1
|
Zhang Y, Zhu Y, Li M, Zhang M, Shou D, Tong P. A promising approach to diabetic osteoporosis: oxymatrine's effects on gut microbiota and osteoblasts. Nutr Diabetes 2025; 15:19. [PMID: 40328755 PMCID: PMC12055986 DOI: 10.1038/s41387-025-00374-x] [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: 04/24/2024] [Revised: 04/25/2025] [Accepted: 04/28/2025] [Indexed: 05/08/2025] Open
Abstract
OBJECTIVES Oxymatrine (OMT), a quinolizidine alkaloid derived from Sophora flavescens Ait., has demonstrated therapeutic potential in type 2 diabetes mellitus (T2DM). This study aimed to investigate its effects on diabetic osteoporosis (DOP) and explore the underlying mechanisms involving gut microbiota and osteogenic regulation. METHODS In a rat model of T2DM, intragastric Oxymatrine was used to study trabecular bone repair through bone microstructure and histopathology analyses. Changes in gut microbiota, especially Gram-negative bacteria releasing lipopolysaccharides (LPS), were assessed via 16S rRNA sequencing. miRNA sequencing on LPS-induced rat osteoblasts, with and without Oxymatrine, explored osteoblast proliferation, mineralization, and the miR-539-5p/OGN/Runx2 pathway. RESULTS The administration of OMT resulted in an enhancement of diabetic osteopathy by reversing trabecular bone loss and modifying the composition of gut microbiota, specifically affecting Gram-negative bacteria that release LPS into the bloodstream. miRNA sequencing revealed that miR-539-5p, which was upregulated in LPS-induced ROBs, was downregulated following OMT treatment. Furthermore, OMT was found to promote osteoblast proliferation and mineralization under conditions of LPS exposure and modulate the miR-539-5p/OGN/Runx2 signaling pathway. CONCLUSIONS OMT improves diabetic osteoporosis by altering gut microbiota, decreasing LPS release, and enhancing osteoblast growth and differentiation through the miR-539-5p/OGN/Runx2 pathway, suggesting its potential as a treatment.
Collapse
Affiliation(s)
- Yang Zhang
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yiwen Zhu
- The First Clinical School, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Mengying Li
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Minjie Zhang
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Dan Shou
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Peijian Tong
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310053, China.
- The First Clinical School, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
2
|
Zhang X, Huang F, Liu J, Zhou Z, Yuan S, Jiang H. Molecular Mechanism of Ginsenoside Rg3 Alleviation in Osteoporosis via Modulation of KPNA2 and the NF-κB Signalling Pathway. Clin Exp Pharmacol Physiol 2025; 52:e70019. [PMID: 39821958 DOI: 10.1111/1440-1681.70019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 10/27/2024] [Accepted: 12/05/2024] [Indexed: 01/19/2025]
Abstract
Osteoporosis is mainly caused by an imbalance in osteoclast and osteoblast regulation, resulting in an imbalance in bone homeostasis. Ginsenoside Rg3 (Rg3) has been reported to have a therapeutic effect on alleviating osteoporosis. Nonetheless, the underlying mechanisms have not been completely elucidated. Herein, the molecular mechanism of Rg3 alleviation in osteoporosis was further explored. An in vitro model was established utilising the receptor activator of nuclear factor-kappaB ligand (RANKL) to induce osteoclast differentiation of RAW264.7 cells. RNA-sequencing results showed that karyopherin subunit alpha 2 (KPNA2) is one of the significantly differentially expressed genes regulated by Rg3 in RANKL-induced RAW264.7 cells. Basic experiments further suggested that KPNA2 is up-regulated in a time-dependent manner in the RANKL-induced RAW264.7 cells, while Rg3 treatment reduced its expression in a dose- and time-dependent manner. Knockdown of KPNA2 inhibited osteoclast formation and the expression of related molecules, including those in the nuclear factor kappa-B (NF-κB) pathway. The NF-κB inhibitor, JSH-23, partially abolished the impact of KPNA2 overexpression on osteoclast formation, indicating KPNA2 activates NF-κB. Furthermore, KPNA2 overexpression partially abolished the inhibitory impact of Rg3 on osteoclast formation, indicating that KPNA2 is a target of Rg3. These results suggest that KPNA2 plays a role in how Rg3 influences on osteoclast differentiation and osteoporosis through the NF-κB pathway.
Collapse
Affiliation(s)
- Xiaonan Zhang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
- Department of Orthopedics, Shenzhen Third People's Hospital, Shenzhen, China
| | - Fenglan Huang
- Department of Outpatient, Shenzhen University General Hospital, Shenzhen, China
| | - Jinzhu Liu
- Department of Orthopedics, Shenzhen Third People's Hospital, Shenzhen, China
| | - Zhenzhong Zhou
- Department of Orthopedics, Shenzhen Third People's Hospital, Shenzhen, China
| | - Shanyou Yuan
- Department of Orthopedics, Shenzhen Third People's Hospital, Shenzhen, China
| | - Haoli Jiang
- Department of Orthopedics, Shenzhen Third People's Hospital, Shenzhen, China
| |
Collapse
|
3
|
Tian X, Gao Y, Li C, Ma W, Zhang J, Ju Y, Ding J, Zeng S, Hameed HMA, Aung HL, Zhong N, Cook GM, Hu J, Zhang T. A novel non-invasive murine model for rapidly testing drug activity via inhalation administration against Mycobacterium tuberculosis. Front Pharmacol 2025; 15:1400436. [PMID: 39830329 PMCID: PMC11739085 DOI: 10.3389/fphar.2024.1400436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 12/03/2024] [Indexed: 01/22/2025] Open
Abstract
The efficacy of many compounds against Mycobacterium tuberculosis is often limited when administered via conventional oral or injection routes due to suboptimal pharmacokinetic characteristics. Inhalation-based delivery methods have been investigated to achieve high local therapeutic doses in the lungs. However, previous models, typically employing wild-type M. tuberculosis strains, were intricate, time-consuming, labor-intensive, and with poor reproducibility. In this study, we developed an autoluminescence-based inhalation administration model to evaluate drug activity by quantifying relative light units (RLUs) emitted from live mice infected with autoluminescent M. tuberculosis. This novel approach offers several advantages: (1) it eliminates the need for anesthesia in mice during administration and simplifies the instrument manipulation; (2) it is cost-effective by utilizing mice instead of larger animals; (3) it shortens the time from several months to 16 or 17 days for obtaining result; (4) it is non-invasive by directly measuring the live RLUs of mice as a surrogate marker for colony-forming units for in vivo drug activity testing; (5) up to six mice can be administrated daily and simultaneously, even 2-3 times/day; (6) results are relatively objective and reproducible results minimizing human factors. Proof-of-concept experiments demonstrated that inhalable rifampicin, isoniazid, and ethambutol showed anti-M. tuberculosis activity at concentrations as low as 0.5, 0.5, and 0.625 mg/mL, respectively, as evidenced by comparing the live RLUs of mice. Furthermore, consistency between RLUs and colony-forming units of the autoluminescent M. tuberculosis in lungs reaffirms the reliability of RLUs as an indicator of drug efficacy, highlighting the potential of this approach for accurately assessing anti-M. tuberculosis activity in vivo. This autoluminescence-based, non-invasive inhalation model offers a substantial reduction in the time, effort, and cost required for evaluating the efficacy of screening new drugs and repurposing old drugs in vivo via inhalation administration.
