09/17/2021] [Indexed: 02/08/2023]
Abstract
The growth of blood vessels and osteogenesis are coupled in bone tissue. A specialized subset of CD31hiEndomucinhi (CD31hiEmcnhi) vascular endothelium in bone has been identified to positively regulate bone formation. Pulsed electromagnetic field (PEMF) can promote the facture healing and reverse the loss of bone mass. However, the underlying mechanisms mediating in the positive effects of PEMF on bone mass accrual remain unclear. In the ovariectomized (OVX) osteoporotic mouse model, PEMF with specific parameters was administrated after 12 weeks of surgery and continued for 8 weeks. μCT analysis, quantitative PCR and Elisa assays were used to assess the PEMF-induced the osteogenesis, while immunostaining and flow cytometry were used to evaluate the abundance of CD31hiEmcnhi endothelium in the metaphysis near the growth plate. Administration of PEMF substantially countered OVX-induced bone loss as shown by greater trabecular bone, higher expression of Osterix, PDGFB and Col-1a1 transcripts, and modulation of bone anabolic and catabolic activity. The PEMF-induced osteogenesis was coupled by the expansion of CD31hiEmcnhi endothelium as demonstrated by CD31 and Endomucin double-positive immunostaining and flow cytometry. Concurrently, the higher level of HIF-α was found in PEMF-treated mice than in vehicle controls. Notably, inhibition of HIF-1α considerably reduced PEMF-induced osteogenesis, and led to a remarkable decrease of CD31hiEmcnhi vessels in the PEMF-treated OVX mice. The present study demonstrated the PEMF-induced coupling promotion of osteogenesis and CD31hiEmcnhi endothelial cells in a mouse model of postmenopausal osteoporosis. This coupling effect might be mediated in HIF-1α signaling in CD31hiEmcnhi endothelium. These findings open up new directions of al that might enable therapeutic improvement of osteogenesis in patients with osteoporosis.
Collapse
Affiliation(s)
- Qian Wang
- Center of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Rehabilitation Key Laboratory of Sichuan Province, Sichuan University, Chengdu, Sichuan, China
| | - Jun Zhou
- Department of Rehabilitation, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Xiangxiu Wang
- Center of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Rehabilitation Key Laboratory of Sichuan Province, Sichuan University, Chengdu, Sichuan, China
| | - Yang Xu
- Center of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Rehabilitation Key Laboratory of Sichuan Province, Sichuan University, Chengdu, Sichuan, China
| | - Zhejun Liang
- Center of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Rehabilitation Key Laboratory of Sichuan Province, Sichuan University, Chengdu, Sichuan, China
| | - Xintong Gu
- Center of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Rehabilitation Key Laboratory of Sichuan Province, Sichuan University, Chengdu, Sichuan, China
| | - Chengqi He
- Center of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Rehabilitation Key Laboratory of Sichuan Province, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
20
|
Huang J, Li Y, Wang L, He C. Combined Effects of Low-Frequency Pulsed Electromagnetic Field and Melatonin on Ovariectomy-Induced Bone Loss in Mice. Bioelectromagnetics 2021; 42:616-628. [PMID: 34516671 DOI: 10.1002/bem.22372] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 08/12/2021] [Accepted: 09/01/2021] [Indexed: 02/05/2023]
Abstract
Pulsed electromagnetic field (PEMF) therapy and melatonin (MEL) supplementation are expected to be important strategies for the treatment of osteoporosis. The aim of the current study was to investigate the efficacy of PEMF therapy, MEL supplementation, a combination of PEMF therapy, and MEL supplementation (PEMF + MEL) in mice with bilateral ovariectomy (OVX)-induced osteoporosis. Forty 12-week-old female C57/BL mice were randomly assigned to five groups (n = 8/group): OVX, PEMF, MEL, PEMF + MEL, and sham-operation (sham) groups. All mice in the first four groups were subjected to OVX. The mice in the PEMF and PEMF + MEL groups were exposed to PEMF (75 Hz, 1.6 mT, 1 h/day for 12 weeks), while those in the MEL and PEMF + MEL groups were administered MEL (50 mg/kg, i.p.). Body mass, micro-computed tomography, histology, immunohistochemistry, and real-time polymerase chain reaction were performed. PEMF + MEL treatment enhanced bone volume fraction (BV/TV) 2.2-fold over OVX control (P < 0.001) and increased expression levels of collagen type I (COL1) 1.9-fold and bone morphogenetic protein 2 (BMP2) 2.5-fold. PEMF + MEL also reduced the ratio of bone surface/bone volume (BS/BV) by 40% (P < 0.05) and appeared to reduce the number of osteoclasts in the metaphysis area. Preservation of bone value and bone microarchitecture in the combined therapy group were found to be superior to those in the single treatment groups. However, there were no apparent differences between the PEMF and MEL groups. The use of a combination of PEMF therapy and MEL supplementation may be an effective method to treat osteoporosis. © 2021 Bioelectromagnetics Society.
Collapse
Affiliation(s)
- Jinming Huang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Li
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Liqiong Wang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Chengqi He
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
21
|
Zhou X, Guan Z, Jin X, Zhao J, Chen G, Ding J, Ren Y, Zhai X, Zhou Q, Guan Z. Reversal of alopecia areata, osteoporosis follow treatment with activation of Tgr5 in mice. Biosci Rep 2021; 41:BSR20210609. [PMID: 34196345 PMCID: PMC8292761 DOI: 10.1042/bsr20210609] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 04/28/2021] [Accepted: 04/30/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Alopecia areata is an autoimmune hair loss disease with infiltration of pro-inflammatory cells into hair follicles. The role of Tgr5 in dermatitis has attracted considerable attention. The present study aimed to investigate the effect of Tgr5 in the development of Alopecia areata. METHODS The study utilized a comparison control group design with four groups of wild-type group, wild-type+INT777 group, Tgr5-/- group, and Tgr5-/-+INT777 group. The mice were treated with INT777 (30 mg/kg/day) or the carrier solution (DMSO) intraperitoneally for 7 weeks, and the back skin was collected and analyzed by histology and immunohistochemistry staining. The lumbar vertebrae 4 has also been analyzed by DXA and Micro-CT. RESULTS Tgr5-/- mice displayed the decreasingly significant in hair area and length, skin thickness, and the ratio of anagen and telogen, collagen, and mast cell number and loss the bone mass than WT group. After treating with INT777, the appearance of alopecia areata and bone microstructure has improved. Immunohistochemistry and qPCR analysis showed that activation of Tgr5 can down-regulate the express of JAK1, STAT3, IL-6, TNF-α, and VEGF. CONCLUSION These findings indicate that activation of Tgr5 mediated amelioration of alopecia areata and osteoporosis by down-regulated JAK1-STAT3 signaling pathway.
Collapse
Affiliation(s)
- Xiaohui Zhou
- Qinghai Provincial People’s Hospital, Xining, Qinghai, P.R. China
| | - Zhiqiang Guan
- Department of Dermatology, The First People's Hospital of Xuzhou, Xuzhou, Jiangsu 221002, P.R. China
| | - Xiao Jin
- Department of Rheumatology and Immunology, The First People’s Hospital of Xuzhou, Xuzhou, Jiangsu 221002, P.R. China
| | - Jianbin Zhao
- Department of Dermatology, The First People's Hospital of Xuzhou, Xuzhou, Jiangsu 221002, P.R. China
| | - Guisheng Chen
- Department of Dermatology, Traditional Chinese Medicine Hospital of Xuzhou Jiangsu 221002, P.R. China
| | - Jicun Ding
- Department of Dermatology, The First People's Hospital of Xuzhou, Xuzhou, Jiangsu 221002, P.R. China
| | - Yile Ren
- Department of Rheumatology and Immunology, The First People’s Hospital of Xuzhou, Xuzhou, Jiangsu 221002, P.R. China
| | - Xiaoxiang Zhai
- Department of Dermatology, The Seventh People’s Hospital of Shanghai, Shanghai 200137, P.R. China
| | - Qiyun Zhou
- Qinghai Provincial People’s Hospital, Xining, Qinghai, P.R. China
| | - Zhiyuan Guan
- Department of Orthopedics, The Shanghai Tenth People's Hospital of Tongji University, Shanghai, P.R. China
- Department of Orthopedics, Peking University Third Hospital, Beijing, P.R. China
| |
Collapse
|
22
|
Wang L, Xie S, Zhu S, Gao C, He C. Efficacy of Pulsed Electromagnetic Fields on Experimental Osteopenia in Rodents: A Systematic Review. Bioelectromagnetics 2021; 42:415-431. [PMID: 34004034 DOI: 10.1002/bem.22348] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 04/27/2021] [Accepted: 05/01/2021] [Indexed: 02/05/2023]
Abstract
Osteoporosis leads to increased bone fragility and risk of fractures. Different strategies have been employed to reduce bone loss, including the use of a pulsed electromagnetic field (PEMF). Although many experimental studies have demonstrated the effect of PEMF on reduction of bone loss, the outcomes studied are varied and insufficient, and the quality of evidence is unknown. Therefore, the aim of this review was to assess the preclinical evidence on the effect of PEMF on bone loss. The existing challenges were also evaluated, and suggestions were provided to strengthen the quality of evidence in future studies. All original articles concerning the effect of PEMF on osteoporosis in animal models were included. Twenty-four studies met the inclusion criteria, 23 of which suggested that PEMF was effective in reducing bone loss, while one study failed to demonstrate any benefit. Risk of bias analysis suggested that information on key measures to reduce bias was frequently not reported. Animal models for osteoporosis, PEMF intervention regimens, outcomes, and specific bone detection sites seemed to influence the efficacy of PEMF in osteoporosis. Our results indicate the potential benefits of PEMF selection in animal models of osteoporosis. However, due to the heterogeneity of the parameters and the quality of the included literature, comprehensive studies using standardized protocols are warranted to confirm the results. © 2021 Bioelectromagnetics Society.
