1
|
Zhang X, Zhang L, Li D, Wang Q, Wang L, Zheng Z, Xie Y. Computational exploration of Eucommia ulmoides flavonoids as potential RANKL inhibitors via molecular docking and dynamics simulations. Sci Rep 2025; 15:17175. [PMID: 40382406 PMCID: PMC12085681 DOI: 10.1038/s41598-025-01913-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 05/09/2025] [Indexed: 05/20/2025] Open
Abstract
Osteoporosis, characterized by excessive osteoclast activation, is mediated through the RANKL/RANK/OPG signaling axis. While flavonoids from Eucommia ulmoides (EU) have demonstrated anti-osteoclastogenic activity, their atomic-level mechanisms remain elusive. Here, we investigated six EU-derived flavonoids (cyrtominetin, quercetin, syringetin, genistein, ombuin, and kaempferol) targeting RANKL using integrated computational approaches. Molecular docking revealed strong binding affinities (Total_Score > 4.0) for all compounds, with cyrtominetin exhibiting the highest affinity (-50.205 kJ/mol via MM-PBSA), primarily through hydrogen bonds with Gly178, His180, Lys181, and Asn295. Moreover, most flavonoids interacted with RANKL by forming strong hydrogen bonds with Gly178 and Asn295, exhibiting higher binding affinity that was identified as essential for the activity. All-atom molecular dynamics simulations (100 ns) confirmed complex stability, demonstrating: low RMSD fluctuations (< 4.0 Å) and compact Rg values (16.0-17.0 Å). Notably, binding free energy decomposition identified both electrostatic and van der Waals contributions as critical for stabilization. These results identify cyrtominetin as a promising lead compound for RANKL inhibition, providing structural insights for designing flavonoid-based therapeutics against osteoporosis.
Collapse
Affiliation(s)
- Xiaofei Zhang
- Department of Laboratory Medicine, Northwest Womens and Childrens Hospital, 1616 Yanxiang Road, Xi'an, 710061, Shaanxi, China
| | - Lixia Zhang
- Department of Clinical Laboratory, Shaanxi Provincial Peoples Hospital, Xi'an, China
| | - Dan Li
- Department of Laboratory Medicine, Northwest Womens and Childrens Hospital, 1616 Yanxiang Road, Xi'an, 710061, Shaanxi, China
| | - Qi Wang
- Department of Clinical Laboratory, Second Affiliated Hospital of Xian Jiaotong University, Xi'an, Shaanxi, China
| | - Libin Wang
- Department of Laboratory Medicine, Northwest Womens and Childrens Hospital, 1616 Yanxiang Road, Xi'an, 710061, Shaanxi, China
| | - Ziqi Zheng
- College of Life Sciences, Northwest University, 229 North Taibai Road, Xi'an, Shaanxi, 710069, People's Republic of China
| | - Yun Xie
- Department of Laboratory Medicine, Northwest Womens and Childrens Hospital, 1616 Yanxiang Road, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
2
|
Zhao R, Fu J, Jiang Y, Wu L, Yan Z, Liu Y. Maltol promotes osteoclastogenesis and exacerbates periodontitis via TRAF6/JNK/Nfatc1 pathway. Biochem Biophys Res Commun 2025; 770:151855. [PMID: 40373380 DOI: 10.1016/j.bbrc.2025.151855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 04/09/2025] [Accepted: 04/18/2025] [Indexed: 05/17/2025]
Abstract
OBJECTIVE The aim of this study was to investigate the role of maltol in osteoclast differentiation and its mechanism, and to provide evidence for the effect of common sweeteners on periodontal tissue destruction and the prevention of periodontitis. METHODS BMMNCs were treated with maltol, M-CSF and RANKL to observe their osteoclast potential. The differentiation of osteoclasts was observed by TRAP staining, Western blotting and RT-PCR analysis. Further investigations into the relevant signaling pathways were carried out. In vivo, periodontitis was established by ligating the maxillary second molars of mice with silk thread (n = 8 in each group). After that, we evaluated the effect of maltol on bone resorption by oral gavage. RESULTS Maltol significantly promoted osteoclast differentiation of rankl stimulated BMMNCs. This is mediated by modulation of the tumor necrosis factor receptor-related factor 6 (TRAF6)/C-Jun n-terminal kinase (JNK)/activated T cell nuclear factor 1 (Nfatc1) signaling pathway. In addition, maltol can significantly promote bone resorption in animal models of periodontitis. CONCLUSIONS Maltol promotes OC differentiation of bone marrow mesenchymal stem cells induced by rankl through TRAF/JNK pathway and upregulates NFATc1 expression. Maltol promotes bone resorption by promoting osteoclast differentiation in experimental periodontitis model of mice.