Collapse
Affiliation(s)
- Xirong Tian
- State Key Laboratory of Respiratory Disease, Joint School of Life Sciences, Guangzhou Chest Hospital, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China
- University of Chinese Academy of Sciences (UCAS), Beijing, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yamin Gao
- State Key Laboratory of Respiratory Disease, Joint School of Life Sciences, Guangzhou Chest Hospital, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China
- University of Chinese Academy of Sciences (UCAS), Beijing, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Chunyu Li
- State Key Laboratory of Respiratory Disease, Joint School of Life Sciences, Guangzhou Chest Hospital, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China
- University of Chinese Academy of Sciences (UCAS), Beijing, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Wanli Ma
- State Key Laboratory of Respiratory Disease, Joint School of Life Sciences, Guangzhou Chest Hospital, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China
- University of Chinese Academy of Sciences (UCAS), Beijing, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Jingran Zhang
- State Key Laboratory of Respiratory Disease, Joint School of Life Sciences, Guangzhou Chest Hospital, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China
- University of Chinese Academy of Sciences (UCAS), Beijing, China
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Yanan Ju
- State Key Laboratory of Respiratory Disease, Joint School of Life Sciences, Guangzhou Chest Hospital, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China
- University of Chinese Academy of Sciences (UCAS), Beijing, China
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Jie Ding
- State Key Laboratory of Respiratory Disease, Joint School of Life Sciences, Guangzhou Chest Hospital, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China
- University of Chinese Academy of Sciences (UCAS), Beijing, China
- Institute of Physical Science and Information Technology, Anhui University, Hefei, China
| | - Sanshan Zeng
- State Key Laboratory of Respiratory Disease, Joint School of Life Sciences, Guangzhou Chest Hospital, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China
- University of Chinese Academy of Sciences (UCAS), Beijing, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - H. M. Adnan Hameed
- State Key Laboratory of Respiratory Disease, Joint School of Life Sciences, Guangzhou Chest Hospital, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China
- University of Chinese Academy of Sciences (UCAS), Beijing, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Htin Lin Aung
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Department of Microbiology and Immunology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | | | - Gregory M. Cook
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Department of Microbiology and Immunology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
- Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| | - Jinxing Hu
- State Key Laboratory of Respiratory Disease, Joint School of Life Sciences, Guangzhou Chest Hospital, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Tianyu Zhang
- State Key Laboratory of Respiratory Disease, Joint School of Life Sciences, Guangzhou Chest Hospital, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, China
- Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences (CAS), Guangzhou, China
- University of Chinese Academy of Sciences (UCAS), Beijing, China
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
| |
Collapse
|
4
|
Chen H, Weng Z, Kalinowska M, Xiong L, Wang L, Song H, Xiao J, Wang F, Shen X. Anti-osteoporosis effect of bioactives in edible medicinal plants: a comprehensive review. Crit Rev Food Sci Nutr 2024:1-17. [PMID: 39093554 DOI: 10.1080/10408398.2024.2386449] [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: 08/04/2024]
Abstract
Current treatments for osteoporosis include a calcium-rich diet, adequate exercise, and medication. Many synthetic drugs, although fast-acting, can cause a range of side effects for patients when taken over a long period, such as irritation of the digestive tract and a burden on the kidneys. As the world's population ages, the prevalence of osteoporosis is increasing, and the development of safe and effective treatments is urgently needed. Active compounds in edible and medicinal homologous plants have been used for centuries to improve bone quality. It is possible to employ them as dietary supplements to prevent osteoporosis. In this review, we analyze the influencing factors of osteoporosis and systematically summarize the research progress on the anti-osteoporosis effects of active compounds in edible and medicinal homologous plants. The literature suggests that some naturally occurring active compounds in edible and medicinal homologous plants can inhibit bone loss, prevent the degeneration of bone cell microstructure, and reduce bone fragility through alleviating oxidative stress, regulating autophagy, anti-inflammation, improving gut flora, and regulating estrogen level with little side effects. Our review provides useful guidance for the use of edible and medicinal homologous plants and the development of safer novel anti-osteoporosis dietary supplements.
Collapse
Affiliation(s)
- Huiling Chen
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Zebin Weng
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Monika Kalinowska
- Department of Chemistry, Biology and Biotechnology, Bialystok University of Technology, Bialystok, Poland
| | - Ling Xiong
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Luanfeng Wang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Haizhao Song
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Jianbo Xiao
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
- Department of Analytical Chemistry and Food Science, Instituto de Agroecoloxía e Alimentación (IAA) - CITEXVI, Universidade de Vigo, Nutrition and Bromatology Group, Vigo, Spain
| | - Fang Wang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
- Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, Massachusetts, USA
| | - Xinchun Shen
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| |
Collapse
|
5
|
Lin Z, Zhou Z, Ye J, Wei J, Chen S, Zhou W, Bi Y, Zhou Z, Xie G, Yuan G, Yao G. Trifolirhizin protects ovariectomy-induced bone loss in mice by inhibiting osteoclast differentiation and bone resorption. Heliyon 2024; 10:e34250. [PMID: 39130482 PMCID: PMC11315080 DOI: 10.1016/j.heliyon.2024.e34250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/05/2024] [Accepted: 07/05/2024] [Indexed: 08/13/2024] Open
Abstract
Background Osteoporosis is a debilitating condition characterized by reduced bone density and microstructure, leading to increased susceptibility to fractures and increased mortality, particularly among older individuals. Despite the availability of drugs for osteoporosis treatment, the need for targeted and innovative agents with fewer adverse effects persists. Trifolirhizin, a natural pterostalin derived from the root of Sophora flavescens, has been previously studied for its effects on certain anticancer and antiinflammatory. The impact of trifolirhizin on the formation and function of osteoclasts remain unclear. Purpose Herein, the possible roles of trifolirhizin the formation and function of osteoclasts and the underlying mechanism were explored. Methods: Bone marrow-derived macrophages (BMMs) were employed to evaluate the roles of trifolirhizin on steoclastogenesis, bone absorption and the underlying mechanism in vitro. Bone loss model was established by ovariectomy(OVX) in mice in vivo. Results Trifolirhizin repressed osteoclastogenesis, bone resorption induced by receptor activator of nuclear factor kappa B ligand (RANKL) in vitro. Mechanistically, trifolirhizin inhibits RANKL-induced MAPK signal transduction and NFATc1 expression. Moreover, trifolirhizin inhibited osteoclast marker gene expression, including NFATc1, CTSK, MMP9, DC-STAMP, ACP5, and V-ATPase-D2. Additionally, trifolirhizin was found to protect against ovariectomy(OVX)-induced bone loss in mice. Conclusion Trifolirhizin can effectively inhibit osteoclast production and bone resorption activity. The results of our study provide evidence for trifolirhizin as a potential drug for the prevention and treatment of osteoporosis and other osteolytic diseases.