Collapse
Affiliation(s)
- Liqiong Wang
- Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, P.R. China.,Rehabilitation Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, P.R. China.,Institute for Disaster Management and Reconstruction, Sichuan University-The Hong Kong Polytechnic University, Chengdu, P.R. China
| | - Suhang Xie
- Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, P.R. China.,Rehabilitation Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Siyi Zhu
- Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, P.R. China.,Rehabilitation Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Chengfei Gao
- The Affiliated Hospital of Qingdao University, Qingdao, P.R. China
| | - Chengqi He
- Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, P.R. China.,Rehabilitation Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, P.R. China.,Institute for Disaster Management and Reconstruction, Sichuan University-The Hong Kong Polytechnic University, Chengdu, P.R. China
| |
Collapse
|
23
|
Zhou J, Gao YH, Zhu BY, He WF, Wang G, Xian CJ, Chen KM. The frequency window effect of sinusoidal electromagnetic fields in promoting osteogenic differentiation and bone formation involves extension of osteoblastic primary cilia and activation of protein kinase A. Cell Biol Int 2021; 45:1685-1697. [PMID: 33811714 DOI: 10.1002/cbin.11606] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 03/19/2021] [Accepted: 04/01/2021] [Indexed: 01/27/2023]
Abstract
Electromagnetic fields (EMFs) have emerged as a versatile means for osteoporosis treatment and prevention. However, its optimal application parameters are still elusive. Here, we optimized the frequency parameter first by cell culture screening and then by animal experiment validation. Osteoblasts isolated from newborn rats (ROBs) were exposed 90 min/day to 1.8 mT SEMFs at different frequencies (ranging from 10 to 100 Hz, interval of 10 Hz). SEMFs of 1.8 mT inhibited ROB proliferation at 30, 40, 50, 60 Hz, but increased proliferation at 10, 70, 80 Hz. SEMFs of 10, 50, and 70 Hz promoted ROB osteogenic differentiation and mineralization as shown by alkaline phosphatase (ALP) activity, calcium content, and osteogenesis-related molecule expression analyses, with 50 Hz showing greater effects than 10 and 70 Hz. Treatment of young rats with 1.8 mT SEMFs at 10, 50, or 100 Hz for 2 months significantly increased whole-body bone mineral density (BMD) and femur microarchitecture, with the 50 Hz group showing the greatest effect. Furthermore, 1.8 mT SEMFs extended primary cilia lengths of ROBs and increased protein kinase A (PKA) activation also in a frequency-dependent manner, again with 50 Hz SEMFs showing the greatest effect. Pretreatment of ROBs with the PKA inhibitor KT5720 abolished the effects of SEMFs to increase primary cilia length and promote osteogenic differentiation/mineralization. These results indicate that 1.8 mT SEMFs have a frequency window effect in promoting osteogenic differentiation/mineralization in ROBs and bone formation in growing rats, which involve osteoblast primary cilia length extension and PKA activation.
Collapse
Affiliation(s)
- Jian Zhou
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou, China.,Institute of Orthopaedics, The 940th Hospital of Joint Logistic Support Force, People's Liberation Army of China, Lanzhou, China
| | - Yu Hai Gao
- Institute of Orthopaedics, The 940th Hospital of Joint Logistic Support Force, People's Liberation Army of China, Lanzhou, China
| | - Bao Ying Zhu
- Institute of Orthopaedics, The 940th Hospital of Joint Logistic Support Force, People's Liberation Army of China, Lanzhou, China
| | - Wen Fang He
- Institute of Orthopaedics, The 940th Hospital of Joint Logistic Support Force, People's Liberation Army of China, Lanzhou, China
| | - Gang Wang
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou, China
| | - Cory J Xian
- UniSA Clinical & Health Science, University of South Australia, Adelaide, South Australia, Australia
| | - Ke Ming Chen
- Institute of Orthopaedics, The 940th Hospital of Joint Logistic Support Force, People's Liberation Army of China, Lanzhou, China
| |
Collapse
|
24
|
Xu YH, Sun YC, Liu J, Li HX, Huang CY, Pang YY, Wu T, Hu X. Serum Pharmacochemistry Analysis Combined with Network Pharmacology Approach to Investigate the Antiosteoporosis Effect of Xianlinggubao Capsule in vivo. PHARMACEUTICAL FRONTS 2021. [DOI: 10.1055/s-0041-1726301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
AbstractXianlinggubao capsule (XLGB) is a traditional Chinese medicine multi-component herbal prescription and has been widely used in osteoporosis (OP) treatment. However, the underlying anti-OP mechanisms of XLGB have not been fully studied. In this study, an ovariectomized rat model of OP was established. The OP rats were orally administrated with XLGB, and then the main absorbed components in serum sample were assessed based on liquid chromatography-tandem mass spectrometry (LC-MS/MS). Subsequently, the potential anti-OP markers in XLGB were screened based on a network pharmacology strategy. Molecular docking analysis was used for confirmation. LC-MS showed 22 absorbed components in the serum sample of OP rat with XLGB treatment. Network pharmacology and pathway analysis suggested 19 potential anti-OP markers in XLGB. According to molecular docking process, most of the potential markers displayed strong interactions with the 22 absorbed components mentioned above. Besides, an absorbed component–potential marker–pathway network was further established. In conclusion, our data suggested the possible mechanisms for XLGB in OP treatment, in which the “multicomponents, multitargets, and multipathways” participated. Our article provided possible direction for drug discovery in OP and could help for exploring novel application of XLGB in clinical setting.
Collapse
Affiliation(s)
- Yun-Hui Xu
- State Key Lab. of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
| | - Yi-Chun Sun
- Sinopharm Group Tongjitang (Guizhou) Pharmaceutical Co., Ltd., Guiyang, People's Republic of China
- Guangdong Efong Pharmaceutical Co., Ltd, Guangzhou, People's Republic of China
| | - Jie Liu
- State Key Lab. of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
| | - Hui-Xin Li
- Sinopharm Group Tongjitang (Guizhou) Pharmaceutical Co., Ltd., Guiyang, People's Republic of China
| | - Chun-Yue Huang
- State Key Lab. of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
| | - Yuan-Yuan Pang
- Sinopharm Group Tongjitang (Guizhou) Pharmaceutical Co., Ltd., Guiyang, People's Republic of China
| | - Tong Wu
- State Key Lab. of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
| | - Xiao Hu
- State Key Lab. of New Drug and Pharmaceutical Process, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
- Sinopharm Group Tongjitang (Guizhou) Pharmaceutical Co., Ltd., Guiyang, People's Republic of China
| |
Collapse
|
25
|
Matrix Vesicles: Role in Bone Mineralization and Potential Use as Therapeutics. Pharmaceuticals (Basel) 2021; 14:ph14040289. [PMID: 33805145 PMCID: PMC8064082 DOI: 10.3390/ph14040289] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/18/2021] [Accepted: 03/23/2021] [Indexed: 12/14/2022] Open
Abstract
Bone is a complex organ maintained by three main cell types: osteoblasts, osteoclasts, and osteocytes. During bone formation, osteoblasts deposit a mineralized organic matrix. Evidence shows that bone cells release extracellular vesicles (EVs): nano-sized bilayer vesicles, which are involved in intercellular communication by delivering their cargoes through protein–ligand interactions or fusion to the plasma membrane of the recipient cell. Osteoblasts shed a subset of EVs known as matrix vesicles (MtVs), which contain phosphatases, calcium, and inorganic phosphate. These vesicles are believed to have a major role in matrix mineralization, and they feature bone-targeting and osteo-inductive properties. Understanding their contribution in bone formation and mineralization could help to target bone pathologies or bone regeneration using novel approaches such as stimulating MtV secretion in vivo, or the administration of in vitro or biomimetically produced MtVs. This review attempts to discuss the role of MtVs in biomineralization and their potential application for bone pathologies and bone regeneration.