Collapse
Affiliation(s)
- Rui Zhao
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, 100070, PR China
| | - Jingfei Fu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, 100070, PR China
| | - Yiyang Jiang
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, 100070, PR China
| | - Lili Wu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, 100070, PR China
| | - Ziqi Yan
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, 100070, PR China
| | - Yi Liu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, 100070, PR China.
| |
Collapse
|
3
|
Yun HM, Kim B, Kim E, Park KR. Rhusflavone Modulates Osteoclastogenesis Through RANKL-Induced AKT Signaling in Bone Marrow-Derived Macrophages. Int J Mol Sci 2025; 26:3025. [PMID: 40243668 PMCID: PMC11988637 DOI: 10.3390/ijms26073025] [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: 03/06/2025] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
Osteoclast differentiation inhibition is a viable treatment strategy for osteoporosis because osteoclasts play a vital role in disease progression. Rhusflavone (Rhus), a biflavonoid, exhibits a sedative-hypnotic effect via the positive allosteric modulation of GABA(A) receptors. Although several biflavonoids possess activities that help prevent bone loss, the potential effects of Rhus on osteoclastogenesis have not been reported yet. In this study, we investigated the effects and underlying biological mechanisms of Rhus isolated from the dried roots of Rhus succedanea on osteoclastogenesis in primary cultured bone marrow-derived macrophages. No cytotoxicity was observed in bone marrow macrophages (BMMs) or during osteoclast differentiation. However, Rhus reduced the number of tartrate-resistant acid phosphatase (TRAP)-positive multinuclear osteoclasts during receptor activator of nuclear factor kappa B ligand (RANKL)-induced osteoclastogenesis. The results of F-actin ring formation demonstrated that Rhus suppresses the bone resorption activity of osteoclasts. Additionally, Rhus inhibits the expression of osteoclast differentiation marker proteins, specifically c-Fos and NF-ATc1. Western blot analysis revealed that Rhus primarily attenuated RANKL-mediated key signaling pathways, particularly the AKT signaling pathway. Furthermore, we found that the AKT activator and inhibitor pharmacologically abolished and enhanced the inhibitory effects of Rhus on osteoclast differentiation, respectively. Taken together, our findings provide evidence that Rhus is a promising biologically active compound that regulates osteoclast differentiation by inhibiting the AKT signaling pathway, which may contribute to future drug development.
Collapse
Affiliation(s)
- Hyung-Mun Yun
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Bomi Kim
- National Institute for Korean Medicine Development, Gyeongsan 38540, Republic of Korea; (B.K.); (E.K.)
| | - Eonmi Kim
- National Institute for Korean Medicine Development, Gyeongsan 38540, Republic of Korea; (B.K.); (E.K.)
| | - Kyung-Ran Park
- Honam Regional Center, Korea Basic Science Institute (KBSI), Gwangju 61751, Republic of Korea
| |
Collapse
|
4
|
Liu T, Li J, Duan M, Wang Y, Jiang Z, Gan C, Xiang Z, Sheng J, Wang X, Xu H. Stephanine Protects Against Osteoporosis by Suppressing Osteoclastogenesis via Inhibition of the RANKL-RANK Interaction. J Cell Mol Med 2024; 28:e70256. [PMID: 39636143 PMCID: PMC11619157 DOI: 10.1111/jcmm.70256] [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: 07/24/2024] [Revised: 10/29/2024] [Accepted: 11/20/2024] [Indexed: 12/07/2024] Open
Abstract
The interaction between the receptor activator of nuclear factor-κB ligand (RANKL) and its receptor RANK is known to regulate osteoclastogenesis in bone remodelling and has become an important therapeutic target for the treatment of osteoporosis. Stephanine (SA), an isoquinoline aporphine-type alkaloid isolated from Stephania plants, possesses excellent anti-inflammatory effects and can be used for rheumatoid arthritis treatment. However, its specific role in osteoclastogenesis and osteoporosis remains unknown. In this study, we investigated the influence of SA on osteoclastogenesis in RANKL-stimulated RAW 264.7 cells and osteoporosis in an ovariectomised (OVX) mouse model and elucidated the underlying molecular mechanism. In vitro, SA can bind to RANK and RANKL with the KD values of 3.7 and 76.47 μM, respectively, and disrupt the RANKL-RANK interaction, which inhibits RANKL-stimulated RANK-tumour necrosis factor receptor associated factor 6 (TRAF6) binding and RANK signalling pathways activation, downregulates the expression of key osteoclastogenesis-related regulatory factors in osteoclast precursors, ultimately suppresses osteoclast differentiation and activation. In vivo, SA significantly ameliorated bone loss through inhibiting osteoclastogenesis in OVX mice because of the decreased number of osteoclasts and the increased trabecular bone area. SA markedly inhibited the serum levels of tartrate-resistant acid phosphatase 5b (TRACP-5b), c-telopeptide of type I collagen (CTX-I), and RANKL, whereas it increased that of osteoprotegerin (OPG) in OVX mice. Additionally, SA strikingly downregulated the OVX-induced expression of osteoclast-specific genes and proteins. Taken together, this study elucidated that SA can effectively protect against osteoporosis by suppressing osteoclastogenesis via inhibition of the RANKL-RANK interaction, which supports the potential application of SA as a natural therapeutic agent for osteoporosis.
