1
|
Wang H, Sun X, Cui K, Wang Y, Sun Y. Ubiquitin specific peptidase 13 protects against inflammation-associated joint injury in collagen-induced rheumatoid arthritis mice by targeting TRAF6. Int Immunopharmacol 2025; 156:114617. [PMID: 40252462 DOI: 10.1016/j.intimp.2025.114617] [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: 08/08/2024] [Revised: 04/01/2025] [Accepted: 04/02/2025] [Indexed: 04/21/2025]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease with complex pathogenesis. Ubiquitin-specific protease 13 (USP13), a member of the deubiquitinating enzyme (DUB) superfamily, plays diverse roles in cellular events. This study investigates the role of USP13 in RA, revealing its significant downregulation in peripheral blood mononuclear cells (PBMCs) from RA patients compared to healthy individuals. USP13 expression negatively correlates with RA characteristics, including erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), and rheumatoid factor (RF). Consistent with these findings, USP13 levels were reduced in knee joint synovial tissues of collagen-induced arthritis (CIA) mice and lipopolysaccharide (LPS)-stimulated bone marrow-derived macrophages (BMDMs). Mechanistically, USP13 directly interacts with tumor necrosis factor receptor-associated factor 6 (TRAF6), reducing its K63-linked polyubiquitination and thereby inhibiting TRAF6 expression. This interaction restrains nuclear factor κB (NF-κB) signaling, leading to a marked attenuation of LPS-induced inflammatory responses in BMDMs. Importantly, the anti-inflammatory effects of USP13 over-expression are largely dependent on TRAF6 suppression. In vitro, USP13 over-expression in BMDMs inhibits the proliferation of fibroblast-like synoviocytes (FLS) and osteoclastogenesis. In addition, USP13-overexpressing BMDMs in CIA mice significantly alleviates RA development, as evidenced by reduced synovial hyperplasia, inflammatory cell infiltration, cartilage destruction, and bone loss. These findings highlight the essential role of USP13 in macrophages during RA progression and reveal its therapeutic potential by targeting TRAF6 signaling.
Collapse
Affiliation(s)
- Haili Wang
- Department of General Medince, Affiliated Hospital of Beihua University, Jilin 132011, China
| | - Xinyi Sun
- Department of General Medince, Affiliated Hospital of Beihua University, Jilin 132011, China
| | - Kai Cui
- Department of Oncology, Affiliated Hospital of Beihua University, Jilin 132011, China
| | - Yan Wang
- Geriatric Department, Jilin Integrated Traditional Chinese and Western Medicine Hospital of Jilin, Jilin 132011, China
| | - Yuman Sun
- Department of General Medince, Affiliated Hospital of Beihua University, Jilin 132011, China.
| |
Collapse
|
2
|
Wang K, Gao Q, Bai Y, Yu R, Luo Q. The Expression Levels of Transforming Growth Factor β1 and Tumor Necrosis Factor Receptor Associated Factor 6 in Allergic Rhinitis Patients and Their Potential Relationship with Epithelial - Mesenchymal Transition: A Pilot Prospective Observational Study. J Asthma Allergy 2024; 17:1083-1092. [PMID: 39502932 PMCID: PMC11537165 DOI: 10.2147/jaa.s474445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 10/14/2024] [Indexed: 11/08/2024] Open
Abstract
Objective To study the role of transforming growth factor beta 1 (TGF-β1) and tumor necrosis factor receptor related factor 6 (TRAF6) in the progression of epithelial mesenchymal transformation (EMT) in allergic rhinitis (AR). Methods A total of 30 patients underwent nasal endoscopic surgery at our Hospital were selected for 15 patients in each group based on their allergy status. Inferior turbinate mucosa tissue was obtained and analyzed using immunohistochemical (IHC) tests, real-time quantitative PCR (qRT-PCR) detection, and Western blotting (WB) tests to measure TGF-β1, TRAF6, E-cadherin, Vimentin, and α-Smooth Muscle Actin (α-SMA) expression levels. Results The expression levels of TGF-β1, TRAF6, Vimentin, and α-SMA were significantly higher in the AR group compared to the control group as shown by IHC, qRT-PCR, and WB (P < 0.05). E-cadherin expression was significantly lower group than in the control group (P < 0.05). Protein expression of TGF-β1 showed significantly positive correlations with TRAF6 (r = 0.8188, P = 0.0002), α-SMA (r = 0.8076, P = 0.0003), and Vimentin (r = 0.6917, P = 0.0043). There was a significantly negative correlation between protein expression of TGF-β1 and E-cadherin (r = -0.8032, P = 0.0003). Protein expression of TRAF6 showed a significantly negative correlation with E-cadherin (r = -0.6405, P = 0.0101) but positive correlations with α-SMA (r = 0.5809, P = 0.0231) and Vimentin (r = 0.555, P = 0.0318). Conclusion TGF-β1, TRAF6, and EMT-related markers (Vimentin, α-SMA) were highly expressed in the nasal mucosa of AR patients. TGF-β1 and TRAF6 may be involved in the epithelial-mesenchymal transition in allergic rhinitis.
Collapse
Affiliation(s)
- Kai Wang
- Department of Otorhinolaryngology, FirstAffiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
- Department of Otorhinolaryngology, the 908th Hospital of Chinese People’s Liberation Army Joint Logistic Support Force, Nanchang, Jiangxi, People’s Republic of China
| | - Qin Gao
- Department of Otorhinolaryngology, Central Theater Command General Hospital of the People’s Liberation Army of China, Wuhan, Hubei, People’s Republic of China
| | - Yelong Bai
- Department of Otorhinolaryngology, FirstAffiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Rong Yu
- Department of Otorhinolaryngology, FirstAffiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Qing Luo
- Department of Otorhinolaryngology, FirstAffiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| |
Collapse
|
3
|
Iwanowska M, Kochman M, Szatko A, Zgliczyński W, Glinicki P. Bone Disease in Primary Hyperparathyroidism-Changes Occurring in Bone Metabolism and New Potential Treatment Strategies. Int J Mol Sci 2024; 25:11639. [PMID: 39519190 PMCID: PMC11546563 DOI: 10.3390/ijms252111639] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/15/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024] Open
Abstract
Primary hyperparathyroidism (PHPT) is a common endocrinopathy, predominantly caused by a single parathyroid adenoma that is responsible for the excessive secretion of parathyroid hormone (PTH)-the hallmark of disease. Excess of this hormone causes remarkable changes in bone metabolism, including an increased level of bone remodeling with a predominance of bone resorption. Those changes lead to deterioration of bone structure and density, especially in cortical bone. The main treatment for PHPT is surgical removal of the adenoma, which normalizes PTH levels and terminates the progression of bone disease and leads to its regeneration. However, because not all the patients are suitable candidates for surgery, alternative therapies are needed. Current non-surgical treatments targeting bone disease secondary to PHPT include bisphosphonates and denosumab. Those antiresorptives prevent further bone loss, but they lack the ability to regenerate already degraded bone. There is ongoing research to find targeted drugs capable of halting resorption alongside stimulating bone formation. This review presents the advancements in understanding the molecular mechanisms responsible for bone disease in PHPT and assesses the efficacy of new potential therapeutic approaches (e.g., allosteric inhibitors of the PTH receptor, V-ATPase, or cathepsin inhibitors) aimed at mitigating bone loss and enhancing bone regeneration in affected patients.
Collapse
Affiliation(s)
- Mirella Iwanowska
- Department of Endocrinology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland
| | - Magdalena Kochman
- Department of Endocrinology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland
| | - Alicja Szatko
- Department of Endocrinology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland
- EndoLab Laboratory, Centre of Postgraduate Medical Education, 01-809 Warsaw, Poland
| | - Wojciech Zgliczyński
- Department of Endocrinology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland
| | - Piotr Glinicki
- Department of Endocrinology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland
- EndoLab Laboratory, Centre of Postgraduate Medical Education, 01-809 Warsaw, Poland
| |
Collapse
|
4
|
Mukhopadhyay U, Levantovsky S, Carusone TM, Gharbi S, Stein F, Behrends C, Bhogaraju S. A ubiquitin-specific, proximity-based labeling approach for the identification of ubiquitin ligase substrates. SCIENCE ADVANCES 2024; 10:eadp3000. [PMID: 39121224 PMCID: PMC11313854 DOI: 10.1126/sciadv.adp3000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 07/03/2024] [Indexed: 08/11/2024]
Abstract
Over 600 E3 ligases in humans execute ubiquitination of specific target proteins in a spatiotemporal manner to elicit desired signaling effects. Here, we developed a ubiquitin-specific proximity-based labeling method to selectively biotinylate substrates of a given ubiquitin ligase. By fusing the biotin ligase BirA and an Avi-tag variant to the candidate E3 ligase and ubiquitin, respectively, we were able to specifically enrich bona fide substrates of a ligase using a one-step streptavidin pulldown under denaturing conditions. We applied our method, which we named Ub-POD, to the really interesting new gene (RING) E3 ligase RAD18 and identified proliferating cell nuclear antigen and several other critical players in the DNA damage repair pathway. Furthermore, we successfully applied Ub-POD to the RING ubiquitin ligase tumor necrosis factor receptor-associated factor 6 and a U-box-type E3 ubiquitin ligase carboxyl terminus of Hsc70-interacting protein. We anticipate that our method could be widely adapted to all classes of ubiquitin ligases to identify substrates.