Collapse
|
26
|
Zhai M, Yan X, Liu J, Long Z, Zhao S, Li W, Liu Y, Hai C. Electromagnetic Fields Ameliorate Insulin Resistance and Hepatic Steatosis by Modulating Redox Homeostasis and SREBP-1c Expression in db/db Mice. Diabetes Metab Syndr Obes 2021; 14:1035-1042. [PMID: 33727836 PMCID: PMC7954280 DOI: 10.2147/dmso.s294020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 02/24/2021] [Indexed: 12/11/2022] Open
Abstract
PURPOSE The prevalence of nonalcoholic fatty liver disease (NAFLD), which has recently become known as metabolic-associated fatty liver disease (MAFLD), has risen. However, pharmacotherapies for this disease have not been approved. Electromagnetic fields (EMFs) have excellent bioeffects on multiple diseases. However, the effects of EMFs on NAFLD are unknown. This study investigated the bioeffects of EMF exposure on insulin resistance, liver redox homeostasis and hepatic steatosis in db/db mice. METHODS Animals were sacrificed after EMF exposure for 8 weeks. The fasting blood glucose and insulin levels in the serum were tested. The homeostatic model assessment of insulin resistance (HOMA-IR) was calculated by a formula. The levels of MDA, GSSG and GSH, biomarkers of redox, were assessed. The activities of CAT, SOD and GSH-Px were assessed. The body and liver weights were measured. Hepatic lipid accumulation was observed by Oil Red O staining. Hepatic CAT, GR, GSH-Px, SOD1, SOD2 and SREBP-1 expression was determined by Western blotting. RESULTS EMF exposure ameliorated insulin resistance and oxidative stress in the liver by downregulating the MDA and GSSG levels, increasing the reduced GSH levels, and promoting the GSH-Px levels in db/db mice. In addition, liver weight and triglyceride (TG) levels were reduced by EMF exposure. Simultaneously, EMF exposure improved hepatic steatosis by downregulating the protein expression of SREBP-1c. CONCLUSION The present findings suggest that EMF exposure has positive effects in the treatment of NAFLD.
Collapse
Affiliation(s)
- Mingming Zhai
- Department of Biomedical Engineering, Air Force Medical University, Xi’an, People’s Republic of China
| | - Xi Yan
- Department of Dermatology, The Second Affiliated Hospital, Air Force Medical University, Xi’an, People’s Republic of China
| | - Jiangzheng Liu
- Department of Toxicology, Shanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University, Xi’an, People’s Republic of China
| | - Zi Long
- Department of Toxicology, Shanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University, Xi’an, People’s Republic of China
| | - Siyan Zhao
- Institute of Nuclear Biological and Chemical Defence, Beijing, People’s Republic of China
| | - Wendan Li
- Institute of Nuclear Biological and Chemical Defence, Beijing, People’s Republic of China
| | - Ying Liu
- Institute of Nuclear Biological and Chemical Defence, Beijing, People’s Republic of China
- Ying Liu Institute of Nuclear Biological and Chemical Defence, No. 1, Yangfang Zhongxin North Street, Beijing, 102205, People’s Republic of China Email
| | - Chunxu Hai
- Department of Toxicology, Shanxi Provincial Key Lab of Free Radical Biology and Medicine, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Air Force Medical University, Xi’an, People’s Republic of China
- Correspondence: Chunxu Hai Air Force Medical University (AFMU), No. 169 Changle West Road, Xi’an, Shaanxi, 710032, People’s Republic of ChinaTel +86-29-84774879 Email
| |
Collapse
|
27
|
Shi F, Ni L, Gao YM. Tetrandrine Attenuates Cartilage Degeneration, Osteoclast Proliferation, and Macrophage Transformation through Inhibiting P65 Phosphorylation in Ovariectomy-induced Osteoporosis. Immunol Invest 2020; 51:465-479. [PMID: 33140671 DOI: 10.1080/08820139.2020.1837864] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND Osteoporosis is a common metabolic bone disease with high prevalence. Tetrandrine (TET) suppressed osteoclastogenesis, while the roles of TET in osteoporosis regulation remained unclear. Thus, the study aimed to investigate the effect of TET on osteoporosis and the underlying mechanism. METHODS The osteoporosis rabbit model was established through anterior cruciate ligament transection (ACLT) and bilateral ovariectomy (OVX). The degeneration of articular cartilage was assessed using HE staining and Alcian blue staining. The liver and kidney tissue injury was determined using HE staining. The activity of osteoclasts was evaluated using Tartrate-resistant acid phosphatase (TRAP) staining. The changes in bone structural parameters were determined through measuring the BMD, BV/TV, Tb.Th, Tb.N, and Tb.Sp, and the serum levels of calcium and phosphorus. Macrophage polarization was determined using Flow cytometry. RESULTS The bone structural parameters including BMD, BV/TV, Tb.N, Tb.Th and Tb.Sp were changed in osteoporosis rabbit, which was reversed by TET. Besides, TET suppressed the increased serum levels of calcium and phosphorus in osteoporosis rabbit. Furthermore, TET inhibited the degeneration of articular cartilage and the activity of osteoclasts induced by osteoporosis. Moreover, TET inhibited the levels of MMP-9, PPAR-γ, RANKL, β-CTX and TRACP-5b, and increased the levels of OPG, ALP and osteocalcin (OC) in osteoporosis. Additionally, TET promoted macrophage transformation from M1 to M2 in osteoporotic and inhibited the production of IL-1β, TNF-α, and IL-6. TET also inhibited the p65 phosphorylation in osteoporosis. Besides, TET reversed RANKL-induced osteoclasts proliferation, p65 phosphorylation, and the expression changes of RANKL, Ki67, PPAR-γ, ALP, OPG. CONCLUSION TET attenuated bone structural parameters including BMD, BV/TV, Tb.N, Tb.Th and Tb.Sp, inhibited articular cartilage degeneration, promoted bone formation, inhibited the inflammatory response, and promoted macrophage transformation from M1 to M2 via NF-κB inactivation in osteoporosis. TET may be a promising drug for osteoporosis therapy. ABBREVIATION TET: Tetrandrine; ACLT: anterior cruciate ligament transection; OVX: ovariectomy; TRAP: Tartrate-resistant acid phosphatase; BMD: bone mineral density; BV/TV: Bone volume/total volume; Tb.Th: trabecular thickness; Tb.N: trabecular number; Tb.Sp: trabecular separation; MMP-9: Matrix metallopeptidase 9; PPAR-γ: Peroxisome proliferator-activated receptor gamma; RANKL: Receptor activator of nuclear factor kappa-B ligand; OPG: Osteoprotegerin; ALP: alkaline phosphatase; OC: osteocalcin; β-CTX: β isomer of C-terminal telopeptide of type Ⅰ collagen; TRACP-5b: Tartrate-resistant acid phosphatase 5b; TNF-α: tumor necrosis factor-α; IL-1β: interleukin-1β; IL-6: interleukin 6; NF-κB: Nuclear factor kappa B; PKC-α: Protein kinase C alpha; qRT-PCR: Quantitative real-time polymerase chain reaction.
Collapse
Affiliation(s)
- Fang Shi
- Department of Traditional Chinese Medicine, Beijing JiShuitan Hospital, Beijing, China
| | - Lei Ni
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Ye-Mei Gao
- Department of Traditional Chinese Medicine, Beijing JiShuitan Hospital, Beijing, China
| |
Collapse
|
28
|
Harris D, Garrett K, Uppuganti S, Creecy A, Nyman JS. The BALB/c mouse as a preclinical model of the age-related deterioration in the lumbar vertebra. Bone 2020; 137:115438. [PMID: 32480022 PMCID: PMC7354228 DOI: 10.1016/j.bone.2020.115438] [Citation(s) in RCA: 8] [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: 04/03/2020] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 02/08/2023]
Abstract
The likelihood of experiencing an osteoporotic fracture of one or more vertebral bodies increases with age, and this increase is not solely due to sex steroid deficiency. For the purpose of assessing the effectiveness of novel therapeutic strategies in the prevention of vertebral fractures among the elderly, we hypothesized that the BALB/c mouse model of aging phenocopies the age-related decrease in human VB strength. To test this hypothesis, we assessed the age-related changes in trabecular architecture of the L6 VB, with respect to those in the distal femur metaphysis, between 6-mo. (young adulthood, n = 20/sex) and 20-mo. of age (old age, n = 18/sex) and then determined how well the architectural characteristics, volumetric bone mineral density (vBMD), and predicted failure force from μCT-derived finite element analysis (μFEA) with linear elastic failure criteria explained the age-related variance in VB strength, which was the ultimate force during quasi-static loading of the VB in compression. While there was a pronounced age-related deterioration in trabecular architecture in the distal femur metaphysis of female and male BALB/c mice, the decrease in trabecular bone volume fraction and trabecular number between 6-mo. and 20-mo. of age occurred in male mice, but not in female mice. As such, the VB strength was lower with age in males only. Nonetheless, BV/TV and volumetric bone mineral density (vBMD) positively correlated with the ultimate compressive force of the L6 VB for both females and males. Whether using a fixed homogeneous distribution of tissue modulus (Et = 18 GPa) or a heterogeneous distribution of Et based on a positive relationship with TMD, the predicted failure force of the VB was not independent of age, thereby suggesting linear μFEA may not be a suitable replacement for mechanical-based measurements of strength with respect to age-related changes. Overall, the BALB/c mouse model of aging mimics the age-related in decline in human VB strength when comparing 6-mo. and 20-mo. old male mice. The decrease in VB strength in female mice may occur over a different age range.
