1
|
Yao F, Bao Y, Meng Q, Chen Y, Zhao L, Wang P, Zhou B. Periprosthetic osteolysis: Mechanisms and potential treatment strategies. Cell Signal 2025; 131:111758. [PMID: 40132773 DOI: 10.1016/j.cellsig.2025.111758] [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] [Received: 12/11/2024] [Revised: 02/26/2025] [Accepted: 03/19/2025] [Indexed: 03/27/2025]
Abstract
Periprosthetic osteolysis is a common bone-related disorder that often occurs after total hip arthroplasty. The implants can cause damage to bone and bone-related cells due to mechanical stress and micromotions, resulting in the generation of a large number of wear particles. These wear particles trigger inflammation and oxidative stress in the surrounding tissues, disrupting the delicate balance maintained by osteoblasts and osteoclasts, ultimately leading to bone loss around the implant. Clinical investigations have demonstrated that Epimedium prenylflavonoids, miR-19a-3p, stem cell-derived exosomes, and certain non-PPO category pharmaceuticals have regulatory effects on bone homeostasis through distinct molecular pathways. Notably, this phenomenon reflects inherent biological rationality rather than stochastic occurrence. Extensive research has revealed that multiple natural compounds, non-coding RNAs, exosomes, and non-PPO therapeutics not only exert modulatory influences on critical pathophysiological processes including inflammatory cascades, oxidative stress responses, and tissue regeneration mechanisms, but also effectively regulate bone-related cellular functions to inhibit PPO progression. Therefore, this review comprehensively and systematically summarizes the main pathogenic mechanisms of periprosthetic osteolysis. Furthermore, it delves deeper into the research progress on the applications of currently reported natural products, ncRNAs, exosomes, and non-PPO medications in the treatment of periprosthetic osteolysis. Based on this, we hope that this paper can provide new perspectives and references for the future development of drugs targeting periprosthetic osteolysis.
Collapse
Affiliation(s)
- Fang Yao
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Yue Bao
- Department of Nursing, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Qian Meng
- Outpatient Department, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Yanrong Chen
- Department of Orthopaedics, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Luxi Zhao
- Department of Anesthesiology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Pingmei Wang
- Department of Orthopaedics, The People's Hospital of Shimen County, Shimen 415399, China
| | - Bin Zhou
- Department of Orthopaedics, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| |
Collapse
|
2
|
Dai G, Yang Y, Mu W, Li Y, Tu Y. Integrating thermal proteome profiling and virtual screening to reveal the mechanism of Bletilla striata against osteoclast-driven osteoporosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156735. [PMID: 40220420 DOI: 10.1016/j.phymed.2025.156735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 03/30/2025] [Accepted: 04/05/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Osteoclast (OC) overactivation and increased bone resorption are central factors contributing to osteoporosis (OP). Bletilla striata (Thunb.) Reichb. f. is known for its hemostatic properties, however, its therapeutic potential and underlying mechanisms in OC-dependent OP remain unclear. PURPOSE This study aimed to evaluate the anti-OP effects of Bletilla striata extract (BSE) and elucidate its mechanisms of action (MoAs) through an innovative integrated strategy combining isothermal thermal proteome profiling (TPP) and structure-based virtual screening (SBVS). RESULTS BSE treatment significantly decreased bone loss in ovariectomized rats, as evidenced by micro-CT and pathological analyses. This effect was attributed to inhibition of OC formation by BSE. BSE markedly blocked RANKL-induced OC differentiation, inhibited F-actin ring formation, and decreased the expression of marker genes and proteins including NFATc1, CTSK, and MMP-9. Using an isothermal TPP strategy, 110 proteins were identified as the direct targets of BSE for OC inhibition based on proteome-level thermal stability analysis (fold change > 2.0). Subsequently, using label-free quantitative proteomics, 159 proteins were identified as indirect response proteins resulting from the interaction between BSE and direct targets. The SBVS approach was employed to establish a one-to-one correspondence between the chemical ingredients in BSE and the direct targets revealed by isothermal TPP. Based on these data, an "active ingredient-direct target-indirect response signal" interaction network was constructed, which revealed the key targets (ATM, PIK3R1, BUB1B, and NR3C1) and active ingredients (phenanthrene dimers and steroids) of BSE responsible for inhibiting OC formation. CONCLUSION This study not only highlights the therapeutic potential of BSE for treating OP but also provides a novel methodological framework for interpreting the MoAs of complex Traditional Chinese Medicine (TCM) extracts. By integrating isothermal TPP and SBVS, this study offers new insights into the mechanism of TCM.
Collapse
Affiliation(s)
- Guiyu Dai
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Ying Yang
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region of China
| | - Wen Mu
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Yanfang Li
- School of Chemical Engineering, Sichuan University, Chengdu, 610065, China.
| | - Yanbei Tu
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| |
Collapse
|
3
|
MacDonald E, Johannes L, Wunder C. Acidification on the plasma membrane. Curr Opin Cell Biol 2025; 95:102531. [PMID: 40378645 DOI: 10.1016/j.ceb.2025.102531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 05/19/2025]
Abstract
The pH balance between extracellular and intracellular space is crucial for a multitude of cellular processes. Real-time observation of pH fluctuations in the range 4-9 in live cells and tissues in a sensitive, non-invasive manner has become feasible with advances in pH quantification by organic dyes, genetically encoded fluorescent proteins, and DNA-based probes. We discuss mechanisms through which pH affects cell cycle, transcription, senescence, neurotransmission, glycolipid-lectin driven endocytosis, tissue remodelling, immune responses, and GPCR signalling. Growth factor-stimulated acidification of the extracellular space notably triggers enzymatic reactions like desialylation at the plasma membrane that control processes involving cell migration and bone resorption. Research into the role of pH in cellular physiology continues to be a fertile ground for discovery that underscores its fundamental importance.
Collapse
Affiliation(s)
- Ewan MacDonald
- Montpellier Cell Biology Research Center, CRBM, Université 40 Montpellier, CNRS, Montpellier, France.
| | - Ludger Johannes
- Institut Curie, PSL Research University, U1339 INSERM, UMR3666 CNRS, Chemical Biology of Cancer Unit, Paris, France; Inria Center at University of Rennes, SAIRPICO Team, U1339 INSERM, Institut Curie, Chemical Biology of Cancer Unit, Paris, France.
| | - Christian Wunder
- Institut Curie, PSL Research University, U1339 INSERM, UMR3666 CNRS, Chemical Biology of Cancer Unit, Paris, France; Inria Center at University of Rennes, SAIRPICO Team, U1339 INSERM, Institut Curie, Chemical Biology of Cancer Unit, Paris, France.
| |
Collapse
|
4
|
Wen S, Liao X, Chang R, Wang S. Structures and biological activities of anti-osteoporotic drugs: An overview of promising small-molecule therapeutics for the treatment of osteoporosis. Bioorg Chem 2025; 162:108568. [PMID: 40381465 DOI: 10.1016/j.bioorg.2025.108568] [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: 01/08/2025] [Revised: 04/27/2025] [Accepted: 05/06/2025] [Indexed: 05/20/2025]
Abstract
Osteoporosis is the most common metabolic skeletal disorder of the skeleton, stemming from a cellular imbalance between bone formation by osteoblasts and bone resorption by osteoclasts. This imbalance causes bones to become weak and brittle, thus increasing the risk of fractures. The prevalence of osteoporosis escalates with advancing age, thereby presenting a considerable public health challenge that has elicited substantial public concern. Existing anti-osteoporotic drugs typically act by inhibiting bone resorption, promoting bone formation, and exerting a dual effect. Yet, most of these pharmaceuticals have fundamental limitations, such as targeting only one specific site and a tendency to cause side effects. With advancements in anti-osteoporotic medications, numerous new small-molecule drugs have been developed and synthesized. These novel active compounds exhibit good anti-osteoporotic activity both in vitro and in vivo by enhancing osteoblast differentiation and mineralization, as well as inhibiting osteoclast resorption. The present study seeks to review the development and current status of new compounds exhibiting anti-osteoporotic activity, to establish a solid foundation for preventing and treating osteoporosis, promoting pharmacological research, and aiding in developing new medications.
Collapse
Affiliation(s)
- Shun Wen
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China
| | - Xiaoyan Liao
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China
| | - Rui Chang
- Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun 130021, China
| | - Siyuan Wang
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China.
| |
Collapse
|
5
|
Zhang J, Hu W, Li Y, Kang F, Yao X, Li J, Dong S. MAGI1 attenuates osteoarthritis by regulating osteoclast fusion in subchondral bone through the RhoA-ROCK1 signaling pathway. J Orthop Translat 2025; 52:167-181. [PMID: 40322041 PMCID: PMC12049846 DOI: 10.1016/j.jot.2025.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/30/2025] [Accepted: 04/14/2025] [Indexed: 05/08/2025] Open
Abstract
Background Osteoarthritis (OA) is a chronic joint disorder that predominantly affects middle-aged or elderly individuals. Subchondral bone remodeling due to osteoclast hyperactivation is regarded as a major feature of early OA. During osteoclast fusion and multinucleation, the cytoskeleton reorganization leads to the formation of actin belts and ultimately bone resorption. Membrane-associated guanylate kinase with an inverted repeat member 1 (MAGI1) is a scaffolding protein that is crucial for linking the extracellular environment to intracellular signaling pathways and cytoskeleton. However, the role of MAGI1 in subchondral bone osteoclast fusion remains unclear. Methods In this study, we collected knee joint samples from OA patients and established the OA mouse model to examine the expression of MAGI1. Furthermore, we established the OA rat model and locally injected rAAV9-mediated shMagi1 into the subchondral bone to knock down MAGI1 expression. Micro-CT, histological staining, and immunofluorescence were employed to assess the effects of MAGI1 knockdown on subchondral bone homeostasis and OA process. We isolated and cultured osteoclasts from femoral and tibial bone marrow. Receptor activator of nuclear factor-κB ligand (RANKL)-stimulated osteoclasts served as an in vitro model for OA and underwent RNA sequencing. We employed gain- and loss-of-function experiments using MAGI1-overexpression plasmids and small interfering RNA to explore the role of MAGI1 in osteoclast differentiation. Further molecular experiments, including RT-qPCR, western blotting, immunofluorescence staining, and LC-MS/MS were performed to investigate underlying mechanisms. Results MAGI1 expression was significantly downregulated during RANKL-induced osteoclastogenesis in vitro. Additionally, a progressive decrease in MAGI1 expression was consistently observed in both knee joint samples from OA patients and mouse OA models, correlating with OA progression. Knockdown of MAGI1 in subchondral bone increased osteoclast numbers and worsened subchondral bone microarchitecture and cartilage degeneration; MAGI1 knockdown rats exhibited elevated PDGF-BB, Netrin-1, and CGRP+ sensory innervation. Overexpression and knockdown of MAGI1 suppressed and promoted osteoclast differentiation, respectively. Mechanistically, MAGI1 overexpression decreased the levels of RhoA, ROCK1, and p-p65 in RANKL-treated osteoclasts, which was rescued by the addition of RhoA activator narciclasine. Conclusion Our results demonstrate that MAGI1 suppresses osteoclast fusion through the RhoA/ROCK1 signaling pathway, targeting MAGI1 in subchondral bone osteoclasts may be a promising therapeutic strategy mitigate the advancement of OA. The translational potential of this article This study reveals that the scaffold protein MAGI1 participates in osteoarthritis progression by regulating osteoclast fusion, providing novel theoretical foundations and potential therapeutic targets for osteoarthritis treatment.
Collapse
Affiliation(s)
- Jing Zhang
- College of Bioengineering, Chongqing University, Chongqing, 400044, PR China
| | - Wenhui Hu
- Department of Basic Medicine, Frontier Medical Service Training Brigade, Army Medical University (Third Military Medical University), Changji, Xinjiang, 831200, PR China
- Department of Biomedical Materials Science, College of Biomedical Engineering, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
| | - Yuheng Li
- Department of Biomedical Materials Science, College of Biomedical Engineering, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
| | - Fei Kang
- Department of Biomedical Materials Science, College of Biomedical Engineering, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
| | - Xuan Yao
- Department of Clinical Hematology Faculty of Laboratory Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
| | - Jianmei Li
- Department of Biomedical Materials Science, College of Biomedical Engineering, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
| | - Shiwu Dong
- Department of Biomedical Materials Science, College of Biomedical Engineering, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
- State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing, 400038, PR China
| |
Collapse
|
6
|
Luo P, Zhong Y, Yang X, Lai Q, Huang S, Zhang X, Zhang B, Wei Y. Self-assembled water soluble and bone-targeting phosphorylated quercetin ameliorates postmenopausal osteoporosis in ovariectomy mice. Colloids Surf B Biointerfaces 2025; 249:114495. [PMID: 39798316 DOI: 10.1016/j.colsurfb.2025.114495] [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] [Received: 11/08/2024] [Revised: 12/30/2024] [Accepted: 01/02/2025] [Indexed: 01/15/2025]
Abstract
Natural compounds have shown promising application prospects in preventing or treating various diseases, including osteoporosis on account of their abundant sources, low price, multi-targeting and multiple biological effects. As a bioactive natural product, quercetin (Que) has previously demonstrated to ameliorate osteoporosis (OP), however, its poor bioavailability resulting from low water solubility, poor stability and lack of bone-targeting largely restricted its efficacy and clinical applications. Inspired by the bone-targeting capability of phosphate compounds, we reported a one-step procedure for synthesis of phosphorylated Que (p-Que) by direct phosphorylating phenol groups of Que for the first time. The phosphate groups on p-Que could not only improve the water dispersibility of Que, but also endow p-Que desirable bioavailability and bone-targeting feature. The results from biological assays suggested that p-Que could inhibit osteoclastogenesis and bone resorption and alleviate trabeculae loss in osteoporotic mice. In conclusion, this work demonstrated that phosphorylation strategy can effectively solve low water solubility, lack of bone-targeting capability and poor bioavailability of natural compounds, providing a novel and efficient approach for development of OP nanomedicines.
Collapse
Affiliation(s)
- Peng Luo
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17 Yong Wai Zheng Street, Nanchang, Jiangxi 330006, China; Department of Chemistry, Nanchang University, 999 Xuefu Avenue, Nanchang 330031, China
| | - Yanlong Zhong
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17 Yong Wai Zheng Street, Nanchang, Jiangxi 330006, China; Department of Chemistry, Nanchang University, 999 Xuefu Avenue, Nanchang 330031, China
| | - Xiaowei Yang
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17 Yong Wai Zheng Street, Nanchang, Jiangxi 330006, China; Department of Chemistry, Nanchang University, 999 Xuefu Avenue, Nanchang 330031, China
| | - Qi Lai
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17 Yong Wai Zheng Street, Nanchang, Jiangxi 330006, China; Department of Chemistry, Nanchang University, 999 Xuefu Avenue, Nanchang 330031, China
| | - Shaorong Huang
- Institute of Geriatrics, Jiangxi Provincial People's Hospital & The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi 330006, China.
| | - Xiaoyong Zhang
- Department of Chemistry, Nanchang University, 999 Xuefu Avenue, Nanchang 330031, China.
| | - Bin Zhang
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 17 Yong Wai Zheng Street, Nanchang, Jiangxi 330006, China.
| | - Yen Wei
- Department of Chemistry and the Tsinghua Center for Frontier Polymer Research, Tsinghua University, Beijing 100084, China
| |
Collapse
|
7
|
Seo J, Ko R, Kim M, Seo J, Lee H, Kim D, Jeong W, Kim HS, Lee SY. Pim1 promotes the maintenance of bone homeostasis by regulating osteoclast function. Exp Mol Med 2025; 57:733-744. [PMID: 40164682 PMCID: PMC12046003 DOI: 10.1038/s12276-025-01421-4] [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: 08/14/2024] [Revised: 12/24/2024] [Accepted: 01/05/2025] [Indexed: 04/02/2025] Open
Abstract
The Pim1 (proviral integration site for Moloney leukemia virus 1) protein is a serine/threonine kinase that is essential for cell proliferation, apoptosis and innate immune responses. Here we show that Pim1 promotes osteoclast resorptive function without affecting osteoclast numbers. Specifically, we found that mice lacking Pim1 (Pim1-/-) developed increased trabecular bone mass and indices such as trabecular bone-mass density. This was due to the direct phosphorylation of TRAF6 by Pim1 in mature osteoclasts, which activated the Akt-GSK3β signaling pathway. This, in turn, promoted the acetylation and consequent stabilization of microtubules, which permitted the formation of the osteoclast sealing zone. In vivo experiments then showed that, when mice with lipopolysaccharide-induced bone loss or tumor-induced osteolysis were treated with SGI-1776, a Pim1 inhibitor that is more selective for Pim1, the bone loss was significantly ameliorated. Thus, Pim1 plays an important role in osteoclast function and may be a therapeutic target for bone-related diseases.