Collapse
Affiliation(s)
- Urbi Mukhopadhyay
- European Molecular Biology Laboratory, 71 avenue des Martyrs, 38042 Grenoble, France
| | - Sophie Levantovsky
- Munich Cluster for Systems Neurology, Medical Faculty, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Teresa Maria Carusone
- European Molecular Biology Laboratory, 71 avenue des Martyrs, 38042 Grenoble, France
| | - Sarah Gharbi
- European Molecular Biology Laboratory, 71 avenue des Martyrs, 38042 Grenoble, France
| | - Frank Stein
- Proteomics Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Christian Behrends
- Munich Cluster for Systems Neurology, Medical Faculty, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Sagar Bhogaraju
- European Molecular Biology Laboratory, 71 avenue des Martyrs, 38042 Grenoble, France
| |
Collapse
|
5
|
Ayyasamy R, Fan S, Czernik P, Lecka-Czernik B, Chattopadhyay S, Chakravarti R. 14-3-3ζ suppresses RANKL signaling by destabilizing TRAF6. J Biol Chem 2024; 300:107487. [PMID: 38908751 PMCID: PMC11331427 DOI: 10.1016/j.jbc.2024.107487] [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: 05/12/2024] [Accepted: 05/31/2024] [Indexed: 06/24/2024] Open
Abstract
Macrophages are essential regulators of inflammation and bone loss. Receptor activator of nuclear factor-κβ ligand (RANKL), a pro-inflammatory cytokine, is responsible for macrophage differentiation to osteoclasts and bone loss. We recently showed that 14-3-3ζ-knockout (YwhazKO) rats exhibit increased bone loss in the inflammatory arthritis model. 14-3-3ζ is a cytosolic adaptor protein that actively participates in many signaling transductions. However, the role of 14-3-3ζ in RANKL signaling or bone remodeling is unknown. We investigated how 14-3-3ζ affects osteoclast activity by evaluating its role in RANKL signaling. We utilized 14-3-3ζ-deficient primary bone marrow-derived macrophages obtained from wildtype and YwhazKO animals and RAW264.7 cells generated using CRISPR-Cas9. Our results showed that 14-3-3ζ-deficient macrophages, upon RANKL stimulation, have bigger and stronger tartrate-resistant acid phosphatase-positive multinucleated cells and increased bone resorption activity. The presence of 14-3-3ζ suppressed RANKL-induced MAPK and AKT phosphorylation, transcription factors (NFATC1 and p65) nuclear translocation, and subsequently, gene induction (Rank, Acp5, and Ctsk). Mechanistically, 14-3-3ζ interacts with TRAF6, an essential component of the RANKL receptor complex. Upon RANKL stimulation, 14-3-3ζ-TRAF6 interaction was increased, while RANK-TRAF6 interaction was decreased. Importantly, 14-3-3ζ supported TRAF6 ubiquitination and degradation by the proteasomal pathway, thus dampening the downstream RANKL signaling. Together, we show that 14-3-3ζ regulates TRAF6 levels to suppress inflammatory RANKL signaling and osteoclast activity. To the best of our knowledge, this is the first report on 14-3-3ζ regulation of RANKL signaling and osteoclast activation.
Collapse
Affiliation(s)
- R Ayyasamy
- Department of Physiology & Pharmacology, College of Medicine & Life Sciences, University of Toledo, Toledo, Ohio, USA
| | - S Fan
- Department of Medical Microbiology & Immunology, College of Medicine & Life Sciences, University of Toledo, Toledo, Ohio, USA
| | - P Czernik
- Department of Orthopedics, College of Medicine & Life Sciences, University of Toledo, Toledo, Ohio, USA
| | - B Lecka-Czernik
- Department of Orthopedics, College of Medicine & Life Sciences, University of Toledo, Toledo, Ohio, USA
| | - S Chattopadhyay
- Department of Medical Microbiology & Immunology, College of Medicine & Life Sciences, University of Toledo, Toledo, Ohio, USA; Microbiology, Immunology & Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - R Chakravarti
- Department of Physiology & Pharmacology, College of Medicine & Life Sciences, University of Toledo, Toledo, Ohio, USA.
| |
Collapse
|
6
|
Xie L, Ren X, Yang Z, Zhou T, Zhang M, An W, Guan Z. Exosomal circ_0000722 derived from periodontal ligament stem cells undergoing osteogenic differentiation promotes osteoclastogenesis. Int Immunopharmacol 2024; 128:111520. [PMID: 38199194 DOI: 10.1016/j.intimp.2024.111520] [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: 06/03/2023] [Revised: 01/05/2024] [Accepted: 01/05/2024] [Indexed: 01/12/2024]
Abstract
Periodontal ligament stem cells (PDLSCs), which are considered promising stem cells for regeneration of periodontal bony tissue, can also manipulate alveolar bone remodeling by exosomes. In this study, we investigated interactions between PDLSCs under osteogenic differentiation and osteoclast precursors. The results showed that conditioned medium from PDLSCs under 5d osteogenic induction promoted osteoclastogenesis of RAW264.7 cells. The exosomes extracted from those conditioned media showed similar effects on osteoclastogenesis. Furthermore, exosomes from PDLSCs under 5d of osteogenic induction showed significantly high expression of circ_0000722, compared with exosomes from PDLSCs before osteogenic induction. Downregulation of circ_0000722 significantly attenuated the effect of PDLSC-derived exosomes on the osteoclastogenesis of RAW264.7 cells. Our findings suggested that exosomal circ_0000722 derived from periodontal ligament stem cells undergoing osteogenic differentiation might promote osteoclastogenesis by upregulating TRAF6 expression and activating downstream NF-κB and AKT signaling pathways.
Collapse
Affiliation(s)
- Liangkun Xie
- Department of Oral Implantology, Kunming Medical University School and Hospital of Stomatology, Kunming, Yunnan, China; Yunnan Key Laboratory of Stomatology, Kunming, Yunnan, China
| | - Xuefeng Ren
- Yunnan Key Laboratory of Stomatology, Kunming, Yunnan, China; Department of Periodontology, Kunming Medical University School and Hospital of Stomatology, Kunming, Yunnan, China
| | - Zijie Yang
- Department of Oral Implantology, Kunming Medical University School and Hospital of Stomatology, Kunming, Yunnan, China; Yunnan Key Laboratory of Stomatology, Kunming, Yunnan, China
| | - Ting Zhou
- Yunnan Key Laboratory of Stomatology, Kunming, Yunnan, China
| | - Mingzhu Zhang
- Yunnan Key Laboratory of Stomatology, Kunming, Yunnan, China; Department of Periodontology, Kunming Medical University School and Hospital of Stomatology, Kunming, Yunnan, China
| | - Wei An
- Department of Oral Implantology, Kunming Medical University School and Hospital of Stomatology, Kunming, Yunnan, China; Yunnan Key Laboratory of Stomatology, Kunming, Yunnan, China
| | - Zheng Guan
- Biomedical Research Center, the Affiliated Calmette Hospital of Kunming Medical University (the First Hospital of Kunming), Kunming, Yunnan, China.
| |
Collapse
|
7
|
Jeong C, Lee CH, Lee Y, Seo J, Wang W, Park KH, Oh E, Cho Y, Park C, Son YJ, Yoon Park JH, Kang H, Lee KW. Ulmus macrocarpa Hance trunk bark extracts inhibit RANKL-induced osteoclast differentiation and prevent ovariectomy-induced osteoporosis in mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117285. [PMID: 37839769 DOI: 10.1016/j.jep.2023.117285] [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: 07/30/2023] [Revised: 09/25/2023] [Accepted: 10/04/2023] [Indexed: 10/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ulmus macrocarpa Hance (UmH) bark has been traditionally utilized for medicinal purposes. The bark extract of this plant has diverse health benefits, and its potential role in enhancing bone health is of distinct interest, particularly when considering the substantial health and economic implications of bone-related pathologies, such as osteoporosis. Despite the compelling theoretical implications of UmH bark in fortifying bone health, no definitive evidence at the in vivo level is currently available, thus highlighting the innovative and as-yet-unexplored potential of this field of study. AIM OF THE STUDY Primarily, our study aims to conduct a meticulous analysis of the disparity in the concentration of active compounds in the UmH root bark (Umrb) and trunk bark (Umtb) extracts and confirm UmH bark's efficacy in enhancing bone health in vivo, illuminating the cellular mechanisms involved. MATERIALS AND METHODS The Umrb and Umtb extracts were subjected to component analysis using high-performance liquid chromatography and then assessed for their inhibitory effects on osteoclast differentiation through the TRAP assay. An ovariectomized (OVX) mouse model replicates postmenopausal conditions commonly associated with osteoporosis. Micro-CT was used to analyze bone structure parameters, and enzyme-linked immunosorbent assay and staining were used to assess bone formation markers and osteoclast activity. Furthermore, this study investigated the impact of the extract on the expression of pivotal proteins and genes involved in bone formation and resorption using mouse bone marrow-derived macrophages (BMMs). RESULTS The findings of our study reveal a significant discrepancy in the concentration of active constituents between Umrb and Umtb, establishing Umtb as a superior source for promoting bone health. I addition, a standardized pilot-scale procedure was conducted for credibility. The bone health benefits of Umtb were verified using an OVX model. This validation involved the assessment of various parameters, including BMD, BV/TV, and BS/TV, using micro-CT imaging. Additionally, the activation of osteoblasts was evaluated by Umtb by measuring specific factors such as ALP, OCN, OPG in blood samples and through IHC staining. In the same investigations, diminished levels of osteoclast differentiation factors, such as TRAP, NFATc1, were also observed. The observed patterns exhibited consistency in vitro BMM investigations. CONCLUSIONS Through verification at both in vitro levels using BMMs and in vivo levels using the OVX-induced mouse model, our research demonstrates that Umtb is a more effective means of improving bone health in comparison to Umrb. These findings pave the way for developing health-functional foods or botanical drugs targeting osteoporosis and other bone-related disorders and enhance the prospects for future research extensions, including clinical studies, in extract applications.