Collapse
Affiliation(s)
- Zihong Lin
- Department of Orthopedics, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Department of Shantou Central Hospital, Shantou, Guangdong, China
| | - Zhigao Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Shantou University Medical College, Shantou, Guangdong, China
| | - Jiajie Ye
- Department of Orthopedics, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Shantou University Medical College, Shantou, Guangdong, China
| | - Jinfu Wei
- Shantou University Medical College, Shantou, Guangdong, China
- Department of Orthopedics, The Sixth Affiliated Hospital,School of Medicine, South China University of Technology, Foshan, Guangdong, China
| | - Shaozhe Chen
- Department of Orthopedics, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Shantou University Medical College, Shantou, Guangdong, China
| | - Wenyun Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Shantou University Medical College, Shantou, Guangdong, China
| | - Yonghao Bi
- Department of Orthopedics, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Shantou University Medical College, Shantou, Guangdong, China
| | - Zibin Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Shantou University Medical College, Shantou, Guangdong, China
| | - Gang Xie
- Shantou University Medical College, Shantou, Guangdong, China
| | - Guixin Yuan
- Department of Orthopedics, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Guanfeng Yao
- Department of Orthopedics, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
6
|
Chen W, Wang Q, Tao H, Lu L, Zhou J, Wang Q, Huang W, Yang X. Subchondral osteoclasts and osteoarthritis: new insights and potential therapeutic avenues. Acta Biochim Biophys Sin (Shanghai) 2024; 56:499-512. [PMID: 38439665 DOI: 10.3724/abbs.2024017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024] Open
Abstract
Osteoarthritis (OA) is the most common joint disease, and good therapeutic results are often difficult to obtain due to its complex pathogenesis and diverse causative factors. After decades of research and exploration of OA, it has been progressively found that subchondral bone is essential for its pathogenesis, and pathological changes in subchondral bone can be observed even before cartilage lesions develop. Osteoclasts, the main cells regulating bone resorption, play a crucial role in the pathogenesis of subchondral bone. Subchondral osteoclasts regulate the homeostasis of subchondral bone through the secretion of degradative enzymes, immunomodulation, and cell signaling pathways. In OA, osteoclasts are overactivated by autophagy, ncRNAs, and Rankl/Rank/OPG signaling pathways. Excessive bone resorption disrupts the balance of bone remodeling, leading to increased subchondral bone loss, decreased bone mineral density and consequent structural damage to articular cartilage and joint pain. With increased understanding of bone biology and targeted therapies, researchers have found that the activity and function of subchondral osteoclasts are affected by multiple pathways. In this review, we summarize the roles and mechanisms of subchondral osteoclasts in OA, enumerate the latest advances in subchondral osteoclast-targeted therapy for OA, and look forward to the future trends of subchondral osteoclast-targeted therapies in clinical applications to fill the gaps in the current knowledge of OA treatment and to develop new therapeutic strategies.
Collapse
Affiliation(s)
- Wenlong Chen
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou 215000, China
- Gusu School, Nanjing Medical University, Suzhou 215000, China
| | - Qiufei Wang
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215000, China
| | - Huaqiang Tao
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215000, China
| | - Lingfeng Lu
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou 215000, China
- Gusu School, Nanjing Medical University, Suzhou 215000, China
| | - Jing Zhou
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou 215000, China
- Gusu School, Nanjing Medical University, Suzhou 215000, China
| | - Qiang Wang
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215000, China
| | - Wei Huang
- Department of Orthopaedics, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Xing Yang
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou 215000, China
- Gusu School, Nanjing Medical University, Suzhou 215000, China
| |
Collapse
|
7
|
Ma X, Luo L, Karrar E, Zhang L, Li J. Comparative Study on the Absorption and Metabolism of Pinoresinol and Pinoresinol-4-O-β-D-Glucopyranoside in Mice. Mol Nutr Food Res 2023; 67:e2300536. [PMID: 37891711 DOI: 10.1002/mnfr.202300536] [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: 07/27/2023] [Revised: 09/20/2023] [Indexed: 10/29/2023]
Abstract
SCOPE Lignans are a group of phenolic compounds commonly found in plants, often in the form of glycosides. This study investigates the differences in the digestion, absorption, and metabolism of lignans and their glucosides using pinoresinol (PIN) and pinoresinol-4-O-β-D-glucopyranoside (PMG). METHODS AND RESULTS After oral administration mice PIN and PMG with a dose of 0.1 µmol kg-1 . The results showed that the stomach and small intestine rapidly absorbe PIN and PMG in their prototype form. After oral administration of 0.25 h, serum levels of PIN and PMG reach peak values of 61.14 and 52.97 ng mL-1 , respectively. This indicates a faster PIN absorption rate than PMG, likely due to the glycosides attach to the parent compound, with concentrations of 1574.14 and 876.75 ng g-1 , respectively. Pharmacokinetic analysis reveals that PIN has a greater area under the curve and a longer half-life than PMG in serum and liver. Moreover, mice in the PIN group exhibit higher metabolite levels in the serum and liver compared to those in the PMG group. CONCLUSION The deglycosylation process that occurs during the pickling of white radish facilitates the absorption and metabolism of the lignans fraction in the body.
Collapse
Affiliation(s)
- Xiaoyang Ma
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, 361021, China
| | - Lianzhong Luo
- Engineering Research Center of Marine Biopharmaceutical Resource, Fujian Province University, Xiamen Medical College, Xiamen, 361021, China
| | - Emad Karrar
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, 361021, China
| | - Lingyu Zhang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, 361021, China
| | - Jian Li
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, 361021, China
| |
Collapse
|
8
|
Li J, Ma X, Luo L, Tang D, Zhang L. The What and Who of Dietary Lignans in Human Health: Special Attention to Estrogen Effects and Safety Evaluation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:16419-16434. [PMID: 37870451 DOI: 10.1021/acs.jafc.3c02680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
Lignans are a group of phenolic compounds found in plant-based diets. The human body can obtain lignans through diet, which are then metabolized into enterolignans. The enterolignans have been linked to several health benefits, including anticancer, anti-inflammatory, antioxidant effects, and estrogen effects. This review explores the relationship between the estrogenic effects of lignans and health. This review not only considers the estrogen-like activity of lignans but also discusses the safe dosage of lignans at different life stages. In addition, this review also identified other types of bioactive compounds that can act synergistically with lignans to promote health. Studies have shown that lignan administration during pregnancy and lactation reduces the risk of breast cancer in offspring. Further studies are needed to investigate the estrogenic safety effects of lignan on pregnant women and children. Whether lignans combine with other nutrients in complex food substrates to produce synergistic effects remains to be investigated. This review provides a basis for future studies on the safe dose of lignan and recommended dietary intake of lignan. We believe that the acquired as discussed here has implications for developing dietary therapies that can promote host nutrition and modulate estrogenic diseases.
Collapse
Affiliation(s)
- Jian Li
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Xiaoyang Ma
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Lianzhong Luo
- Xiamen Key Laboratory of Marine Medicinal Natural Products Resources, Xiamen Medical College, Xiamen 361023, China
| | - Danqing Tang
- The School of Foreign Languages of Jimei University, Xiamen 361021, China
| | - Lingyu Zhang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| |
Collapse
|
9
|
Jia M, Luo J, Gao B, Huangfu Y, Bao Y, Li D, Jiang S. Preparation of synbiotic milk powder and its effect on calcium absorption and the bone microstructure in calcium deficient mice. Food Funct 2023; 14:3092-3106. [PMID: 36919678 DOI: 10.1039/d2fo04092a] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Calcium deficiency can lead to osteoporosis. Adequate calcium intake can improve calcium deficiency and prevent osteoporosis. Milk powder is the best source of dietary calcium supplements. Probiotics and prebiotics are considered to be beneficial substances for promoting calcium absorption. In this study, synbiotic milk powder (SMP) was prepared by combining the three, and its calcium supplementation effect and osteogenic activity were evaluated in calcium deficient mice. Through prebiotic screening experiments in vitro, after adding 1.2% iso-malto-oligosaccharide, the number of viable bacteria and the calcium enrichment of Lactobacillus plantarum JJBYG12 increased by 8.15% and 94.53% compared with those of the control group. Long-term calcium deficiency led to a significant reduction in calcium absorption and bone calcium content in mice, accompanied by structural deterioration of bone trabeculae. SMP significantly improved apparent calcium absorption, increased serum calcium and phosphorus levels, and decreased ALP activity and CTX-1 levels. In the meantime, the bone mineral density increased significantly, and the number of bone trabeculae and the proliferation and differentiation of osteoblasts also increased. SMP has good dietary calcium supplementation capacity and bone remodeling ability without significant side effects on major organs. These findings provide insights into using SMP as a dietary calcium source to improve bone health.