Collapse
Affiliation(s)
- Jeongin Seo
- Department of Life Science, Ewha Womans University, Seoul, South Korea
- Multitasking Macrophage Research Center, Ewha Womans University, Seoul, South Korea
- Brain Korea 21 FOUR Program, LIFE Talent Development for Future Response, Ewha Womans University, Seoul, South Korea
| | - Ryeojin Ko
- Department of Life Science, Ewha Womans University, Seoul, South Korea
- Multitasking Macrophage Research Center, Ewha Womans University, Seoul, South Korea
| | - Minhee Kim
- Department of Life Science, Ewha Womans University, Seoul, South Korea
- Multitasking Macrophage Research Center, Ewha Womans University, Seoul, South Korea
- Brain Korea 21 FOUR Program, LIFE Talent Development for Future Response, Ewha Womans University, Seoul, South Korea
| | - Jeongmin Seo
- Department of Life Science, Ewha Womans University, Seoul, South Korea
- Multitasking Macrophage Research Center, Ewha Womans University, Seoul, South Korea
| | - Hana Lee
- Department of Biomedical Engineering, Yonsei University, Wonju, South Korea
| | - Doyong Kim
- Department of Biomedical Engineering, Yonsei University, Wonju, South Korea
| | - Woojin Jeong
- Department of Life Science, Ewha Womans University, Seoul, South Korea
- Multitasking Macrophage Research Center, Ewha Womans University, Seoul, South Korea
- Brain Korea 21 FOUR Program, LIFE Talent Development for Future Response, Ewha Womans University, Seoul, South Korea
| | - Han Sung Kim
- Department of Biomedical Engineering, Yonsei University, Wonju, South Korea
| | - Soo Young Lee
- Department of Life Science, Ewha Womans University, Seoul, South Korea.
- Multitasking Macrophage Research Center, Ewha Womans University, Seoul, South Korea.
- Brain Korea 21 FOUR Program, LIFE Talent Development for Future Response, Ewha Womans University, Seoul, South Korea.
| |
Collapse
|
8
|
Park S, Yoon K, Hong E, Kim MW, Kang MG, Mizuno S, Kim HJ, Lee MJ, Choi HJ, Heo JS, Bae JB, An H, Park N, Park H, Kim P, Son M, Pang K, Park JY, Takahashi S, Kwon YJ, Kang DW, Kim SJ. Tm4sf19 inhibition ameliorates inflammation and bone destruction in collagen-induced arthritis by suppressing TLR4-mediated inflammatory signaling and abnormal osteoclast activation. Bone Res 2025; 13:40. [PMID: 40128226 PMCID: PMC11933450 DOI: 10.1038/s41413-025-00419-y] [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: 08/28/2024] [Revised: 01/02/2025] [Accepted: 02/20/2025] [Indexed: 03/26/2025] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by inflammation and abnormal osteoclast activation, leading to bone destruction. We previously demonstrated that the large extracellular loop (LEL) of Tm4sf19 is important for its function in osteoclast differentiation, and LEL-Fc, a competitive inhibitor of Tm4sf19, effectively suppresses osteoclast multinucleation and prevent bone loss associated with osteoporosis. This study aimed to investigate the role of Tm4sf19 in RA, an inflammatory and abnormal osteoclast disease, using a mouse model of collagen-induced arthritis (CIA). Tm4sf19 expression was observed in macrophages and osteoclasts within the inflamed synovium, and Tm4sf19 expression was increased together with inflammatory genes in the joint bones of CIA-induced mice compared with the sham control group. Inhibition of Tm4sf19 by LEL-Fc demonstrated both preventive and therapeutic effects in a CIA mouse model, reducing the CIA score, swelling, inflammation, cartilage damage, and bone damage. Knockout of Tm4sf19 gene or inhibition of Tm4sf19 activity by LEL-Fc suppressed LPS/IFN-γ-induced TLR4-mediated inflammatory signaling in macrophages. LEL-Fc disrupted not only the interaction between Tm4sf19 and TLR4/MD2, but also the interaction between TLR4 and MD2. μCT analysis showed that LEL-Fc treatment significantly reduced joint bone destruction and bone loss caused by hyperactivated osteoclasts in CIA mice. Taken together, these findings suggest that LEL-Fc may be a potential treatment for RA and RA-induced osteoporosis by simultaneously targeting joint inflammation and bone destruction caused by abnormal osteoclast activation.
Collapse
Affiliation(s)
- Sujin Park
- GILO Institute, GILO Foundation, Seoul, Republic of Korea
| | | | - Eunji Hong
- GILO Institute, GILO Foundation, Seoul, Republic of Korea
| | | | - Min Gi Kang
- GILO Institute, GILO Foundation, Seoul, Republic of Korea
| | - Seiya Mizuno
- Laboratory Animal Resource Center in Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | | | | | | | - Jin Sun Heo
- GILO Institute, GILO Foundation, Seoul, Republic of Korea
| | | | - Haein An
- GILO Institute, GILO Foundation, Seoul, Republic of Korea
| | - Naim Park
- Medpacto Inc., Seoul, Republic of Korea
| | - Hyeyeon Park
- GILO Institute, GILO Foundation, Seoul, Republic of Korea
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Republic of Korea
| | - Pyunggang Kim
- GILO Institute, GILO Foundation, Seoul, Republic of Korea
| | - Minjung Son
- GILO Institute, GILO Foundation, Seoul, Republic of Korea
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Republic of Korea
| | - Kyoungwha Pang
- GILO Institute, GILO Foundation, Seoul, Republic of Korea
| | - Je Yeun Park
- GILO Institute, GILO Foundation, Seoul, Republic of Korea
| | - Satoru Takahashi
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yong Jung Kwon
- GILO Institute, GILO Foundation, Seoul, Republic of Korea
| | | | - Seong-Jin Kim
- GILO Institute, GILO Foundation, Seoul, Republic of Korea.
- Medpacto Inc., Seoul, Republic of Korea.
| |
Collapse
|
9
|
MacDonald E, Forrester A, Valades-Cruz CA, Madsen TD, Hetmanski JHR, Dransart E, Ng Y, Godbole R, Shp AA, Leconte L, Chambon V, Ghosh D, Pinet A, Bhatia D, Lombard B, Loew D, Larsen MR, Leffler H, Lefeber DJ, Clausen H, Blangy A, Caswell P, Shafaq-Zadah M, Mayor S, Weigert R, Wunder C, Johannes L. Growth factor-triggered de-sialylation controls glycolipid-lectin-driven endocytosis. Nat Cell Biol 2025; 27:449-463. [PMID: 39984654 DOI: 10.1038/s41556-025-01616-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 01/09/2025] [Indexed: 02/23/2025]
Abstract
Glycolipid-lectin-driven endocytosis controls the formation of clathrin-independent carriers and the internalization of various cargos such as β1 integrin. Whether this process is regulated in a dynamic manner remained unexplored. Here we demonstrate that, within minutes, the epidermal growth factor triggers the galectin-driven endocytosis of cell-surface glycoproteins, such as integrins, that are key regulators of cell adhesion and migration. The onset of this process-mediated by the Na+/H+ antiporter NHE1 as well as the neuraminidases Neu1 and Neu3-requires the pH-triggered enzymatic removal of sialic acids whose presence otherwise prevents galectin binding. De-sialylated glycoproteins are then retrogradely transported to the Golgi apparatus where their glycan make-up is reset to regulate EGF-dependent invasive-cell migration. Further evidence is provided for a role of neuraminidases and galectin-3 in acidification-dependent bone resorption. Glycosylation at the cell surface thereby emerges as a dynamic and reversible regulatory post-translational modification that controls a highly adaptable trafficking pathway.
Collapse
Affiliation(s)
- Ewan MacDonald
- Cellular and Chemical Biology Unit, Institut Curie, Université PSL, U1143 INSERM, UMR3666 CNRS, Paris, France
- Cellular Organization and Signaling Group, National Centre for Biological Sciences, Bangalore, India
- Montpellier Cell Biology Research Center, CRBM, Université de Montpellier, CNRS, Montpellier, France
| | - Alison Forrester
- Cellular and Chemical Biology Unit, Institut Curie, Université PSL, U1143 INSERM, UMR3666 CNRS, Paris, France
- WEL Research Institute, Wavre, Belgium
- Université de Namur ASBL, Namur, Belgium
| | - Cesar A Valades-Cruz
- Cellular and Chemical Biology Unit, Institut Curie, Université PSL, U1143 INSERM, UMR3666 CNRS, Paris, France
- SERPICO Project Team, Inria-UMR144 CNRS Institut Curie, PSL Research University, Paris, France
- SERPICO Project Team, Inria Centre Rennes-Bretagne Atlantique, Rennes, France
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Thomas D Madsen
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- Department for Cellular and Molecular Medicine, Copenhagen Center for Glycomics, University of Copenhagen, Copenhagen, Denmark
| | - Joseph H R Hetmanski
- Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
- Division of Biosciences, Department of Life Sciences, Centre for Genome Engineering and Maintenance, Brunel University London, London, UK
| | - Estelle Dransart
- Cellular and Chemical Biology Unit, Institut Curie, Université PSL, U1143 INSERM, UMR3666 CNRS, Paris, France
- SAIRPICO Project Team, Inria Center at University of Rennes, U1143 INSERM, Institut Curie, UMR3666 CNRS, PSL Research University, Paris, France
| | - Yeap Ng
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Rashmi Godbole
- Cellular Organization and Signaling Group, National Centre for Biological Sciences, Bangalore, India
- The University of Trans-disciplinary Health Sciences and Technology (TDU), Bangalore, India
| | - Ananthan Akhil Shp
- Cellular and Chemical Biology Unit, Institut Curie, Université PSL, U1143 INSERM, UMR3666 CNRS, Paris, France
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Ludovic Leconte
- SERPICO Project Team, Inria-UMR144 CNRS Institut Curie, PSL Research University, Paris, France
- SERPICO Project Team, Inria Centre Rennes-Bretagne Atlantique, Rennes, France
| | - Valérie Chambon
- Cellular and Chemical Biology Unit, Institut Curie, Université PSL, U1143 INSERM, UMR3666 CNRS, Paris, France
| | - Debarpan Ghosh
- Cellular and Chemical Biology Unit, Institut Curie, Université PSL, U1143 INSERM, UMR3666 CNRS, Paris, France
| | - Alexis Pinet
- Cellular and Chemical Biology Unit, Institut Curie, Université PSL, U1143 INSERM, UMR3666 CNRS, Paris, France
| | - Dhiraj Bhatia
- Cellular and Chemical Biology Unit, Institut Curie, Université PSL, U1143 INSERM, UMR3666 CNRS, Paris, France
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Gujarat, India
| | - Bérangère Lombard
- CurieCoreTech Spectrométrie de Masse Protéomique, Institut Curie, Université PSL, Paris, France
| | - Damarys Loew
- CurieCoreTech Spectrométrie de Masse Protéomique, Institut Curie, Université PSL, Paris, France
| | - Martin R Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Hakon Leffler
- Section MIG (Microbiology, Immunology, Glycobiology), Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Dirk J Lefeber
- Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Henrik Clausen
- Department for Cellular and Molecular Medicine, Copenhagen Center for Glycomics, University of Copenhagen, Copenhagen, Denmark
| | - Anne Blangy
- Montpellier Cell Biology Research Center (CRBM), Université de Montpellier, CNRS, Montpellier, France
| | - Patrick Caswell
- Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Massiullah Shafaq-Zadah
- Cellular and Chemical Biology Unit, Institut Curie, Université PSL, U1143 INSERM, UMR3666 CNRS, Paris, France
- SAIRPICO Project Team, Inria Center at University of Rennes, U1143 INSERM, Institut Curie, UMR3666 CNRS, PSL Research University, Paris, France
| | - Satyajit Mayor
- Cellular Organization and Signaling Group, National Centre for Biological Sciences, Bangalore, India
- Centre for Mechanochemical Cell Biology, Warwick Medical School, University of Warwick, Warwick, UK
| | - Roberto Weigert
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| | - Christian Wunder
- Cellular and Chemical Biology Unit, Institut Curie, Université PSL, U1143 INSERM, UMR3666 CNRS, Paris, France.
- SAIRPICO Project Team, Inria Center at University of Rennes, U1143 INSERM, Institut Curie, UMR3666 CNRS, PSL Research University, Paris, France.
| | - Ludger Johannes
- Cellular and Chemical Biology Unit, Institut Curie, Université PSL, U1143 INSERM, UMR3666 CNRS, Paris, France.
- SAIRPICO Project Team, Inria Center at University of Rennes, U1143 INSERM, Institut Curie, UMR3666 CNRS, PSL Research University, Paris, France.
| |
Collapse
|
10
|
Zhang X, Feng C, Yuan T, Wang Y, Wang H, Lu Q, Lv Y, Li Z, Fu C, Sun S. Inhibition of protein disulfide isomerase mitigates steroid-induced osteonecrosis of the femoral head by suppressing osteoclast activity through the reduction of cellular oxidative stress. Chem Biol Interact 2024; 404:111263. [PMID: 39393751 DOI: 10.1016/j.cbi.2024.111263] [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] [Received: 05/13/2024] [Revised: 07/21/2024] [Accepted: 10/08/2024] [Indexed: 10/13/2024]
Abstract
Osteonecrosis of the femoral head (ONFH) is a devastating and irreversible hip disease usually associated with increased oxidative stress due to the clinical application of high-dose or long-term glucocorticoids (GCs). Previous publications have demonstrated protein disulfide isomerase (PDI) plays a critical role in regulating cellular production of reactive oxygen species (ROS). We therefore ask whether interfering PDI could affect GCs-stimulated osteoclastogenesis. To test the hypothesis, we conducted bioinformatics and network analysis based on potential gene targets of steroid-induced osteonecrosis of the femoral head (SIONFH) in light of multiple databases and concomitantly verified the associated biological effect via the in vitro model of dexamethasone (DEX)-stimulated osteoclastogenesis. The results revealed 70 potential gene targets for SIONFH intervention, including the P4HB gene that encodes PDI. Further analysis based on network topology-based analysis techniques (NTA), protein-protein interaction (PPI) networks, and mouse cell atlas database identified the importance of PDI in regulating the cellular redox state of osteoclast during ONFH. Western blotting (WB) validations also indicated that PDI may be a positive regulator in the process of DEX-stimulated osteoclastogenesis. Hence, various PDI inhibitors were subjected to molecular docking with PDI and their performances were analyzed, including 3-Methyltoxoflavin (3 M) which inhibits PDI expression, and ribostamycin sulfate (RS) which represses PDI chaperone activity. The binding energies of DEX, 3 M, and RS to PDI were -5.3547, -4.2324, and -5.9917 kcal/mol, respectively. The Protein-Ligand Interaction Profiler (PLIP) analysis demonstrated that both hydrogen bonds and hydrophobic interactions were the key contributions to the DEX-PDI and 3M-PDI complexes, while only hydrogen bonds were identified as the predominant driving forces in the RS-PDI complex. Subsequent experiments showed that both 3 M and RS reduced osteoclast differentiation and bone resorption activity by stifling the expression of osteoclastic markers. This reduction was primarily due to the PDI inhibitors boosting the antioxidant system, thereby reducing the production of intracellular ROS. In conclusion, our results supported PDI's involvement in SIONFH progression by regulating ROS in osteoclasts and highlighted PDI inhibitors may serve as potential options for SIONFH treatment.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Orthopaedic Research Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Changgong Feng
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Orthopaedic Research Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Tao Yuan
- Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yi Wang
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Orthopaedic Research Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Haojue Wang
- Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Qizhen Lu
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Orthopaedic Research Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - YongShuang Lv
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Orthopaedic Research Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Ziqing Li
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Orthopaedic Research Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Chuanyun Fu
- Department of Stomatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; School of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Shui Sun
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Orthopaedic Research Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China; Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
11
|
Dufrancais O, Verdys P, Plozza M, Métais A, Juzans M, Sanchez T, Bergert M, Halper J, Panebianco CJ, Mascarau R, Gence R, Arnaud G, Neji MB, Maridonneau-Parini I, Cabec VL, Boerckel JD, Pavlos NJ, Diz-Muñoz A, Lagarrigue F, Blin-Wakkach C, Carréno S, Poincloux R, Burkhardt JK, Raynaud-Messina B, Vérollet C. Moesin controls cell-cell fusion and osteoclast function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.13.593799. [PMID: 38798563 PMCID: PMC11118517 DOI: 10.1101/2024.05.13.593799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Cell-cell fusion is an evolutionarily conserved process that is essential for many functions, including fertilisation and the formation of placenta, muscle and osteoclasts, multinucleated cells that are unique in their ability to resorb bone. The mechanisms of osteoclast multinucleation involve dynamic interactions between the actin cytoskeleton and the plasma membrane that are still poorly characterized. Here, we found that moesin, a cytoskeletal linker protein member of the Ezrin/Radixin/Moesin (ERM) protein family, is activated during osteoclast maturation and plays an instrumental role in both osteoclast fusion and function. In mouse and human osteoclast precursors, moesin inhibition favors their ability to fuse into multinucleated osteoclasts. Accordingly, we demonstrated that moesin depletion decreases membrane-to-cortex attachment and enhances the formation of tunneling nanotubes (TNTs), F-actin-based intercellular bridges that we reveal here to trigger cell-cell fusion. Moesin also controls HIV-1- and inflammation-induced cell fusion. In addition, moesin regulates the formation of the sealing zone, the adhesive structure determining osteoclast bone resorption area, and thus controls bone degradation, via a β3-integrin/RhoA/SLK pathway. Supporting our results, moesin - deficient mice present a reduced density of trabecular bones and increased osteoclast abundance and activity. These findings provide a better understanding of the regulation of cell-cell fusion and osteoclast biology, opening new opportunities to specifically target osteoclast activity in bone disease therapy.