Collapse
Affiliation(s)
- Chanhyeok Jeong
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Chang Hyung Lee
- Bio-MAX Institute, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Yongjin Lee
- Department of Pharmacy, Sunchon National University, Suncheon, 57922, Republic of Korea.
| | - Jiwon Seo
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Weihong Wang
- Laboratory of Marine Drugs, School of Earth and Environmental Sciences, NS-80, Seoul National University, Seoul, 08826, Republic of Korea; Research Institute of Oceanography, Seoul National University, NS-80, Seoul, 08826, Republic of Korea.
| | - Kyu-Hyung Park
- Laboratory of Marine Drugs, School of Earth and Environmental Sciences, NS-80, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Eunseok Oh
- Laboratory of Marine Drugs, School of Earth and Environmental Sciences, NS-80, Seoul National University, Seoul, 08826, Republic of Korea; Research Institute of Oceanography, Seoul National University, NS-80, Seoul, 08826, Republic of Korea.
| | - Youbin Cho
- Laboratory of Marine Drugs, School of Earth and Environmental Sciences, NS-80, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Chanyoon Park
- Bio-MAX Institute, Seoul National University, Seoul, 08826, Republic of Korea; Laboratory of Marine Drugs, School of Earth and Environmental Sciences, NS-80, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Young-Jin Son
- Department of Pharmacy, Sunchon National University, Suncheon, 57922, Republic of Korea.
| | - Jung Han Yoon Park
- Bio-MAX Institute, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Heonjoong Kang
- Bio-MAX Institute, Seoul National University, Seoul, 08826, Republic of Korea; Laboratory of Marine Drugs, School of Earth and Environmental Sciences, NS-80, Seoul National University, Seoul, 08826, Republic of Korea; Research Institute of Oceanography, Seoul National University, NS-80, Seoul, 08826, Republic of Korea; Interdisciplinary Graduate Program in Genetic Engineering, Seoul National University, NS-80, Seoul, 08826, Republic of Korea.
| | - Ki Won Lee
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea; Bio-MAX Institute, Seoul National University, Seoul, 08826, Republic of Korea; Advanced Institutes of Convergence Technology, Seoul National University, Suwon, 16229, Republic of Korea; Institutes of Green Bio Science & Technology, Seoul National University, Pyeongchang, 25354, Republic of Korea; Department of Agricultural Biotechnology and Center for Food and Bio Convergence, Seoul National. University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
8
|
Jin C, Zheng J, Yang Q, Jia Y, Li H, Liu X, Xu Y, Chen Z, He L. Morusin Inhibits RANKL-induced Osteoclastogenesis and Ovariectomized Osteoporosis. Comb Chem High Throughput Screen 2024; 27:1358-1370. [PMID: 37807416 DOI: 10.2174/0113862073252310230925062415] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 07/18/2023] [Accepted: 08/09/2023] [Indexed: 10/10/2023]
Abstract
BACKGROUND Postmenopausal osteoporosis (PMOP) is a classic type of osteoporosis that has gradually become a significant health problem worldwide. There is an urgent need for a safe alternative therapeutic agent considering the poor therapeutic strategies currently available for this disease. The roots and bark of the Morus australis tree (Moraceae) are used to make a traditional Chinese medicine known as "Morusin", and accumulating evidence has demonstrated its multiple activities, such as anti-inflammatory and anti-tumor effects. OBJECTIVE In this study, we aim to explore the effect of Morusin on mouse osteoclasts and its mechanism. METHODS In this study, we explored the inhibitory effects of Morusin on murine osteoclasts in vitro and its mechanism, and the protective effect of Morusin on an ovariectomy (OVX)-induced osteoporosis model in vivo. RESULTS The results showed that Morusin prevented OVX-induced bone loss and dramatically decreased RANKL-induced osteoclastogenesis. Morusin interfered with RANKL-activated NF- κB, MAPK, and PI3K/AKT signaling pathways. The expression of three master factors that control osteoclast differentiation, c-Fos, NFATc1, and c-Jun, was reduced by Morusin treatment. Collectively, in vitro results indicated that Morusin has a protective effect on OVX-induced bone loss in a mouse model. CONCLUSION Our data provide encouraging evidence that Morusin may be an effective treatment for PMOP.
Collapse
Affiliation(s)
- Cong Jin
- Department of Orthopaedics, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China
| | - Jiewen Zheng
- Department of Orthopaedics, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China
- Shaoxing University School of Medicine, Shaoxing, Zhejiang, 312000, China
| | - Qichang Yang
- Department of Orthopaedics, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Yewei Jia
- Department of Orthopaedics, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China
| | - Haibo Li
- Department of Orthopaedics, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China
| | - Xuewen Liu
- Department of Orthopaedics, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China China
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Yangjun Xu
- Department of Orthopaedics, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China
- Shaoxing University School of Medicine, Shaoxing, Zhejiang, 312000, China
| | - Zhuolin Chen
- Department of Orthopaedics, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China
- Department of Orthopaedics, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310000, China
| | - Lei He
- Department of Orthopaedics, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China
| |
Collapse
|
9
|
Xie Z, Wu Y, Shen Y, Guo J, Yuan P, Ma Q, Wang S, Jie Z, Zhou H, Fan S, Chen S. USP7 Inhibits Osteoclastogenesis via Dual Effects of Attenuating TRAF6/TAK1 Axis and Stimulating STING Signaling. Aging Dis 2023; 14:2267-2283. [PMID: 37199589 PMCID: PMC10676781 DOI: 10.14336/ad.2023.0325-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 03/25/2023] [Indexed: 05/19/2023] Open
Abstract
Ubiquitination is a reversible post-translational modification implicated in cell differentiation, homeostasis, and organ development. Several deubiquitinases (DUBs) decrease protein ubiquitination through the hydrolysis of ubiquitin linkages. However, the role of DUBs in bone resorption and formation is still unclear. In this study, we identified DUB ubiquitin-specific protease 7 (USP7) as a negative regulator of osteoclast formation. USP7 combines with tumor necrosis factor receptor-associated factor 6 (TRAF6) and inhibits its ubiquitination by impairing the Lys63-linked polyubiquitin chain. Such impairment leads to the suppression of receptor activator of NF-κB ligand (RANKL)-mediated nuclear factor-κB (NF-κB) and mitogen-activated protein kinases (MAPKs) activation without affecting TRAF6 stability. USP7 also protects the stimulator of interferon genes (STING) against degradation, inducing interferon-β (IFN-β) expression in osteoclast formation, thereby inhibiting osteoclastogenesis cooperatively with the classical TRAF6 pathway. Furthermore, USP7 inhibition accelerates osteoclast differentiation and bone resorption both in vitro and in vivo. Contrarily, USP7 overexpression impairs osteoclast differentiation and bone resorption in vitro and in vivo. Additionally, in ovariectomy (OVX) mice, USP7 levels are lower than those in sham-operated mice, suggesting that USP7 plays a role in osteoporosis. Altogether, our data reveal the dual effect of USP7-mediated TRAF6 signal transduction and USP7-mediated protein degradation of STING in osteoclast formation.
Collapse
Affiliation(s)
- Ziang Xie
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China.
| | - Yizheng Wu
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China.
| | - Yang Shen
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China.
| | - Jiandong Guo
- Department of Orthopedic Surgery, Ninth people’s Hospital of Hangzhou, Hangzhou, China.
| | - Putao Yuan
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China.
| | - Qingliang Ma
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China.
| | - Shiyu Wang
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China.
| | - Zhiwei Jie
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China.
| | - Hongyi Zhou
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China.
| | - Shunwu Fan
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China.
| | - Shuai Chen
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
10
|
Chen Z, Joseph D, Ding M, Bhujbal SP, Rajan RPS, Kim E, Park SW, Lee S, Lee TH. Synthesis and evaluation of 2-NMPA derivatives as potential agents for prevention of osteoporosis in vitro and in vivo. Eur J Med Chem 2023; 260:115767. [PMID: 37651877 DOI: 10.1016/j.ejmech.2023.115767] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/25/2023] [Accepted: 08/25/2023] [Indexed: 09/02/2023]
Abstract
Abnormal osteoclast differentiation causes various bone disorders such as osteoporosis. Targeting the formation and activation of osteoclasts has been recognized as an effective approach for preventing osteoporosis. Herein, we synthesized eleven 2-NMPA derivatives which are (2-(2-chlorophenoxy)-N-(4-alkoxy-2-morpholinophenyl) acetamides, and evaluated their suppression effects on osteoclastogenesis in vitro by using TRAP-staining assay. Among the synthesized eleven novel 2-NMPAs, 4-(2-(2-chlorophenoxy)acetamido)-3-morpholinophenyl trifluoromethanesulfonate (11b), 4-(2-(2-chlorophenoxy) acetamido)-3-morpholinophenyl-3-(N-(2-oxo-2-((2-(phenylthio) phenyl) amino) ethyl)methylsulfonamido)benzoate (11d), and 4-(2-(2-chlorophenoxy) acetamido)-3-morpholinophenyl 4-acetamidobenzenesulfonate (11h) displayed highly inhibitory bioactivity on the differentiation of primary osteoclasts. 11h was selected for further investigation of the inhibitory effects and potential mechanism involved in the suppression of osteoclastogenesis. In vitro analysis suggested that 11h inhibited osteoclastogenesis with an IC50 of 358.29 nM, decreased the formation of F-action belts and bone resorption, without interfering cell viability and osteoblast differentiation. Furthermore, the mRNA expressions of osteoclast-specific genes such as Acp5, Nfatc1, Dc-stamp, Atp6v0d2, Mmp9, and Ctsk significantly decreased following 11h treatment. RANKL-induced osteoclast-specific proteins analysis demonstrated that 11h suppressed osteoclast differentiation by downregulating of RANKL-mediated TRAF6 expression, followed by inactivation of PI3K/AKT and IκBα/NF-κB signaling pathways. Finally, 11h inhibited ovariectomy-induced bone loss in vivo. Therefore, the current work highlighted the therapeutic potential of 11h as an anti-osteoporosis lead compound.