Collapse
Affiliation(s)
- Titi Liu
- College of ScienceYunnan Agricultural UniversityKunmingChina
- Key Laboratory of Pu‐Er Tea Science, Ministry of EducationYunnan Agricultural UniversityKunmingChina
| | - Jin Li
- College of ScienceYunnan Agricultural UniversityKunmingChina
- Key Laboratory of Pu‐Er Tea Science, Ministry of EducationYunnan Agricultural UniversityKunmingChina
| | - Meiyan Duan
- Key Laboratory of Pu‐Er Tea Science, Ministry of EducationYunnan Agricultural UniversityKunmingChina
- College of Food Science and TechnologyYunnan Agricultural UniversityKunmingChina
| | - Ya Wang
- College of ScienceYunnan Agricultural UniversityKunmingChina
- Key Laboratory of Pu‐Er Tea Science, Ministry of EducationYunnan Agricultural UniversityKunmingChina
| | - Zhe Jiang
- Key Laboratory of Pu‐Er Tea Science, Ministry of EducationYunnan Agricultural UniversityKunmingChina
- College of Food Science and TechnologyYunnan Agricultural UniversityKunmingChina
| | - Chunxia Gan
- Key Laboratory of Pu‐Er Tea Science, Ministry of EducationYunnan Agricultural UniversityKunmingChina
| | - Zemin Xiang
- Key Laboratory of Pu‐Er Tea Science, Ministry of EducationYunnan Agricultural UniversityKunmingChina
- College of Food Science and TechnologyYunnan Agricultural UniversityKunmingChina
| | - Jun Sheng
- Key Laboratory of Pu‐Er Tea Science, Ministry of EducationYunnan Agricultural UniversityKunmingChina
| | - Xuanjun Wang
- Key Laboratory of Pu‐Er Tea Science, Ministry of EducationYunnan Agricultural UniversityKunmingChina
| | - Huanhuan Xu
- College of ScienceYunnan Agricultural UniversityKunmingChina
- Key Laboratory of Pu‐Er Tea Science, Ministry of EducationYunnan Agricultural UniversityKunmingChina
| |
Collapse
|
5
|
Huang J, Song D, Xu M, Gan K, Wang C, Chen L, Huang Q, Chen J, Su Y, Xu J, Zhao J, Liu Q. Trifolirhizin reduces osteoclast formation and prevents inflammatory osteolysis by inhibiting RANKL-induced activation of NF-κB and MAPK signaling pathways and ROS. Phytother Res 2024; 38:4650-4666. [PMID: 39098645 DOI: 10.1002/ptr.8299] [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: 10/18/2023] [Revised: 06/25/2024] [Accepted: 07/08/2024] [Indexed: 08/06/2024]
Abstract
Inflammatory osteolysis is often caused by the excessive activation of osteoclasts stimulated by bacterial products such as lipopolysaccharide. The natural flavonoid trifolirhizin (TRI) has anti-inflammatory properties; however, its function in inflammatory bone lysis remains unclear. This study aimed to elucidate the potential regulatory mechanisms of TRI in osteoclasts.Tartrate-resistant acid phosphatase (TRAP) staining, acid secretion assays, podosomal actin belt fluorescence staining, and bone resorption assays were used to investigate the effects of TRI on osteoclast differentiation and bone resorption. A reactive oxygen species (ROS) measurement kit was used to detect the effect of TRI on ROS levels in osteoclasts. The effects of TRI on genes and signaling pathways related to osteoclast differentiation were determined by quantitative polymerase chain reaction (qPCR) and western blotting. A mouse model of lipopolysaccharide-mediated inflammatory osteolysis was established, and the effects of TRI treatment on bone mass were observed using micro-CT and histological examination. Mechanistically, TRI reduced ROS production by inhibiting receptor activator of nuclear factor-κB ligand (RANKL)-induced activation of the nuclear factor-κB (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways, and by upregulating the expression levels of the anti-ROS enzymes heme oxygenase-1 (HO-1) and catalase (CAT), which contributed to the degradation of ROS, ultimately leading to a decrease in osteoclastogenesis. TRI inhibited osteoclast formation and ameliorated lipopolysaccharide (LPS)-mediated inflammatory osteolysis. Thus, TRI may be a candidate agent for anti-inflammatory osteolysis.