Collapse
|
12
|
de Oliveira G, de Andrade Rodrigues L, Souza da Silva AA, Gouvea LC, Silva RCL, Sasso-Cerri E, Cerri PS. Reduction of osteoclast formation and survival following suppression of cytokines by diacerein in periodontitis. Biomed Pharmacother 2024; 177:117086. [PMID: 39013222 DOI: 10.1016/j.biopha.2024.117086] [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] [Received: 04/23/2024] [Revised: 06/29/2024] [Accepted: 07/01/2024] [Indexed: 07/18/2024] Open
Abstract
Periodontitis causes an increase in several bioactive agents such as interleukins (IL), tumor necrosis factor (TNF)-α and receptor activator of NF-kB ligand (RANKL), which induce the osteoclast formation and activity. Since diacerein exerts anti-TNF-α and anti-IL-1 effects, alleviating bone destruction in osteoarthritis, we investigated whether this drug inhibits the formation and survival of osteoclast in the periodontitis. Rats were distributed into 3 groups: 1) group with periodontitis treated with 100 mg/kg diacerein (PDG), 2) group with periodontitis treated with saline (PSG) and group control (CG) without any treatment. After 7, 15 and 30 days, the maxillae were collected for light and transmission electron microscopy analyses. Gingiva samples were collected to evaluate the mRNA levels for Tnf, Il1b, Tnfsf11 and Tnfrsf11b by RT-qPCR. In PDG, the expression of Tnf and Il1b genes reduced significantly compared to PSG, except for Tnf expression at 7 days. The number of osteoclasts reduced significantly in the PDG in comparison with PSG at 7 and 15 days. In all periods, the IL-6 immunoexpression, RANKL/OPG immunoexpression and mRNA levels of Tnfsf11/Tnfrsf11b ratio were significantly lower in PDG than in PSG. PDG exhibited significantly higher frequency of TUNEL-positive osteoclasts than in PSG and CG at all time points. Osteoclasts with caspase-3-immunolabelled cytoplasm and nuclei with masses of condensed chromatin were observed in PDG, confirming osteoclast apoptosis. Diacerein inhibits osteoclastogenesis by decreasing Tnf and Il1b mRNA levels, resulting in decreased RANKL/OPG ratio, and induces apoptosis in osteoclasts of alveolar process of rat molars with periodontitis.
Collapse
Affiliation(s)
- Gabriella de Oliveira
- São Paulo State University (UNESP), School of Dentistry, Laboratory of Histology and Embryology, Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry, Araraquara, SP, Brazil
| | - Lucas de Andrade Rodrigues
- São Paulo State University (UNESP), School of Dentistry, Laboratory of Histology and Embryology, Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry, Araraquara, SP, Brazil
| | | | - Lays Cristina Gouvea
- São Paulo State University (UNESP), School of Dentistry, Laboratory of Histology and Embryology, Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry, Araraquara, SP, Brazil
| | - Renata Cristina Lima Silva
- São Paulo State University (UNESP), School of Dentistry, Laboratory of Histology and Embryology, Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry, Araraquara, SP, Brazil
| | - Estela Sasso-Cerri
- São Paulo State University (UNESP), School of Dentistry, Laboratory of Histology and Embryology, Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry, Araraquara, SP, Brazil
| | - Paulo Sérgio Cerri
- São Paulo State University (UNESP), School of Dentistry, Laboratory of Histology and Embryology, Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry, Araraquara, SP, Brazil.
| |
Collapse
|
13
|
Hu S, Liang Y, Pan X. Exosomes: A promising new strategy for treating osteoporosis in the future. J Drug Deliv Sci Technol 2024; 97:105571. [DOI: 10.1016/j.jddst.2024.105571] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
14
|
Ding ZB, Chen Y, Zheng YR, Wang YY, Deng WD, Zheng JH, Yang Q, Chen ZY, Li LH, Jiang H, Li XJ. Inhibition of PPP1R15A alleviates osteoporosis via suppressing RANKL-induced osteoclastogenesis. Acta Pharmacol Sin 2024; 45:790-802. [PMID: 38191913 PMCID: PMC10943029 DOI: 10.1038/s41401-023-01209-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 11/28/2023] [Indexed: 01/10/2024]
Abstract
Osteoporosis results from overactivation of osteoclasts. There are currently few drug options for treatment of this disease. Since the successful development of allosteric inhibitors, phosphatases have become attractive therapeutic targets. Protein phosphatase 1, regulatory subunit 15 A (PPP1R15A), is a stress-responsive protein, which promotes the UPR (unfolded protein response) and restores protein homeostasis. In this study we investigated the role of PPP1R15A in osteoporosis and osteoclastogenesis. Ovariectomy (OVX)-induced osteoporosis mouse model was established, osteoporosis was evaluated in the left femurs using micro-CT. RANKL-stimulated osteoclastogenesis was used as in vitro models. We showed that PPP1R15A expression was markedly increased in BMMs derived from OVX mice and during RANKL-induced osteoclastogenesis in vitro. Knockdown of PPP1R15A or application of Sephin1 (a PPP1R15A allosteric inhibitor in a phase II clinical trial) significantly inhibited osteoclastogenesis in vitro. Sephin1 (0.78, 3.125 and 12.5 μM) dose-dependently mitigated the changes in NF-κB, MAPK, and c-FOS and the subsequent nuclear factor of activated T cells 1 (NFATc1) translocation in RANKL-stimulated BMMs. Both Sephin1 and PPP1R15A knockdown increased the phosphorylated form of eukaryotic initiation factor 2α (eIF2α); knockdown of eIF2α reduced the inhibitory effects of Sephin1 on NFATc1-luc transcription and osteoclast formation. Furthermore, Sephin1 or PPP1R15A knockdown suppressed osteoclastogenesis in CD14+ monocytes from osteoporosis patients. In OVX mice, injection of Sephin1 (4, 8 mg/kg, i.p.) every two days for 6 weeks significantly inhibited bone loss, and restored bone destruction and decreased TRAP-positive cells. This study has identified PPP1R15A as a novel target for osteoclast differentiation, and genetic inhibition or allosteric inhibitors of PPP1R15A, such as Sephin1, can be used to treat osteoporosis. This study revealed that PPP1R15A expression was increased in osteoporosis in both human and mice. Inhibition of PPP1R15A by specific knockdown or an allosteric inhibitor Sephin1 mitigated murine osteoclast formation in vitro and attenuated ovariectomy-induced osteoporosis in vivo. PPP1R15A inhibition also suppressed pathogenic osteoclastogenesis in CD14+ monocytes from osteoporosis patients. These results identify PPP1R15A as a novel regulator of osteoclastogenesis and a valuable therapeutic target for osteoporosis.
Collapse
Affiliation(s)
- Zong-Bao Ding
- Laboratory of Anti-inflammatory and Immunomodulatory Pharmacology, Innovation Program of Drug Research on Inflammatory and Immune Diseases, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China
| | - Yan Chen
- Laboratory of Anti-inflammatory and Immunomodulatory Pharmacology, Innovation Program of Drug Research on Inflammatory and Immune Diseases, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yu-Rong Zheng
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yi-Yuan Wang
- Laboratory of Anti-inflammatory and Immunomodulatory Pharmacology, Innovation Program of Drug Research on Inflammatory and Immune Diseases, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Wen-de Deng
- Laboratory of Anti-inflammatory and Immunomodulatory Pharmacology, Innovation Program of Drug Research on Inflammatory and Immune Diseases, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jie-Huang Zheng
- Laboratory of Anti-inflammatory and Immunomodulatory Pharmacology, Innovation Program of Drug Research on Inflammatory and Immune Diseases, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Qin Yang
- Laboratory of Anti-inflammatory and Immunomodulatory Pharmacology, Innovation Program of Drug Research on Inflammatory and Immune Diseases, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zi-Ye Chen
- Laboratory of Anti-inflammatory and Immunomodulatory Pharmacology, Innovation Program of Drug Research on Inflammatory and Immune Diseases, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Li-Hong Li
- Laboratory of Anti-inflammatory and Immunomodulatory Pharmacology, Innovation Program of Drug Research on Inflammatory and Immune Diseases, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Hui Jiang
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Xiao-Juan Li
- Laboratory of Anti-inflammatory and Immunomodulatory Pharmacology, Innovation Program of Drug Research on Inflammatory and Immune Diseases, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
15
|
Wang Y, Yuan T, Wang H, Meng Q, Li H, Feng C, Li Z, Sun S. Inhibition of Protein Disulfide Isomerase Attenuates Osteoclast Differentiation and Function via the Readjustment of Cellular Redox State in Postmenopausal Osteoporosis. Inflammation 2024; 47:626-648. [PMID: 38055120 DOI: 10.1007/s10753-023-01933-z] [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: 08/29/2023] [Revised: 10/16/2023] [Accepted: 11/13/2023] [Indexed: 12/07/2023]
Abstract
Due to the accumulation of reactive oxygen species (ROS) and heightened activity of osteoclasts, postmenopausal osteoporosis could cause severe pathological bone destruction. Protein disulfide isomerase (PDI), an endoplasmic prototypic thiol isomerase, plays a central role in affecting cellular redox state. To test whether suppression of PDI could inhibit osteoclastogenesis through cellular redox regulation, bioinformatics network analysis was performed on the causative genes, followed by biological validation on the osteoclastogenesis in vitro and ovariectomy (OVX) mice model in vivo. The analysis identified PDI as one of gene targets for postmenopausal osteoporosis, which was positively expressed during osteoclastogenesis. Therefore, PDI expression inhibitor and chaperone activity inhibitor were used to verify the effects of PDI inhibitors on osteoclastogenesis. Results demonstrated that PDI inhibitors could reduce osteoclast number and inhibit resorption function via suppression on osteoclast marker genes. The mechanisms behind the scenes were the PDI inhibitors-caused intracellular ROS reduction via enhancement of the antioxidant system. Micro-CT and histological results indicated PDI inhibitors could effectively alleviate or even prevent bone loss in OVX mice. In conclusion, our findings unveiled the suppressive effects of PDI inhibitors on osteoclastogenesis by reducing intracellular ROS, providing new therapeutic options for postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Yi Wang
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Tao Yuan
- Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Haojue Wang
- Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Qi Meng
- Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Haoyang Li
- Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Changgong Feng
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Ziqing Li
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China.
| | - Shui Sun
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China.
| |
Collapse
|
16
|
Lou E, Vérollet C, Winkler F, Zurzolo C, Valdebenito-Silva S, Eugenin E. Tunneling nanotubes and tumor microtubes-Emerging data on their roles in intercellular communication and pathophysiology: Summary of an International FASEB Catalyst Conference October 2023. FASEB J 2024; 38:e23514. [PMID: 38466151 DOI: 10.1096/fj.202302551] [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: 12/09/2023] [Accepted: 02/12/2024] [Indexed: 03/12/2024]
Abstract
In the past decade, there has been a steady rise in interest in studying novel cellular extensions and their potential roles in facilitating human diseases, including neurologic diseases, viral infectious diseases, cancer, and others. One of the exciting new aspects of this field is improved characterization and understanding of the functions and potential mechanisms of tunneling nanotubes (TNTs), which are actin-based filamentous protrusions that are structurally distinct from filopodia. TNTs form and connect cells at long distance and serve as direct conduits for intercellular communication in a wide range of cell types in vitro and in vivo. More researchers are entering this field and investigating the role of TNTs in mediating cancer cell invasion and drug resistance, cellular transfer of proteins, RNA or organelles, and intercellular spread of infectious agents, such as viruses, bacteria, and prions. Even further, the elucidation of highly functional membrane tubes called "tumor microtubes" (TMs) in incurable gliomas has further paved a new path for understanding how and why the tumor type is highly invasive at the cellular level and also resistant to standard therapies. Due to the wide-ranging and rapidly growing applicability of TNTs and TMs in pathophysiology across the spectrum of biology, it has become vital to bring researchers in the field together to discuss advances and the future of research in this important niche of protrusion biology.
Collapse
Affiliation(s)
- Emil Lou
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Christel Vérollet
- Institut de Pharmacologie et Biologie Structurale (IPBS), Université de Toulouse, Centre National de la Recherche Scientifique, Université Toulouse III - Paul Sabatier (UPS), Toulouse, France
- International Research Laboratory (IRP) CNRS "IM-TB/HIV", Toulouse, France
- International Research Laboratory (IRP) CNRS "IM-TB/HIV", Buenos Aires, Argentina
| | - Frank Winkler
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Chiara Zurzolo
- Institut Pasteur, Université Paris Cité, CNRS UMR 3691, Membrane Traffic and Pathogenesis, Paris, France
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | | | - Eliseo Eugenin
- Department of Neurobiology, The University of Texas Medical Branch (UTMB), Galveston, Texas, USA
| |
Collapse
|
17
|
Lee HG, Hur J, Won JP, Seo HG. Ginseng (Panax ginseng) leaf extract modulates the expression of heme oxygenase-1 to attenuate osteoclast differentiation. Fitoterapia 2024; 173:105831. [PMID: 38278423 DOI: 10.1016/j.fitote.2024.105831] [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] [Received: 09/12/2023] [Revised: 01/23/2024] [Accepted: 01/23/2024] [Indexed: 01/28/2024]
Abstract
Osteoporosis is an aging disease characterized by an imbalance between bone formation and resorption. However, drugs that inhibit bone resorption have various adverse effects. Ginseng (Panax ginseng), a prominent herbal medicine in East Asia for >2000 years, is renowned for its manifold beneficial properties, including antioxidant, anti-cancer, anti-diabetic, and anti-adipogenic activities. Despite its long history of use, the pharmacological functions of ginseng leaves are not yet fully comprehended. In this study, we evaluated the potential effects of ginseng leaf extract (GLE) on receptor activator of nuclear factor κB ligand (RANKL)-induced osteoclast differentiation in RAW264.7 macrophage cells. Tartrate-resistant acid phosphatase (TRAP) staining revealed that GLE had significant anti-osteoclastogenic activity. GLE significantly reduced mRNA levels of osteoclast differentiation markers including TRAP, nuclear factor of activated T cell cytoplasmic 1, and cathepsin K. It also suppressed the production of reactive oxygen species (ROS) and secretion of high mobility group box-1 (HMGB1) in RANKL-treated RAW264.7 cells. In addition, GLE upregulated dose- and time-dependently the expression of heme oxygenase-1 (HO-1), eventually suppressing ROS production and HMGB1 secretion. This effects of GLE were significantly reversed by Tin Protoporphyrin IX dichloride, an inhibitor of HO-1, and HO-1 shRNA, indicating that HO-1 potently inhibits RANKL-induced osteoclast differentiation by inhibiting ROS production and HMGB1 secretion. Taken together, these observations suggest that GLE could have therapeutic potential as a natural product-derived medicine for the treatment of bone disorders.