Collapse
Affiliation(s)
- Zhihao Chen
- BioMedical Sciences Graduate Program (BMSGP), Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Devaneyan Joseph
- Department of Chemistry, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Mina Ding
- BioMedical Sciences Graduate Program (BMSGP), Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Swapnil Pandurang Bhujbal
- Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, 426-791, Republic of Korea
| | | | - Eunae Kim
- Department of Pharmacy, College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
| | - Sang-Wook Park
- Department of Oral Biochemistry, Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Sunwoo Lee
- Department of Chemistry, Chonnam National University, Gwangju, 61186, Republic of Korea.
| | - Tae-Hoon Lee
- Department of Oral Biochemistry, Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
11
|
Kim C. Extracellular Signal-Regulated Kinases Play Essential but Contrasting Roles in Osteoclast Differentiation. Int J Mol Sci 2023; 24:15342. [PMID: 37895023 PMCID: PMC10607827 DOI: 10.3390/ijms242015342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Bone homeostasis is regulated by the balanced actions of osteoblasts that form the bone and osteoclasts (OCs) that resorb the bone. Bone-resorbing OCs are differentiated from hematopoietic monocyte/macrophage lineage cells, whereas osteoblasts are derived from mesenchymal progenitors. OC differentiation is induced by two key cytokines, macrophage colony-stimulating factor (M-CSF), a factor essential for the proliferation and survival of the OCs, and receptor activator of nuclear factor kappa-B ligand (RANKL), a factor for responsible for the differentiation of the OCs. Mitogen-activated protein kinases (MAPKs), including extracellular signal-regulated kinases (ERKs), p38, and c-Jun N-terminal kinases, play an essential role in regulating the proliferation, differentiation, and function of OCs. ERKs have been known to play a critical role in the differentiation and activation of OCs. In most cases, ERKs positively regulate OC differentiation and function. However, several reports present conflicting conclusions. Interestingly, the inhibition of OC differentiation by ERK1/2 is observed only in OCs differentiated from RAW 264.7 cells. Therefore, in this review, we summarize the current understanding of the conflicting actions of ERK1/2 in OC differentiation.
Collapse
Affiliation(s)
- Chaekyun Kim
- BK21 Program in Biomedical Science & Engineering, Laboratory for Leukocyte Signaling Research, Department of Pharmacology, College of Medicine, Inha University, Incheon 22212, Republic of Korea
| |
Collapse
|
12
|
Yuan W, Hu Y, Lu C, Zhang J, Liu Y, Li X, Jia K, Huang Y, Li Z, Chen X, Wang F, Yi X, Che X, Xiong H, Cheng B, Ma J, Zhao Y, Lu H. Propineb induced notochord deformity, craniofacial malformation, and osteoporosis in zebrafish through dysregulated reactive oxygen species generation. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2023; 261:106596. [PMID: 37290275 DOI: 10.1016/j.aquatox.2023.106596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/28/2023] [Accepted: 05/30/2023] [Indexed: 06/10/2023]
Abstract
Dithiocarbamate (DTC) fungicides are contaminants that are ubiquitous in the environment. Exposure to DTC fungicides has been associated with a variety of teratogenic developmental effects. Propineb, a member of DTCs, was evaluated for the toxicological effects on notochord and craniofacial development, osteogenesis in zebrafish model. Embryos at 6 hours post-fertilization (hpf) were exposed to propineb at dosages of 1 and 4 μM. Morphological parameters were evaluated at exposure times of 24, 48, 72, and 120 hpf after propineb exposure. The survival and hatching rates as well as body length decreased at 1 and 4 μmol/L groups. Besides, transgenic zebrafish exposed to propineb showed abnormal vacuole biogenesis in notochord cells at the early stage of development. The expression of collagen type 2 alpha 1a (col2a1a), sonic hedgehog (shh), and heat shock protein family B member 11 (hspb11) measured by quantitative PCR and in situ hybridization experiment of col8a1a gene have consolidated the proposal process. Besides, Alcian blue, calcein, and alizarin red staining profiles displayed craniofacial malformations and osteoporosis were induced following propineb exposure. PPB exposure induced the changes in oxidative stress and reactive oxygen species inhibitor alleviated the deformities of PPB. Collectively, our data suggested that propineb exposure triggered bone abnormalities in different phenotypes of zebrafish. Therefore, propineb is a potential toxicant of high priority concern for aquatic organisms.
Collapse
Affiliation(s)
- Wei Yuan
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Ying Hu
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Chen Lu
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Jun Zhang
- Nanjing Institute of Environmental Sciences, Ministry of Ecology and Environment of the People's Republic of China, Nanjing, 210042, Jiangsu, China
| | - Ye Liu
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Xinran Li
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Kun Jia
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Yong Huang
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Zekun Li
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Xiaomei Chen
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Fei Wang
- The First Clinical College of Gannan Medical Uinversity, Ganzhou, 341000, Jiangxi, China
| | - Xiaokun Yi
- The First Clinical College of Gannan Medical Uinversity, Ganzhou, 341000, Jiangxi, China
| | - Xiaofang Che
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Haibin Xiong
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Bo Cheng
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Jinze Ma
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Yan Zhao
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Huiqiang Lu
- Ganzhou Key Laboratory for Drug Screening and Discovery, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China; Affiliated Hospital of Jinggangshan University, Jinggangshan University, Ji'an, 343009, Jiangxi, China..
| |
Collapse
|
13
|
Hong SJ, Jung S, Jang JS, Mo S, Kwon JO, Kim MK, Kim HH. PARK2 Induces Osteoclastogenesis through Activation of the NF-κB Pathway. Mol Cells 2022; 45:749-760. [PMID: 36047447 PMCID: PMC9589368 DOI: 10.14348/molcells.2022.0058] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/25/2022] [Accepted: 06/20/2022] [Indexed: 11/27/2022] Open
Abstract
Osteoclast generation from monocyte/macrophage lineage precursor cells needs to be tightly regulated to maintain bone homeostasis and is frequently over-activated in inflammatory conditions. PARK2, a protein associated with Parkinson's disease, plays an important role in mitophagy via its ubiquitin ligase function. In this study, we investigated whether PARK2 is involved in osteoclastogenesis. PARK2 expression was found to be increased during the receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast differentiation. PARK2 gene silencing with siRNA significantly reduced osteoclastogenesis induced by RANKL, LPS (lipopolysaccharide), TNFα (tumor necrosis factor α), and IL-1β (interleukin-1β). On the other hand, overexpression of PARK2 promoted osteoclastogenesis. This regulation of osteoclastogenesis by PARK2 was mediated by IKK (inhibitory κB kinase) and NF-κB activation while MAPK (mitogen-activated protein kinases) activation was not involved. Additionally, administration of PARK2 siRNA significantly reduced osteoclastogenesis and bone loss in an in vivo model of inflammatory bone erosion. Taken together, this study establishes a novel role for PARK2 as a positive regulator in osteoclast differentiation and inflammatory bone destruction.
Collapse
Affiliation(s)
- Seo Jin Hong
- Department of Cell and Developmental Biology, BK21 Program and Dental Research Institute (DRI), School of Dentistry, Seoul National University, Seoul 03080, Korea
| | - Suhan Jung
- Department of Cell and Developmental Biology, BK21 Program and Dental Research Institute (DRI), School of Dentistry, Seoul National University, Seoul 03080, Korea
| | - Ji Sun Jang
- Department of Cell and Developmental Biology, BK21 Program and Dental Research Institute (DRI), School of Dentistry, Seoul National University, Seoul 03080, Korea
| | - Shenzheng Mo
- Department of Cell and Developmental Biology, BK21 Program and Dental Research Institute (DRI), School of Dentistry, Seoul National University, Seoul 03080, Korea
| | - Jun-Oh Kwon
- Department of Cell and Developmental Biology, BK21 Program and Dental Research Institute (DRI), School of Dentistry, Seoul National University, Seoul 03080, Korea
| | - Min Kyung Kim
- Department of Cell and Developmental Biology, BK21 Program and Dental Research Institute (DRI), School of Dentistry, Seoul National University, Seoul 03080, Korea
| | - Hong-Hee Kim
- Department of Cell and Developmental Biology, BK21 Program and Dental Research Institute (DRI), School of Dentistry, Seoul National University, Seoul 03080, Korea
| |
Collapse
|
14
|
E3 Ubiquitin Ligases: Potential Therapeutic Targets for Skeletal Pathology and Degeneration. Stem Cells Int 2022; 2022:6948367. [PMID: 36203882 PMCID: PMC9532118 DOI: 10.1155/2022/6948367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/06/2022] [Accepted: 09/03/2022] [Indexed: 11/18/2022] Open
Abstract
The ubiquitination-proteasome system (UPS) is crucial in regulating a variety of cellular processes including proliferation, differentiation, and survival. Ubiquitin protein ligase E3 is the most critical molecule in the UPS system. Dysregulation of the UPS system is associated with many conditions. Over the past few decades, there have been an increasing number of studies focusing on the UPS system and how it affects bone metabolism. Multiple E3 ubiquitin ligases have been found to mediate osteogenesis or osteolysis through a variety of pathways. In this review, we describe the mechanisms of UPS, especially E3 ubiquitin ligases on bone metabolism. To date, many E3 ubiquitin ligases have been found to regulate osteogenesis or osteoclast differentiation. We review the classification of these E3 enzymes and the mechanisms that influence upstream and downstream molecules and transduction pathways. Finally, this paper reviews the discovery of the relevant UPS inhibitors, drug molecules, and noncoding RNAs so far and prospects the future research and treatment.