Collapse
Affiliation(s)
- Jian Huang
- Guangxi Key Laboratory of 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, Life Sciences Institute, Guangxi Medical University, Nanning, China
| | - Dezhi Song
- Guangxi Key Laboratory of 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, Life Sciences Institute, Guangxi Medical University, Nanning, China
| | - Minglian Xu
- Guangxi Key Laboratory of 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, Life Sciences Institute, Guangxi Medical University, Nanning, China
| | - Kai Gan
- Guangxi Key Laboratory of 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, Life Sciences Institute, Guangxi Medical University, Nanning, China
| | - Chaofeng Wang
- Guangxi Key Laboratory of 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, Life Sciences Institute, Guangxi Medical University, Nanning, China
| | - Liuyuan Chen
- Guangxi Key Laboratory of 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, Life Sciences Institute, Guangxi Medical University, Nanning, China
| | - Qian Huang
- Guangxi Key Laboratory of 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, Life Sciences Institute, Guangxi Medical University, Nanning, China
| | - Junchun Chen
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical University, Nanning, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yuangang Su
- Guangxi Key Laboratory of 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, Life Sciences Institute, Guangxi Medical University, Nanning, China
| | - Jiake Xu
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical University, Nanning, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jinmin Zhao
- Guangxi Key Laboratory of 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, Life Sciences Institute, Guangxi Medical University, Nanning, China
| | - Qian Liu
- Guangxi Key Laboratory of 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, Life Sciences Institute, Guangxi Medical University, Nanning, China
| |
Collapse
|
6
|
Joseph AG, Biji M, Murali VP, Sherin DR, Valsan A, Sukumaran VP, Radhakrishnan KV, Maiti KK. A comprehensive apoptotic assessment of niloticin in cervical cancer cells: a tirucallane-type triterpenoid from Aphanamixis polystachya (Wall.) Parker. RSC Med Chem 2024:d4md00318g. [PMID: 39246746 PMCID: PMC11378019 DOI: 10.1039/d4md00318g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 08/01/2024] [Indexed: 09/10/2024] Open
Abstract
Pharmacologically active small organic molecules derived from natural resources are prominent drug candidates due to their inherent structural diversity. Herein, we explored one such bioactive molecule, niloticin, which is a tirucallane-type triterpenoid isolated from the stem barks of Aphanamixis polystachya (Wall.) Parker. After initial screening with other isolated compounds from the same plant, niloticin demonstrated selective cytotoxicity against cervical cancer cells (HeLa) with an IC50 value of 11.64 μM. Whereas the compound exhibited minimal cytotoxicity in normal epithelial cell line MCF-10A, with an IC50 value of 83.31 μM. Subsequently, in silico molecular docking studies of niloticin based on key apoptotic proteins such as p53, Fas, FasL, and TNF β revealed striking binding affinity, reflecting docking scores of -7.2, -7.1, -6.8, and -7.2. Thus, the binding stability was evaluated through molecular dynamic simulation. In a downstream process, the apoptotic capability of niloticin was effectively validated through in vitro fluorimetric assays, encompassing nuclear fragmentation. Additionally, an insightful approach involving surface-enhanced Raman spectroscopy (SERS) re-establishes the occurrence of DNA cleavage during cellular apoptosis. Furthermore, niloticin was observed to induce apoptosis through both intrinsic and extrinsic pathways. This was evidenced by the upregulation of upstream regulatory molecules such as CD40 and TNF, which facilitate the activation of caspase 8. Concurrently, niloticin-induced p53 activation augmented the expression of proapoptotic proteins Bax and Bcl-2 and downregulation of IAPs, leading to the release of cytochrome C and subsequent activation of caspase 9. Therefore, the reflection of mitochondrial-mediated apoptosis is in good agreement with molecular docking studies. Furthermore, the anti-metastatic potential was evidenced by wound area closure and Ki67 expression patterns. This pivotal in vitro assessment confirms the possibility of niloticin being a potent anti-cancer drug candidate, and to the best of our knowledge, this is the first comprehensive anticancer assessment of niloticin in HeLa cells.
Collapse
Affiliation(s)
- Anuja Gracy Joseph
- CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Chemical Sciences and Technology Division (CSTD), Organic Chemistry Section Industrial Estate Thiruvananthapuram 695019 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Mohanan Biji
- CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Chemical Sciences and Technology Division (CSTD), Organic Chemistry Section Industrial Estate Thiruvananthapuram 695019 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Vishnu Priya Murali
- CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Chemical Sciences and Technology Division (CSTD), Organic Chemistry Section Industrial Estate Thiruvananthapuram 695019 India
| | - Daisy R Sherin
- School of Digital Sciences, Kerala University of Digital Sciences, Innovation and Technology Thiruvananthapuram-695317 India
| | - Alisha Valsan
- CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Chemical Sciences and Technology Division (CSTD), Organic Chemistry Section Industrial Estate Thiruvananthapuram 695019 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Vimalkumar P Sukumaran
- CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Chemical Sciences and Technology Division (CSTD), Organic Chemistry Section Industrial Estate Thiruvananthapuram 695019 India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Kokkuvayil Vasu Radhakrishnan
- CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Chemical Sciences and Technology Division (CSTD), Organic Chemistry Section Industrial Estate Thiruvananthapuram 695019 India
| | - Kaustabh Kumar Maiti
- CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Chemical Sciences and Technology Division (CSTD), Organic Chemistry Section Industrial Estate Thiruvananthapuram 695019 India
| |
Collapse
|
7
|
Liao J, Lu L, Chu X, Xiong Y, Zhou W, Cao F, Cheng P, Shahbazi MA, Liu G, Mi B. Cell membrane coated nanoparticles: cutting-edge drug delivery systems for osteoporosis therapy. NANOSCALE 2024; 16:8236-8255. [PMID: 38584466 DOI: 10.1039/d3nr06264c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Osteoporosis, characterized by a reduction in bone mineral density, represents a prevalent skeletal disorder with substantial global health implications. Conventional therapeutic strategies, exemplified by bisphosphonates and hormone replacement regimens, though effective, encounter inherent limitations and challenges. Recent years have witnessed the surge of cell-membrane-coated nanoparticles (CMNPs) as a promising intervention for osteoporosis, leveraging their distinct attributes including refined biocompatibility, heightened pharmaceutical payload capacity, as well as targeted drug release kinetics. However, a comprehensive review consolidating the application of CMNPs-based therapy for osteoporosis remains absent within the existing literature. In this review, we provide a concise overview of the distinctive pathogenesis associated with osteoporosis, alongside an in-depth exploration of the physicochemical attributes intrinsic to CMNPs derived from varied cellular sources. Subsequently, we explore the potential utility of CMNPs, elucidating emerging trends in their deployment for osteoporosis treatment through multifaceted therapeutic approaches. By linking the notable attributes of CMNPs with their roles in mitigating osteoporosis, this review serves as a catalyst for further advances in the design of advanced CMNPs tailored for osteoporosis management. Ultimately, such progress is promising for enhancing outcomes in anti-bone loss interventions, paving the way for clinical translation in the near future.