Collapse
Affiliation(s)
- Dominique Harris
- Meharry Medical College, 1005 Dr. DB Todd Jr. Blvd., Nashville, TN 37208, USA
| | - Kate Garrett
- Department of Orthopaedic Surgery, Vanderbilt University Medical Center, 1215 21(st) Ave. S., Suite 4200, Nashville, TN 37232, USA
| | - Sasidhar Uppuganti
- Department of Orthopaedic Surgery, Vanderbilt University Medical Center, 1215 21(st) Ave. S., Suite 4200, Nashville, TN 37232, USA; Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Amy Creecy
- Department of Orthopaedic Surgery, Vanderbilt University Medical Center, 1215 21(st) Ave. S., Suite 4200, Nashville, TN 37232, USA; Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, TN 37232, USA
| | - Jeffry S Nyman
- Department of Orthopaedic Surgery, Vanderbilt University Medical Center, 1215 21(st) Ave. S., Suite 4200, Nashville, TN 37232, USA; Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, TN 37232, USA; Department of Veterans Affairs, Tennessee Valley Healthcare System, 1310 24(th) Ave. S., Nashville, TN 37212, USA.
| |
Collapse
|
29
|
Liang Y, Zhu B, Li S, Zhai Y, Yang Y, Bai Z, Zeng Y, Li D. Curcumin protects bone biomechanical properties and microarchitecture in type 2 diabetic rats with osteoporosis via the TGFβ/Smad2/3 pathway. Exp Ther Med 2020; 20:2200-2208. [PMID: 32765696 PMCID: PMC7401480 DOI: 10.3892/etm.2020.8943] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 04/24/2020] [Indexed: 12/21/2022] Open
Abstract
Type 2 diabetic osteoporosis (T2DOP) has become a common secondary cause of osteoporosis that accelerates bone loss and leads to bone fractures. The aim of the current study was to investigate the association between the anti-osteoporotic effect of curcumin (Cur) and the transforming growth factor (TGF)β/Smads signaling pathway. Male Sprague-Dawley rats were used in the experiments. The type 2 diabetes mellitus (T2DM) animals were treated with Cur for 8 weeks and blood lipid markers, bone microstructure and bone biomechanics were then evaluated. The mRNA expression levels of TGFβ1, type I TGFβ receptor (TβRI), TβRII and Smad2/3 were determined using reverse transcription-quantitative PCR (RT-qPCR) and immunohistochemistry. The body weight of rats with type 2 diabetes-induced osteoporosis increased (P<0.05), while the lipid (total cholesterol, triglyceride and low-density lipoprotein) and fasting blood glucose levels were decreased by Cur (P<0.05). In addition, Cur significantly improved bone biomechanical properties (maximum load, breaking load, elastic load and the bone rigidity coefficient) and preserved bone microarchitecture (P<0.05). The RT-qPCR and IHC results revealed that Cur increased TGFβ1, TβRI, TβRII and Smad2/3 expression levels and promoted Smad2/3 phosphorylation in bones. The present results also indicated that Cur regulated lipid and glucose levels, improved bone biomechanical properties and preserved bone microarchitecture, and that these effects may be mediated via TGFβ/Smad2/3 pathway activation.
Collapse
Affiliation(s)
- Yanlong Liang
- Department of Basic Medicine, Zhaoqing Medical College, Zhaoqing, Guangdong 526020, P.R. China.,School of Computer Science and Software, Zhaoqing College, Zhaoqing, Guangdong 526061, P.R. China
| | - Benben Zhu
- Department of Pharmacy, Affiliated People's Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, P.R. China
| | - Shuhui Li
- Key Laboratory of Tropical Diseases, Faculty of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Yun Zhai
- Department of Basic Medicine, Zhaoqing Medical College, Zhaoqing, Guangdong 526020, P.R. China
| | - Yiqiu Yang
- Department of Basic Medicine, Zhaoqing Medical College, Zhaoqing, Guangdong 526020, P.R. China
| | - Zaixian Bai
- Department of Pharmacy, Affiliated People's Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, P.R. China
| | - Yuan Zeng
- Department of Pharmacy, People's Hospital of Nanxiong, Shaoguan, Guangdong 512400, P.R. China
| | - Dawei Li
- Department of Orthopedics and Traumatology, The Fourth Affiliated Hospital of Inner Mongolia Medical University, Baotou, Inner Mongolia 014030, P.R. China
| |
Collapse
|
30
|
Lin CC, Chang YT, Lin RW, Chang CW, Wang GJ, Lai KA. Single pulsed electromagnetic field restores bone mass and microarchitecture in denervation/disuse osteopenic mice. Med Eng Phys 2020; 80:52-59. [DOI: 10.1016/j.medengphy.2019.10.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 07/09/2019] [Accepted: 10/14/2019] [Indexed: 12/25/2022]
|
31
|
Li S, Wei C, Lv Y. Preparation and Application of Magnetic Responsive Materials in Bone Tissue Engineering. Curr Stem Cell Res Ther 2020; 15:428-440. [PMID: 31893995 DOI: 10.2174/1574888x15666200101122505] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/01/2019] [Accepted: 12/06/2019] [Indexed: 11/22/2022]
Abstract
At present, many kinds of materials are used for bone tissue engineering, such as polymer materials, metals, etc., which in general have good biocompatibility and mechanical properties. However, these materials cannot be controlled artificially after implantation, which may result in poor repair performance. The appearance of the magnetic response material enables the scaffolds to have the corresponding ability to the external magnetic field. Within the magnetic field, the magnetic response material can achieve the targeted release of the drug, improve the performance of the scaffold, and further have a positive impact on bone formation. This paper first reviewed the preparation methods of magnetic responsive materials such as magnetic nanoparticles, magnetic polymers, magnetic bioceramic materials and magnetic alloys in recent years, and then introduced its main applications in the field of bone tissue engineering, including promoting osteogenic differentiation, targets release, bioimaging, cell patterning, etc. Finally, the mechanism of magnetic response materials to promote bone regeneration was introduced. The combination of magnetic field treatment methods will bring significant progress to regenerative medicine and help to improve the treatment of bone defects and promote bone tissue repair.
Collapse
Affiliation(s)
- Song Li
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing, China
| | - Changling Wei
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing, China
| | - Yonggang Lv
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing, China
| |
Collapse
|
32
|
Suryani L, Too JH, Hassanbhai AM, Wen F, Lin DJ, Yu N, Teoh SH. Effects of Electromagnetic Field on Proliferation, Differentiation, and Mineralization of MC3T3 Cells. Tissue Eng Part C Methods 2020; 25:114-125. [PMID: 30661463 DOI: 10.1089/ten.tec.2018.0364] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
IMPACT STATEMENT We present the study about how the parameters of pulsed electromagnetic field (PEMF) stimulus affected calvarial osteoblast precursor cell in terms of growth, viability, and differentiation. This research provides insight and foundation to clinical application of noninvasive therapy using PEMF to improve bone regeneration.
Collapse
Affiliation(s)
- Luvita Suryani
- 1 School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore.,2 Centre for Developmental Biology, Tissue Engineering, Regenerative Medicine and Innovation, Singapore, Singapore
| | - Jian Hui Too
- 3 National Dental Centre Singapore, Singapore, Singapore
| | - Ammar Mansoor Hassanbhai
- 1 School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore.,2 Centre for Developmental Biology, Tissue Engineering, Regenerative Medicine and Innovation, Singapore, Singapore
| | - Feng Wen
- 1 School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore.,2 Centre for Developmental Biology, Tissue Engineering, Regenerative Medicine and Innovation, Singapore, Singapore
| | - Daryl Jimian Lin
- 1 School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | - Na Yu
- 3 National Dental Centre Singapore, Singapore, Singapore.,4 Duke-NUS Medical School Singapore, Singapore, Singapore
| | - Swee-Hin Teoh
- 1 School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore.,2 Centre for Developmental Biology, Tissue Engineering, Regenerative Medicine and Innovation, Singapore, Singapore.,5 Lee Kong Chian School of Medicine Singapore, Singapore, Singapore
| |
Collapse
|
33
|
Li J, Li X, Liu D, Hamamura K, Wan Q, Na S, Yokota H, Zhang P. eIF2α signaling regulates autophagy of osteoblasts and the development of osteoclasts in OVX mice. Cell Death Dis 2019; 10:921. [PMID: 31801950 PMCID: PMC6892793 DOI: 10.1038/s41419-019-2159-z] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 11/05/2019] [Accepted: 11/11/2019] [Indexed: 12/22/2022]
Abstract
Bone loss in postmenopausal osteoporosis is induced chiefly by an imbalance of bone-forming osteoblasts and bone-resorbing osteoclasts. Salubrinal is a synthetic compound that inhibits de-phosphorylation of eukaryotic translation initiation factor 2 alpha (eIF2α). Phosphorylation of eIF2α alleviates endoplasmic reticulum (ER) stress, which may activate autophagy. We hypothesized that eIF2α signaling regulates bone homeostasis by promoting autophagy in osteoblasts and inhibiting osteoclast development. To test the hypothesis, we employed salubrinal to elevate the phosphorylation of eIF2α in an ovariectomized (OVX) mouse model and cell cultures. In the OVX model, salubrinal prevented abnormal expansion of rough ER and decreased the number of acidic vesiculars. It regulated ER stress-associated signaling molecules such as Bip, p-eIF2α, ATF4 and CHOP, and promoted autophagy of osteoblasts via regulation of eIF2α, Atg7, LC3, and p62. Salubrinal markedly alleviated OVX-induced symptoms such as reduction of bone mineral density and bone volume fraction. In primary bone-marrow-derived cells, salubrinal increased the differentiation of osteoblasts, and decreased the formation of osteoclasts by inhibiting nuclear factor of activated T-cells cytoplasmic 1 (NFATc1). Live cell imaging and RNA interference demonstrated that suppression of osteoclastogenesis is in part mediated by Rac1 GTPase. Collectively, this study demonstrates that ER stress-autophagy axis plays an important role in OVX mice. Bone-forming osteoblasts are restored by maintaining phosphorylation of eIF2α, and bone-resorbing osteoclasts are regulated by inhibiting NFATc1 and Rac1 GTPase.