Collapse
|
15
|
Wu MH, Hsu WB, Chen MH, Shi CS. Inhibition of Neddylation Suppresses Osteoclast Differentiation and Function In Vitro and Alleviates Osteoporosis In Vivo. Biomedicines 2022; 10:2355. [PMID: 36289618 PMCID: PMC9598818 DOI: 10.3390/biomedicines10102355] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 09/20/2023] Open
Abstract
Neddylation, or the covalent addition of NEDD8 to specific lysine residue of proteins, is a reversible posttranslational modification, which regulates numerous biological functions; however, its involvement and therapeutic significance in osteoporosis remains unknown. Our results revealed that during the soluble receptor activator of nuclear factor-κB ligand (sRANKL)-stimulated osteoclast differentiation, the neddylation and expression of UBA3, the NEDD8-activating enzyme (NAE) catalytic subunit, were dose- and time-dependently upregulated in RAW 264.7 macrophages. UBA3 knockdown for diminishing NAE activity or administering low doses of the NAE inhibitor MLN4924 significantly suppressed sRANKL-stimulated osteoclast differentiation and bone-resorbing activity in the macrophages by inhibiting sRANKL-stimulated neddylation and tumor necrosis factor receptor-associated factor 6 (TRAF6)-activated transforming growth factor-β-activated kinase 1 (TAK1) downstream signaling for diminishing nuclear factor-activated T cells c1 (NFATc1) expression. sRANKL enhanced the interaction of TRAF6 with the neddylated proteins and the polyubiquitination of TRAF6's lysine 63, which activated TAK1 downstream signaling; however, this process was inhibited by MLN4924. MLN4924 significantly reduced osteoporosis in an ovariectomy- and sRANKL-induced osteoporosis mouse model in vivo. Our novel finding was that NAE-mediated neddylation participates in RANKL-activated TRAF6-TAK1-NFATc1 signaling during osteoclast differentiation and osteoporosis, suggesting that neddylation may be a new target for treating osteoporosis.
Collapse
Affiliation(s)
- Meng-Huang Wu
- Department of Orthopaedics, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Department of Orthopedics, Taipei Medical University Hospital, Taipei 11031, Taiwan
- TMU Biodesign Center, Taipei Medical University, Taipei 11031, Taiwan
| | - Wei-Bin Hsu
- Sports Medicine Center, Chang Gung Memorial Hospital, Puzi 61301, Taiwan
| | - Mei-Hsin Chen
- Sports Medicine Center, Chang Gung Memorial Hospital, Puzi 61301, Taiwan
| | - Chung-Sheng Shi
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33332, Taiwan
- Colon and Rectal Surgery, Department of Surgery, Chiayi Chang Gung Memorial Hospital, Puzi 61301, Taiwan
| |
Collapse
|
16
|
Osteostatin Inhibits M-CSF+RANKL-Induced Human Osteoclast Differentiation by Modulating NFATc1. Int J Mol Sci 2022; 23:ijms23158551. [PMID: 35955685 PMCID: PMC9369336 DOI: 10.3390/ijms23158551] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/24/2022] [Accepted: 07/29/2022] [Indexed: 01/27/2023] Open
Abstract
Parathyroid hormone-related protein (PTHrP) C-terminal peptides regulate the metabolism of bone cells. PHTrP [107–111] (osteostatin) promotes bone repair in animal models of bone defects and prevents bone erosion in inflammatory arthritis. In addition to its positive effects on osteoblasts, osteostatin may inhibit bone resorption. The aim of this study was to determine the effects of osteostatin on human osteoclast differentiation and function. We used macrophage colony-stimulating factor (M-CSF) and receptor activator of nuclear factor κB ligand (RANKL) to induce the osteoclast differentiation of adherent human peripheral blood mononuclear cells. Tartrate-resistant acid phosphatase (TRAP) staining was performed for the detection of the osteoclasts. The function of mature osteoclasts was assessed with a pit resorption assay. Gene expression was evaluated with qRT-PCR, and nuclear factor of activated T cells, cytoplasmic 1 (NFATc1) nuclear translocation was studied by immunofluorescence. We observed that osteostatin (100, 250 and 500 nM) decreased the differentiation of osteoclasts in a concentration-dependent manner, but it did not modify the resorptive ability of mature osteoclasts. In addition, osteostatin decreased the mRNA levels of cathepsin K, osteoclast associated Ig-like receptor (OSCAR) and NFATc1. The nuclear translocation of the master transcription factor in osteoclast differentiation NFATc1 was reduced by osteostatin. Our results suggest that the anti-resorptive effects of osteostatin may be dependent on the inhibition of osteoclastogenesis. This study has shown that osteostatin controls human osteoclast differentiation in vitro through the downregulation of NFATc1.
Collapse
|
17
|
Yin X, Liu Q, Liu F, Tian X, Yan T, Han J, Jiang S. Emerging Roles of Non-proteolytic Ubiquitination in Tumorigenesis. Front Cell Dev Biol 2022; 10:944460. [PMID: 35874839 PMCID: PMC9298949 DOI: 10.3389/fcell.2022.944460] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 06/15/2022] [Indexed: 12/13/2022] Open
Abstract
Ubiquitination is a critical type of protein post-translational modification playing an essential role in many cellular processes. To date, more than eight types of ubiquitination exist, all of which are involved in distinct cellular processes based on their structural differences. Studies have indicated that activation of the ubiquitination pathway is tightly connected with inflammation-related diseases as well as cancer, especially in the non-proteolytic canonical pathway, highlighting the vital roles of ubiquitination in metabolic programming. Studies relating degradable ubiquitination through lys48 or lys11-linked pathways to cellular signaling have been well-characterized. However, emerging evidence shows that non-degradable ubiquitination (linked to lys6, lys27, lys29, lys33, lys63, and Met1) remains to be defined. In this review, we summarize the non-proteolytic ubiquitination involved in tumorigenesis and related signaling pathways, with the aim of providing a reference for future exploration of ubiquitination and the potential targets for cancer therapies.
Collapse
Affiliation(s)
- Xiu Yin
- Clinical Medical Laboratory Center, Jining First People's Hospital, Jining Medical University, Jining, China
| | - Qingbin Liu
- Clinical Medical Laboratory Center, Jining First People's Hospital, Jining Medical University, Jining, China
| | - Fen Liu
- Clinical Medical Laboratory Center, Jining First People's Hospital, Jining Medical University, Jining, China
| | - Xinchen Tian
- Clinical Medical Laboratory Center, Jining First People's Hospital, Jining Medical University, Jining, China.,Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tinghao Yan
- Clinical Medical Laboratory Center, Jining First People's Hospital, Jining Medical University, Jining, China.,Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jie Han
- Department of Thyroid and Breast Surgery, Jining First People's Hospital, Jining Medical University, Jining, China
| | - Shulong Jiang
- Clinical Medical Laboratory Center, Jining First People's Hospital, Jining Medical University, Jining, China
| |
Collapse
|
18
|
Read OJ, Harrison DJ. Silencing Itch in human peripheral blood monocytes promotes their differentiation into osteoclasts. Mol Biol Rep 2022; 49:9113-9119. [PMID: 35793050 PMCID: PMC9463264 DOI: 10.1007/s11033-022-07726-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/15/2022] [Indexed: 11/24/2022]
Abstract
Introduction Two clinical case reports of humans with mutations in Itch reported distinct morphological defects such as stunted growth, macrocephaly, and dysmorphic features indicating a role for Itch in bone remodelling. Studies in mice have found that the encoded E3 ubiquitin ligase acts as a negative regulator of osteoclastogenesis, however no studies have investigated whether this is translatable to a human model. Experimental procedures Human peripheral blood monocytes were separated from whole blood and grown in M-CSF containing media. Media was later supplemented with RANKL to promote osteoclast differentiation. Transient siRNA-mediated Itch knockdown (si-Itch) in monocytes was verified by qPCR and western blot to confirm reduction in both Itch mRNA and protein respectively. Monocytes were aliquoted onto 96-well plates where confluence and osteoclast formation were analysed using automated cytometry analysis before and after staining for tartrate resistant acid phosphatase activity (TRAP). Cells were also stained with Hoechst33342 to look for multinucleate cells. Results Cells treated with si-Itch showed an 80% knockdown in Itch mRNA and > 75% reduction in protein. Following the 7-day differentiation period, si-Itch caused a 47% increase in multinucleate cells and a 17% increase in numbers of large cellular bodies and, indicating an overall increase in mature osteoclast formation. Conclusions Our preliminary data shows silencing Itch expression increases the potential of primary human monocytes to differentiate into osteoclast-like cells in vitro. Supplementary Information The online version contains supplementary material available at 10.1007/s11033-022-07726-1.
Collapse
Affiliation(s)
- O J Read
- Pathology Department, School of Medicine, University of St Andrews, St Andrews, UK.
| | - D J Harrison
- Pathology Department, School of Medicine, University of St Andrews, St Andrews, UK
| |
Collapse
|
19
|
Jianwei W, Ye T, Hongwei W, Dachuan L, Fei Z, Jianyuan J, Hongli W. The Role of TAK1 in RANKL-Induced Osteoclastogenesis. Calcif Tissue Int 2022; 111:1-12. [PMID: 35286417 DOI: 10.1007/s00223-022-00967-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/28/2022] [Indexed: 12/31/2022]
Abstract
Bone remodelling is generally a dynamic process orchestrated by bone-resorbing osteoclasts and bone-forming osteoblasts. Osteoclasts are the only cell type capable of bone resorption to maintain bone homeostasis in the human body. However, excessive osteoclastogenesis can lead to osteolytic diseases. The receptor activator of nuclear factor-κB (NF-κB) ligand (RANKL) has been widely considered to be an important modulator of osteoclastogenesis thereby participating in the pathogenesis of osteolytic diseases. Transforming growth factor β-activated kinase 1 (TAK1), a member of the mitogen-activated protein kinase kinase kinase family, is an important intracellular molecule that regulates multiple signalling pathways, such as NF-κB and mitogen-activated protein kinase to mediate multiple physiological processes, including cell survival, inflammation, and tumourigenesis. Furthermore, increasing evidence has demonstrated that TAK1 is intimately involved in RANKL-induced osteoclastogenesis. Moreover, several detailed mechanisms by which TAK1 regulates RANKL-induced osteoclastogenesis have been clarified, and some potential approaches targeting TAK1 for the treatment of osteolytic diseases have emerged. In this review, we discuss how TAK1 functions in RANKL-mediated signalling pathways and highlight the significant role of TAK1 in RANKL-induced osteoclastogenesis. In addition, we discuss the potential clinical implications of TAK1 inhibitors for the treatment of osteolytic diseases.