Collapse
Affiliation(s)
- Jiewen Liao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Li Lu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Xiangyu Chu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Yuan Xiong
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Wu Zhou
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Faqi Cao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Peng Cheng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Mohammad-Ali Shahbazi
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| | - Guohui Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Bobin Mi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| |
Collapse
|
8
|
Jeong S, Kim IK, Moon H, Kim H, Song BW, Choi JW, Kim SW, Lee S, Chae DS, Lim S. A 70% Ethanol Neorhodomela munita Extract Attenuates RANKL-Induced Osteoclast Activation and H 2O 2-Induced Osteoblast Apoptosis In Vitro. Molecules 2024; 29:1741. [PMID: 38675559 PMCID: PMC11052068 DOI: 10.3390/molecules29081741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
The rapid aging of the population worldwide presents a significant social and economic challenge, particularly due to osteoporotic fractures, primarily resulting from an imbalance between osteoclast-mediated bone resorption and osteoblast-mediated bone formation. While conventional therapies offer benefits, they also present limitations and a range of adverse effects. This study explores the protective impact of Neorhodomela munita ethanol extract (EN) on osteoporosis by modulating critical pathways in osteoclastogenesis and apoptosis. Raw264.7 cells and Saos-2 cells were used for in vitro osteoclast and osteoblast models, respectively. By utilizing various in vitro methods to detect osteoclast differentiation/activation and osteoblast death, it was demonstrated that the EN's potential to inhibit RANKL induced osteoclast formation and activation by targeting the MAPKs-NFATc1/c-Fos pathway and reducing H2O2-induced cell death through the downregulation of apoptotic signals. This study highlights the potential benefits of EN for osteoporosis and suggests that EN is a promising natural alternative to traditional treatments.
Collapse
Affiliation(s)
- Seongtae Jeong
- The Interdisciplinary Graduate Program in Integrative Biotechnology, Yonsei University, Seoul 03722, Republic of Korea;
| | - Il-Kwon Kim
- Department of Convergence Science, College of Medicine, Catholic Kwandong University, International St. Mary’s Hospital, Incheon 22711, Republic of Korea;
| | - Hanbyeol Moon
- Department of Integrated Omics for Biomedical Sciences, Graduate School, Yonsei University, Seoul 03722, Republic of Korea;
| | - Hojin Kim
- Department for Medical Science, College of Medicine, Catholic Kwandong University, Gangneung-si 25601, Republic of Korea;
| | - Byeong-Wook Song
- Department of Convergence Science, College of Medicine, Catholic Kwandong University, Gangneung-si 25601, Republic of Korea; (B.-W.S.); (S.W.K.); (S.L.)
| | - Jung-Won Choi
- Medical Science Research Institute, College of Medicine, Catholic Kwandong University, Incheon Metropolitan City 22711, Republic of Korea;
| | - Sang Woo Kim
- Department of Convergence Science, College of Medicine, Catholic Kwandong University, Gangneung-si 25601, Republic of Korea; (B.-W.S.); (S.W.K.); (S.L.)
| | - Seahyoung Lee
- Department of Convergence Science, College of Medicine, Catholic Kwandong University, Gangneung-si 25601, Republic of Korea; (B.-W.S.); (S.W.K.); (S.L.)
| | - Dong-Sik Chae
- Department of Orthopedic Surgery, International St. Mary’s Hospital, Catholic Kwandong University, Gangneung-si 25601, Republic of Korea
| | - Soyeon Lim
- Department of Convergence Science, College of Medicine, Catholic Kwandong University, Gangneung-si 25601, Republic of Korea; (B.-W.S.); (S.W.K.); (S.L.)