Collapse
Affiliation(s)
- Hyuk Gyoon Lee
- Department of Animal Food Resources, College of Sang-Huh Life Sciences, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jinwoo Hur
- Department of Animal Food Resources, College of Sang-Huh Life Sciences, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jun Pil Won
- Department of Animal Food Resources, College of Sang-Huh Life Sciences, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Han Geuk Seo
- Department of Animal Food Resources, College of Sang-Huh Life Sciences, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| |
Collapse
|
18
|
Luo N, Zhang L, Xiu C, Luo X, Hu S, Ji K, Liu Q, Chen J. Piperlongumine, a Piper longum-derived amide alkaloid, protects mice from ovariectomy-induced osteoporosis by inhibiting osteoclastogenesis via suppression of p38 and JNK signaling. Food Funct 2024; 15:2154-2169. [PMID: 38311970 DOI: 10.1039/d3fo03830k] [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: 02/06/2024]
Abstract
Postmenopausal osteoporosis (PMOP) is a metabolic bone disease that results from overproduction and hyperactivation of osteoclasts caused by insufficient estrogen in women after menopause. Current therapeutic strategies are mainly focused on treating PMOP patients who have already developed severe bone loss or even osteoporotic fractures. Obviously, a better strategy is to prevent PMOP from occurring in the first place. However, such reagents are largely lacking. Piperlongumine (PLM), an amide alkaloid extracted from long pepper Piper longum, exhibits the anti-osteoclastogenic effect in normal bone marrow macrophages (BMMs) and the protective effect against osteolysis induced by titanium particles in mice. This study examined the preventive effect of PLM on PMOP and explored the potential mechanism of this effect using both ovariectomized mice and their primary cells. The result showed that PLM (5 and 10 mg kg-1) administered daily for 6 weeks ameliorated ovariectomy-induced bone loss and osteoclast formation in mice. Further cell experiments showed that PLM directly suppressed osteoclast formation, F-actin ring formation, and osteoclastic resorption pit formation in BMMs derived from osteoporotic mice, but did not obviously affect osteogenic differentiation of bone marrow stromal cells (BMSCs) from these mice. Western blot analysis revealed that PLM attenuated maximal activation of p38 and JNK pathways by RANKL stimulation without affecting acute activation of NF-κB, AKT, and ERK signaling. Furthermore, PLM inhibited expression of key osteoclastogenic transcription factors NFATc1/c-Fos and their target genes (Dcstamp, Atp6v0d2, Acp5, and Oscar). Taken together, our findings suggest that PLM inhibits osteoclast formation and function by suppressing RANKL-induced activation of the p38/JNK-cFos/NFATc1 signaling cascade, thereby preventing ovariectomy-induced osteoporosis in mice. Thus, PLM can potentially be used as an anti-resorption drug or dietary supplement for the prevention of PMOP.
Collapse
Affiliation(s)
- Na Luo
- Department of Clinical Medicine, Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, 51 Huzhou Street, Gongshu District, Hangzhou, Zhejiang 310015, China.
- Orthopedic Institute, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Lei Zhang
- Orthopedic Institute, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Chunmei Xiu
- Department of Clinical Medicine, Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, 51 Huzhou Street, Gongshu District, Hangzhou, Zhejiang 310015, China.
| | - Xi Luo
- Orthopedic Institute, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Siyuan Hu
- Orthopedic Institute, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Kaizhong Ji
- Department of Clinical Medicine, Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, 51 Huzhou Street, Gongshu District, Hangzhou, Zhejiang 310015, China.
| | - Qingbai Liu
- Department of Orthopaedics, Lianshui County People's Hospital, The Affiliated Lianshui County People's Hospital of Kangda College of Nanjing Medical University, Huai'an, Jiangsu, China.
| | - Jianquan Chen
- Department of Clinical Medicine, Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, 51 Huzhou Street, Gongshu District, Hangzhou, Zhejiang 310015, China.
- Orthopedic Institute, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
19
|
Liu Y, Li TQ, Bai J, Liu WL, Wang ZR, Feng C, Pu LL, Wang XX, Liu H. Isoquercitrin attenuates the osteoclast-mediated bone loss in rheumatoid arthritis via the Nrf2/ROS/NF-κB pathway. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166977. [PMID: 38065271 DOI: 10.1016/j.bbadis.2023.166977] [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] [Received: 09/02/2023] [Revised: 11/20/2023] [Accepted: 11/28/2023] [Indexed: 12/17/2023]
Abstract
An excess of osteoclastogenesis significantly contributes to the development of rheumatoid arthritis (RA). Activation of the nuclear factor erythroid-2 related factor 2 (Nrf2) and nuclear factor kappa B (NF-κB) ligand (RANKL)-induced reactive oxygen species (ROS)-to-NF-κB signaling cascade are important mechanisms regulating osteoclastogenesis; however, whether Nrf2 is involved in RANKL-induced NF-κB activation is controversial. Isoquercitrin, a natural flavonoid compound, has been shown to have Nrf2-dependent antioxidant effects inprevious studies. We sought to verify whether isoquercitrin could modulate RANKL-induced NF-κB activation by activating Nrf2, thereby affecting osteoclastogenesis. Tartrate-resistant acid phosphatase staining, F-actin ring staining and resorption pit assay suggested that isoquercitrin significantly inhibited osteoclastogenesis and osteolytic function. Mitosox staining showed that RANKL-induced ROS generation was significantly inhibited by isoquercitrin from day 3 of the osteoclast differentiation cycle. Quantitative real-time PCR, Western blot, and immunofluorescence indicated that isoquercitrin activated the Nrf2 signaling pathway and inhibited NF-κB expression. And when we used the Nrf2-specific inhibitor ML385, the inhibition of NF-κB by isoquercitrin disappeared. Moreover, we found that Nrf2 is not uninvolved in RANKL-induced NF-κB activation and may be related to the timing of ROS regulation. When we limited isoquercitrin administration to 2 days, Nrf2 remained activated and the inhibition of NF-κB disappeared. In vivo experiments suggested that isoquercitrin attenuated RA modeling-induced bone loss. Overall, isoquercitrin-activated Nrf2 blocked the RANKL-induced ROS-to-NF-κB signaling cascade response, thereby inhibiting osteoclastogenesis and bone loss. These findings provide new ideas for the treatment of RA.
Collapse
Affiliation(s)
- Yan Liu
- Lanzhou University, Lanzhou 730000, China; Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Tian-Qi Li
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Jin Bai
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Wei-Li Liu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Zi-Rou Wang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Chong Feng
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Ling-Ling Pu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| | - Xin-Xing Wang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| | - Hui Liu
- Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
20
|
Park S, Heo JS, Mizuno S, Kim M, An H, Hong E, Kang MG, Kim J, Yun R, Park H, Noh EH, Lee MJ, Yoon K, Kim P, Son M, Pang K, Lee J, Park J, Ooshima A, Kim TJ, Park JY, Yang KM, Myung SJ, Bae H, Lee KM, Letterio J, Park SH, Takahashi S, Kim SJ. Tm4sf19 deficiency inhibits osteoclast multinucleation and prevents bone loss. Metabolism 2024; 151:155746. [PMID: 38016540 DOI: 10.1016/j.metabol.2023.155746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 11/30/2023]
Abstract
BACKGROUND Multinucleation is a hallmark of osteoclast formation and has a unique ability to resorb bone matrix. During osteoclast differentiation, the cytoskeleton reorganization results in the generation of actin belts and eventual bone resorption. Tetraspanins are involved in adhesion, migration and fusion in various cells. However, its function in osteoclast is still unclear. In this study, we identified Tm4sf19, a member of the tetraspanin family, as a regulator of osteoclast function. MATERIALS AND METHODS We investigate the effect of Tm4sf19 deficiency on osteoclast differentiation using bone marrow-derived macrophages obtained from wild type (WT), Tm4sf19 knockout (KO) and Tm4sf19 LELΔ mice lacking the large extracellular loop (LEL). We analyzed bone mass of young and aged WT, KO and LELΔ mice by μCT analysis. The effects of Tm4sf19 LEL-Fc fusion protein were accessed in osteoclast differentiation and osteoporosis animal model. RESULTS We found that deficiency of Tm4sf19 inhibited osteoclast function and LEL of Tm4sf19 was responsible for its function in osteoclasts in vitro. KO and LELΔ mice exhibited higher trabecular bone mass compared to WT mice. We found that Tm4sf19 interacts with integrin αvβ3 through LEL, and that this binding is important for cytoskeletal rearrangements in osteoclast by regulating signaling downstream of integrin αvβ3. Treatment with LEL-Fc fusion protein inhibited osteoclast function in vitro and administration of LEL-Fc prevented bone loss in an osteoporosis mouse model in vivo. CONCLUSION We suggest that Tm4sf19 regulates osteoclast function and that LEL-Fc may be a promising drug to target bone destructive diseases caused by osteoclast hyper-differentiation.
Collapse
Affiliation(s)
- Sujin Park
- GILO Institute, GILO Foundation, Seoul, Republic of Korea
| | - Jin Sun Heo
- GILO Institute, GILO Foundation, Seoul, Republic of Korea
| | - Seiya Mizuno
- Laboratory Animal Resource Center in Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Minwoo Kim
- Medpacto Inc., Seoul, Republic of Korea; Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Haein An
- GILO Institute, GILO Foundation, Seoul, Republic of Korea; Department of Biological Sciences, Sungkyunkwan University, Suwon, Republic of Korea
| | - Eunji Hong
- GILO Institute, GILO Foundation, Seoul, Republic of Korea; Department of Biological Sciences, Sungkyunkwan University, Suwon, Republic of Korea
| | - Min Gi Kang
- GILO Institute, GILO Foundation, Seoul, Republic of Korea; Department of Biological Sciences, Sungkyunkwan University, Suwon, Republic of Korea
| | - Junil Kim
- School of Systems Biomedical Science, Soongsil University, Seoul, Republic of Korea
| | - Rebecca Yun
- GILO Institute, GILO Foundation, Seoul, Republic of Korea; Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyeyeon Park
- GILO Institute, GILO Foundation, Seoul, Republic of Korea; Department of Biological Sciences, Sungkyunkwan University, Suwon, Republic of Korea
| | | | | | | | - Pyunggang Kim
- GILO Institute, GILO Foundation, Seoul, Republic of Korea
| | - Minjung Son
- GILO Institute, GILO Foundation, Seoul, Republic of Korea; Department of Biological Sciences, Sungkyunkwan University, Suwon, Republic of Korea
| | - Kyoungwha Pang
- GILO Institute, GILO Foundation, Seoul, Republic of Korea
| | - Jihee Lee
- GILO Institute, GILO Foundation, Seoul, Republic of Korea
| | - Jinah Park
- GILO Institute, GILO Foundation, Seoul, Republic of Korea; Amoris Bio Inc., Seoul, Republic of Korea
| | - Akira Ooshima
- GILO Institute, GILO Foundation, Seoul, Republic of Korea
| | - Tae-Jin Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Republic of Korea
| | - Je Yeon Park
- GILO Institute, GILO Foundation, Seoul, Republic of Korea
| | | | - Seung-Jae Myung
- Department of Gastroenterology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Hyun Bae
- Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kyung-Mi Lee
- Department of Biochemistry and Molecular Biology, College of Medicine, Korea University, Seoul, Republic of Korea
| | - John Letterio
- Case Comprehensive Cancer Center, Case Western Reserve University and Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, USA; The Angie Fowler Adolescent & Young Adult Cancer Institute, University Hospitals Rainbow Babies & Children's Hospital, Cleveland, OH, USA
| | - Seok Hee Park
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Republic of Korea
| | - Satoru Takahashi
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Seong-Jin Kim
- GILO Institute, GILO Foundation, Seoul, Republic of Korea; Medpacto Inc., Seoul, Republic of Korea.
| |
Collapse
|
21
|
Morel A, Douat C, Blangy A, Vives V. Bone resorption by osteoclasts involves fine tuning of RHOA activity by its microtubule-associated exchange factor GEF-H1. Front Physiol 2024; 15:1342024. [PMID: 38312316 PMCID: PMC10834693 DOI: 10.3389/fphys.2024.1342024] [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: 11/21/2023] [Accepted: 01/08/2024] [Indexed: 02/06/2024] Open
Abstract
Bone health is controlled by the balance between bone formation by osteoblasts and degradation by osteoclasts. A disequilibrium in favor of bone resorption leads to osteolytic diseases characterized by decreased bone density. Osteoclastic resorption is dependent on the assembly of an adhesion structure: the actin ring, also called podosome belt or sealing zone, which is composed of a unique patterning of podosomes stabilized by microtubules. A better understanding of the molecular mechanisms regulating the crosstalk between actin cytoskeleton and microtubules network is key to find new treatments to inhibit bone resorption. Evidence points to the importance of the fine tuning of the activity of the small GTPase RHOA for the formation and maintenance of the actin ring, but the underlying mechanism is not known. We report here that actin ring disorganization upon microtubule depolymerization is mediated by the activation of the RHOA-ROCK signaling pathway. We next show the involvement of GEF-H1, one of RHOA guanine exchange factor highly expressed in osteoclasts, which has the particularity of being negatively regulated by sequestration on microtubules. Using a CRISPR/Cas9-mediated GEF-H1 knock-down osteoclast model, we demonstrate that RHOA activation upon microtubule depolymerization is mediated by GEF-H1 release. Interestingly, although lower levels of GEF-H1 did not impact sealing zone formation in the presence of an intact microtubule network, sealing zone was smaller leading to impaired resorption. Altogether, these results suggest that a fine tuning of GEF-H1 through its association with microtubules, and consequently of RHOA activity, is essential for osteoclast sealing zone stability and resorption function.
Collapse
Affiliation(s)
- Anne Morel
- CRBM (Montpellier cell Biology Research Center), Univ Montpellier, CNRS (National Center for Scientific Research), Montpellier, France
| | - Christophe Douat
- CRBM (Montpellier cell Biology Research Center), Univ Montpellier, CNRS (National Center for Scientific Research), Montpellier, France
| | - Anne Blangy
- CRBM (Montpellier cell Biology Research Center), Univ Montpellier, CNRS (National Center for Scientific Research), Montpellier, France
| | - Virginie Vives
- CRBM (Montpellier cell Biology Research Center), Univ Montpellier, CNRS (National Center for Scientific Research), Montpellier, France
| |
Collapse
|
22
|
Kempf K, Capello Y, Melhem R, Lescoat C, Kempf O, Cornu A, Fremaux I, Chaignepain S, Groppi A, Nikolski M, Deffieux D, Génot E, Quideau S. Systemic Convergent Multitarget Interactions of Plant Polyphenols Revealed by Affinity-Based Protein Profiling of Bone Cells Using C-Glucosidic Vescal(ag)in-Bearing Chemoproteomic Probes. ACS Chem Biol 2023; 18:2495-2505. [PMID: 37948120 DOI: 10.1021/acschembio.3c00440] [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: 11/12/2023]
Abstract
The ellagitannins vescalagin and vescalin, known as actin-dependent inhibitors of osteoclastic bone resorption, were mounted onto chemical probes to explore their interactions with bone cell proteins by means of affinity-based chemoproteomics and bioinformatics. The chemical reactivity of the pyrogallol units of these polyphenols toward oxidation into electrophilic ortho-quinones was exploited using NaIO4 to promote the covalent capture of target proteins, notably those expressed at lower abundance and those interacting with polyphenols at low-to-moderate levels of affinity. Different assays revealed the multitarget nature of both ellagitannins, with 100-370 statistically significant proteins captured by their corresponding probes. A much higher number of proteins were captured from osteoclasts than from osteoblasts. Bioinformatic analyses unveiled a preference for the capture of proteins having phosphorylated ligands and GTPase regulators and enabled the identification of 33 potential target proteins with systemic relevance to osteoclast differentiation and activity, as well as to the regulation of actin dynamics.