Collapse
Affiliation(s)
- Jie Li
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, 300070, Tianjin, China
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, 300070, Tianjin, China
| | - Xinle Li
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, 300070, Tianjin, China
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, 300070, Tianjin, China
| | - Daquan Liu
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, 300070, Tianjin, China
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, 300070, Tianjin, China
| | - Kazunori Hamamura
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Qiaoqiao Wan
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Sungsoo Na
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Hiroki Yokota
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Ping Zhang
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, 300070, Tianjin, China.
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, 300070, Tianjin, China.
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN, 46202, USA.
- Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin Medical University, 300052, Tianjin, China.
| |
Collapse
|
34
|
Ehnert S, Schröter S, Aspera-Werz RH, Eisler W, Falldorf K, Ronniger M, Nussler AK. Translational Insights into Extremely Low Frequency Pulsed Electromagnetic Fields (ELF-PEMFs) for Bone Regeneration after Trauma and Orthopedic Surgery. J Clin Med 2019; 8:2028. [PMID: 31756999 PMCID: PMC6947624 DOI: 10.3390/jcm8122028] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/15/2019] [Accepted: 11/18/2019] [Indexed: 02/07/2023] Open
Abstract
The finding that alterations in electrical potential play an important role in the mechanical stimulation of the bone provoked hype that noninvasive extremely low frequency pulsed electromagnetic fields (ELF-PEMF) can be used to support healing of bone and osteochondral defects. This resulted in the development of many ELF-PEMF devices for clinical use. Due to the resulting diversity of the ELF-PEMF characteristics regarding treatment regimen, and reported results, exposure to ELF-PEMFs is generally not among the guidelines to treat bone and osteochondral defects. Notwithstanding, here we show that there is strong evidence for ELF-PEMF treatment. We give a short, confined overview of in vitro studies investigating effects of ELF-PEMF treatment on bone cells, highlighting likely mechanisms. Subsequently, we summarize prospective and blinded studies, investigating the effect of ELF-PEMF treatment on acute bone fractures and bone fracture non-unions, osteotomies, spinal fusion, osteoporosis, and osteoarthritis. Although these studies favor the use of ELF-PEMF treatment, they likewise demonstrate the need for more defined and better controlled/monitored treatment modalities. However, to establish indication-oriented treatment regimen, profound knowledge of the underlying mechanisms in the sense of cellular pathways/events triggered is required, highlighting the need for more systematic studies to unravel optimal treatment conditions.
Collapse
Affiliation(s)
- Sabrina Ehnert
- Siegfried Weller Institute for Trauma Research, Depterment of Trauma and Reconstructive Surgery, BG Unfallklinik Tübingen, Eberhard Karls Universität Tübingen, D-72076 Tübingen, Germany; (S.S.); (R.H.A.-W.); (W.E.); (A.K.N.)
| | - Steffen Schröter
- Siegfried Weller Institute for Trauma Research, Depterment of Trauma and Reconstructive Surgery, BG Unfallklinik Tübingen, Eberhard Karls Universität Tübingen, D-72076 Tübingen, Germany; (S.S.); (R.H.A.-W.); (W.E.); (A.K.N.)
| | - Romina H. Aspera-Werz
- Siegfried Weller Institute for Trauma Research, Depterment of Trauma and Reconstructive Surgery, BG Unfallklinik Tübingen, Eberhard Karls Universität Tübingen, D-72076 Tübingen, Germany; (S.S.); (R.H.A.-W.); (W.E.); (A.K.N.)
| | - Wiebke Eisler
- Siegfried Weller Institute for Trauma Research, Depterment of Trauma and Reconstructive Surgery, BG Unfallklinik Tübingen, Eberhard Karls Universität Tübingen, D-72076 Tübingen, Germany; (S.S.); (R.H.A.-W.); (W.E.); (A.K.N.)
| | - Karsten Falldorf
- Sachtleben GmbH, Hamburg, Haus Spectrum am UKE, Martinistraße 64, D-20251 Hamburg, Germany; (K.F.); (M.R.)
| | - Michael Ronniger
- Sachtleben GmbH, Hamburg, Haus Spectrum am UKE, Martinistraße 64, D-20251 Hamburg, Germany; (K.F.); (M.R.)
| | - Andreas K. Nussler
- Siegfried Weller Institute for Trauma Research, Depterment of Trauma and Reconstructive Surgery, BG Unfallklinik Tübingen, Eberhard Karls Universität Tübingen, D-72076 Tübingen, Germany; (S.S.); (R.H.A.-W.); (W.E.); (A.K.N.)
| |
Collapse
|
35
|
Wu X, Ding J, Xu X, Wang X, Liu J, Jiang J, Liu Q, Kong G, Huang Z, Yang Z, Zhu Q. Ketogenic diet compromises vertebral microstructure and biomechanical characteristics in mice. J Bone Miner Metab 2019; 37:957-966. [PMID: 30968187 DOI: 10.1007/s00774-019-01002-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 03/19/2019] [Indexed: 12/31/2022]
Abstract
Ketogenic diet (KD) compromised the microstructure of cancellous bone and the mechanical property in the appendicular bone of mice, while the effects of KD on the axial bone have not been reported. This study aimed to compare the changes in the microstructure and mechanical properties of the forth lumbar (L4) vertebra in KD and ovariectomized (OVX) mice. Forty eight-week-old female C57BL/6J mice were assigned into four groups: SD (standard diet) + Sham, SD + OVX, KD + Sham, and KD + OVX groups. L4 vertebra was scanned by micro-CT to examine the microstructure of cancellous bone, after which simulative compression tests were performed using finite element (FE) analysis. Vertebral compressive test and histological staining of the L4 and L5 vertebrae were performed to observe the biomechanical and histomorphologic changes. The KD + Sham and SD + OVX exhibited a remarkable declination in the parameters of cancellous bone compared with the SD + Sham group, while KD + OVX demonstrated the most serious bone loss in the four groups. The stiffness was significantly higher in the SD + Sham group than the other three groups, but no difference was found between the remaining groups. The trabecular parameters were significantly correlated with the stiffness. Meanwhile, the OVX + Sham and KD + OVX groups showed a significant decrease in the failure load of compressive test, while there was no difference between the KD + Sham and SD + Sham groups. These findings suggest that KD may compromise the vertebral microstructure and compressive stiffness to a similar level as OVX did, indicating adverse effects of KD on the axial bone of the mice.