| |
Collapse
|
9
|
Zheng H, Liu Y, Deng Y, Li Y, Liu S, Yang Y, Qiu Y, Li B, Sheng W, Liu J, Peng C, Wang W, Yu H. Recent advances of NFATc1 in rheumatoid arthritis-related bone destruction: mechanisms and potential therapeutic targets. Mol Med 2024; 30:20. [PMID: 38310228 PMCID: PMC10838448 DOI: 10.1186/s10020-024-00788-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] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 01/22/2024] [Indexed: 02/05/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory disease characterized by inflammation of the synovial tissue and joint bone destruction, often leading to significant disability. The main pathological manifestation of joint deformity in RA patients is bone destruction, which occurs due to the differentiation and proliferation of osteoclasts. The transcription factor nuclear factor-activated T cell 1 (NFATc1) plays a crucial role in this process. The regulation of NFATc1 in osteoclast differentiation is influenced by three main factors. Firstly, NFATc1 is activated through the upstream nuclear factor kappa-B ligand (RANKL)/RANK signaling pathway. Secondly, the Ca2+-related co-stimulatory signaling pathway amplifies NFATc1 activity. Finally, negative regulation of NFATc1 occurs through the action of cytokines such as B-cell Lymphoma 6 (Bcl-6), interferon regulatory factor 8 (IRF8), MAF basic leucine zipper transcription factor B (MafB), and LIM homeobox 2 (Lhx2). These three phases collectively govern NFATc1 transcription and subsequently affect the expression of downstream target genes including TRAF6 and NF-κB. Ultimately, this intricate regulatory network mediates osteoclast differentiation, fusion, and the degradation of both organic and inorganic components of the bone matrix. This review provides a comprehensive summary of recent advances in understanding the mechanism of NFATc1 in the context of RA-related bone destruction and discusses potential therapeutic agents that target NFATc1, with the aim of offering valuable insights for future research in the field of RA. To assess their potential as therapeutic agents for RA, we conducted a drug-like analysis of potential drugs with precise structures.
Collapse
Affiliation(s)
- Hao Zheng
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Innovative Materia Medica Research Institute, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Yuexuan Liu
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Innovative Materia Medica Research Institute, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Yasi Deng
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Innovative Materia Medica Research Institute, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Yunzhe Li
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Innovative Materia Medica Research Institute, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Shiqi Liu
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Innovative Materia Medica Research Institute, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Yong Yang
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Innovative Materia Medica Research Institute, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Yun Qiu
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Innovative Materia Medica Research Institute, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Bin Li
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Innovative Materia Medica Research Institute, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Wenbing Sheng
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Innovative Materia Medica Research Institute, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Jinzhi Liu
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Innovative Materia Medica Research Institute, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Caiyun Peng
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Innovative Materia Medica Research Institute, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Wei Wang
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Innovative Materia Medica Research Institute, Hunan University of Chinese Medicine, Changsha, 410208, China.
| | - Huanghe Yu
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Innovative Materia Medica Research Institute, Hunan University of Chinese Medicine, Changsha, 410208, China.
| |
Collapse
|
10
|
Hou J, Xu P, Zhong Y, Zhou Z, Zhang W. Interleukin-21 knockout reduces bone loss in ovariectomized mice by inhibiting osteoclastogenesis. Biosci Biotechnol Biochem 2023; 87:1265-1273. [PMID: 37708033 DOI: 10.1093/bbb/zbad103] [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: 04/17/2023] [Accepted: 07/27/2023] [Indexed: 09/16/2023]
Abstract
Estrogen deficiency accelerates osteoporosis in elderly women. However, the role of IL-21 in postmenopausal osteoporosis remains unclear. Female wild-type (WT) C57BL/6 and IL-21 knockout (KO) mice were used for ovariectomy (OVX). Here, IL-21 levels were significantly increased in the serum and bone tissues of WT-OVX mice. The trabecular bone space of the femur was significantly increased, and the bone mass was reduced in OVX mice, accompanied by a significant decrease in the maximum load, energy absorption, and elastic modulus indices. In contrast, IL-21 knockout effectively alleviated the effects of OVX on bone mass. Serum TRACP-5b and receptor activator of nuclear factor kappa B ligand (RANKL) levels and osteoclastogenesis were significantly higher in OVX mice than in sham mice, while serum TRACP-5b and RANKL levels and osteoclastogenesis were significantly decreased in IL-21 KO + OVX mice compared to WT + OVX mice. IL-21 knockdown reduces TRACP-5b, RANKL, and osteoclastogenesis, effectively preventing bone resorption and alleviating the progression of OVX-induced osteoporosis.