Collapse
Affiliation(s)
- Karl Kempf
- Univ. Bordeaux, ISM (CNRS-UMR 5255), 351 Cours de la Libération, 33405 Talence, Cedex, France
- Department of Safety and Quality of Meat, Max Rubner-Institut (MRI), E.-C.-Baumann-Straße 20, 95326 Kulmbach, Germany
| | - Yoan Capello
- Univ. Bordeaux, ISM (CNRS-UMR 5255), 351 Cours de la Libération, 33405 Talence, Cedex, France
| | - Rana Melhem
- Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux (INSERM U1045), 2 Rue Robert Escarpit, 33607 Pessac, Cedex, France
| | - Claire Lescoat
- Univ. Bordeaux, IBGC (CNRS-UMR 5095), Centre de Bioinformatique de Bordeaux, 146 Rue Léo Saignat, 33076 Bordeaux, Cedex, France
| | - Oxana Kempf
- Univ. Bordeaux, ISM (CNRS-UMR 5255), 351 Cours de la Libération, 33405 Talence, Cedex, France
| | - Anaëlle Cornu
- Univ. Bordeaux, ISM (CNRS-UMR 5255), 351 Cours de la Libération, 33405 Talence, Cedex, France
| | - Isabelle Fremaux
- Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux (INSERM U1045), 2 Rue Robert Escarpit, 33607 Pessac, Cedex, France
| | - Stéphane Chaignepain
- Univ. Bordeaux, CBMN (CNRS-UMR 5248), Centre de Génomique Fonctionnelle de Bordeaux, 146 Rue Léo Saignat, 33076 Bordeaux, Cedex, France
| | - Alexis Groppi
- Univ. Bordeaux, IBGC (CNRS-UMR 5095), Centre de Bioinformatique de Bordeaux, 146 Rue Léo Saignat, 33076 Bordeaux, Cedex, France
| | - Macha Nikolski
- Univ. Bordeaux, IBGC (CNRS-UMR 5095), Centre de Bioinformatique de Bordeaux, 146 Rue Léo Saignat, 33076 Bordeaux, Cedex, France
| | - Denis Deffieux
- Univ. Bordeaux, ISM (CNRS-UMR 5255), 351 Cours de la Libération, 33405 Talence, Cedex, France
| | - Elisabeth Génot
- Univ. Bordeaux, Centre de Recherche Cardio-Thoracique de Bordeaux (INSERM U1045), 2 Rue Robert Escarpit, 33607 Pessac, Cedex, France
| | - Stéphane Quideau
- Univ. Bordeaux, ISM (CNRS-UMR 5255), 351 Cours de la Libération, 33405 Talence, Cedex, France
- Institut Universitaire de France, 1 Rue Descartes, 75231 Paris, Cedex 05, France
| |
Collapse
|
23
|
Yin Z, Gong G, Liu X, Yin J. Mechanism of regulating macrophages/osteoclasts in attenuating wear particle-induced aseptic osteolysis. Front Immunol 2023; 14:1274679. [PMID: 37860014 PMCID: PMC10582964 DOI: 10.3389/fimmu.2023.1274679] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/22/2023] [Indexed: 10/21/2023] Open
Abstract
Joint replacement surgery is the most effective treatment for end-stage arthritis. Aseptic loosening caused by periprosthetic osteolysis is a common complication after joint replacement. Inflammation induced by wear particles derived from prosthetic biomaterials is a major cause of osteolysis. We emphasize that bone marrow-derived macrophages and their fusion-derived osteoclasts play a key role in this pathological process. Researchers have developed multiple intervention approaches to regulate macrophage/osteoclast activation. Aiming at wear particle-induced periprosthetic aseptic osteolysis, this review separately discusses the molecular mechanism of regulation of ROS formation and inflammatory response through intervention of macrophage/osteoclast RANKL-MAPKs-NF-κB pathway. These molecular mechanisms regulate osteoclast activation in different ways, but they are not isolated from each other. There is also a lot of crosstalk among the different mechanisms. In addition, other bone and joint diseases related to osteoclast activation are also briefly introduced. Therefore, we discuss these new findings in the context of existing work with a view to developing new strategies for wear particle-associated osteolysis based on the regulation of macrophages/osteoclasts.
Collapse
Affiliation(s)
- Zhaoyang Yin
- Department of Orthopedics, The Affiliated Lianyungang Hospital of Xuzhou Medical University (The First People’s Hospital of Lianyungang), Lianyungang, China
| | - Ge Gong
- Department of Geriatrics, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xinhui Liu
- Department of Orthopedics, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, China
| | - Jian Yin
- Department of Orthopedics, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, China
| |
Collapse
|
24
|
Sun R, Hai N, Yang B, Chen J, Li J, Li Q, Zhao J, Xu J, Liu Q, Zhou B. Pteryxin suppresses osteoclastogenesis and prevents bone loss via inhibiting the MAPK/Ca 2+ signaling pathways mediated by ROS. Biomed Pharmacother 2023; 165:114898. [PMID: 37352699 DOI: 10.1016/j.biopha.2023.114898] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 05/09/2023] [Accepted: 05/16/2023] [Indexed: 06/25/2023] Open
Abstract
Osteoporosis, as a severe public health problem worldwide, causes systemic damage to bone mass, strength, and microstructure with an increased propensity for fragility fractures. Given the inherent adverse effects associated with long-term use of current prescription medications for osteoporosis treatment, identifying natural alternatives to existing treatment methods is imperative. Pteryxin (PTX), a natural coumarin, is isolated from the Peucedanum species belonging to the family Apiaceae. PTX has been reported to have antioxidant, anti-inflammatory and anti-obesity properties. However, its effect on osteoporosis has not been clarified. Our study confirmed that PTX could attenuate the formation of osteoclasts and bone resorption on a dose-dependent basis in vitro. Consistently, in vivo ovariectomy (OVX)-induced osteoporosis models simulating the physiological characteristics of postmenopausal women showed that PTX could partially reverse the bone loss caused by OVX. Further study of its mechanism revealed that PTX might block the MAPK and Ca2+-calcineurin-NFATc1 signaling pathways by decreasing the reactive oxygen species (ROS) level in osteoclasts to dampen the expression of critical transcriptional NFATc1 and downstream osteoclast-specific genes. Overall, PTX may present a new or alternative treatment option for osteoporosis.
Collapse
Affiliation(s)
- Ran Sun
- 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; Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Na Hai
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Biao Yang
- 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; Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - JunChun Chen
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Jing Li
- Neusoft Institute Guangdong, Foshan, Guangdong, China
| | - Qiufei Li
- 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; Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Jinmin Zhao
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China; Research Centre for Regenerative Medicine, Orthopedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jiake Xu
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Qian Liu
- Research Centre for Regenerative Medicine, Orthopedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.
| | - Bo Zhou
- 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; Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
25
|
Krasnova O, Neganova I. Assembling the Puzzle Pieces. Insights for in Vitro Bone Remodeling. Stem Cell Rev Rep 2023; 19:1635-1658. [PMID: 37204634 DOI: 10.1007/s12015-023-10558-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/08/2023] [Indexed: 05/20/2023]
Abstract
As a highly dynamic organ, bone changes during throughout a person's life. This process is referred to as 'bone remodeling' and it involves two stages - a well-balanced osteoclastic bone resorption and an osteoblastic bone formation. Under normal physiological conditions bone remodeling is highly regulated that ensures tight coupling between bone formation and resorption, and its disruption results in a bone metabolic disorder, most commonly osteoporosis. Though osteoporosis is one of the most prevalent skeletal ailments that affect women and men aged over 40 of all races and ethnicities, currently there are few, if any safe and effective therapeutic interventions available. Developing state-of-the-art cellular systems for bone remodeling and osteoporosis can provide important insights into the cellular and molecular mechanisms involved in skeletal homeostasis and advise better therapies for patients. This review describes osteoblastogenesis and osteoclastogenesis as two vital processes for producing mature, active bone cells in the context of interactions between cells and the bone matrix. In addition, it considers current approaches in bone tissue engineering, pointing out cell sources, core factors and matrices used in scientific practice for modeling bone diseases and testing drugs. Finally, it focuses on the challenges that bone regenerative medicine is currently facing.
Collapse
Affiliation(s)
- O Krasnova
- Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg, Russia
| | - I Neganova
- Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg, Russia.
| |
Collapse
|
26
|
Kikyo N. Circadian Regulation of Macrophages and Osteoclasts in Rheumatoid Arthritis. Int J Mol Sci 2023; 24:12307. [PMID: 37569682 PMCID: PMC10418470 DOI: 10.3390/ijms241512307] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
Rheumatoid arthritis (RA) represents one of the best examples of circadian fluctuations in disease severity. Patients with RA experience stiffness, pain, and swelling in afflicted joints in the early morning, which tends to become milder toward the afternoon. This has been primarily explained by the higher blood levels of pro-inflammatory hormones and cytokines, such as melatonin, TNFα, IL-1, and IL-6, in the early morning than in the afternoon as well as insufficient levels of anti-inflammatory cortisol, which rises later in the morning. Clinical importance of the circadian regulation of RA symptoms has been demonstrated by the effectiveness of time-of-day-dependent delivery of therapeutic agents in chronotherapy. The primary inflammatory site in RA is the synovium, where increased macrophages, T cells, and synovial fibroblasts play central roles by secreting pro-inflammatory cytokines, chemokines, and enzymes to stimulate each other, additional immune cells, and osteoclasts, ultimately leading to cartilage and bone erosion. Among these central players, macrophages have been one of the prime targets for the study of the link between circadian rhythms and inflammatory activities. Gene knockout experiments of various core circadian regulators have established that disruption of any core circadian regulators results in hyper- or hypoactivation of inflammatory responses by macrophages when challenged by lipopolysaccharide and bacteria. Although these stimulations are not directly linked to RA etiology, these findings serve as a foundation for further study by providing proof of principle. On the other hand, circadian regulation of osteoclasts, downstream effectors of macrophages, remain under-explored. Nonetheless, circadian expression of the inducers of osteoclastogenesis, such as TNFα, IL-1, and IL-6, as well as the knockout phenotypes of circadian regulators in osteoclasts suggest the significance of the circadian control of osteoclast activity in the pathogenesis of RA. More detailed mechanistic understanding of the circadian regulation of macrophages and osteoclasts in the afflicted joints could add novel local therapeutic options for RA.
Collapse
Affiliation(s)
- Nobuaki Kikyo
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA;
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
27
|
Fu J, Li S, Ma H, Yang J, Pagnotti GM, Brown LM, Weiss SJ, Mapara MY, Lentzsch S. The checkpoint inhibitor PD-1H/VISTA controls osteoclast-mediated multiple myeloma bone disease. Nat Commun 2023; 14:4271. [PMID: 37460553 PMCID: PMC10352288 DOI: 10.1038/s41467-023-39769-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 06/27/2023] [Indexed: 07/20/2023] Open
Abstract
Multiple myeloma bone disease is characterized by the development of osteolytic bone lesions. Recent work identified matrix metalloproteinase 13 as a myeloma-derived fusogen that induces osteoclast activation independent of its proteolytic activity. We now identify programmed death-1 homolog, PD-1H, as the bona fide MMP-13 receptor on osteoclasts. Silencing PD-1H or using Pd-1h-/- bone marrow cells abrogates the MMP-13-enhanced osteoclast fusion and bone-resorptive activity. Further, PD-1H interacts with the actin cytoskeleton and plays a necessary role in supporting c-Src activation and sealing zone formation. The critical role of PD-1H in myeloma lytic bone lesions was confirmed using a Pd-1h-/- myeloma bone disease mouse model wherein myeloma cells injected into Pd-1h-/-Rag2-/- results in attenuated bone destruction. Our findings identify a role of PD-1H in bone biology independent of its known immunoregulatory functions and suggest that targeting the MMP-13/PD-1H axis may represent a potential approach for the treatment of myeloma associated osteolysis.
Collapse
Grants
- S10 OD020056 NIH HHS
- P30 CA013696 NCI NIH HHS
- R01 AR075168 NIAMS NIH HHS
- R01 CA252756 NCI NIH HHS
- R01 CA175313 NCI NIH HHS
- S10 RR027050 NCRR NIH HHS
- UL1 TR002529 NCATS NIH HHS
- TL1 TR002531 NCATS NIH HHS
- NIH/NCI R01CA175313 (S. Lentzsch) NIH/NCI R01CA252756 (S. Lentzsch, J. Fu, M. Mapara) Emerson Collective Grant (S.Lentzsch) CRI/ Wade F. B. Thompson CLIP Grant (CRI4395) (S. Lentzsch) Leukemia & Lymphoma Society Translational Research Program (TRP) Grant (S. Lentzsch). Flow cytometry sorting was performed in the CCTI Flow Cytometry Core, supported in part by the Office of the Director of the National Institutes of Health under awards S10RR027050 and S10OD020056. This study used the resources of the Herbert Irving Comprehensive Cancer Center Confocal and Specialized Microscopy Shared Resource funded in part through Center Grant P30CA013696.
- NIH/NCI R01CA252756 (S. Lentzsch, J. Fu, M. Mapara) DoD Award W81XWH-21-1-0607 (J. Fu) International Myeloma Society (IMS) and Paula and Rodger Riney Foundation Translational Research Award (J. Fu)
- International Myeloma Society (IMS) and Paula and Rodger Riney Foundation Translational Research Award (S. Li)
- Indiana Clinical and Translational Sciences Institute TL1 Grant TR002531 (A. Shekhar, PI), UL1 TR002529 (A. Shekhar, PI), Cancer Prevention and Research Institute of Texas Grant RR190108 (Guise, PI), and Dive into the Pink (Guise, PI).
- The Synapt G2 mass spectrometry system was funded under NY State Contract # C023061, New York State Department of Heath NYSTEM (New York State Stem Cell Science) with LMB. The Q Exactive HF mass spectrometer was funded under a contract to LMB (NYSTEM # C029159 (New York State Stem Cell Science Board)) with matching funds from Columbia University and the Columbia Stem Cell Initiative.
Collapse
Affiliation(s)
- Jing Fu
- Columbia University Irving Medical Center, Department of Medicine, New York, NY, USA
| | - Shirong Li
- Columbia University Irving Medical Center, Department of Medicine, New York, NY, USA
| | - Huihui Ma
- Columbia University Irving Medical Center, Department of Medicine, New York, NY, USA
- Columbia Center for Translational Immunology, New York, NY, USA
| | - Jun Yang
- Columbia University Irving Medical Center, Department of Medicine, New York, NY, USA
| | - Gabriel M Pagnotti
- Indiana University, Indianapolis, IN, USA
- University of Texas-MD Anderson Cancer Center, Houston, TX, USA
| | - Lewis M Brown
- Quantitative Proteomics and Metabolomics Center, Columbia University, New York, NY, USA
| | - Stephen J Weiss
- Department of Internal Medicine, Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Markus Y Mapara
- Columbia University Irving Medical Center, Department of Medicine, New York, NY, USA
- Columbia Center for Translational Immunology, New York, NY, USA
| | - Suzanne Lentzsch
- Columbia University Irving Medical Center, Department of Medicine, New York, NY, USA.
| |
Collapse
|
28
|
Mao L, Wang L, Xu J, Zou J. The role of integrin family in bone metabolism and tumor bone metastasis. Cell Death Discov 2023; 9:119. [PMID: 37037822 PMCID: PMC10086008 DOI: 10.1038/s41420-023-01417-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/21/2023] [Accepted: 03/24/2023] [Indexed: 04/12/2023] Open
Abstract
Integrins have been the research focus of cell-extracellular matrix adhesion (ECM) and cytokine receptor signal transduction. They are involved in the regulation of bone metabolism of bone precursor cells, mesenchymal stem cells (MSCs), osteoblasts (OBs), osteoclasts (OCs), and osteocytes. Recent studies expanded and updated the role of integrin in bone metabolism, and a large number of novel cytokines were found to activate bone metabolism pathways through interaction with integrin receptors. Integrins act as transducers that mediate the regulation of bone-related cells by mechanical stress, fluid shear stress (FSS), microgravity, hypergravity, extracellular pressure, and a variety of physical factors. Integrins mediate bone metastasis of breast, prostate, and lung cancer by promoting cancer cell adhesion, migration, and survival. Integrin-mediated targeted therapy showed promising prospects in bone metabolic diseases. This review emphasizes the latest research results of integrins in bone metabolism and bone metastasis and provides a vision for treatment strategies.
Collapse
Affiliation(s)
- Liwei Mao
- School of Kinesiology, Shanghai University of Sport, 200438, Shanghai, China
| | - Lian Wang
- School of Kinesiology, Shanghai University of Sport, 200438, Shanghai, China
| | - Jiake Xu
- School of Biomedical Sciences, The University of Western Australia, WA, 6009, Perth, Australia
| | - Jun Zou
- School of Kinesiology, Shanghai University of Sport, 200438, Shanghai, China.
| |
Collapse
|
29
|
Zhu L, Tang Y, Li XY, Kerk SA, Lyssiotis CA, Sun X, Wang Z, Cho JS, Ma J, Weiss SJ. Proteolytic regulation of a galectin-3/Lrp1 axis controls osteoclast-mediated bone resorption. J Cell Biol 2023; 222:e202206121. [PMID: 36880731 PMCID: PMC9998966 DOI: 10.1083/jcb.202206121] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 12/18/2022] [Accepted: 01/23/2023] [Indexed: 03/08/2023] Open
Abstract
Bone-resorbing osteoclasts mobilize proteolytic enzymes belonging to the matrix metalloproteinase (MMP) family to directly degrade type I collagen, the dominant extracellular matrix component of skeletal tissues. While searching for additional MMP substrates critical to bone resorption, Mmp9/Mmp14 double-knockout (DKO) osteoclasts-as well as MMP-inhibited human osteoclasts-unexpectedly display major changes in transcriptional programs in tandem with compromised RhoA activation, sealing zone formation and bone resorption. Further study revealed that osteoclast function is dependent on the ability of Mmp9 and Mmp14 to cooperatively proteolyze the β-galactoside-binding lectin, galectin-3, on the cell surface. Mass spectrometry identified the galectin-3 receptor as low-density lipoprotein-related protein-1 (Lrp1), whose targeting in DKO osteoclasts fully rescues RhoA activation, sealing zone formation and bone resorption. Together, these findings identify a previously unrecognized galectin-3/Lrp1 axis whose proteolytic regulation controls both the transcriptional programs and the intracellular signaling cascades critical to mouse as well as human osteoclast function.