Collapse
Affiliation(s)
- Mingjie Jia
- School of Forestry, Northeast Forestry University, #26Hexing Road, Harbin 150040, PR China.
| | - Jiayuan Luo
- School of Forestry, Northeast Forestry University, #26Hexing Road, Harbin 150040, PR China.
| | - Bo Gao
- School of Forestry, Northeast Forestry University, #26Hexing Road, Harbin 150040, PR China.
| | - Yunpeng Huangfu
- School of Forestry, Northeast Forestry University, #26Hexing Road, Harbin 150040, PR China.
| | - Yihong Bao
- School of Forestry, Northeast Forestry University, #26Hexing Road, Harbin 150040, PR China. .,Key Laboratory of Forest Food Resources Utilization of Heilongjiang Province, Harbin 150040, PR China
| | - Dehai Li
- School of Forestry, Northeast Forestry University, #26Hexing Road, Harbin 150040, PR China. .,Key Laboratory of Forest Food Resources Utilization of Heilongjiang Province, Harbin 150040, PR China
| | - Shilong Jiang
- Heilongjiang Feihe Dairy Company Limited, Beijing 100015, PR China
| |
Collapse
|
10
|
Sharma A, Michels LV, Pitsillides AA, Greeves J, Plotkin LI, Cardo V, Sims NA, Clarkin CE. Sexing Bones: Improving Transparency of Sex Reporting to Address Bias Within Preclinical Studies. J Bone Miner Res 2023; 38:5-13. [PMID: 36301601 PMCID: PMC10099537 DOI: 10.1002/jbmr.4729] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/11/2022] [Accepted: 10/20/2022] [Indexed: 01/10/2023]
Abstract
Despite knowledge that sexually dimorphic mechanisms regulate bone homeostasis, sex often remains unreported and unconsidered in preclinical experimental design. Failure to report sex could lead to inappropriate generalizations of research findings and less effective translation into clinical practice. Preclinical sex bias (preferential selection of one sex) is present across other fields, including neuroscience and immunology, but remains uninvestigated in skeletal research. For context, we first summarized key literature describing sexually dimorphic bone phenotypes in mice. We then investigated sex reporting practices in skeletal research, specifically how customary it is for murine sex to be included in journal article titles or abstracts and then determined whether any bias in sex reporting exists. Because sex hormones are important regulators of bone health (gonadectomy procedures, ie, ovariectomy [OVX] and orchidectomy [ORX], are common yet typically not reported with sex), we incorporated reporting of OVX and ORX terms, representing female and male mice, respectively, into our investigations around sex bias. Between 1999 and 2020, inclusion of sex in titles or abstracts was low in murine skeletal studies (2.6%-4.06%). Reporting of OVX and ORX terms was low (1.44%-2.64%) and reporting of OVX and ORX with sex uncommon (0.4%-0.3%). When studies were combined to include both sexes and OVX (representing female) and ORX terms (representing male), a bias toward reporting of female mice was evident. However, when the terms OVX and ORX were removed, a bias toward the use of male mice was identified. Thus, studies focusing on sex hormones are biased toward female reporting with all other studies biased in reporting of male mice. We now call upon journal editors to introduce consistent guidance for transparent and accessible reporting of murine sex in skeletal research to better monitor preclinical sex bias, to diversify development of treatments for bone health, and to enable global skeletal health equity. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Aikta Sharma
- School of Biological Sciences, University of Southampton, Southampton, UK
| | - Lysanne V Michels
- School of Biological Sciences, University of Southampton, Southampton, UK
| | - Andrew A Pitsillides
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Julie Greeves
- Army Health and Performance Research, Ministry of Defence, Andover, UK
| | - Lillian I Plotkin
- Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Valentina Cardo
- Winchester School of Art, University of Southampton, Winchester, UK
| | - Natalie A Sims
- Department of Medicine at St. Vincent's Hospital, St. Vincent's Institute of Medical Research and The University of Melbourne, Fitzroy, Australia
| | - Claire E Clarkin
- School of Biological Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
11
|
Jang WY, Kim MY, Cho JY. Antioxidant, Anti-Inflammatory, Anti-Menopausal, and Anti-Cancer Effects of Lignans and Their Metabolites. Int J Mol Sci 2022; 23:ijms232415482. [PMID: 36555124 PMCID: PMC9778916 DOI: 10.3390/ijms232415482] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/01/2022] [Accepted: 12/04/2022] [Indexed: 12/12/2022] Open
Abstract
Since chronic inflammation can be seen in severe, long-lasting diseases such as cancer, there is a high demand for effective methods to modulate inflammatory responses. Among many therapeutic candidates, lignans, absorbed from various plant sources, represent a type of phytoestrogen classified into secoisolariciresionol (Seco), pinoresinol (Pino), matairesinol (Mat), medioresinol (Med), sesamin (Ses), syringaresinol (Syr), and lariciresinol (Lari). Lignans consumed by humans can be further modified into END or ENL by the activities of gut microbiota. Lignans are known to exert antioxidant and anti-inflammatory activities, together with activity in estrogen receptor-dependent pathways. Lignans may have therapeutic potential for postmenopausal symptoms, including cardiovascular disease, osteoporosis, and psychological disorders. Moreover, the antitumor efficacy of lignans has been demonstrated in various cancer cell lines, including hormone-dependent breast cancer and prostate cancer, as well as colorectal cancer. Interestingly, the molecular mechanisms of lignans in these diseases involve the inhibition of inflammatory signals, including the nuclear factor (NF)-κB pathway. Therefore, we summarize the recent in vitro and in vivo studies evaluating the biological effects of various lignans, focusing on their values as effective anti-inflammatory agents.
Collapse
Affiliation(s)
- Won Young Jang
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Mi-Yeon Kim
- School of Systems Biomedical Science, Soongsil University, Seoul 06978, Republic of Korea
- Correspondence: (M.-Y.K.); (J.Y.C.); Tel.: +82-2-820-0458 (M.-Y.K.); +82-31-290-7868 (J.Y.C.)
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Correspondence: (M.-Y.K.); (J.Y.C.); Tel.: +82-2-820-0458 (M.-Y.K.); +82-31-290-7868 (J.Y.C.)
| |
Collapse
|
12
|
Wang M, An M, Fan MS, Zhang SS, Sun Z, Zhao Y, Xiang ZM, Sheng J. FAEE exerts a protective effect against osteoporosis by regulating the MAPK signalling pathway. PHARMACEUTICAL BIOLOGY 2022; 60:467-478. [PMID: 35180021 PMCID: PMC8865110 DOI: 10.1080/13880209.2022.2039216] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
CONTEXT Ferulic acid ethyl ester (FAEE) is abundant in Ligusticum chuanxiong Hort. (Apiaceae) and grains, and possesses diverse biological activities; but the effects of FAEE on osteoporosis has not been reported. OBJECTIVE This study investigated whether FAEE can attenuate osteoclastogenesis and relieve ovariectomy-induced osteoporosis via attenuating mitogen-activated protein kinase (MAPK). MATERIALS AND METHODS We stimulated RAW 264.7 cells with receptor activator of NF-κB ligand (RANKL) followed by FAEE. The roles of FAEE in osteoclast production and osteogenic resorption of mature osteoclasts were evaluated by tartrate resistant acid phosphatase (TRAP) staining, expression of osteoclast-specific genes, proteins and MAPK. Ovariectomized (OVX) female Sprague-Dawley rats were administered FAEE (20 mg/kg/day) for 12 weeks to explore its potential in vivo, and then histology was undertaken in combination with cytokines analyses. RESULTS FAEE suppressed RANKL-induced osteoclast formation (96 ± 0.88 vs. 15 ± 1.68) by suppressing the expression of osteoclast-specific genes, proteins and MAPK signalling pathway related proteins (p-ERK/ERK, p-JNK/JNK and p-P38/P38) in vitro. In addition, OVX rats exposed to FAEE maintained their normal calcium (Ca) (2.72 ± 0.02 vs. 2.63 ± 0.03, p < 0.05) balance, increased oestradiol levels (498.3 ± 9.43 vs. 398.7 ± 22.06, p < 0.05), simultaneously reduced levels of bone mineral density (BMD) (0.159 ± 0.0016 vs. 0.153 ± 0.0025, p < 0.05) and bone mineral content (BMC) (0.8 ± 0.0158 vs. 0.68 ± 0.0291, p < 0.01). DISCUSSION AND CONCLUSIONS These findings suggested that FAEE could be used to ameliorate osteoporosis by the MAPK signalling pathway, suggesting that FAEE could be a potential therapeutic candidate for osteoporosis.