Collapse
Affiliation(s)
- Junlong Hou
- Department of Orthopaedics, Jieyang People's Hospital, Jieyang, China
| | - Ping Xu
- Spinal Trauma Area 2, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yanheng Zhong
- Spinal Trauma Area 2, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Zhigang Zhou
- Spinal Trauma Area 2, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Department of Orthopaedics, The Fifth Affiliated Hospital of Jinan University, Heyuan, China
| | - Wencai Zhang
- Spinal Trauma Area 2, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
11
|
Wang SJ, Zhang J, Zhang JZ, Ning RN, Li CC, Xu X, Jiang M, Qiu WW. Synthesis and Biological Evaluation of Heterocyclic Ring-Fused 20( S)-Protopanaxadiol Derivatives as Potent Antiosteoporosis Agents. J Med Chem 2023; 66:11965-11984. [PMID: 37597216 DOI: 10.1021/acs.jmedchem.3c00601] [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: 08/21/2023]
Abstract
A series of heterocyclic ring-fused derivatives of 20(S)-protopanaxadiol (PPD) were synthesized and evaluated for their inhibitory effects on RANKL-induced osteoclastogenesis. Among these compounds, 33 (SH491, IC50 = 11.8 nM) showed the highest potency with 100% inhibition at 0.1 μM and 44.4% inhibition at an even lower concentration of 0.01 μM, which was much more potent than the lead compound PPD (IC50 = 10.3 μM). Cytotoxicity tests indicated that the inhibitory effect of these compounds on RANKL-induced osteoclast differentiation was not due to their cytotoxicity. Interestingly, SH491 also exhibited a notable impact on the osteoblastogenesis of MC3T3-E1 preosteoblasts. Mechanistic studies revealed that SH491 inhibits the expression of osteoclastogenesis-related marker genes and proteins, including TRAP, CTSK, MMP-9, and ATPase v0d2. In vivo, SH491 could dramatically decrease the ovariectomy-induced osteoclast activity and relieve osteoporosis obviously. Thus, these PPD derivatives could be served as promising leads for the development of novel antiosteoporosis agents.
Collapse
Affiliation(s)
- Shuan-Jing Wang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Jiahui Zhang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, 200025 Shanghai, China
| | - Jing-Zan Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Ruo-Nan Ning
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, 200025 Shanghai, China
| | - Chen-Chen Li
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Xing Xu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, 200025 Shanghai, China
| | - Min Jiang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, 200025 Shanghai, China
| | - Wen-Wei Qiu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| |
Collapse
|
12
|
Deng M, Wang Z, Luo J, Cao H, Li Y, Chen L, Liu G. CircZNF367 promotes osteoclast differentiation and osteoporosis by interacting with FUS to maintain CRY2 mRNA stability. J Orthop Surg Res 2023; 18:492. [PMID: 37434265 DOI: 10.1186/s13018-023-03955-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 06/23/2023] [Indexed: 07/13/2023] Open
Abstract
BACKGROUND Osteoporosis, characterized by reduced bone mass and deterioration of bone quality, is a significant health concern for postmenopausal women. Considering that the specific role of circRNAs in osteoporosis and osteoclast differentiation remains poorly understood, this study aims to shed light on their involvement in these processes to enhance our understanding and potentially contribute to improved treatment strategies for osteoporosis. METHODS An osteoporotic model was constructed in vivo in ovariectomized mouse. In vitro, we induced osteoclast formation in bone marrow-derived macrophages (BMDMs) using M-CSF + RANKL. To assess osteoporosis in mice, we conducted HE staining. We used MTT and TRAP staining to measure cell viability and osteoclast formation, respectively, and also evaluated their mRNA and protein expression levels. In addition, RNA pull-down, RIP and luciferase reporter assays were performed to investigate interactions, and ChIP assay was used to examine the impact of circZNF367 knockdown on the binding between FUS and CRY2. RESULTS We observed increased expression of CircZNF367, FUS and CRY2 in osteoporotic mice and M-CSF + RANKL-induced BMDMs. Functionally, knocking down circZNF367 inhibited osteoporosis in vivo. Furthermore, interference with circZNF367 suppressed osteoclast proliferation and the expression of TRAP, NFATc1, and c-FOS. Mechanistically, circZNF367 interacted with FUS to maintain CRY2 mRNA stability. Additionally, knocking down CRY2 rescued M-CSF + RANKL-induced osteoclast differentiation in BMDMs promoted by circZNF367 and FUS. CONCLUSION This study reveals that the circZNF367/FUS axis may accelerate osteoclasts differentiation by upregulating CRY2 in osteoporosis and suggests that targeting circZNF367 may have potential therapeutic effects on osteoporosis.
Collapse
Affiliation(s)
- Mingsi Deng
- Department of Stomatology, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, People's Republic of China
- Department of Orthodontics, Changsha Stomatology Hospital, Changsha, 410005, Hunan, People's Republic of China
| | - Zhengguang Wang
- Department of Spine Surgery, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, People's Republic of China
| | - Jia Luo
- Changsha Blood Center, Changsha, 410001, Hunan, People's Republic of China
| | - Heng Cao
- The Department of Wound Joint Surgery, Affiliated Hospital of Yiyang Medical College, Yiyang, 413000, Hunan, People's Republic of China
| | - Yong Li
- Department of Emergency, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, People's Republic of China
| | - Liangjian Chen
- Department of Stomatology, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, People's Republic of China
| | - Gengyan Liu
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, No.138, Tongzipo Road, Yuelu District, Changsha, 410013, Hunan, People's Republic of China.