Collapse
Affiliation(s)
- Lingxin Zhu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Ann Arbor, MI, USA
- Life Sciences Institute, University of Michigan, Ann Arbor, Ann Arbor, MI, USA
| | - Yi Tang
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Ann Arbor, MI, USA
- Life Sciences Institute, University of Michigan, Ann Arbor, Ann Arbor, MI, USA
| | - Xiao-Yan Li
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Ann Arbor, MI, USA
- Life Sciences Institute, University of Michigan, Ann Arbor, Ann Arbor, MI, USA
| | - Samuel A. Kerk
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, Ann Arbor, MI, USA
- Doctoral Program in Cancer Biology, University of Michigan, Ann Arbor, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Ann Arbor, MI, USA
| | - Costas A. Lyssiotis
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, Ann Arbor, MI, USA
| | - Xiaoyue Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zijun Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jung-Sun Cho
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Ann Arbor, MI, USA
- Life Sciences Institute, University of Michigan, Ann Arbor, Ann Arbor, MI, USA
| | - Jun Ma
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Ann Arbor, MI, USA
- Life Sciences Institute, University of Michigan, Ann Arbor, Ann Arbor, MI, USA
| | - Stephen J. Weiss
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Ann Arbor, MI, USA
- Life Sciences Institute, University of Michigan, Ann Arbor, Ann Arbor, MI, USA
| |
Collapse
|
30
|
Zhu L, Tang Y, Li XY, Kerk SA, Lyssiotis CA, Feng W, Sun X, Hespe GE, Wang Z, Stemmler MP, Brabletz S, Brabletz T, Keller ET, Ma J, Cho JS, Yang J, Weiss SJ. A Zeb1/MtCK1 metabolic axis controls osteoclast activation and skeletal remodeling. EMBO J 2023; 42:e111148. [PMID: 36843552 PMCID: PMC10068323 DOI: 10.15252/embj.2022111148] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 01/29/2023] [Accepted: 02/02/2023] [Indexed: 02/28/2023] Open
Abstract
Osteoclasts are bone-resorbing polykaryons responsible for skeletal remodeling during health and disease. Coincident with their differentiation from myeloid precursors, osteoclasts undergo extensive transcriptional and metabolic reprogramming in order to acquire the cellular machinery necessary to demineralize bone and digest its interwoven extracellular matrix. While attempting to identify new regulatory molecules critical to bone resorption, we discovered that murine and human osteoclast differentiation is accompanied by the expression of Zeb1, a zinc-finger transcriptional repressor whose role in normal development is most frequently linked to the control of epithelial-mesenchymal programs. However, following targeting, we find that Zeb1 serves as an unexpected regulator of osteoclast energy metabolism. In vivo, Zeb1-null osteoclasts assume a hyperactivated state, markedly decreasing bone density due to excessive resorptive activity. Mechanistically, Zeb1 acts in a rheostat-like fashion to modulate murine and human osteoclast activity by transcriptionally repressing an ATP-buffering enzyme, mitochondrial creatine kinase 1 (MtCK1), thereby controlling the phosphocreatine energy shuttle and mitochondrial respiration. Together, these studies identify a novel Zeb1/MtCK1 axis that exerts metabolic control over bone resorption in vitro and in vivo.
Collapse
Affiliation(s)
- Lingxin Zhu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.,Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Yi Tang
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.,Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Xiao-Yan Li
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.,Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Samuel A Kerk
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.,Doctoral Program in Cancer Biology, University of Michigan, Ann Arbor, MI, USA.,Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Costas A Lyssiotis
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.,Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.,Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Wenqing Feng
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Xiaoyue Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Geoffrey E Hespe
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.,Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA.,Department of Surgery, Section of Plastic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Zijun Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Marc P Stemmler
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, FAU University Erlangen-Nürnberg, Erlangen, Germany
| | - Simone Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, FAU University Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, FAU University Erlangen-Nürnberg, Erlangen, Germany
| | - Evan T Keller
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA.,Department of Urology and the Institute of Gerontology, University of Michigan, Ann Arbor, MI, USA
| | - Jun Ma
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.,Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Jung-Sun Cho
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.,Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Jingwen Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Stephen J Weiss
- Division of Genetic Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.,Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
31
|
Abstract
Osteoclasts are multinucleated cells with the unique ability to resorb bone matrix. Excessive production or activation of osteoclasts leads to skeletal pathologies that affect a significant portion of the population. Although therapies that effectively target osteoclasts have been developed, they are associated with sometimes severe side effects, and a fuller understanding of osteoclast biology may lead to more specific treatments. Along those lines, a rich body of work has defined essential signaling pathways required for osteoclast formation, function, and survival. Nonetheless, recent studies have cast new light on long-held views regarding the origin of these cells during development and homeostasis, their life span, and the cellular sources of factors that drive their production and activity during homeostasis and disease. In this review, we discuss these new findings in the context of existing work and highlight areas of ongoing and future investigation.
Collapse
Affiliation(s)
- Deborah J Veis
- Division of Bone and Mineral Diseases, Musculoskeletal Research Center; and Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA; .,Shriners Hospitals for Children, St. Louis, Missouri, USA
| | - Charles A O'Brien
- Center for Musculoskeletal Disease Research, Division of Endocrinology, and Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.,Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA
| |
Collapse
|
32
|
Inguito KL, Schofield MM, Faghri AD, Bloom ET, Heino M, West VC, Ebron KMM, Elliott DM, Parreno J. Stress deprivation of tendon explants or Tpm3.1 inhibition in tendon cells reduces F-actin to promote a tendinosis-like phenotype. Mol Biol Cell 2022; 33:ar141. [PMID: 36129771 PMCID: PMC9727789 DOI: 10.1091/mbc.e22-02-0067] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 08/29/2022] [Accepted: 09/12/2022] [Indexed: 02/04/2023] Open
Abstract
Actin is a central mediator between mechanical force and cellular phenotype. In tendons, it is speculated that mechanical stress deprivation regulates gene expression by reducing filamentous (F)-actin. However, the mechanisms regulating tenocyte F-actin remain unclear. Tropomyosins (Tpms) are master regulators of F-actin. There are more than 40 Tpm isoforms, each having the unique capability to stabilize F-actin subpopulations. We investigated F-actin polymerization in stress-deprived tendons and tested the hypothesis that stress fiber-associated Tpm(s) stabilize F-actin to regulate cellular phenotype. Stress deprivation of mouse tail tendon down-regulated tenogenic and up-regulated protease (matrix metalloproteinase-3) mRNA levels. Concomitant with mRNA modulation were increases in G/F-actin, confirming reduced F-actin by tendon stress deprivation. To investigate the molecular regulation of F-actin, we identified that tail, Achilles, and plantaris tendons express three isoforms in common: Tpm1.6, 3.1, and 4.2. Tpm3.1 associates with F-actin in native and primary tenocytes. Tpm3.1 inhibition reduces F-actin, leading to decreases in tenogenic expression, increases in chondrogenic expression, and enhancement of protease expression in mouse and human tenocytes. These expression changes by Tpm3.1 inhibition are consistent with tendinosis progression. A further understanding of F-actin regulation in musculoskeletal cells could lead to new therapeutic interventions to prevent alterations in cellular phenotype during disease progression.
Collapse
Affiliation(s)
- Kameron L. Inguito
- Departments of Biological Sciences, University of Delaware, Newark, DE 19716
| | - Mandy M. Schofield
- Departments of Biological Sciences, University of Delaware, Newark, DE 19716
| | - Arya D. Faghri
- Departments of Biological Sciences, University of Delaware, Newark, DE 19716
| | - Ellen T. Bloom
- Biomedical Engineering, University of Delaware, Newark, DE 19716
| | - Marissa Heino
- Departments of Biological Sciences, University of Delaware, Newark, DE 19716
- Biomedical Engineering, University of Delaware, Newark, DE 19716
| | - Valerie C. West
- Biomedical Engineering, University of Delaware, Newark, DE 19716
| | | | - Dawn M. Elliott
- Biomedical Engineering, University of Delaware, Newark, DE 19716
| | - Justin Parreno
- Departments of Biological Sciences, University of Delaware, Newark, DE 19716
- Biomedical Engineering, University of Delaware, Newark, DE 19716
| |
Collapse
|
33
|
Cheng Y, Liu H, Li J, Ma Y, Song C, Wang Y, Li P, Chen Y, Zhang Z. Monascin abrogates RANKL-mediated osteoclastogenesis in RAW264.7 cells via regulating MAPKs signaling pathways. Front Pharmacol 2022; 13:950122. [PMID: 35910375 PMCID: PMC9337785 DOI: 10.3389/fphar.2022.950122] [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: 05/22/2022] [Accepted: 06/28/2022] [Indexed: 11/16/2022] Open
Abstract
Osteoclasts (OCs) are multinucleated cells that play a major role in osteolytic diseases such as osteoporosis. Monascin (Ms) is one of the active substances in the traditional Chinese medicine red yeast rice. Studies have found that red yeast rice can maintain bone health. In this study, the anti-osteoclastogenesis effects of Ms on RANKL-induced RAW264.7 cells were assessed, and the underlying mechanism was investigated. Ms exhibited inhibitory effects on OC differentiation and formation in a dose-dependent manner and suppressed the bone-resorbing activity of mature OCs. Ms blocked OCs-typical genes (c-Fos, NFATc1, CSTK, MMP-9, TRAP, ITG-β3, OSCAR and DC-STAMP). Furthermore, Ms treatment considerably inhibited the activation of MAPKs, JNK and p38. Taken together, Ms suppresses RANKL-induced osteoclastogenesis of RAW264.7 cells by restraining MAPKs signaling pathways and is a potential therapeutic option as a novel OC inhibitor to mitigate bone erosion.
Collapse
Affiliation(s)
- Yin Cheng
- Institute of Basic Theory, China Academy of Chinese Medical Sciences, Beijing, China
| | - Haixia Liu
- Institute of Basic Theory, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jing Li
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences; Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy (BZ0381), Beijing, China
| | - Yujie Ma
- Institute of Basic Theory, China Academy of Chinese Medical Sciences, Beijing, China
| | - Changheng Song
- Institute of Basic Theory, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuhan Wang
- Institute of Basic Theory, China Academy of Chinese Medical Sciences, Beijing, China
| | - Pei Li
- Institute of Basic Theory, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanjing Chen
- Institute of Basic Theory, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Yanjing Chen, ; Zhiguo Zhang,
| | - Zhiguo Zhang
- Institute of Basic Theory, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Yanjing Chen, ; Zhiguo Zhang,
| |
Collapse
|
34
|
Das BK, Wang L, Fujiwara T, Zhou J, Aykin-Burns N, Krager KJ, Lan R, Mackintosh SG, Edmondson R, Jennings ML, Wang X, Feng JQ, Barrientos T, Gogoi J, Kannan A, Gao L, Xing W, Mohan S, Zhao H. Transferrin receptor 1-mediated iron uptake regulates bone mass in mice via osteoclast mitochondria and cytoskeleton. eLife 2022; 11:73539. [PMID: 35758636 PMCID: PMC9352353 DOI: 10.7554/elife.73539] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 06/25/2022] [Indexed: 11/13/2022] Open
Abstract
Increased intracellular iron spurs mitochondrial biogenesis and respiration to satisfy high-energy demand during osteoclast differentiation and bone-resorbing activities. Transferrin receptor 1 (Tfr1) mediates cellular iron uptake through endocytosis of iron-loaded transferrin, and its expression increases during osteoclast differentiation. Nonetheless, the precise functions of Tfr1 and Tfr1-mediated iron uptake in osteoclast biology and skeletal homeostasis remain incompletely understood. To investigate the role of Tfr1 in osteoclast lineage cells in vivo and in vitro, we crossed Tfrc (encoding Tfr1)-floxed mice with Lyz2 (LysM)-Cre and Cathepsin K (Ctsk)-Cre mice to generate Tfrc conditional knockout mice in myeloid osteoclast precursors (Tfr1ΔLysM) or differentiated osteoclasts (Tfr1ΔCtsk), respectively. Skeletal phenotyping by µCT and histology unveiled a significant increase in trabecular bone mass with normal osteoclast number in long bones of 10-week-old young and 6-month-old adult female but not male Tfr1ΔLysM mice. Although high trabecular bone volume in long bones was observed in both male and female Tfr1ΔCtsk mice, this phenotype was more pronounced in female knockout mice. Consistent with this gender-dependent phenomena, estrogen deficiency induced by ovariectomy decreased trabecular bone mass in Tfr1ΔLysM mice. Mechanistically, disruption of Tfr1 expression attenuated mitochondrial metabolism and cytoskeletal organization in mature osteoclasts in vitro by attenuating mitochondrial respiration and activation of the Src-Rac1-WAVE regulatory complex axis, respectively, leading to decreased bone resorption with little impact on osteoclast differentiation. These results indicate that Tfr1-mediated iron uptake is specifically required for osteoclast function and is indispensable for bone remodeling in a gender-dependent manner.
Collapse
Affiliation(s)
- Bhaba K Das
- Long Beach VA Healthcare System, Southern California Institute for Research and Education, Long Beach, United States
| | - Lei Wang
- Department of Orthopedics, Anhui Medical University, Hefei, China
| | - Toshifumi Fujiwara
- Department of Orthopedic Surgery, Kyushu University Hospital, Fukuoka, Japan
| | - Jian Zhou
- Department of Orthopedics, Anhui Medical University, HeFei, China
| | - Nukhet Aykin-Burns
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, United States
| | - Kimberly J Krager
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, United States
| | - Renny Lan
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, United States
| | - Samuel G Mackintosh
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, United States
| | - Ricky Edmondson
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, United States
| | - Michael L Jennings
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, United States
| | - Xiaofang Wang
- Department of Biomedical Sciences, Texas A&M University, Dallas, United States
| | - Jian Q Feng
- Department of Biomedical Sciences, Texas A&M University, Dallas, United States
| | | | - Jyoti Gogoi
- Long Beach VA Healthcare System, Southern California Institute for Research and Education, Long Beach, United States
| | - Aarthi Kannan
- Long Beach VA Healthcare System, Southern California Institute for Research and Education, Long Beach, United States
| | - Ling Gao
- Long Beach VA Healthcare System, Southern California Institute for Research and Education, Long Beach, United States
| | - Weirong Xing
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, United States
| | - Subburaman Mohan
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, United States
| | - Haibo Zhao
- Long Beach VA Healthcare System, Southern California Institute for Research and Education, Long Beach, United States
| |
Collapse
|
35
|
Matsubara T, Yasuda K, Mizuta K, Kawaue H, Kokabu S. Tyrosine Kinase Src Is a Regulatory Factor of Bone Homeostasis. Int J Mol Sci 2022; 23:ijms23105508. [PMID: 35628319 PMCID: PMC9146043 DOI: 10.3390/ijms23105508] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 11/16/2022] Open
Abstract
Osteoclasts, which resorb the bone, and osteoblasts, which form the bone, are the key cells regulating bone homeostasis. Osteoporosis and other metabolic bone diseases occur when osteoclast-mediated bone resorption is increased and bone formation by osteoblasts is decreased. Analyses of tyrosine kinase Src-knockout mice revealed that Src is essential for bone resorption by osteoclasts and suppresses bone formation by osteoblasts. Src-knockout mice exhibit osteopetrosis. Therefore, Src is a potential target for osteoporosis therapy. However, Src is ubiquitously expressed in many tissues and is involved in various biological processes, such as cell proliferation, growth, and migration. Thus, it is challenging to develop effective osteoporosis therapies targeting Src. To solve this problem, it is necessary to understand the molecular mechanism of Src function in the bone. Src expression and catalytic activity are maintained at high levels in osteoclasts. The high activity of Src is essential for the attachment of osteoclasts to the bone matrix and to resorb the bone by regulating actin-related molecules. Src also inhibits the activity of Runx2, a master regulator of osteoblast differentiation, suppressing bone formation in osteoblasts. In this paper, we introduce the molecular mechanisms of Src in osteoclasts and osteoblasts to explore its potential for bone metabolic disease therapy.