Collapse
Affiliation(s)
- Xiuhua Wu
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China
| | - Jianyang Ding
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China
| | - Xiaolin Xu
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China
| | - Xiaomeng Wang
- Department of Spinal Surgery, LongYan First Hospital, Longyan, Fujian, China
| | - Junhao Liu
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China
| | - Jie Jiang
- Department of Spinal Surgery, Hongdu Hospital of TCM, Nanchang, Jiangxi, China
| | - Qi Liu
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China
| | - Ganggang Kong
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China
| | - Zucheng Huang
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China
| | - Zhou Yang
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China
| | - Qingan Zhu
- Department of Spinal Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, China.
| |
Collapse
|
36
|
Bernardo R, Rodrigues A, Soares Dos Santos MP, Carneiro P, Lopes A, Sequeira Amaral J, Sequeira Amaral V, Morais R. Novel magnetic stimulation methodology for low-current implantable medical devices. Med Eng Phys 2019; 73:77-84. [PMID: 31477429 DOI: 10.1016/j.medengphy.2019.07.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 05/10/2019] [Accepted: 07/28/2019] [Indexed: 12/15/2022]
Abstract
Recent studies highlight the ability of inductive architectures to deliver therapeutic magnetic stimuli to target tissues and to be embedded into small-scale intracorporeal medical devices. However, to date, current micro-scale biomagnetic devices require very high electric current excitations (usually exceeding 1 A) to ensure the delivery of efficient magnetic flux densities. This is a critical problem as advanced implantable devices demand self-powering, stand-alone and long-term operation. This work provides, for the first time, a novel small-scale magnetic stimulation system that requires up to 50-fold lower electric current excitations than required by relevant biomagnetic technology recently proposed. Computational models were developed to analyse the magnetic stimuli distributions and densities delivered to cellular tissues during in vitro experiments, such that the feasibility of this novel stimulator can be firstly evaluated on cell culture tests. The results demonstrate that this new stimulative technology is able to deliver osteogenic stimuli (0.1-7 mT range) by current excitations in the 0.06-4.3 mA range. Moreover, it allows coil designs with heights lower than 1 mm without significant loss of magnetic stimuli capability. Finally, suitable core diameters and stimulator-stimulator distances allow to define heterogeneity or quasi-homogeneity stimuli distributions. These results support the design of high-sophisticated biomagnetic devices for a wide range of therapeutic applications.
Collapse
Affiliation(s)
- Rodrigo Bernardo
- Department of Mechanical Engineering, University of Aveiro, Aveiro, Portugal
| | - André Rodrigues
- Department of Mechanical Engineering, University of Aveiro, Aveiro, Portugal
| | - Marco P Soares Dos Santos
- Department of Mechanical Engineering, University of Aveiro, Aveiro, Portugal; Centre for Mechanical Technology and Automation (TEMA), University of Aveiro, Aveiro, Portugal; Associated Laboratory for Energy, Transports and Aeronautics (LAETA), Portugal.
| | - Pedro Carneiro
- Department of Mechanical Engineering, University of Aveiro, Aveiro, Portugal
| | - António Lopes
- Department of Physics, University of Aveiro, Aveiro, Portugal; Aveiro Institute of Materials, Aveiro, Portugal
| | - João Sequeira Amaral
- Department of Physics, University of Aveiro, Aveiro, Portugal; Aveiro Institute of Materials, Aveiro, Portugal
| | - Vítor Sequeira Amaral
- Department of Physics, University of Aveiro, Aveiro, Portugal; Aveiro Institute of Materials, Aveiro, Portugal
| | - Raul Morais
- University of Trás-os-Montes e Alto Douro, Vila Real, Portugal; Institute for Systems and Computer Engineering, Technology and Science (INESC TEC), Porto, Portugal
| |
Collapse
|
37
|
Galli C, Colangelo M, Pedrazzi G, Guizzardi S. The Response of Osteoblasts and Bone to Sinusoidal Electromagnetic Fields: Insights from the Literature. Calcif Tissue Int 2019; 105:127-147. [PMID: 30997574 DOI: 10.1007/s00223-019-00554-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 04/11/2019] [Indexed: 12/23/2022]
Abstract
Electromagnetic fields (EMFs) have been proposed as a tool to ameliorate bone formation and healing. Despite their promising results, however, they have failed to enter routine clinical protocols to treat bone conditions where higher bone mass has to be achieved. This is no doubt also due to a fundamental lack of knowledge and understanding on their effects and the optimal settings for attaining the desired therapeutic effects. This review analysed the available in vitro and in vivo studies that assessed the effects of sinusoidal EMFs (SEMFs) on bone and bone cells, comparing the results and investigating possible mechanisms of action by which SEMFs interact with tissues and cells. The effects of SEMFs on bone have not been as thoroughly investigated as pulsed EMFs; however, abundant evidence shows that SEMFs affect the proliferation and differentiation of osteoblastic cells, acting on multiple cellular mechanisms. SEMFs have also proven to increase bone mass in rodents under normal conditions and in osteoporotic animals.
Collapse
Affiliation(s)
- C Galli
- Department of Medicine and Surgery, University of Parma, Parma, Italy.
| | - M Colangelo
- Department of Medicine and Surgery, Histology and Embryology Lab, University of Parma, Parma, Italy
| | - G Pedrazzi
- Department of Medicine and Surgery, Neuroscience Unit, University of Parma, Via Volturno 39, 43126, Parma, Italy
| | - S Guizzardi
- Department of Medicine and Surgery, Histology and Embryology Lab, University of Parma, Parma, Italy
| |
Collapse
|
38
|
Zhou J, Gao YH, Zhu BY, Shao JL, Ma HP, Xian CJ, Chen KM. Sinusoidal Electromagnetic Fields Increase Peak Bone Mass in Rats by Activating Wnt10b/β-Catenin in Primary Cilia of Osteoblasts. J Bone Miner Res 2019; 34:1336-1351. [PMID: 30779853 DOI: 10.1002/jbmr.3704] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 01/31/2019] [Accepted: 02/12/2019] [Indexed: 12/22/2022]
Abstract
Extremely low-frequency electromagnetic fields have been considered a potential candidate for the prevention and treatment of osteoporosis; however, their action mechanism and optimal magnetic flux density (intensity) parameter are still elusive. The present study found that 50-Hz sinusoidal electromagnetic fields (SEMFs) at 1.8 mT increased the peak bone mass of young rats by increasing bone formation. Gene array expression studies with femoral bone samples showed that SEMFs increased the expression levels of collagen-1α1 and Wnt10b, a critical ligand of the osteogenic Wnt/β-catenin pathway. Consistently, SEMFs promoted osteogenic differentiation and maturation of rat calvarial osteoblasts (ROBs) in vitro through activating the Wnt10b/β-catenin pathway. This osteogenesis-promoting effect of SEMFs via Wnt10b/β-catenin signaling was found to depend on the functional integrity of primary cilia in osteoblasts. When the primary cilia were abrogated by small interfering RNA (siRNA) targeting IFT88, the ability of SEMFs to promote the osteogenic differentiation of ROBs through activating Wnt10b/β-catenin signaling was blocked. Although the knockdown of Wnt10b expression with RNA interference had no effect on primary cilia, it significantly suppressed the promoting effect of SEMFs on osteoblastic differentiation/maturation. Wnt10b was normally localized at the bases of primary cilia, but it disappeared (or was released) from the cilia upon SEMF treatment. Interestingly, primary cilia were elongated to different degrees by different intensities of 50-Hz SEMFs, with the window effect observed at 1.8 mT, and the expression level of Wnt10b increased in accord with the lengths of primary cilia. These results indicate that 50-Hz 1.8-mT SEMFs increase the peak bone mass of growing rats by promoting osteogenic differentiation/maturation of osteoblasts, which is mediated, at least in part, by Wnt10b at the primary cilia and the subsequent activation of Wnt/β-catenin signaling. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Jian Zhou
- Institute of Orthopaedics, Lanzhou General Hospital, CPLA, Lanzhou 730050, People's Republic of China
| | - Yu-Hai Gao
- Institute of Orthopaedics, Lanzhou General Hospital, CPLA, Lanzhou 730050, People's Republic of China
| | - Bao-Ying Zhu
- Institute of Orthopaedics, Lanzhou General Hospital, CPLA, Lanzhou 730050, People's Republic of China
| | - Jia-Le Shao
- Institute of Orthopaedics, Lanzhou General Hospital, CPLA, Lanzhou 730050, People's Republic of China
| | - Hui-Ping Ma
- Department of Pharmacy, Lanzhou General Hospital, CPLA, Lanzhou 730050, People's Republic of China
| | - Cory J Xian
- School of Pharmacy and Medical Sciences, and UniSA Cancer Research Institute, University of South Australia, Adelaide, SA 5001, Australia
| | - Ke-Ming Chen
- Institute of Orthopaedics, Lanzhou General Hospital, CPLA, Lanzhou 730050, People's Republic of China
| |
Collapse
|
39
|
Galli C, Pedrazzi G, Guizzardi S. The cellular effects of Pulsed Electromagnetic Fields on osteoblasts: A review. Bioelectromagnetics 2019; 40:211-233. [PMID: 30908726 DOI: 10.1002/bem.22187] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 03/08/2019] [Indexed: 12/12/2022]
Abstract
Electromagnetic fields (EMFs) have long been known to interact with living organisms and their cells and to bear the potential for therapeutic use. Among the most extensively investigated applications, the use of Pulsed EMFs (PEMFs) has proven effective to ameliorate bone healing in several studies, although the evidence is still inconclusive. This is due in part to our still-poor understanding of the mechanisms by which PEMFs act on cells and affect their functions and to an ongoing lack of consensus on the most effective parameters for specific clinical applications. The present review has compared in vitro studies on PEMFs on different osteoblast models, which elucidate potential mechanisms of action for PEMFs, up to the most recent insights into the role of primary cilia, and highlight the critical issues underlying at least some of the inconsistent results in the available literature. Bioelectromagnetics. 2019;9999:XX-XX. © 2019 Bioelectromagnetics Society.