| |
Collapse
|
13
|
FTY720 Attenuates LPS-Induced Inflammatory Bone Loss by Inhibiting Osteoclastogenesis via the NF- κB and HDAC4/ATF Pathways. J Immunol Res 2023; 2023:8571649. [PMID: 36644540 PMCID: PMC9839404 DOI: 10.1155/2023/8571649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 12/14/2022] [Accepted: 12/22/2022] [Indexed: 01/07/2023] Open
Abstract
Osteoclast (OC) abnormalities lead to many osteolytic diseases, such as osteoporosis, inflammatory bone erosion, and tumor-induced osteolysis. Exploring effective strategies to remediate OCs dysregulation is essential. FTY720, also known as fingolimod, has been approved for the treatment of multiple sclerosis and has anti-inflammatory and immunosuppressive effects. Here, we found that FTY720 inhibited osteoclastogenesis and OC function by inhibiting nuclear factor kappa-B (NF-κB) signaling. Interestingly, we also found that FTY720 inhibited osteoclastogenesis by upregulating histone deacetylase 4 (HDAC4) expression levels and downregulating activating transcription factor 4 (ATF4) expression levels. In vivo, FTY720 treatment prevented lipopolysaccharide- (LPS-) induced calvarial osteolysis and significantly reduced the number of tartrate-resistant acid phosphatase- (TRAP-) positive OCs. Taken together, these results demonstrate that FTY720 can inhibit osteoclastogenesis and ameliorate inflammation-induced bone loss. Which may provide evidence of a new therapeutic target for skeletal diseases caused by OC abnormalities.
Collapse
|
14
|
Gal M, Kim O, Tran PT, Huong LT, Nhiem NX, Van Kiem P, Dang NH, Lee JH. Mussaendoside O, a N-triterpene cycloartane saponin, attenuates RANKL-induced osteoclastogenesis and inhibits lipopolysaccharide-induced bone loss. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 105:154378. [PMID: 35961265 DOI: 10.1016/j.phymed.2022.154378] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/29/2022] [Accepted: 08/02/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Elevated activity of osteoclasts (OCs) is linked to osteolytic bone diseases, such as osteoporosis and rheumatoid arthritis. Developing natural anti-osteoclastogenic compounds with greater efficacy and fewer adverse effects is crucial for preventing or treating osteolytic bone diseases. N-triterpene cycloartane saponins (NTCSs) are rarely found in nature, and their inhibitory effects on OC differentiation in vitro and in vivo have not yet been explored. PURPOSE This study was aimed to investigate the effect of mussaendoside O, an NTCS isolated from Mussaenda pubescens, on RANKL-induced OC differentiation and its underlying mechanism in vitro, and lipopolysaccharide (LPS)-induced bone resorption in a mouse model. METHODS The content of mussaendoside O in methanol extract of M. pubescens was determined by HPLC. The inhibitory effects of mussaendoside O on RANKL-induced OC formation were assessed using TRAP staining, western blotting, immunofluorescence staining, and real-time qPCR. Meanwhile, the effects of mussaendoside O on LPS-induced inflammatory responses were assessed using a Griess reagent and qPCR. The effects of mussaendoside O on LPS-induced bone resorption in a mouse model were evaluated using micro-CT and immunohistochemical staining. RESULTS Mussaendoside O inhibited RANKL-induced TRAP-positive multinucleated OC formation in a concentration-dependent manner without affecting cell viability. However, mussaendoside O did not inhibit LPS-induced mRNA expression of COX-2, iNOS, and TNF-α. Mice orally administrated with mussaendoside O exhibited significant protection from LPS-induced bone resorption and OC formation. At the molecular level, mussaendoside O suppressed RANKL-activated phosphorylation of p38 MAPK and JNK, as well as c-Fos expression. In addition, mussaendoside O suppressed RANKL-induced NFATc1 activation and the expression of its target genes, including OSCAR, DC-STAMP, CtsK, and TRAP. CONCLUSION Mussaendoside O attenuates OC differentiation in vitro and LPS-induced bone resorption in a mouse model by inhibiting the RANKL-activated c-Fos/NFATc1 signaling pathways. Therefore, mussaendoside O may be a valuable lead compound for preventing or treating of osteolytic bone diseases.
Collapse
Affiliation(s)
- Minju Gal
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, Gangwon-Do 24341, Republic of Korea
| | - Okwha Kim
- Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon, Gangwon-Do 24341, Republic of Korea
| | - Phuong Thao Tran
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, Gangwon-Do 24341, Republic of Korea
| | - Le Thanh Huong
- University of Science and Technology of Hanoi, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam
| | - Nguyen Xuan Nhiem
- Institute of Marine Biochemistry, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam
| | - Phan Van Kiem
- Institute of Marine Biochemistry, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam
| | - Nguyen Hai Dang
- University of Science and Technology of Hanoi, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam.
| | - Jeong-Hyung Lee
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, Gangwon-Do 24341, Republic of Korea; Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon, Gangwon-Do 24341, Republic of Korea.
| |
Collapse
|