Collapse
Affiliation(s)
- Ming‑Yue Wang
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, P. R. China
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, P. R. China
| | - Meng‑Fei An
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, P. R. China
- College of Science, Yunnan Agricultural University, Kunming, P. R. China
| | - Mao-Si Fan
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, P. R. China
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, P. R. China
| | - Shao-Shi Zhang
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, P. R. China
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, P. R. China
| | - Ze‑Rui Sun
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, P. R. China
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, P. R. China
| | - Yun‑Li Zhao
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education; Yunnan Provincial Center for Research & Development of Natural Products; School of Chemical Science and Technology, Yunnan University, Kunming, P. R. China
- Yun‑Li Zhao
| | - Ze-Min Xiang
- College of Science, Yunnan Agricultural University, Kunming, P. R. China
- Ze-Min Xiang
| | - Jun Sheng
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, Yunnan Agricultural University, Kunming, P. R. China
- College of Science, Yunnan Agricultural University, Kunming, P. R. China
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Kunming, P. R. China
- CONTACT Jun Sheng
| |
Collapse
|
13
|
Thapa S, Nandy A, Rendina-Ruedy E. Endocrinal metabolic regulation on the skeletal system in post-menopausal women. Front Physiol 2022; 13:1052429. [PMID: 36439254 PMCID: PMC9691779 DOI: 10.3389/fphys.2022.1052429] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 10/27/2022] [Indexed: 08/13/2023] Open
Abstract
Osteoporosis is a common endocrinologic disorder characterized as a chronic bone loss condition. Sexual dimorphism is ubiquitous in the incidence of osteoporosis with post-menopausal women being acutely affected. Gonadal sex hormones including estrogen act as crucial regulators of bone mass; therefore, loss of such hormones leads to an imbalance in skeletal turnover leading to osteoporosis. Estrogen can influence both bone formation as well as resorption by reducing osteoblast activity and enhancing osteoclastogenesis. Additionally, estrogen is a potent regulator of systemic metabolism. Recent studies have provided clues that estrogenic effect on bone might also involve alterations in bone cell metabolism and bioenergetic potential. While direct effects of gonadal hormones ability to alter intracellular metabolism of bone cells has not been studied, there is precedence within the literature that this is occurring and contributing to post-menopausal bone loss. This review aims to serve as a perspective piece detailing the prospective role of gonadal hormones regulating bone cell metabolic potential.
Collapse
Affiliation(s)
- Santosh Thapa
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Ananya Nandy
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Elizabeth Rendina-Ruedy
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States
- Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
14
|
Qi G, Jiang Z, Lu W, Li D, Chen W, Yang X, Ding L, Yuan H. Berbamine inhibits RANKL- and M-CSF-mediated osteoclastogenesis and alleviates ovariectomy-induced bone loss. Front Pharmacol 2022; 13:1032866. [PMID: 36408260 PMCID: PMC9666778 DOI: 10.3389/fphar.2022.1032866] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/21/2022] [Indexed: 10/06/2023] Open
Abstract
Osteoporosis is a common public health problem characterized by decreased bone mass, increased bone brittleness and damage to the bone microstructure. Excessive bone resorption by osteoclasts is the main target of the currently used drugs or treatment for osteoporosis. Effective antiresorptive drugs without side effects following long-term administration have become a major focus of anti-osteoporotic drugs. In the present study, we investigated the effect of berbamine, a small molecule natural product from Berberis amurensis Rupr, a traditional Chinese medicine, on RANKL-induced osteoclast differentiation in vitro and ovariectomy-induced bone loss in vivo. The results demonstrated that berbamine at a safe and effective dose inhibited osteoclastogenesis and bone resorption function in vitro by suppressing the nuclear factor-κB signaling pathway. In addition, berbamine protected against osteoporosis by inhibiting osteoclastogenesis and bone resorption function without affecting osteogenesis in the ovariectomy mouse model. These findings revealed that berbamine has a protective role against osteoporosis and may represent a novel promising treatment strategy for osteoporosis.
Collapse
Affiliation(s)
- Guobin Qi
- Department of Orthopaedics, Shanghai Sixth People’s Hospital, Shanghai, China
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zengxin Jiang
- Department of Orthopaedics, Shanghai Sixth People’s Hospital, Shanghai, China
| | - Wei Lu
- Department of Orthopedic Surgery, Shanghai TCM-Integrated Hospital Shanghai University of TCM, Shanghai, China
| | - Defang Li
- Department of Orthopedic Surgery, Jinshan Hospital, Fudan University, Shanghai, China
| | - Weibing Chen
- Department of Orthopedic Surgery, Jinshan Hospital, Fudan University, Shanghai, China
| | - Xiuying Yang
- Department of Radiology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Lei Ding
- Department of Orthopedic Surgery, Jinshan Hospital, Fudan University, Shanghai, China
| | - Hengfeng Yuan
- Department of Orthopaedics, Shanghai Sixth People’s Hospital, Shanghai, China
| |
Collapse
|
15
|
Wang D, Liu L, Qu Z, Zhang B, Gao X, Huang W, Feng M, Gong Y, Kong L, Wang Y, Yan L. Hypoxia-inducible factor 1α enhances RANKL-induced osteoclast differentiation by upregulating the MAPK pathway. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1227. [PMID: 36544674 PMCID: PMC9761153 DOI: 10.21037/atm-22-4603] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/21/2022] [Indexed: 11/30/2022]
Abstract
Background Hypoxia (low-oxygen tension) and excessive osteoclast activation are common conditions in many bone loss diseases, such as osteoporosis, rheumatoid arthritis (RA), and pathologic fractures. Hypoxia-inducible factor 1 alpha (HIF1α) regulates cellular responses to hypoxic conditions. However, it is not yet known how HIF1α directly affects osteoclast differentiation and activation. This study sought to. explore the effects of HIF1α on osteoclast differentiation and it's molecular mechanisms. Methods L-mimosine, a prolyl hydroxylase (PHDs) domain inhibitor, was used to stabilize HIF1α in normoxia. In the presence of receptor activator of nuclear factor-kB (NF-kB) ligand (RANKL), RAW264.7 cells were cultured and stimulated by treatment with L-mimosine at several doses to maintain various levels of intracellular HIF1α. The multi-nucleated cells were assessed by a tartrate-resistant acid phosphatase (TRAP) and F-actin ring staining assays. The osteoclast-specific genes, such as Cathepsin K, β3-Integrin, TRAP, c-Fos, nuclear factor of activated T cells, cytoplasmic 1 (NFATc1), and matrix metallo-proteinase 9 (MMP9), were analyzed by real time-polymerase chain reaction (RT-PCR). The expression of relevant proteins was analyzed by Western blot. Results L-mimosine increased the content of intracellular HIF1α in a dose-dependent manner, which in turn promoted RANKL-induced osteoclast formation and relevant protein expression by upregulating the mitogen-activated protein kinase (MAPK) pathways. Conclusions Our findings suggest that HIF1α directly increases the osteoclast differentiation of RANKL-mediated RAW264.7 cells in vitro by upregulating the MAPK pathways.