Collapse
Affiliation(s)
- Carlo Galli
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Giuseppe Pedrazzi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Stefano Guizzardi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| |
Collapse
|
40
|
Soares Dos Santos MP, Coutinho J, Marote A, Sousa B, Ramos A, Ferreira JAF, Bernardo R, Rodrigues A, Marques AT, Cruz E Silva OABD, Furlani EP, Simões JAO, Vieira SI. Capacitive technologies for highly controlled and personalized electrical stimulation by implantable biomedical systems. Sci Rep 2019; 9:5001. [PMID: 30899061 PMCID: PMC6428833 DOI: 10.1038/s41598-019-41540-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 02/28/2019] [Indexed: 11/09/2022] Open
Abstract
Cosurface electrode architectures are able to deliver personalized electric stimuli to target tissues. As such, this technology holds potential for a variety of innovative biomedical devices. However, to date, no detailed analyses have been conducted to evaluate the impact of stimulator architecture and geometry on stimuli features. This work characterizes, for the first time, the electric stimuli delivered to bone cellular tissues during in vitro experiments, when using three capacitive architectures: stripped, interdigitated and circular patterns. Computational models are presented that predict the influence of cell confluence, cosurface architecture, electrodes geometry, gap size between electrodes and power excitation on the stimuli delivered to cellular layers. The results demonstrate that these stimulators are able to deliver osteoconductive stimuli. Significant differences in stimuli distributions were observed for different stimulator designs and different external excitations. The thickness specification was found to be of utmost importance. In vitro experiments using an osteoblastic cell line highlight that cosurface stimulation at a low frequency can enhance osteoconductive responses, with some electrode-specific differences being found. A major feature of this type of work is that it enables future detailed analyses of stimuli distribution throughout more complex biological structures, such as tissues and organs, towards sophisticated biodevice personalization.
Collapse
Affiliation(s)
- Marco P Soares Dos Santos
- Centre for Mechanical Technology & Automation (TEMA), University of Aveiro, Aveiro, Portugal.
- Department of Mechanical Engineering, University of Aveiro, Aveiro, Portugal.
- Associated Laboratory for Energy, Transports and Aeronautics (LAETA), Porto, Portugal.
| | - J Coutinho
- Department of Mechanical Engineering, University of Aveiro, Aveiro, Portugal
| | - Ana Marote
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Bárbara Sousa
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - A Ramos
- Centre for Mechanical Technology & Automation (TEMA), University of Aveiro, Aveiro, Portugal
- Department of Mechanical Engineering, University of Aveiro, Aveiro, Portugal
| | - Jorge A F Ferreira
- Centre for Mechanical Technology & Automation (TEMA), University of Aveiro, Aveiro, Portugal
- Department of Mechanical Engineering, University of Aveiro, Aveiro, Portugal
| | - Rodrigo Bernardo
- Department of Mechanical Engineering, University of Aveiro, Aveiro, Portugal
| | - André Rodrigues
- Department of Mechanical Engineering, University of Aveiro, Aveiro, Portugal
| | - A Torres Marques
- Associated Laboratory for Energy, Transports and Aeronautics (LAETA), Porto, Portugal
- Mechanical Engineering Department, University of Porto, 4200-465, Porto, Portugal
| | - Odete A B da Cruz E Silva
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Edward P Furlani
- Department of Chemical and Biological Engineering, Department of Electrical Engineering, University at Buffalo, SUNY, Buffalo, NY, USA
| | - José A O Simões
- Department of Mechanical Engineering, University of Aveiro, Aveiro, Portugal
| | - Sandra I Vieira
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
41
|
Huang X, Das R, Patel A, Nguyen TD. Physical Stimulations for Bone and Cartilage Regeneration. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2018; 4:216-237. [PMID: 30740512 PMCID: PMC6366645 DOI: 10.1007/s40883-018-0064-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 06/07/2018] [Indexed: 12/26/2022]
Abstract
A wide range of techniques and methods are actively invented by clinicians and scientists who are dedicated to the field of musculoskeletal tissue regeneration. Biological, chemical, and physiological factors, which play key roles in musculoskeletal tissue development, have been extensively explored. However, physical stimulation is increasingly showing extreme importance in the processes of osteogenic and chondrogenic differentiation, proliferation and maturation through defined dose parameters including mode, frequency, magnitude, and duration of stimuli. Studies have shown manipulation of physical microenvironment is an indispensable strategy for the repair and regeneration of bone and cartilage, and biophysical cues could profoundly promote their regeneration. In this article, we review recent literature on utilization of physical stimulation, such as mechanical forces (cyclic strain, fluid shear stress, etc.), electrical and magnetic fields, ultrasound, shock waves, substrate stimuli, etc., to promote the repair and regeneration of bone and cartilage tissue. Emphasis is placed on the mechanism of cellular response and the potential clinical usage of these stimulations for bone and cartilage regeneration.
Collapse
|
42
|
Catalano A, Loddo S, Bellone F, Pecora C, Lasco A, Morabito N. Pulsed electromagnetic fields modulate bone metabolism via RANKL/OPG and Wnt/β-catenin pathways in women with postmenopausal osteoporosis: A pilot study. Bone 2018; 116:42-46. [PMID: 30010081 DOI: 10.1016/j.bone.2018.07.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 07/01/2018] [Accepted: 07/12/2018] [Indexed: 11/21/2022]
Abstract
Pulsed electromagnetic fields (PEMFs) have been proven to enhance in vitro and in vivo osteogenesis with unknown mechanism. Aim of our study was to explore whether RANKL/OPG and Wnt/β-Catenin pathways could be involved in bone response to PEMFs in a setting of postmenopausal osteoporotic women. Forty-three women (mean age 62.8 ± 4.5 yr.) were randomized into two groups. The PEMFs group received PEMFs treatment (50 min treatment session/day, 6 treatment sessions/week, for a total of 25 times), by wearing a specific gilet applied to the trunk and connected to the electromagnetic device (Biosalus, by HSD Srl, Serravalle RSM), while women assigned to control group received sham PEMFs with the same device. BSAP as bone formation and CTX as bone resorption markers, RANKL, OPG, β-Catenin, DKK-1 and sclerostin were obtained at baseline, after 30 and 60 days. In PEMFs group, BSAP levels significantly increased after 30 and 60 days while CTX concentrations decreased at day 60. RANKL levels significantly decreased after 60 days. OPG was not significantly changed, but the RANKL/OPG ratio significantly decreased at day 30. DKK-1 levels decreased, while β-catenin concentrations increased after 30 and 60 days (P < 0.05). No significant changes of calcium, phosphorus, creatinine and sclerostin were detected. In the PEMFs group, at day 30, Δsclerostin was associated with ΔRANKL/OPG ratio (r = -0.5, P = 0.03) and ΔDKK-1 was associated with Δβ-Catenin (r = -0.47, P = 0.02). In women with postmenopausal osteoporosis, our data provide evidence of a PEMFs modulation of RANKL/OPG and Wnt/β-Catenin signaling pathways able to explain the metabolic effects of PEMFs on bone.
Collapse
Affiliation(s)
- Antonino Catalano
- Department of Clinical and Experimental Medicine, University Hospital of Messina, Messina, Italy.
| | - Saverio Loddo
- Department of Clinical and Experimental Medicine, University Hospital of Messina, Messina, Italy
| | - Federica Bellone
- Department of Clinical and Experimental Medicine, University Hospital of Messina, Messina, Italy
| | - Carmelo Pecora
- Vertebral Surgery Section, Carmona Clinic, Messina, Italy
| | - Antonino Lasco
- Department of Clinical and Experimental Medicine, University Hospital of Messina, Messina, Italy
| | - Nunziata Morabito
- Department of Clinical and Experimental Medicine, University Hospital of Messina, Messina, Italy
| |
Collapse
|
43
|
The Use of Pulsed Electromagnetic Fields to Promote Bone Responses to Biomaterials In Vitro and In Vivo. Int J Biomater 2018; 2018:8935750. [PMID: 30254677 PMCID: PMC6140132 DOI: 10.1155/2018/8935750] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/09/2018] [Indexed: 12/14/2022] Open
Abstract
Implantable biomaterials are extensively used to promote bone regeneration or support endosseous prosthesis in orthopedics and dentistry. Their use, however, would benefit from additional strategies to improve bone responses. Pulsed Electromagnetic Fields (PEMFs) have long been known to act on osteoblasts and bone, affecting their metabolism, in spite of our poor understanding of the underlying mechanisms. Hence, we have the hypothesis that PEMFs may also ameliorate cell responses to biomaterials, improving their growth, differentiation, and the expression of a mature phenotype and therefore increasing the tissue integration of the implanted devices and their clinical success. A broad range of settings used for PEMFs stimulation still represents a hurdle to better define treatment protocols and extensive research is needed to overcome this issue. The present review includes studies that investigated the effects of PEMFs on the response of bone cells to different classes of biomaterials and the reports that focused on in vivo investigations of biomaterials implanted in bone.