Collapse
Affiliation(s)
- Wu Jianwei
- Department of Orthopaedics, Huashan Hospital, Fudan University, No. 12 Middle Wulumuqi Road, Shanghai City, 200040, Shanghai, China
| | - Tian Ye
- Department of Orthopaedics, Huashan Hospital, Fudan University, No. 12 Middle Wulumuqi Road, Shanghai City, 200040, Shanghai, China
| | - Wang Hongwei
- Department of Orthopaedics, Huashan Hospital, Fudan University, No. 12 Middle Wulumuqi Road, Shanghai City, 200040, Shanghai, China
| | - Li Dachuan
- Department of Orthopaedics, Huashan Hospital, Fudan University, No. 12 Middle Wulumuqi Road, Shanghai City, 200040, Shanghai, China
| | - Zou Fei
- Department of Orthopaedics, Huashan Hospital, Fudan University, No. 12 Middle Wulumuqi Road, Shanghai City, 200040, Shanghai, China
| | - Jiang Jianyuan
- Department of Orthopaedics, Huashan Hospital, Fudan University, No. 12 Middle Wulumuqi Road, Shanghai City, 200040, Shanghai, China.
| | - Wang Hongli
- Department of Orthopaedics, Huashan Hospital, Fudan University, No. 12 Middle Wulumuqi Road, Shanghai City, 200040, Shanghai, China.
| |
Collapse
|
20
|
RANKL Impairs the TLR4 Pathway by Increasing TRAF6 and RANK Interaction in Macrophages. BIOMED RESEARCH INTERNATIONAL 2022; 2022:7740079. [PMID: 35463988 PMCID: PMC9019442 DOI: 10.1155/2022/7740079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 03/02/2022] [Accepted: 03/16/2022] [Indexed: 11/17/2022]
Abstract
High serum levels of osteoprotegerin (OPG) are found in patients with obesity, type 2 diabetes, sepsis, or septic shock and are associated with a high mortality rate in stroke. The primary known function of OPG is to bind to the receptor activator of NF-κB ligand (RANKL), and by doing so, it inhibits the binding between RANKL and its receptor (RANK). TLR4 signaling in macrophages involves TRAF6 recruitment and contributes to low-grade chronic inflammation in adipose tissue. LPS is a classical activator of the TLR4 pathway and induces the expression of inflammatory cytokines in macrophages. We have previously observed that in the presence of RANKL, there is no LPS-induced activation of TLR4 in macrophages. In this study, we investigated the crosstalk between RANK and TLR4 pathways in macrophages stimulated with both RANKL and LPS to unveil the role of OPG in inflammatory processes. We found that RANKL inhibits TLR4 activation by binding to RANK, promoting the binding between TRAF6 and RANK, lowering TLR4 activation and the expression of proinflammatory mediators. Furthermore, high OPG levels aggravate inflammation by inhibiting RANKL. Our findings elect RANKL as a candidate for drug development as a way to mitigate the impact of obesity-induced inflammation in patients.
Collapse
|
21
|
Wu L, Liang J, Li J, Xu Y, Chen J, Su Y, Xian Y, Wei J, Xu J, Zhao J, Liu Q, Yang Y. Onc201 reduces osteoclastogenesis and prevents ovariectomy-induced bone loss via inhibiting RANKL-induced NFATc1 activation and the integrin signaling pathway. Eur J Pharmacol 2022; 923:174908. [PMID: 35405113 DOI: 10.1016/j.ejphar.2022.174908] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/17/2022] [Accepted: 03/17/2022] [Indexed: 11/03/2022]
Abstract
Osteoporosis is an osteolytic disease with a disrupted balance between the resorption and formation of bone as well as bone microstructure degeneration, leading to bone loss and increased fracture risk, which greatly affects patients' quality of life. Currently, inhibition of osteoclast bone resorption remains the mainstream treatment for osteoporosis. Onc201, a new compound, induces the gene expression of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and has an efficient anticancer effect in clinical trials. However, its effects on osteolytic disease and the mechanism of action are unclear. We examined the effect of Onc201 on nuclear factor κB ligand-receptor activator (RANKL)-induced osteoclasts via Cell Counting Kit-8, bone resorption assay, luciferase reporter assay, immunofluorescence staining, calcium ion intensity assay and employed an ovariectomy model to investigate the effect of Onc201 on osteoporosis in the mice. Results showed that Onc201 inhibited the function and formation of osteoclasts induced by RANKL in a manner that was dependent on time and concentration, and did not cause cytotoxicity. Mechanistically, Onc201 inhibited osteoclast-relevant genes and NFATc1 expression, the main transcriptional regulatory factor of the formation of osteoclasts induced by RANKL; meanwhile, downregulating the expressions of the osteoclast cytoskeleton key signal molecules integrin αvβ3, focal adhesion kinase (FAK), c-Src, and spleen-associated tyrosine kinase (SYK). In addition, Onc201 had a protective effect on the mouse model of bone loss caused by ovariectomy-induced estrogen deficiency, which is consistent with the in vitro results. Our findings suggest that the new small-molecular compound Onc201 has the potential to prevent osteoclast-related osteolytic diseases.
Collapse
Affiliation(s)
- Liwei Wu
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, People's Republic of China; Collaborative Innovation Center of Regenerative Medicine and Medical Biological Resources Development and Application, Guangxi Medical University, Nanning, Guangxi, 530021, People's Republic of China
| | - Jiamin Liang
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, People's Republic of China; Collaborative Innovation Center of Regenerative Medicine and Medical Biological Resources Development and Application, Guangxi Medical University, Nanning, Guangxi, 530021, People's Republic of China
| | - Jing Li
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, People's Republic of China; Collaborative Innovation Center of Regenerative Medicine and Medical Biological Resources Development and Application, Guangxi Medical University, Nanning, Guangxi, 530021, People's Republic of China
| | - Yang Xu
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, People's Republic of China
| | - Junchun Chen
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, People's Republic of China; Collaborative Innovation Center of Regenerative Medicine and Medical Biological Resources Development and Application, Guangxi Medical University, Nanning, Guangxi, 530021, People's Republic of China
| | - Yuangang Su
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, People's Republic of China; Collaborative Innovation Center of Regenerative Medicine and Medical Biological Resources Development and Application, Guangxi Medical University, Nanning, Guangxi, 530021, People's Republic of China
| | - Yansi Xian
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, People's Republic of China; Collaborative Innovation Center of Regenerative Medicine and Medical Biological Resources Development and Application, Guangxi Medical University, Nanning, Guangxi, 530021, People's Republic of China
| | - Jiyong Wei
- Department of Orthopedics, The First People's Hospital of Nanning, Nanning, Guangxi, 530016, People's Republic of China
| | - Jiake Xu
- School of Biomedical Sciences, University of Western Australia, Perth, WA, 6009, Australia
| | - Jinmin Zhao
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, People's Republic of China; Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, People's Republic of China
| | - Qian Liu
- Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, People's Republic of China.
| | - Yuan Yang
- Department of Orthopedics, Kaiyuan Langdong Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, 530028, People's Republic of China; Research Centre for Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, People's Republic of China.
| |
Collapse
|
22
|
Wang G, Ma C, Chen K, Wang Z, Qiu H, Chen D, He J, Zhang C, Guo D, Lai B, Zhang S, Huang L, Yang F, Yuan J, Chen L, He W, Xu J. Cycloastragenol Attenuates Osteoclastogenesis and Bone Loss by Targeting RANKL-Induced Nrf2/Keap1/ARE, NF-κB, Calcium, and NFATc1 Pathways. Front Pharmacol 2022; 12:810322. [PMID: 35126144 PMCID: PMC8812338 DOI: 10.3389/fphar.2021.810322] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 12/20/2021] [Indexed: 10/21/2023] Open
Abstract
Osteoporosis, which typically affects postmenopausal women, is an osteolytic disease due to over-activation of osteoclasts. However, current drugs targeting osteoclast inhibition face various side effects, making natural compounds with great interest as alternative treatment options. Cycloastragenol (CAG) is a triterpenoid with multiple biological activities. Previously, CAG's activity against aging-related osteoporosis was reported, but the mechanisms of actions for the activities were not understood. This study demonstrated that CAG dose-dependently inhibited osteoclast formation in receptor activator of nuclear factor-κB ligand (RANKL)-stimulated bone marrow macrophage (BMMs). Mechanism studies showed that CAG inhibited NF-κB, calcium, and nuclear factor of activated T cells 1 (NFATc1) pathways. Additionally, CAG also promoted the nuclear factor-erythroid 2-related factor 2 (Nrf2)/Kelch-like ECH-associated protein 1 (Keap1)/anti-oxidative response element (ARE) pathway that scavenges reactive oxygen species (ROS). Furthermore, CAG was also found to prevent bone loss of postmenopausal osteoporosis (PMO) in a preclinical model of ovariectomized (OVX) mice. Collectively, our research confirms that CAG inhibits the formation and function of osteoclasts by regulating RANKL-induced intracellular signaling pathways, which may represent a promising alternative for the therapy of osteoclast-related disease.