Collapse
Affiliation(s)
- Dong Wang
- Department of Orthopedic Surgery, Xi’an Honghui Hospital, Xi’an Jiaotong University, School of Medicine, Xi’an, China;,School of Medicine, Yanan University, Yanan, China
| | - Lin Liu
- Department of Orthopedic Surgery, Xi’an Honghui Hospital, Xi’an Jiaotong University, School of Medicine, Xi’an, China
| | - Zechao Qu
- Department of Orthopedic Surgery, Xi’an Honghui Hospital, Xi’an Jiaotong University, School of Medicine, Xi’an, China;,School of Medicine, Yanan University, Yanan, China
| | - Bo Zhang
- Department of Orthopedic Surgery, Xi’an Honghui Hospital, Xi’an Jiaotong University, School of Medicine, Xi’an, China;,School of Medicine, Yanan University, Yanan, China
| | - Xiangcheng Gao
- Department of Orthopedic Surgery, Xi’an Honghui Hospital, Xi’an Jiaotong University, School of Medicine, Xi’an, China;,School of Medicine, Yanan University, Yanan, China
| | - Wangli Huang
- Department of Orthopedic Surgery, Xi’an Honghui Hospital, Xi’an Jiaotong University, School of Medicine, Xi’an, China;,School of Medicine, Yanan University, Yanan, China
| | - Mingzhe Feng
- Department of Orthopedic Surgery, Xi’an Honghui Hospital, Xi’an Jiaotong University, School of Medicine, Xi’an, China;,School of Medicine, Yanan University, Yanan, China
| | - Yining Gong
- Department of Orthopedic Surgery, Xi’an Honghui Hospital, Xi’an Jiaotong University, School of Medicine, Xi’an, China
| | - Lingbo Kong
- Department of Orthopedic Surgery, Xi’an Honghui Hospital, Xi’an Jiaotong University, School of Medicine, Xi’an, China
| | - Yanjun Wang
- Department of Orthopedic Surgery, Xi’an Honghui Hospital, Xi’an Jiaotong University, School of Medicine, Xi’an, China
| | - Liang Yan
- Department of Orthopedic Surgery, Xi’an Honghui Hospital, Xi’an Jiaotong University, School of Medicine, Xi’an, China
| |
Collapse
|
16
|
Yang B, Su Y, Han S, Chen R, Sun R, Rong K, Long F, Teng H, Zhao J, Liu Q, Qin A. Aminooxyacetic acid hemihydrochloride inhibits osteoclast differentiation and bone resorption by attenuating oxidative phosphorylation. Front Pharmacol 2022; 13:980678. [PMID: 36249744 PMCID: PMC9561130 DOI: 10.3389/fphar.2022.980678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 09/14/2022] [Indexed: 11/13/2022] Open
Abstract
Osteoclasts undergo active metabolic reprogramming to acquire the energy needed during differentiation and bone resorption. Compared with immature osteoclasts, mature osteoclasts comprise higher levels of electron transport chain enzymes and more metabolically active mitochondria. Of all energy metabolism pathways, oxidative phosphorylation is considered to be the most efficient in supplying energy to osteoclasts. We found that the malate-aspartate shuttle inhibitor aminooxyacetic acid hemihydrochloride inhibits osteoclastogenesis and bone resorption by inhibiting exchange of reducing equivalents between the cytosol and the mitochondrial matrix and attenuating mitochondrial oxidative phosphorylation in vitro. The weakening of the oxidative phosphorylation pathway resulted in reduced mitochondrial function and inadequate energy supply along with reduced reactive oxygen species production. Furthermore, treatment with aminooxyacetic acid hemihydrochloride helped recover bone loss in ovariectomized mice. Our findings highlight the potential of interfering with the osteoclast intrinsic energy metabolism pathway as a treatment for osteoclast-mediated osteolytic diseases.
Collapse
Affiliation(s)
- Biao Yang
- Guangxi Key Laboratory of Regenerative Medicine, Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, China
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Yuangang Su
- Guangxi Key Laboratory of Regenerative Medicine, Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, China
| | - Shuai Han
- Guangxi Key Laboratory of Regenerative Medicine, Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, China
| | | | - Ran Sun
- Guangxi Key Laboratory of Regenerative Medicine, Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, China
| | - Kewei Rong
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Long
- Guangxi Key Laboratory of Regenerative Medicine, Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, China
| | - Hailong Teng
- Guangxi Key Laboratory of Regenerative Medicine, Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, China
| | - Jinmin Zhao
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Qian Liu
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
- *Correspondence: An Qin, ; Qian Liu,
| | - An Qin
- Guangxi Key Laboratory of Regenerative Medicine, Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, China
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: An Qin, ; Qian Liu,
| |
Collapse
|
17
|
Chen J, Song D, Xu Y, Wu L, Tang L, Su Y, Xie X, Zhao J, Xu J, Liu Q. Anti-Osteoclast Effect of Exportin-1 Inhibitor Eltanexor on Osteoporosis Depends on Nuclear Accumulation of IκBα–NF-κB p65 Complex. Front Pharmacol 2022; 13:896108. [PMID: 36110547 PMCID: PMC9468713 DOI: 10.3389/fphar.2022.896108] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
Osteoporosis affects around 200 million people globally, with menopausal women accounting for the bulk of cases. In the occurrence and development of osteoporosis, a key role is played by osteoclasts. Excessive osteoclast-mediated bone resorption activity reduces bone mass and increases bone fragility, resulting in osteoporosis. Thus, considerable demand exists for designing effective osteoporosis treatments based on targeting osteoclasts. Eltanexor (Elt; KPT-8602) is a selective nuclear-export inhibitor that covalently binds to and blocks the function of the nuclear-export protein exportin-1 (XPO1), which controls the nucleus-to-cytoplasm transfer of certain critical proteins related to growth regulation and tumor suppression, such as p53, IκBα [nuclear factor-κB (NF-κB) inhibitor α] and FOXO1; among these proteins, IκBα, a critical component of the NF-κB signaling pathway that primarily governs NF-κB activation and transcription. How Elt treatment affects osteoclasts remains poorly elucidated. Elt inhibited the growth and activity of RANKL-induced osteoclasts in vitro in a dose-dependent manner, and Elt exerted no cell-killing effect within the effective inhibitory concentration. Mechanistically, Elt was found to trap IκBα in the nucleus and thus protect IκBα from proteasome degradation, which resulted in the blocking of the translocation of IκBα and NF-κB p65 and the consequent inhibition of NF-κB activity. The suppression of NF-κB activity, in turn, inhibited the activity of two transcription factors (NFATc1 and c-Fos) essential for osteoclast formation and led to the downregulation of genes and proteins related to bone resorption. Our study thus provides a newly identified mechanism for targeting in the treatment of osteoporosis.
Collapse
Affiliation(s)
- Junchun Chen
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, Guangxi Medical University, Nanning, China
| | - Dezhi Song
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Yang Xu
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Liwei Wu
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, Guangxi Medical University, Nanning, China
| | - Lili Tang
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, Guangxi Medical University, Nanning, China
| | - YuanGang Su
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Xiaoxiao Xie
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, Guangxi Medical University, Nanning, China
| | - Jinmin Zhao
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Jiake Xu
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
- *Correspondence: Qian Liu, ; Jiake Xu,
| | - Qian Liu
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- *Correspondence: Qian Liu, ; Jiake Xu,
| |
Collapse
|
18
|
Qu Z, An H, Feng M, Huang W, Wang D, Zhang Z, Yan L. Urolithin B suppresses osteoclastogenesis via inhibiting RANKL-induced signalling pathways and attenuating ROS activities. J Cell Mol Med 2022; 26:4428-4439. [PMID: 35781786 PMCID: PMC9357644 DOI: 10.1111/jcmm.17467] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 04/06/2022] [Accepted: 06/15/2022] [Indexed: 12/14/2022] Open
Abstract
Osteoporosis (OP) has severely affected human health, which is characterized by abnormal differentiation of osteoclasts. Urolithin B (UB), as a potential natural drug, has been reported to exhibit numerous biological activities including antioxidant and anti‐inflammatory but its effects on OP, especially on RANKL‐stimulated osteoclast formation and activation, are still understood. In our study, we have demonstrated for the first time that UB inhibits RANKL‐induced osteoclast differentiation and explored its potential mechanisms of action. The RAW264.7 cells were cultured and induced with RANKL followed by UB treatment. Then, the effects of UB on mature osteoclast differentiation were evaluated by counting tartrate‐resistant acid phosphatase (TRAP)‐positive multinucleated cells and F‐actin ring analysis. Moreover, the effects of UB on RANKL‐induced reactive oxygen species (ROS) were measured by 2′, 7′‐dichlorodihydrofluorescein diacetate (DCFH‐DA) staining. Further, we explored the potential mechanisms of these downregulation effects by performing Western blotting and quantitative RT‐PCR examination. We found that UB represses osteoclastogenesis, F‐actin belts formation, osteoclast‐specific gene expressions and ROS activity in a time‐ and concentration‐dependent manner. Mechanistically, UB attenuates intracellular ROS levels by upregulation of Nrf2 and other ROS scavenging enzymes activation. Furthermore, UB also inhibited RANKL‐induced NF‐κB, MAPK and Akt signalling pathway and suppressed expression of c‐Fos and nuclear factor of activated T cells 1 (NFATc1), which is the master transcription factor of osteoclast differentiation. Taken together, our findings confirm that UB is a polyphenolic compound that can be a potential therapeutic treatment for osteoclast‐related bone diseases such as osteoporosis.