Collapse
|
44
|
Shuai C, Yang W, Peng S, Gao C, Guo W, Lai Y, Feng P. Physical stimulations and their osteogenesis-inducing mechanisms. Int J Bioprint 2018; 4:138. [PMID: 33102916 PMCID: PMC7581999 DOI: 10.18063/ijb.v4i2.138] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 05/09/2018] [Indexed: 12/27/2022] Open
Abstract
Physical stimulations such as magnetic, electric and mechanical stimulation could enhance cell activity and promote bone formation in bone repair process via activating signal pathways, modulating ion channels, regulating bonerelated gene expressions, etc. In this paper, bioeffects of physical stimulations on cell activity, tissue growth and bone healing were systematically summarized, which especially focused on their osteogenesis-inducing mechanisms. Detailedly, magnetic stimulation could produce Hall effect which improved the permeability of cell membrane and promoted the migration of ions, especially accelerating the extracellular calcium ions to pass through cell membrane. Electric stimulation could induce inverse piezoelectric effect which generated electric signals, accordingly up-regulating intracellular calcium levels and growth factor synthesis. And mechanical stimulation could produce mechanical signals which were converted into corresponding biochemical signals, thus activating various signaling pathways on cell membrane and inducing a series of gene expressions. Besides, bioeffects of physical stimulations combined with bone scaffolds which fabricated using 3D printing technology on bone cells were discussed. The equipments of physical stimulation system were described. The opportunities and challenges of physical stimulations were also presented from the perspective of bone repair.
Collapse
Affiliation(s)
- Cijun Shuai
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, China.,Jiangxi University of Science and Technology, Ganzhou, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
| | - Wenjing Yang
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, China
| | - Shuping Peng
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Chengde Gao
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, China
| | - Wang Guo
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, China
| | - Yuxiao Lai
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, China
| | - Pei Feng
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, China
| |
Collapse
|
45
|
Extremely low frequency electromagnetic fields promote mesenchymal stem cell migration by increasing intracellular Ca 2+ and activating the FAK/Rho GTPases signaling pathways in vitro. Stem Cell Res Ther 2018; 9:143. [PMID: 29784011 PMCID: PMC5963142 DOI: 10.1186/s13287-018-0883-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 04/03/2018] [Accepted: 04/20/2018] [Indexed: 12/11/2022] Open
Abstract
Background The ability of mesenchymal stem cells (MSCs) to migrate to the desired tissues or lesions is crucial for stem cell-based regenerative medicine and tissue engineering. Optimal therapeutics for promoting MSC migration are expected to become an effective means for tissue regeneration. Electromagnetic fields (EMF), as a noninvasive therapy, can cause a lot of biological changes in MSCs. However, whether EMF can promote MSC migration has not yet been reported. Methods We evaluated the effects of EMF on cell migration in human bone marrow-derived MSCs. With the use of Helmholtz coils and an EMF stimulator, 7.5, 15, 30, 50, and 70 Hz/1 mT EMF was generated. Additionally, we employed the l-type calcium channel blocker verapamil and the focal adhesion kinase (FAK) inhibitor PF-573228 to investigate the role of intracellular calcium content, cell adhesion proteins, and the Rho GTPase protein family (RhoA, Rac1, and Cdc42) in EMF-mediated MSC migration. Cell adhesion proteins (FAK, talin, and vinculin) were detected by Western blot analysis. The Rho GTPase protein family activities were assessed by G-LISA, and F-actin levels, which reflect actin cytoskeletal organization, were detected using immunofluorescence. Results All the 7.5, 15, 30, 50, and 70 Hz/1 mT EMF promoted MSC migration. EMF increased MSC migration in an intracellular calcium-dependent manner. Notably, EMF-enhanced migration was mediated by FAK activation, which was critical for the formation of focal contacts, as evidenced by increased talin and vinculin expression. Moreover, RhoA, Rac1, and Cdc42 were activated by FAK to increase cytoskeletal organization, thus promoting cell contraction. Conclusions EMF promoted MSC migration by increasing intracellular calcium and activating the FAK/Rho GTPase signaling pathways. This study provides insights into the mechanisms of MSC migration and will enable the rational design of targeted therapies to improve MSC engraftment. Electronic supplementary material The online version of this article (10.1186/s13287-018-0883-4) contains supplementary material, which is available to authorized users.
Collapse
|
46
|
Lei Y, Su J, Xu H, Yu Q, Zhao M, Tian J. Pulsed electromagnetic fields inhibit osteoclast differentiation in RAW264.7 macrophages via suppression of the protein kinase B/mammalian target of rapamycin signaling pathway. Mol Med Rep 2018; 18:447-454. [PMID: 29749519 DOI: 10.3892/mmr.2018.8999] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Accepted: 04/17/2018] [Indexed: 11/06/2022] Open
Abstract
When bone resorption, aided by the activity of osteoclasts, exceeds bone formation induced by osteoblasts, bone metabolism loses equilibration, which results in the development of bone diseases, including osteoporosis. Pulsed electromagnetic fields (PEMFs) are known to be involved in various biological processes, including cell proliferation, differentiation and apoptosis. However, the exact mechanism of action of osteoclasts remains poorly understood. In the present study, the effects of PEMFs on osteoclast differentiation and associated signaling pathways were systematically investigated in RAW264.7 macrophages. RAW264.7 cells were induced by receptor activator of nuclear factor‑κB ligand (RANKL) to obtain osteoclasts in vitro. The results of the present study demonstrated that PEMF exposure decreased osteoclast formation, limited tartrate‑resistant acid phosphatase activity, contracted bone resorption area and inhibited osteoclastic specific gene and protein expression. Furthermore, western blot analysis indicated that PEMFs distinctly abolished the upregulation of phosphorylated‑protein kinase B (Akt), ‑mammalian target of rapamycin (mTOR) and ‑ribosome S6 protein kinase (p70S6K) induced by RANKL, which was consistent with the effects of pharmacological inhibitor perifosine and rapamycin. Therefore, the present study suggested that PEMFs reduced osteoclast formation from RAW264.7 macrophages via inhibition of the Akt/mTOR signaling pathway. These findings provided novel insight into the mechanisms through which PEMFs suppress osteoclast differentiation.
Collapse
Affiliation(s)
- Yutian Lei
- Department of Orthopaedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Jinyu Su
- Department of Pathophysiology, Key Laboratory for Shock and Microcirculation Research of Guangdong, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Haixia Xu
- Department of Orthopaedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Qiang Yu
- Department of Orthopaedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Ming Zhao
- Department of Pathophysiology, Key Laboratory for Shock and Microcirculation Research of Guangdong, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jing Tian
- Department of Orthopaedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| |
Collapse
|
47
|
Li F, Li H, Zhai Q, Li F, Wu T, Sha X, Zhang B, Yang W, Lu Z, Tao H. A new vaccine targeting RANKL, prepared by incorporation of an unnatural Amino acid into RANKL, prevents OVX-induced bone loss in mice. Biochem Biophys Res Commun 2018; 499:648-654. [DOI: 10.1016/j.bbrc.2018.03.205] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 03/27/2018] [Indexed: 01/02/2023]
|
48
|
Guo L, Zhang JP, Zhang KY, Wang HB, Wang H, An GZ, Zhou Y, Meng GL, Ding GR. Effects of 1.8 GHz radiofrequency field on microstructure and bone metabolism of femur in mice. Bioelectromagnetics 2018; 39:386-393. [DOI: 10.1002/bem.22125] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 03/22/2018] [Indexed: 11/10/2022]
Affiliation(s)
- Ling Guo
- Faculty of Preventive Medicine, Department of Radiation Biology; Fourth Military Medical University; Xi′an Shaanxi People's Republic of China
| | - Jun-Ping Zhang
- Faculty of Preventive Medicine, Department of Radiation Biology; Fourth Military Medical University; Xi′an Shaanxi People's Republic of China
| | - Ke-Ying Zhang
- Faculty of Preventive Medicine, Department of Radiation Biology; Fourth Military Medical University; Xi′an Shaanxi People's Republic of China
| | - Huan-Bo Wang
- Brigade of Cadets; Fourth Military Medical University; Xi'an Shaanxi People's Republic of China
| | - Huan Wang
- Faculty of Preventive Medicine, Department of Radiation Biology; Fourth Military Medical University; Xi′an Shaanxi People's Republic of China
| | - Guang-Zhou An
- Faculty of Preventive Medicine, Department of Radiation Biology; Fourth Military Medical University; Xi′an Shaanxi People's Republic of China
| | - Yan Zhou
- Faculty of Preventive Medicine, Department of Radiation Biology; Fourth Military Medical University; Xi′an Shaanxi People's Republic of China
| | - Guo-Lin Meng
- Institute of Orthopaedics; Xijing Hospital; Fourth Military Medical University; Xi'an Shaanxi People's Republic of China
| | - Gui-Rong Ding
- Faculty of Preventive Medicine, Department of Radiation Biology; Fourth Military Medical University; Xi′an Shaanxi People's Republic of China
| |
Collapse
|