Collapse
Affiliation(s)
- Gang Wang
- First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangdong, China
- School of Biomedical Sciences, The University of WA, Perth, WA, Australia
- Guangzhou University of Chinese Medicine, Guangdong, China
| | - Chao Ma
- Guangzhou University of Chinese Medicine, Guangdong, China
| | - Kai Chen
- School of Biomedical Sciences, The University of WA, Perth, WA, Australia
| | - Ziyi Wang
- School of Biomedical Sciences, The University of WA, Perth, WA, Australia
| | - Heng Qiu
- School of Biomedical Sciences, The University of WA, Perth, WA, Australia
| | - Delong Chen
- School of Biomedical Sciences, The University of WA, Perth, WA, Australia
- Guangzhou University of Chinese Medicine, Guangdong, China
| | - Jianbo He
- School of Biomedical Sciences, The University of WA, Perth, WA, Australia
- Guangzhou University of Chinese Medicine, Guangdong, China
| | - Cheng Zhang
- Guangzhou University of Chinese Medicine, Guangdong, China
| | - Ding Guo
- Guangzhou University of Chinese Medicine, Guangdong, China
| | - Boyong Lai
- Guangzhou University of Chinese Medicine, Guangdong, China
| | | | - Linfeng Huang
- Guangzhou University of Chinese Medicine, Guangdong, China
| | - Fan Yang
- Guangzhou University of Chinese Medicine, Guangdong, China
| | - Jinbo Yuan
- School of Biomedical Sciences, The University of WA, Perth, WA, Australia
| | - Leilei Chen
- Guangzhou University of Chinese Medicine, Guangdong, China
- Third Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Wei He
- Guangzhou University of Chinese Medicine, Guangdong, China
- Third Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Jiake Xu
- School of Biomedical Sciences, The University of WA, Perth, WA, Australia
| |
Collapse
|
23
|
Petrova T, Bennett K, Nanda S, Strickson S, Scudamore CL, Prescott AR, Cohen P. Why are the phenotypes of TRAF6 knock-in and TRAF6 knock-out mice so different? PLoS One 2022; 17:e0263151. [PMID: 35157702 PMCID: PMC8843210 DOI: 10.1371/journal.pone.0263151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 01/12/2022] [Indexed: 11/30/2022] Open
Abstract
The expression of TNF-Receptor Associated Factor 6 (TRAF6) is essential for many physiological processes. Here we studied the phenotype of TRAF6[L74H] knock-in mice which are devoid of TRAF6 E3 ligase activity in every cell of the body, but express normal levels of the TRAF6 protein. Remarkably, TRAF6[L74H] mice have none of the phenotypes seen in TRAF6 KO mice. Instead TRAF6[L74H] mice display an entirely different phenotype, exhibiting autoimmunity, and severe inflammation of the skin and modest inflammation of the liver and lungs. Similar to mice with a Treg-specific knockout of TRAF6, or mice devoid of TRAF6 in all T cells, the CD4+ and CD8+ T cells in the spleen and lymph nodes displayed an activated effector memory phenotype with CD44high/CD62Llow expression on the cell surface. In contrast, T cells from WT mice exhibited the CD44low/CD62Lhigh phenotype characteristic of naïve T cells. The onset of autoimmunity and autoinflammation in TRAF6[L74H] mice (two weeks) was much faster than in mice with a Treg-specific knockout of TRAF6 or lacking TRAF6 expression in all T cells (2-3 months) and we discuss whether this may be caused by secondary inflammation of other tissues. The distinct phenotypes of mice lacking TRAF6 expression in all cells appears to be explained by their inability to signal via TNF Receptor Superfamily members, which does not seem to be impaired significantly in TRAF6[L74H] mice.
Collapse
Affiliation(s)
- Tsvetana Petrova
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Kyle Bennett
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom
- Division of Cell Signalling, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Sambit Nanda
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Sam Strickson
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | | | - Alan R. Prescott
- Dundee Imaging Facility and Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Philip Cohen
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, United Kingdom
- * E-mail:
| |
Collapse
|
24
|
Marino S, Hannemann N, Bishop RT, Zeng F, Carrasco G, Meurisse S, Li B, Sophocleous A, Sparatore A, Baeuerle T, Vukicevic S, Auberval M, Mollat P, Bozec A, Idris AI. Anti-inflammatory, but not osteoprotective, effect of the TRAF6/CD40 inhibitor 6877002 in rodent models of local and systemic osteolysis. Biochem Pharmacol 2021; 195:114869. [PMID: 34896056 DOI: 10.1016/j.bcp.2021.114869] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/02/2021] [Accepted: 12/02/2021] [Indexed: 12/19/2022]
Abstract
NFκB plays a key role in inflammation and skeletal disorders. Previously, we reported that pharmacological inhibition of NFκB at the level of TRAF6 suppressed RANKL, CD40L and IL1β-induced osteoclastogenesis and attenuated cancer-induced bone disease. TNFα is also known to regulate TRAF6/NFκB signalling, however the anti-inflammatory and osteoprotective effects associated with inhibition of the TNFα/TRAF6/NFκB axis have not been investigated. Here, we show that in vitro and ex vivo exposure to the verified small-molecule inhibitor of TRAF6, 6877002 prevented TNFα-induced NFκB activation, osteoclastogenesis and calvarial osteolysis, but it had no effects on TNFα-induced apoptosis or growth inhibition in osteoblasts. Additionally, 6877002 disrupted T-cells support for osteoclast formation and synoviocyte motility, without affecting the viability of osteoblasts in the presence of T-cells derived factors. Using the collagen-induced arthritis model, we show that oral and intraperitoneal administration of 6877002 in mice reduced joint inflammation and arthritis score. Unexpectedly, no difference in trabecular and cortical bone parameters were detected between vehicle and 6877002 treated mice, indicating lack of osteoprotection by 6877002 in the arthritis model described. Using two independent rodent models of osteolysis, we confirmed that 6877002 had no effect on trabecular and cortical bone loss in both osteoporotic rats or RANKL- treated mice. In contrast, the classic anti-osteolytic alendronate offered complete osteoprotection in RANKL- treated mice. In conclusion, TRAF6 inhibitors may be of value in the management of the inflammatory component of bone disorders, but may not offer protection against local or systemic bone loss, unless combined with anti-resorptive therapy such as bisphosphonates.
Collapse
Affiliation(s)
- Silvia Marino
- Department of Oncology and Metabolism, University of Sheffield, Medical School, Beech Hill Road, Sheffield S10 2RX, UK; Bone and Cancer Group, Edinburgh Cancer Research Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, EH4 2XR, UK
| | - Nicole Hannemann
- Department of Internal Medicine, 3-Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Ryan T Bishop
- Department of Oncology and Metabolism, University of Sheffield, Medical School, Beech Hill Road, Sheffield S10 2RX, UK
| | - Feier Zeng
- Department of Oncology and Metabolism, University of Sheffield, Medical School, Beech Hill Road, Sheffield S10 2RX, UK
| | - Giovana Carrasco
- Department of Oncology and Metabolism, University of Sheffield, Medical School, Beech Hill Road, Sheffield S10 2RX, UK
| | - Sandrine Meurisse
- Galapagos SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Boya Li
- Department of Oncology and Metabolism, University of Sheffield, Medical School, Beech Hill Road, Sheffield S10 2RX, UK
| | - Antonia Sophocleous
- Department of Life Sciences, School of Sciences, European University Cyprus, 6 Diogenes Street, 1516 Nicosia, Cyprus
| | - Anna Sparatore
- University of Milano, Department of Pharmaceutical Science, Milan, Italy
| | - Tobias Baeuerle
- Institute of Radiology, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum, Erlangen, Bayern, Germany
| | - Slobodan Vukicevic
- Department of Anatomy, Medical School, University of Zagreb, Zagreb, Croatia
| | - Marielle Auberval
- Galapagos SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Patrick Mollat
- Galapagos SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Aline Bozec
- Department of Internal Medicine, 3-Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Aymen I Idris
- Department of Oncology and Metabolism, University of Sheffield, Medical School, Beech Hill Road, Sheffield S10 2RX, UK; Bone and Cancer Group, Edinburgh Cancer Research Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, EH4 2XR, UK.
| |
Collapse
|
25
|
Tang MM, Chen YJ, Wu JY, Amin A, Zhou XM, Jin C, Chen GY, Yu ZL. Ethyl acetate extract of the Musa nana flower inhibits osteoclastogenesis and suppresses NF-κB and MAPK pathways. Food Funct 2021; 12:11586-11598. [PMID: 34713875 DOI: 10.1039/d1fo02204k] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Banana flowers are consumed as a vegetable and traditionally used for managing several health problems including joint pain, a symptom of bone loss. Osteoclasts are key effector cells responsible for bone loss. Some flavonoids in banana flowers, such as quercetin and quercitrin, have been shown to be able to inhibit osteoclastogenesis. Whether banana flowers can inhibit osteoclast formation is unknown. In this study, we prepared the ethyl acetate fraction (FFE-EA) of an ethanolic extract of fresh flowers of Musa nana. Using UPLC-MS/MS analyses, 76 polyphenols were identified in FFE-EA. In RANKL-stimulated RAW264.7 macrophages, FFE-EA inhibited osteoclastogenesis and osteoclastic bone resorption. Mechanistic studies revealed that FFE-EA suppressed NF-κB and MAPK pathways, and lowered mRNA levels of osteoclast formation/function-related genes. These findings suggest that flowers of M. nana could be a source for formulating functional food that benefits bone health.