Collapse
Affiliation(s)
- Zechao Qu
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiao Tong University, Xi'an, China.,Medical College of Yan'an University, Yan'an, China
| | - Hao An
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiao Tong University, Xi'an, China.,Shaanxi University of Chinese Medicine, Xian Yang, China
| | - Mingzhe Feng
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiao Tong University, Xi'an, China.,Medical College of Yan'an University, Yan'an, China
| | - Wangli Huang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiao Tong University, Xi'an, China.,Medical College of Yan'an University, Yan'an, China
| | - Dong Wang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiao Tong University, Xi'an, China.,Medical College of Yan'an University, Yan'an, China
| | - Zhen Zhang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiao Tong University, Xi'an, China
| | - Liang Yan
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiao Tong University, Xi'an, China
| |
Collapse
|
19
|
Tao H, Tao Y, Yang C, Li W, Zhang W, Li X, Gu Y, Hong Y, Yang H, Liu Y, Yang X, Geng D. Gut Metabolite Urolithin A Inhibits Osteoclastogenesis and Senile Osteoporosis by Enhancing the Autophagy Capacity of Bone Marrow Macrophages. Front Pharmacol 2022; 13:875611. [PMID: 35645801 PMCID: PMC9135380 DOI: 10.3389/fphar.2022.875611] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 03/29/2022] [Indexed: 12/16/2022] Open
Abstract
Senile osteoporosis (SOP) is a systemic bone disease that is significantly associated with age and eventually leads to deteriorated bone strength and increased fracture risk. Urolithin A (Uro-A) is a gut microbiome-derived compound that is mainly produced from pomegranates and some nuts. Uro-A has attracted great attention in recent years in view of its protective effects on aging-related diseases, including muscle dysfunction, kidney disease and knee injury. However, its protective influence and possible mechanisms in senile osteoporosis remain unclear. Our study describes the beneficial effect of Uro-A on bone marrow macrophages (BMMs). The in vitro results demonstrated that Uro-A inhibited receptor activator for nuclear factor-κB ligand (RANKL)-induced osteoclastogenesis in BMMs in a concentration-dependent manner. Uro-A significantly reduced the expression of osteoclast-related genes and bone resorption. Mechanistically, we found that the autophagy ability of BMMs was significantly enhanced in the early stage of Uro-A treatment, accompanied by the activation of LC3 and Beclin 1. At the same time, this enhanced autophagy activity was maintained until the later stage after stimulation with RANKL. Furthermore, we found that the MARK signaling pathway was blocked by Uro-A treatment. In a mouse model of aging, Uro-A effectively inhibited bone loss in the proximal femur, spine and tibia of aging mice. These results indicated that Uro-A is a robust and effective treatment for preventing senile osteoporosis bone loss.
Collapse
Affiliation(s)
- Huaqiang Tao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yunxia Tao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chen Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wenming Li
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wei Zhang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xueyan Li
- Anesthesiology Department, Suzhou Municipal Hospital (North District), Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, China
| | - Ye Gu
- Department of Orthopedics, Changshu Hospital Affiliated to Soochow University, First People's Hospital of Changshu City, Changshu, China
| | - Yujing Hong
- Department of Preventive Medicine, Nantong University, Nantong, China
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yu Liu
- Department of Orthopedics, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, China
| | - Xing Yang
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital (North District), Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, China
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Orthopedics, Changshu Hospital Affiliated to Soochow University, First People's Hospital of Changshu City, Changshu, China
| |
Collapse
|
20
|
Huang L, Chen W, Wei L, Su Y, Liang J, Lian H, Wang H, Long F, Yang F, Gao S, Tan Z, Xu J, Zhao J, Liu Q. Lonafarnib Inhibits Farnesyltransferase via Suppressing ERK Signaling Pathway to Prevent Osteoclastogenesis in Titanium Particle-Induced Osteolysis. Front Pharmacol 2022; 13:848152. [PMID: 35300293 PMCID: PMC8921770 DOI: 10.3389/fphar.2022.848152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/10/2022] [Indexed: 11/30/2022] Open
Abstract
Wear debris after total joint arthroplasty can attract the recruitment of macrophages, which release pro-inflammatory substances, triggering the activation of osteoclasts, thereby leading to periprosthetic osteolysis (PPOL) and aseptic loosening. However, the development of pharmacological strategies targeting osteoclasts to prevent periprosthetic osteolysis has not been fruitful. In this study, we worked toward researching the effects and mechanisms of a farnesyltransferase (FTase) inhibitor Lonafarnib (Lon) on receptor activator of nuclear factor κB (NF-κB) ligand (RANKL)-induced osteoclastogenesis and bone resorption, as well as the impacts of Lon on titanium particle-induced osteolysis. To investigate the impacts of Lon on bone resorption and osteoclastogenesis in vitro, bone marrow macrophages were incubated and stimulated with RANKL and macrophage colony-stimulating factor (M-CSF). The influence of Lon on osteolysis prevention in vivo was examined utilizing a titanium particle-induced mouse calvarial osteolysis model. The osteoclast-relevant genes expression was explored by real-time quantitative PCR. Immunofluorescence was used to detect intracellular localization of nuclear factor of activated T cells 1 (NFATc1). SiRNA silence assay was applied to examine the influence of FTase on osteoclasts activation. Related signaling pathways, including NFATc1 signaling, NF-κB, mitogen-activated protein kinases pathways were identified by western blot assay. Lon was illustrated to suppress bone resorptive function and osteoclastogenesis in vitro, and it also reduced the production of pro-inflammatory substances and prevented titanium particle-induced osteolysis in vivo. Lon decreased the expression of osteoclast-relevant genes and suppressed NFATc1 nuclear translocation and auto-amplification. Mechanistically, Lon dampened FTase, and inhibition of FTase reduced osteoclast formation by suppressing ERK signaling. Lon is a promising treatment option for osteoclast-related osteolysis diseases including periprosthetic osteolysis by targeted inhibition of FTase through suppressing ERK signaling.