Collapse
|
36
|
Park HJ, Park JN, Yoon SY, Yu R, Suh JH, Choi HS. Morin Disrupts Cytoskeleton Reorganization in Osteoclasts through an ROS/SHP1/c-Src Axis and Grants Protection from LPS-Induced Bone Loss. Antioxidants (Basel) 2022; 11:963. [PMID: 35624827 PMCID: PMC9137647 DOI: 10.3390/antiox11050963] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 11/16/2022] Open
Abstract
Morin is a naturally occurring flavonoid with anti-inflammatory and antioxidative properties. Therefore, we hypothesized that morin may prevent inflammatory bone loss by reducing oxidative stress. To investigate the effect of morin on inflammatory bone loss, mice were injected with lipopolysaccharide (LPS). Osteoclasts (OCs) were analyzed by tartrate-resistant acid phosphatase (TRAP) staining and actin ring formation. Micro-computerized tomography analysis indicated that morin prevented LPS-induced bone loss in mice. In vivo TRAP staining indicated that morin decreased the number and surface of the OCs that were increased in LPS-treated mice. Furthermore, in vitro experiments indicated that morin decreased the number and activity of OCs upon LPS stimulation. Morin decreased actin ring-containing OCs with decreased activation of c-Src (Y416)/vav guanine nucleotide exchange factor 3/Ras-related C3 botulinum toxin substrate 1 compared with LPS alone. Morin decreased cytosolic reactive oxygen species (ROS), thus preventing the oxidation of Src homology region 2 domain-containing phosphatase 1 (SHP-1), followed by the inactivation of c-Src via direct interaction with SHP1. Conversely, SHP1 knockdown abolished the inhibitory effect of morin on OCs. Therefore, our findings suggest that morin disrupted cytoskeletal reorganization via an ROS/SHP1/c-Src axis in OCs, thereby granting protection from LPS-induced bone loss, which demonstrates its therapeutic potential against inflammatory bone loss.
Collapse
Affiliation(s)
- Hyun-Jung Park
- Department of Biological Sciences (BK21 Program), University of Ulsan, Ulsan 44610, Korea; (H.-J.P.); (J.-N.P.); (S.-Y.Y.)
| | - Jung-Nam Park
- Department of Biological Sciences (BK21 Program), University of Ulsan, Ulsan 44610, Korea; (H.-J.P.); (J.-N.P.); (S.-Y.Y.)
| | - Sun-Young Yoon
- Department of Biological Sciences (BK21 Program), University of Ulsan, Ulsan 44610, Korea; (H.-J.P.); (J.-N.P.); (S.-Y.Y.)
| | - Rina Yu
- Department of Food and Nutrition, University of Ulsan, Ulsan 44610, Korea;
| | - Jae-Hee Suh
- Department of Pathology, Ulsan University Hospital, Ulsan 44030, Korea;
| | - Hye-Seon Choi
- Department of Biological Sciences (BK21 Program), University of Ulsan, Ulsan 44610, Korea; (H.-J.P.); (J.-N.P.); (S.-Y.Y.)
| |
Collapse
|
37
|
Chen C, Hu F, Miao S, Sun L, Jiao Y, Xu M, Huang X, Yang Y, Zhou R. Transcription Factor KLF7 Promotes Osteoclast Differentiation by Suppressing HO-1. Front Genet 2022; 13:798433. [PMID: 35419025 PMCID: PMC8995880 DOI: 10.3389/fgene.2022.798433] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/03/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Osteoporosis is a common orthopedic disease with high prevalence in patients older than 50 years. Osteoporosis is often detected only after the fracture and is hard to treat. Therefore, it is of great significance to explore the molecular mechanism of the occurrence of osteoporosis. Methods: The expression of Heme oxygenase-1 (HO-1) in people with different bone mineral density (BMD) was analyzed based on public databases. GenHacncer and JASPAR databases were adopted to search and verify the upstream transcription factor of HO-1. qRT-PCR, western blot and tartrate-resistant acid phosphatase assays were performed to explore the impact of HO-1 and Kruppel-like factor 7 (KLF7) on osteoclast differentiation. Chromatin immunoprecipitation (ChIP) assay confirmed the binding relationship between KLF7 and HO-1. Finally, Hemin, the agonist of HO-1, was applied in rescue assays, thereby verifying the mechanism of KLF7 modulating osteoclast differentiation by HO-1. Results: Bioinformatics analysis revealed that HO-1 was highly-expressed while KLF7 lowly-expressed in people with high BMD. Besides, a potential binding site of KLF7 was found on the promoter region of HO-1. ChIP assay further manifested the targeting relationship between HO-1 and KLF7. Western blot and TRAP staining unveiled that osteoclast differentiation was suppressed by HO-1, while facilitated by KLF7. Rescue experiments indicated that over-expressed HO-1 could reverse of the promoting effect of KLF7 on osteoclast differentiation. Conclusion: The study confirmed that osteoclast differentiation was promoted by KLF7 constraining HO-1, thereby facilitating osteoporosis. The cognation of the pathogenesis of osteoporosis was further enriched. New treatment could be developed on this basis.
Collapse
Affiliation(s)
- Changhong Chen
- Department of Orthopedics and Injury, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, China
| | - Fei Hu
- Department of Orthopedics and Injury, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, China
| | - Shichang Miao
- Department of Orthopedics and Injury, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, China
| | - Liping Sun
- Department of Orthopedics and Injury, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, China
| | - Yajun Jiao
- Department of Orthopedics and Injury, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, China
| | - Mingwei Xu
- Department of Orthopedics and Injury, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, China
| | - Xin Huang
- Department of Orthopedics and Injury, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, China
| | - Ying Yang
- Department of Orthopedics and Injury, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, China
| | - Rongkui Zhou
- Department of Orthopedics and Injury, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, China
| |
Collapse
|
38
|
Effects and Mechanisms of Rhus chinensis Mill. Fruits on Suppressing RANKL-Induced Osteoclastogenesis by Network Pharmacology and Validation in RAW264.7 Cells. Nutrients 2022; 14:nu14051020. [PMID: 35267996 PMCID: PMC8912277 DOI: 10.3390/nu14051020] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/24/2022] [Accepted: 02/24/2022] [Indexed: 01/11/2023] Open
Abstract
Rhus chinensis Mill. fruits are a kind of widely distributed edible seasoning, which have been documented to possess a variety of biological activities. However, its inhibitory effect on osteoclast formation has not been determined. The objective of this study was to evaluate the effect of the fruits on osteoclast differentiation of RAW264.7 cells, induced by receptor activator of nuclear factor-κB ligand (RANKL) and to illuminate the potential mechanisms using network pharmacology and western blots. Results showed that the extract containing two organic acids and twelve phenolic substances could effectively inhibit osteoclast differentiation in RANKL-induced RAW264.7 cells. Network pharmacology examination and western blot investigation showed that the concentrate essentially decreased the expression levels of osteoclast-specific proteins, chiefly through nuclear factor kappa-B, protein kinase B, and mitogen-activated protein kinase signaling pathways, particularly protein kinase B α and mitogen-activated protein kinase 1 targets. Moreover, the extract likewise directly down regulated the expression of cellular oncogene Fos and nuclear factor of activated T-cells cytoplasmic 1 proteins. Citric acid, quercetin, myricetin-3-O-galactoside, and quercetin-3-O-rhamnoside were considered as the predominant bioactive ingredients. Results of this work may provide a scientific basis for the development and utilization of R. chinensis fruits as a natural edible material to prevent and/or alleviate osteoporosis-related diseases.
Collapse
|
39
|
Garcia-Giralt N, Roca-Ayats N, Abril JF, Martinez-Gil N, Ovejero D, Castañeda S, Nogues X, Grinberg D, Balcells S, Rabionet R. Gene Network of Susceptibility to Atypical Femoral Fractures Related to Bisphosphonate Treatment. Genes (Basel) 2022; 13:genes13010146. [PMID: 35052486 PMCID: PMC8774942 DOI: 10.3390/genes13010146] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 02/01/2023] Open
Abstract
Atypical femoral fractures (AFF) are rare fragility fractures in the subtrocantheric or diaphysis femoral region associated with long-term bisphosphonate (BP) treatment. The etiology of AFF is still unclear even though a genetic basis is suggested. We performed whole exome sequencing (WES) analysis of 12 patients receiving BPs for at least 5 years who sustained AFFs and 4 controls, also long-term treated with BPs but without any fracture. After filtration and prioritization of rare variants predicted to be damaging and present in genes shared among at least two patients, a total of 272 variants in 132 genes were identified. Twelve of these genes were known to be involved in bone metabolism and/or AFF, highlighting DAAM2 and LRP5, both involved in the Wnt pathway, as the most representative. Afterwards, we intersected all mutated genes with a list of 34 genes obtained from a previous study of three sisters with BP-related AFF, identifying nine genes. One of these (MEX3D) harbored damaging variants in two AFF patients from the present study and one shared among the three sisters. Gene interaction analysis using the AFFNET web suggested a complex network among bone-related genes as well as with other mutated genes. BinGO biological function analysis highlighted cytoskeleton and cilium organization. In conclusion, several genes and their interactions could provide genetic susceptibility to AFF, that along with BPs treatment and in some cases with glucocorticoids may trigger this so feared complication.
Collapse
Affiliation(s)
- Natalia Garcia-Giralt
- Musculoskeletal Research Group, IMIM (Hospital del Mar Medical Research Institute), Centro de Investigación Biomédica en Red en Fragilidad y Envejecimiento Saludable (CIBERFES), ISCIII, 08003 Barcelona, Spain; (D.O.); (X.N.)
- Correspondence:
| | - Neus Roca-Ayats
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, CIBERER, IBUB, IRSJD, 08028 Barcelona, Spain; (N.R.-A.); (J.F.A.); (N.M.-G.); (D.G.); (S.B.); (R.R.)
| | - Josep F Abril
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, CIBERER, IBUB, IRSJD, 08028 Barcelona, Spain; (N.R.-A.); (J.F.A.); (N.M.-G.); (D.G.); (S.B.); (R.R.)
| | - Nuria Martinez-Gil
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, CIBERER, IBUB, IRSJD, 08028 Barcelona, Spain; (N.R.-A.); (J.F.A.); (N.M.-G.); (D.G.); (S.B.); (R.R.)
| | - Diana Ovejero
- Musculoskeletal Research Group, IMIM (Hospital del Mar Medical Research Institute), Centro de Investigación Biomédica en Red en Fragilidad y Envejecimiento Saludable (CIBERFES), ISCIII, 08003 Barcelona, Spain; (D.O.); (X.N.)
| | - Santos Castañeda
- Department of Rheumatology, Hospital Universitario de La Princesa, IIS-Princesa, Cátedra UAM-Roche, EPID-Future, Universidad Autónoma de Madrid, 28670 Madrid, Spain;
| | - Xavier Nogues
- Musculoskeletal Research Group, IMIM (Hospital del Mar Medical Research Institute), Centro de Investigación Biomédica en Red en Fragilidad y Envejecimiento Saludable (CIBERFES), ISCIII, 08003 Barcelona, Spain; (D.O.); (X.N.)
| | - Daniel Grinberg
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, CIBERER, IBUB, IRSJD, 08028 Barcelona, Spain; (N.R.-A.); (J.F.A.); (N.M.-G.); (D.G.); (S.B.); (R.R.)
| | - Susanna Balcells
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, CIBERER, IBUB, IRSJD, 08028 Barcelona, Spain; (N.R.-A.); (J.F.A.); (N.M.-G.); (D.G.); (S.B.); (R.R.)
| | - Raquel Rabionet
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, CIBERER, IBUB, IRSJD, 08028 Barcelona, Spain; (N.R.-A.); (J.F.A.); (N.M.-G.); (D.G.); (S.B.); (R.R.)
| |
Collapse
|
40
|
Wang Q, Xie J, Zhou C, Lai W. Substrate stiffness regulates the differentiation profile and functions of osteoclasts via cytoskeletal arrangement. Cell Prolif 2021; 55:e13172. [PMID: 34953003 PMCID: PMC8780927 DOI: 10.1111/cpr.13172] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 11/24/2021] [Accepted: 11/26/2021] [Indexed: 02/05/2023] Open
Abstract
Objectives Aging and common diseases alter the stiffness of bone tissue, causing changes to the microenvironment of the mechanosensitive bone cells. Osteoclasts, the sole bone‐resorbing cells, play a vital role in bone remodeling. This study was performed to elucidate the mechanism through which osteoclasts sense and react to substrate stiffness signals. Materials and methods We fabricated polydimethylsiloxane (PDMS) substrates of different stiffness degrees for osteoclast formation progressed from osteoclast precursors including bone marrow‐derived macrophages (BMMs) and RAW264.7 monocytes. Osteoclast differentiation in response to the stiffness signals was determined by examining the cell morphology, fusion/fission activities, transcriptional profile, and resorption function. Cytoskeletal changes and mechanosensitive adhesion molecules were also assessed. Results Stiffer PDMS substrates accelerated osteoclast differentiation, firstly observed by variations in their morphology and fusion/fission activities. Upregulation of canonical osteoclast markers (Nfatc1, Acp5, Ctsk, Camk2a, Mmp9, Rela, and Traf6) and the fusion master regulator DC‐stamp were detected on stiffer substrates, with similar increases in their bone resorption functions. Additionally, the activation of cytoskeleton‐associated adhesion molecules, including fibronectin and integrin αvβ3, followed by biochemical signaling cascades of paxillin, FAK, PKC, and RhoA, was detected on the stiffer substrates. Conclusions This is the first study to provide evidence proving that extracellular substrate stiffness is a strong determinant of osteoclast differentiation and functions. Higher stiffness upregulated the differentiation profile and activity of osteoclasts, revealing the mechanical regulation of osteoclast activity in bone homeostasis and diseases.
Collapse
Affiliation(s)
- Qingxuan Wang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chenchen Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wenli Lai
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
41
|
Das BK, Gogoi J, Kannan A, Gao L, Xing W, Mohan S, Zhao H. The Cytoplasmic Dynein Associated Protein NDE1 Regulates Osteoclastogenesis by Modulating M-CSF and RANKL Signaling Pathways. Cells 2021; 11:13. [PMID: 35011575 PMCID: PMC8750859 DOI: 10.3390/cells11010013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/15/2021] [Accepted: 12/20/2021] [Indexed: 12/29/2022] Open
Abstract
Cytoskeleton organization and lysosome secretion play an essential role in osteoclastogenesis and bone resorption. The cytoplasmic dynein is a molecular motor complex that regulates microtubule dynamics and transportation of cargos/organelles, including lysosomes along the microtubules. LIS1, NDE1, and NDEL1 belong to an evolutionary conserved pathway that regulates dynein functions. Disruption of the cytoplasmic dynein complex and deletion of LIS1 in osteoclast precursors arrest osteoclastogenesis. Nonetheless, the role of NDE1 and NDEL1 in osteoclast biology remains elusive. In this study, we found that knocking-down Nde1 expression by lentiviral transduction of specific shRNAs markedly inhibited osteoclastogenesis in vitro by attenuating the proliferation, survival, and differentiation of osteoclast precursor cells via suppression of signaling pathways downstream of M-CSF and RANKL as well as osteoclast differentiation transcription factor NFATc1. To dissect how NDEL1 regulates osteoclasts and bone homeostasis, we generated Ndel1 conditional knockout mice in myeloid osteoclast precursors (Ndel1ΔlysM) by crossing Ndel1-floxed mice with LysM-Cre mice on C57BL/6J background. The Ndel1ΔlysM mice developed normally. The µCT analysis of distal femurs and in vitro osteoclast differentiation and functional assays in cultures unveiled the similar bone mass in both trabecular and cortical bone compartments as well as intact osteoclastogenesis, cytoskeleton organization, and bone resorption in Ndel1ΔlysM mice and cultures. Therefore, our results reveal a novel role of NDE1 in regulation of osteoclastogenesis and demonstrate that NDEL1 is dispensable for osteoclast differentiation and function.