Collapse
Affiliation(s)
- Linke Huang
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Department of Orthopaedics, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Weiwei Chen
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Linhua Wei
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China.,The Affiliated Nanning Infectious Disease Hospital of Guangxi Medical University, The Fourth People's Hospital of Nanning, Nanning, China
| | - Yuangang Su
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Jiamin Liang
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Haoyu Lian
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Hui Wang
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Feng Long
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Fan Yang
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Shiyao Gao
- Department of Orthopaedics, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhen Tan
- Department of Orthopaedics, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jiake Xu
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Jinmin Zhao
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Qian Liu
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
21
|
Li D, Luo F, Guo T, Han S, Wang H, Lin Q. Targeting NF-κB pathway by dietary lignans in inflammation: expanding roles of gut microbiota and metabolites. Crit Rev Food Sci Nutr 2022; 63:5967-5983. [PMID: 35068283 DOI: 10.1080/10408398.2022.2026871] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Inflammation is a major factor affecting human health. Nuclear factor-kappa B (NF-κB) plays a vital role in the development of inflammation, and the promoters of most inflammatory cytokine genes have NF-κB-binding sites. Targeting NF-κB could be an exciting route for the prevention and treatment of inflammatory diseases. As important constituents of natural plants, lignans are proved to have numerous biological functions. There are growing pieces of evidence demonstrate that lignans have the potential anti-inflammatory activities. In this work, the type, structure and source of lignans and the influence on mitigating the inflammation are systematically summarized. This review focuses on the targeting NF-κB signaling pathway in the inflammatory response by different lignans and their molecular mechanisms. Lignans also regulate gut microflora and change gut microbial metabolites, which exert novel pathway to prevent NF-κB activation. Taken together, lignans target NF-κB with various mechanisms to inhibit inflammatory cytokine expressions in the inflammatory response. It will provide a scientific theoretical basis for further research on the anti-inflammatory effects of lignans and the development of functional foods.
Collapse
Affiliation(s)
- Dan Li
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Processed Food for Special Medical Purpose, College of Food Science and Engineering, National Engineering Laboratory for Deep Process of Rice and Byproducts, Central South University of Forestry and Technology, Changsha, China
| | - Feijun Luo
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Processed Food for Special Medical Purpose, College of Food Science and Engineering, National Engineering Laboratory for Deep Process of Rice and Byproducts, Central South University of Forestry and Technology, Changsha, China
| | - Tianyi Guo
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Processed Food for Special Medical Purpose, College of Food Science and Engineering, National Engineering Laboratory for Deep Process of Rice and Byproducts, Central South University of Forestry and Technology, Changsha, China
| | - Shuai Han
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Processed Food for Special Medical Purpose, College of Food Science and Engineering, National Engineering Laboratory for Deep Process of Rice and Byproducts, Central South University of Forestry and Technology, Changsha, China
| | - Hanqing Wang
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Processed Food for Special Medical Purpose, College of Food Science and Engineering, National Engineering Laboratory for Deep Process of Rice and Byproducts, Central South University of Forestry and Technology, Changsha, China
| | - Qinlu Lin
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Processed Food for Special Medical Purpose, College of Food Science and Engineering, National Engineering Laboratory for Deep Process of Rice and Byproducts, Central South University of Forestry and Technology, Changsha, China
| |
Collapse
|
22
|
Ding D, Yan J, Feng G, Zhou Y, Ma L, Jin Q. Dihydroartemisinin attenuates osteoclast formation and bone resorption via inhibiting the NF‑κB, MAPK and NFATc1 signaling pathways and alleviates osteoarthritis. Int J Mol Med 2022; 49:4. [PMID: 34738623 PMCID: PMC8589459 DOI: 10.3892/ijmm.2021.5059] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 10/15/2021] [Indexed: 12/29/2022] Open
Abstract
Osteoarthritis (OA) is a chronic, progressive and degenerative disease, and its incidence is increasing on a yearly basis. However, the pathological mechanism of OA at each stage is still unclear. The present study aimed to explore the underlying mechanism of dihydroartemisinin (DHA) in terms of its ability to inhibit osteoclast activation, and to determine its effects on OA in rats. Bone marrow‑derived macrophages were isolated as osteoclast precursors. In the presence or absence of DHA, osteoclast formation was assessed by tartrate‑resistant acid phosphatase (TRAP) staining, cell viability was assessed by Cell Counting Kit‑8 assay, the presence of F‑actin rings was assessed by immunofluorescence, bone resorption was determined by bone slices, luciferase activities of NF‑κB and nuclear factor of activated T cell cytoplasmic 1 (NFATc1) were determined using luciferase assay kits, the protein levels of biomolecules associated with the NF‑κB, MAPK and NFATc1 signaling pathways were determined using western blotting, and the expression of genes involved in osteoclastogenesis were measured using reverse transcription‑quantitative PCR. A knee OA rat model was designed by destabilizing the medial meniscus (DMM). A total of 36 rats were assigned to three groups, namely the sham‑operated, DMM + vehicle and DMM + DHA groups, and the rats were administered DHA or DMSO. At 4 and 8 weeks postoperatively, the microarchitecture of the subchondral bone was analyzed using micro‑CT, the thickness of the cartilage layers was calculated using H&E staining, the extent of cartilage degeneration was scored using Safranin O‑Fast Green staining, TRAP‑stained osteoclasts were counted, and the levels of receptor activator of NF‑κB ligand (RANKL), C‑X‑C‑motif chemokine ligand 12 (CXCL12) and NFATc1 were measured using immunohistochemistry. DHA was found to inhibit osteoclast formation without cytotoxicity, and furthermore, it did not affect bone formation. In addition, DHA suppressed the expression levels of NF‑κB, MAPK, NFATc1 and genes involved in osteoclastogenesis. Progressive cartilage loss was observed at 8 weeks postoperatively. Subchondral bone remodeling was found to be dominated by bone resorption accompanied by increases in the levels of RANKL, CXCL12 and NFATc1 during the first 4 weeks. DHA was found to delay OA progression by inhibiting osteoclast formation and bone resorption during the early phase of OA. Taken together, the results of the present study demonstrated that the mechanism through which DHA could inhibit osteoclast activation may be associated with the NF‑κB, MAPK and NFATc1 signaling pathways, thereby indicating a potential novel strategy for OA treatment.
Collapse
Affiliation(s)
- Dong Ding
- Ningxia Medical University, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Jiangbo Yan
- Ningxia Medical University, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Gangning Feng
- Ningxia Medical University, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Yong Zhou
- Ningxia Medical University, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Long Ma
- Orthopedics Ward 3, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Qunhua Jin
- Ningxia Medical University, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| |
Collapse
|
23
|
An Insight into Sesamolin: Physicochemical Properties, Pharmacological Activities, and Future Research Prospects. Molecules 2021; 26:molecules26195849. [PMID: 34641392 PMCID: PMC8510241 DOI: 10.3390/molecules26195849] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/22/2021] [Accepted: 09/24/2021] [Indexed: 02/08/2023] Open
Abstract
Sesame seeds are rich in lignan content and have been well-known for their health benefits. Unlike the other sesame lignan compounds (i.e., sesamin and sesamol), the study of the pharmacological activity of sesamolin has not been explored widely. This review, therefore, summarizes the information related to sesamolin’s pharmacological activities, and the mechanism of action. Moreover, the influence of its physicochemical properties on pharmacological activity is also discussed. Sesamolin possessed neuroprotective activity against hypoxia-induced reactive oxygen species (ROS) and oxidative stress in neuron cells by reducing the ROS and inhibiting apoptosis. In skin cancer, sesamolin exhibited antimelanogenesis by affecting the expression of the melanogenic enzymes. The anticancer activity of sesamolin based on antiproliferation and inhibition of migration was demonstrated in human colon cancer cells. In addition, treatment with sesamolin could stimulate immune cells to enhance the cytolytic activity to kill Burkitt’s lymphoma cells. However, the toxicity and safety of sesamolin have not been reported. And there is also less information on the experimental study in vivo. The limited aqueous solubility of sesamolin becomes the main problem, which affects its pharmacological activity in the in vitro experiment and clinical efficacy. Therefore, solubility enhancement is needed for further investigation and determination of its pharmacological activity profiles. Since there are fewer reports studying this issue, it could become a future prospective research opportunity.
Collapse
|