Collapse
Affiliation(s)
- Bhaba K. Das
- Southern California Institute for Research and Education, Long Beach VA Healthcare System, Long Beach, CA 90822, USA; (J.G.); (A.K.); (L.G.); (H.Z.)
| | - Jyoti Gogoi
- Southern California Institute for Research and Education, Long Beach VA Healthcare System, Long Beach, CA 90822, USA; (J.G.); (A.K.); (L.G.); (H.Z.)
| | - Aarthi Kannan
- Southern California Institute for Research and Education, Long Beach VA Healthcare System, Long Beach, CA 90822, USA; (J.G.); (A.K.); (L.G.); (H.Z.)
- Department of Dermatology, University of California Irvine, Irvine, CA 92697, USA
| | - Ling Gao
- Southern California Institute for Research and Education, Long Beach VA Healthcare System, Long Beach, CA 90822, USA; (J.G.); (A.K.); (L.G.); (H.Z.)
- Department of Dermatology, University of California Irvine, Irvine, CA 92697, USA
| | - Weirong Xing
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, CA 92357, USA; (W.X.); (S.M.)
| | - Subburaman Mohan
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, CA 92357, USA; (W.X.); (S.M.)
| | - Haibo Zhao
- Southern California Institute for Research and Education, Long Beach VA Healthcare System, Long Beach, CA 90822, USA; (J.G.); (A.K.); (L.G.); (H.Z.)
| |
Collapse
|
42
|
The Neuropeptide VIP Limits Human Osteoclastogenesis: Clinical Associations with Bone Metabolism Markers in Patients with Early Arthritis. Biomedicines 2021; 9:biomedicines9121880. [PMID: 34944693 PMCID: PMC8698638 DOI: 10.3390/biomedicines9121880] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/09/2021] [Accepted: 12/09/2021] [Indexed: 12/19/2022] Open
Abstract
We aimed to evaluate the direct action of VIP on crucial molecules involved in human osteoclast differentiation and function. We also investigated the relationship between VIP serum levels and bone remodeling mediators in early arthritis patients. The expression of VIP receptors and osteoclast gene markers in monocytes and in vitro differentiated osteoclasts was studied by real-time PCR. NFATc1 activity was measured using a TransAM® kit. Osteoclastogenesis was confirmed by quantification of tartrate-resistant acid phosphatase positive multinucleated cells. OsteoAssay® Surface Multiple Well Plate was used to evaluate bone-resorbing activity. The ring-shaped actin cytoskeleton and the VPAC1 and VPAC2 expression were analyzed by immunofluorescence. We described the presence of VIP receptors in monocytes and mature osteoclasts. Osteoclasts that formed in the presence of VIP showed a decreased expression of osteoclast differentiation gene markers and proteolytic enzymes involved in bone resorption. VIP reduced the resorption activity and decreased both β3 integrin expression and actin ring formation. Elevated serum VIP levels in early arthritis patients were associated with lower BMD loss and higher serum OPG concentration. These results demonstrate that VIP exerts an anti-osteoclastogenic action impairing both differentiation and resorption activity mainly through the negative regulation of NFATc1, evidencing its bone-protective effects in humans.
Collapse
|
43
|
Maurin J, Morel A, Guérit D, Cau J, Urbach S, Blangy A, Bompard G. The Beta-Tubulin Isotype TUBB6 Controls Microtubule and Actin Dynamics in Osteoclasts. Front Cell Dev Biol 2021; 9:778887. [PMID: 34869381 PMCID: PMC8639228 DOI: 10.3389/fcell.2021.778887] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 10/11/2021] [Indexed: 11/13/2022] Open
Abstract
Osteoclasts are bone resorbing cells that participate in the maintenance of bone health. Pathological increase in osteoclast activity causes bone loss, eventually resulting in osteoporosis. Actin cytoskeleton of osteoclasts organizes into a belt of podosomes, which sustains the bone resorption apparatus and is maintained by microtubules. Better understanding of the molecular mechanisms regulating osteoclast cytoskeleton is key to understand the mechanisms of bone resorption, in particular to propose new strategies against osteoporosis. We reported recently that β-tubulin isotype TUBB6 is key for cytoskeleton organization in osteoclasts and for bone resorption. Here, using an osteoclast model CRISPR/Cas9 KO for Tubb6, we show that TUBB6 controls both microtubule and actin dynamics in osteoclasts. Osteoclasts KO for Tubb6 have reduced microtubule growth speed with longer growth life time, higher levels of acetylation, and smaller EB1-caps. On the other hand, lack of TUBB6 increases podosome life time while the belt of podosomes is destabilized. Finally, we performed proteomic analyses of osteoclast microtubule-associated protein enriched fractions. This highlighted ARHGAP10 as a new microtubule-associated protein, which binding to microtubules appears to be negatively regulated by TUBB6. ARHGAP10 is a negative regulator of CDC42 activity, which participates in actin organization in osteoclasts. Our results suggest that TUBB6 plays a key role in the control of microtubule and actin cytoskeleton dynamics in osteoclasts. Moreover, by controlling ARHGAP10 association with microtubules, TUBB6 may participate in the local control of CDC42 activity to ensure efficient bone resorption.
Collapse
Affiliation(s)
- Justine Maurin
- Centre de Recherche de Biologie Cellulaire de Montpellier, CNRS, Montpellier University, Montpellier, France
| | - Anne Morel
- Centre de Recherche de Biologie Cellulaire de Montpellier, CNRS, Montpellier University, Montpellier, France
| | - David Guérit
- Centre de Recherche de Biologie Cellulaire de Montpellier, CNRS, Montpellier University, Montpellier, France
| | - Julien Cau
- BioCampus Montpellier, CNRS, INSERM, Montpellier University, Montpellier, France
| | - Serge Urbach
- Institute of Functional Genomics, CNRS, INSERM, Montpellier University, Montpellier, France
| | - Anne Blangy
- Centre de Recherche de Biologie Cellulaire de Montpellier, CNRS, Montpellier University, Montpellier, France
| | - Guillaume Bompard
- Centre de Recherche de Biologie Cellulaire de Montpellier, CNRS, Montpellier University, Montpellier, France
| |
Collapse
|
44
|
Loss of mutual protection between human osteoclasts and chondrocytes in damaged joints initiates osteoclast-mediated cartilage degradation by MMPs. Sci Rep 2021; 11:22708. [PMID: 34811438 PMCID: PMC8608887 DOI: 10.1038/s41598-021-02246-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 11/09/2021] [Indexed: 01/09/2023] Open
Abstract
Osteoclasts are multinucleated, bone-resorbing cells. However, they also digest cartilage during skeletal maintenance, development and in degradative conditions including osteoarthritis, rheumatoid arthritis and primary bone sarcoma. This study explores the mechanisms behind the osteoclast–cartilage interaction. Human osteoclasts differentiated on acellular human cartilage expressed osteoclast marker genes (e.g. CTSK, MMP9) and proteins (TRAP, VNR), visibly damaged the cartilage surface and released glycosaminoglycan in a contact-dependent manner. Direct co-culture with chondrocytes during differentiation increased large osteoclast formation (p < 0.0001) except when co-cultured on dentine, when osteoclast formation was inhibited (p = 0.0002). Osteoclasts cultured on dentine inhibited basal cartilage degradation (p = 0.012). RNA-seq identified MMP8 overexpression in osteoclasts differentiated on cartilage versus dentine (8.89-fold, p = 0.0133), while MMP9 was the most highly expressed MMP. Both MMP8 and MMP9 were produced by osteoclasts in osteosarcoma tissue. This study suggests that bone-resident osteoclasts and chondrocytes exert mutually protective effects on their ‘native’ tissue. However, when osteoclasts contact non-native cartilage they cause degradation via MMPs. Understanding the role of osteoclasts in cartilage maintenance and degradation might identify new therapeutic approaches for pathologies characterized by cartilage degeneration.
Collapse
|
45
|
Abstract
The purpose of this review is to explore self-organizing mechanisms that pattern microtubules (MTs) and spatially organize animal cell cytoplasm, inspired by recent experiments in frog egg extract. We start by reviewing conceptual distinctions between self-organizing and templating mechanisms for subcellular organization. We then discuss self-organizing mechanisms that generate radial MT arrays and cell centers in the absence of centrosomes. These include autocatalytic MT nucleation, transport of minus ends, and nucleation from organelles such as melanosomes and Golgi vesicles that are also dynein cargoes. We then discuss mechanisms that partition the cytoplasm in syncytia, in which multiple nuclei share a common cytoplasm, starting with cytokinesis, when all metazoan cells are transiently syncytial. The cytoplasm of frog eggs is partitioned prior to cytokinesis by two self-organizing modules, protein regulator of cytokinesis 1 (PRC1)-kinesin family member 4A (KIF4A) and chromosome passenger complex (CPC)-KIF20A. Similar modules may partition longer-lasting syncytia, such as early Drosophila embryos. We end by discussing shared mechanisms and principles for the MT-based self-organization of cellular units.
Collapse
Affiliation(s)
- Timothy J Mitchison
- Harvard Medical School, Boston, Massachusetts 02115, USA; ,
- Marine Biological Laboratory, Woods Hole, Massachusetts 02543, USA
| | - Christine M Field
- Harvard Medical School, Boston, Massachusetts 02115, USA; ,
- Marine Biological Laboratory, Woods Hole, Massachusetts 02543, USA
| |
Collapse
|
46
|
Li K, Jin R, Wu X. The role of macrophages and osteoclasts in the progression of leukemia. ACTA ACUST UNITED AC 2021; 26:724-733. [PMID: 34555294 DOI: 10.1080/16078454.2021.1976911] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
ABSTRACTBone marrow microenvironment provides critical regulatory signals for lineage differentiation and maintenance of HSC quiescence, and these signals also contribute to hematological myeloid malignancies. Macrophages exhibit high phenotypic heterogeneity under both physiological and pathological conditions and are mainly divided into proinflammatory M1 and anti-inflammatory M2 macrophages. Furthermore, osteoclasts are multinucleated giant cells that arise by fusion of monocyte/macrophage-like cells, which are commonly known as bone macrophages. Emerging evidence suggests that macrophages and osteoclasts originating from myeloid progenitors lead to two competing differentiation outcomes, and they appear to play an important role in the onset, progression, and bone metastasis of solid cancers. However, little is known about their role in the development of hematological malignancies. In this review, we focus on macrophages and osteoclasts, their role in leukemia, and the potential for targeting these cells in this disease.
Collapse
Affiliation(s)
- Kun Li
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Runming Jin
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xiaoyan Wu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
47
|
Poorhemati H, Komarova SV. Mathematical modeling of the role of bone turnover in pH regulation in bone interstitial fluid. Comput Biol Chem 2021; 94:107564. [PMID: 34455167 DOI: 10.1016/j.compbiolchem.2021.107564] [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: 11/06/2020] [Revised: 07/20/2021] [Accepted: 08/15/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND AND AIMS Bone turnover is strongly affected by pH of surrounding fluid, and in turn plays a role in maintaining systemic pH, however the quantitative contribution of bone processes to pH regulation is not known. Our goal was to develop a mathematical model describing pH regulation in the interstitial fluid and to examine the contribution of hydroxyapatite dissolution and precipitation to pH regulation. MATERIALS AND METHODS We modeled twelve reversible equilibrium reactions of sixteen calcium, phosphate, hydrogen and carbonate species in the interstitial fluid and examined the buffering capacity and range. The effect of hydroxyapatite dissolution and precipitation was modeled by assuming that the calcium, phosphate and hydroxide contained in the bone volume adjacent to the interstitial fluid is instantaneously added to or removed from the interstitial fluid. RESULTS The carbonate buffer was found to dominate electrochemical buffering system of the bone interstitial fluid. Nevertheless, the phosphate added during dissolution of bone hydroxyapatite significantly improved the interstitial fluid buffering capacity. In contrast, hydroxyapatite precipitation had limited effect on the interstitial fluid pH regulation. CONCLUSION This study provides mechanistic insights into the physicochemical processes underlying the known role of bone turnover processes in regulation of body pH homeostasis.
Collapse
Affiliation(s)
- Hossein Poorhemati
- Department of Biological and Biomedical Engineering, McGill University, Montreal, QC, Canada; Shriners Hospital for Children - Canada, Montreal, QC, Canada.
| | - Svetlana V Komarova
- Department of Biological and Biomedical Engineering, McGill University, Montreal, QC, Canada; Shriners Hospital for Children - Canada, Montreal, QC, Canada; Faculty of Dentistry, McGill University, Montreal, QC, Canada.
| |
Collapse
|
48
|
Mounier L, Morel A, Ferrandez Y, Morko J, Vääräniemi J, Gilardone M, Roche D, Cherfils J, Blangy A. Novel 2,7-Diazaspiro[4,4]nonane Derivatives to Inhibit Mouse and Human Osteoclast Activities and Prevent Bone Loss in Ovariectomized Mice without Affecting Bone Formation. J Med Chem 2020; 63:13680-13694. [PMID: 33175535 DOI: 10.1021/acs.jmedchem.0c01201] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Osteoporosis is currently treated with drugs targeting the differentiation or viability osteoclasts, the cells responsible for physiological and pathological bone resorption. Nevertheless, osteoporosis drugs that target only osteoclast activity are expected to preserve bone formation by osteoblasts in contrast to current treatments. We report here the design, synthesis, and biological characterization of a series of novel N-arylsufonamides featuring a diazaspiro[4,4]nonane nucleus to target the guanine nucleotide exchange activity of DOCK5, which is essential for bone resorption by osteoclasts. These compounds can inhibit both mouse and human osteoclast activity. In particular, 4-chlorobenzyl-4-hydroxy-2-phenyl-1-thia-2,7-diazaspiro[4,4]nonane 1,1-dioxide (compound E197) prevented pathological bone loss in mice. Most interestingly, treatment with E197 did not affect osteoclast and osteoblast numbers and hence did not impair bone formation. E197 could represent a lead molecule to develop new antiosteoporotic drugs targeting the mechanism of osteoclast adhesion onto the bone.
Collapse
Affiliation(s)
- Lucile Mounier
- Centre de Recherche en Biologie Cellulaire de Montpellier, Univ Montpellier, CNRS, Montpellier, France, Université de Montpellier, CNRS, 34000 Montpellier, France
| | - Anne Morel
- Centre de Recherche en Biologie Cellulaire de Montpellier, Univ Montpellier, CNRS, Montpellier, France, Université de Montpellier, CNRS, 34000 Montpellier, France
| | - Yann Ferrandez
- Laboratoire de Biologie et Pharmacologie Appliquée, CNRS, Ecole Normale Supérieure Paris-Saclay and Université Paris-Saclay, 91190 Gif-sur-Yvette, France
| | - Jukka Morko
- Pharmatest Services Ltd., Itäinen Pitkäkatu 4, 20520 Turku, Finland
| | - Jukka Vääräniemi
- Pharmatest Services Ltd., Itäinen Pitkäkatu 4, 20520 Turku, Finland
| | | | - Didier Roche
- Edelris, 60 Avenue Rockefeller, 69008 Lyon, France
| | - Jacqueline Cherfils
- Laboratoire de Biologie et Pharmacologie Appliquée, CNRS, Ecole Normale Supérieure Paris-Saclay and Université Paris-Saclay, 91190 Gif-sur-Yvette, France
| | - Anne Blangy
- Centre de Recherche en Biologie Cellulaire de Montpellier, Univ Montpellier, CNRS, Montpellier, France, Université de Montpellier, CNRS, 34000 Montpellier, France
| |
Collapse
|
49
|
Osteoclasts and Microgravity. Life (Basel) 2020; 10:life10090207. [PMID: 32947946 PMCID: PMC7555718 DOI: 10.3390/life10090207] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/10/2020] [Accepted: 09/13/2020] [Indexed: 12/13/2022] Open
Abstract
Astronauts are at risk of losing 1.0% to 1.5% of their bone mass for every month they spend in space despite their adherence to diets and exercise regimens designed to protect their musculoskeletal systems. This loss is the result of microgravity-related impairment of osteocyte and osteoblast function and the consequent upregulation of osteoclast-mediated bone resorption. This review describes the ontogeny of osteoclast hematopoietic stem cells and the contributions macrophage colony stimulating factor, receptor activator of the nuclear factor-kappa B ligand, and the calcineurin pathways make in osteoclast differentiation and provides details of bone formation, the osteoclast cytoskeleton, the immune regulation of osteoclasts, and osteoclast mechanotransduction on Earth, in space, and under conditions of simulated microgravity. The article discusses the need to better understand how osteoclasts are able to function in zero gravity and reviews current and prospective therapies that may be used to treat osteoclast-mediated bone disease.
Collapse
|