Collapse
Affiliation(s)
- Min-Min Tang
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, 571158, China. .,Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, 999077, China. .,Coconut Research Institute of Chinese Academy of Tropical Agricultural Sciences, Wenchang, Hainan, 571339, China.,Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, 571158, China
| | - Ying-Jie Chen
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, 999077, China.
| | - Jia-Ying Wu
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, 999077, China.
| | - Aftab Amin
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, 999077, China.
| | - Xue-Ming Zhou
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, 571158, China. .,Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, 571158, China
| | - Cai Jin
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, 571158, China. .,Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, 571158, China
| | - Guang-Ying Chen
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, 571158, China. .,Key Laboratory of Tropical Medicinal Plant Chemistry of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, 571158, China
| | - Zhi-Ling Yu
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, 999077, China.
| |
Collapse
|
26
|
Xu Q, Zhan P, Li X, Mo F, Xu H, Liu Y, Lai Q, Zhang B, Dai M, Liu X. Bisphosphonate-enoxacin inhibit osteoclast formation and function by abrogating RANKL-induced JNK signalling pathways during osteoporosis treatment. J Cell Mol Med 2021; 25:10126-10139. [PMID: 34651433 PMCID: PMC8572771 DOI: 10.1111/jcmm.16949] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 07/02/2021] [Accepted: 09/19/2021] [Indexed: 01/23/2023] Open
Abstract
Osteoporosis is an age‐related disease characterized by low mineral density, compromised bone strength and increased risk of fragility fracture. Most agents for treating osteoporosis focus primarily on anti‐resorption by inhibiting osteoclast activity. Bisphosphonate (BP) is a potent anti‐resorptive agent that has been used clinically for decades and is proven to be effective. However, BP has a variety of side effects and is far from being an ideal anti‐osteoporosis agent. BP selectively binds to calcium crystals, which are subsequently taken up or released by osteoclasts. Based on the action of BP, we previously demonstrated the inhibitory effect of a novel bone‐targeting BP derivative, bisphosphonate‐enoxacin (BE). In the current study, we used bone marrow‐derived osteoclast cultures to further assess the inhibitory effect of BE on osteoclastogenesis and employed reverse transcription PCR and real‐time PCR to examine expression of osteoclast‐specific genes. Additionally, we used bone resorption and F‐actin immunofluorescence assays to evaluate the effect of BE on osteoclast function and investigated the potential mechanisms affecting osteoclast differentiation and function in vitro. Furthermore, an ovariectomized (OVX) rat model was established to evaluate the therapeutic effects of BE on preventing bone loss. Results showed that BE exerted potent inhibitory effects on osteoclast formation and bone resorption by specifically abrogating RANKL‐induced JNK signalling, and that it preserved OVX rat bone mass in vivo without any notable side effects. Collectively, these results indicated that the BP derivative BE may have significant potential as a treatment for osteoporosis and other osteolytic diseases.
Collapse
Affiliation(s)
- Qiang Xu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| | - Ping Zhan
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| | - Xiaofeng Li
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| | - Fengbo Mo
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| | - Huaen Xu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| | - Yuan Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| | - Qi Lai
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| | - Bin Zhang
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| | - Min Dai
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| | - Xuqiang Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| |
Collapse
|
27
|
Chellaiah MA. L-Plastin Phosphorylation: Possible Regulation by a TNFR1 Signaling Cascade in Osteoclasts. Cells 2021; 10:2432. [PMID: 34572081 PMCID: PMC8464874 DOI: 10.3390/cells10092432] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/25/2021] [Accepted: 09/09/2021] [Indexed: 12/30/2022] Open
Abstract
Tumor necrosis factor-alpha (TNF-α) signaling regulates phosphorylation of L-plastin, which is involved in forming the nascent sealing zone, a precursor zone for the matured sealing ring. This study aimed to illustrate the molecular mechanisms of L-plastin phosphorylation and the subsequent formation of the nascent sealing zone in osteoclasts treated with TNF-α. Here, we report that anti-TNF-receptor 1, inhibitors of signaling proteins (Src, PI3-K, Rho, and Rho-kinase), and siRNA of TRAF-6 attenuated the phosphorylation of LPL and filamentous actin content significantly in the presence of TNF-α. An inhibitor of integrin αvβ3, PKC, or PKA did not inhibit TNF-α-induced L-plastin phosphorylation. Inhibitors of Src and PI3-K and not Rho or Rho-kinase reduced tyrosine phosphorylation of TRAF-6, suggesting that Src and PI3-K regulate TRAF-6 phosphorylation, and Rho and Rho-kinase are downstream of TRAF-6 regulation. Osteoclasts expressing constitutively active or kinase-defective Src proteins were used to determine the role of Src on L-plastin phosphorylation; similarly, the effect of Rho was confirmed by transducing TAT-fused constitutively active (V14) or dominant-negative (N19) Rho proteins into osteoclasts. Pull-down analysis with glutathione S-transferase-fused SH2 and SH3 domains of Src and PI3-K demonstrated coprecipitation of L-plastin and TRAF-6 with the SH3 and SH2 domains of the PI3-K and Src proteins. However, the actual order of the interaction of proteins requires further elucidation; a comprehensive screening should corroborate the initial findings of protein interactions via the SH2/SH3 domains. Ultimately, inhibition of the interaction of proteins with SH2/SH3 could reduce L-plastin phosphorylation and affect NSZ formation and bone resorption in conditions that display osteoclast activation and bone loss.
Collapse
Affiliation(s)
- Meenakshi A Chellaiah
- Department of Oncology and Diagnostic Sciences, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA
| |
Collapse
|
28
|
Liang X, Hou Y, Han L, Yu S, Zhang Y, Cao X, Yan J. ELMO1 Regulates RANKL-Stimulated Differentiation and Bone Resorption of Osteoclasts. Front Cell Dev Biol 2021; 9:702916. [PMID: 34381782 PMCID: PMC8350380 DOI: 10.3389/fcell.2021.702916] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/30/2021] [Indexed: 11/20/2022] Open
Abstract
Bone homeostasis is a metabolic balance between the new bone formation by osteoblasts and old bone resorption by osteoclasts. Excessive osteoclastic bone resorption results in low bone mass, which is the major cause of bone diseases such as rheumatoid arthritis. Small GTPases Rac1 is a key regulator of osteoclast differentiation, but its exact mechanism is not fully understood. ELMO and DOCK proteins form complexes that function as guanine nucleotide exchange factors for Rac activation. Here, we report that ELMO1 plays an important role in differentiation and bone resorption of osteoclasts. Osteoclast precursors derived from bone marrow monocytes (BMMs) of Elmo1–/– mice display defective adhesion and migration during differentiation. The cells also have a reduced activation of Rac1, p38, JNK, and AKT in response to RANKL stimulation. Importantly, we show that bone erosion is alleviated in Elmo1–/– mice in a rheumatoid arthritis mouse model. Taken together, our results suggest that ELMO1, as a regulator of Rac1, regulates osteoclast differentiation and bone resorption both in vitro and in vivo.
Collapse
Affiliation(s)
- Xinyue Liang
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Yafei Hou
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lijuan Han
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Shuxiang Yu
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Yunyun Zhang
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Xiumei Cao
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianshe Yan
- School of Life Sciences, Shanghai University, Shanghai, China.,Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
29
|
Methods to Analyze the Roles of TAK1, TRAF6, and NEMO in the Regulation of NF-κB Signaling by RANK Stimulation During Osteoclastogenesis. Methods Mol Biol 2021. [PMID: 34236644 DOI: 10.1007/978-1-0716-1669-7_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
The skeletal system is constantly undergoing turnover in order to create strong, organized structures, requiring the bone breakdown and building properties by osteoclasts and osteoblasts, respectively. However, in pathological disease states, excessive osteoclast activity can cause bone loss leading to increase in morbidity and mortality. Osteoclasts differentiate from macrophages in the presence of various factors. M-CSF is a cytokine that is required to maintain the survival of macrophages. However, RANKL is the critical factor required for differentiation of osteoclasts. RANKL is produced from a variety of different cell types such as osteoblasts and osteocytes. RANKL binds to RANK, its receptor, on the surface of osteoclast precursors, which activates various signaling pathways to drive the transcription and production of genes important for osteoclast formation. The major signaling pathway activated by RANKL-RANK interaction is the NF-κB pathway. The NF-κB pathway is the principle inflammatory response pathway activated by a variety of stimuli such as inflammatory cytokines, genotoxic stress, and other factors. This likely explains the finding that inflammatory diseases often present with some component of increased osteoclast formation and activity, driving bone loss. Determining the signaling mechanisms downstream of RANKL can provide valuable therapeutic targets for the treatment of bone loss in various disease states.
Collapse
|
30
|
Wang K, Chen Y, Gao S, Wang M, Ge M, Yang Q, Liao M, Xu L, Chen J, Zeng Z, Chen H, Zhang XK, Lin T, Zhou H. Norlichexanthone purified from plant endophyte prevents postmenopausal osteoporosis by targeting ER α to inhibit RANKL signaling. Acta Pharm Sin B 2021; 11:442-455. [PMID: 33643823 PMCID: PMC7893202 DOI: 10.1016/j.apsb.2020.09.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 07/28/2020] [Accepted: 08/21/2020] [Indexed: 12/20/2022] Open
Abstract
Although different types of drugs are available for postmenopausal osteoporosis, the limitations of the current therapies including drug resistances and adverse effects require identification of novel anti-osteoporosis agents. Here, we defined that norlichexanthone (NOR), a natural product, is a ligand of estrogen receptor-alpha (ERα) and revealed its therapeutic potential for postmenopausal osteoporosis. We used mammalian-one hybrid assay to screen for ERα modulators from crude extracts of several plant endophytes. As a result, NOR purified from the extract of endophyte ARL-13 was identified as a selective ERα modulator. NOR directly bound to ERα with an affinity in nanomolar range, revealing that it is a natural ligand of ERα. NOR induced osteoblast formation in MC3T3-E1 precursor cells. Conversely, NOR inhibited receptor activator of nuclear factor-kappa B ligand (RANKL)-induced osteoclast formation in both RAW264.7 macrophages and mouse primary monocytes. Mechanistically, NOR inhibited RANKL-induced association of ERα and TRAF6 to prevent ERα-mediated TRAF6 activation via Lys63-linked ubiquitination. Importantly, NOR exhibited potent anti-osteoporosis efficacy in an ovariectomized mouse model. Comparing to estrogen, NOR was of much less capability in stimulating endometrial hyperplasia and promoting mammalian cancer cell proliferation. Taken together, our study identified NOR as a natural and high affinity ligand of ERα with substantial anti-osteoporosis but less estrogenic activity.
Collapse
Affiliation(s)
- Keqi Wang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Yongyan Chen
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Shuo Gao
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Maosi Wang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Mengmeng Ge
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Qian Yang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Mingkai Liao
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Lin Xu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Junjie Chen
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
- High Throughput Drug Screening Platform, Xiamen University, Xiamen 361102, China
| | - Zhiping Zeng
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
- High Throughput Drug Screening Platform, Xiamen University, Xiamen 361102, China
| | - Haifeng Chen
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
| | - Xiao-kun Zhang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
- High Throughput Drug Screening Platform, Xiamen University, Xiamen 361102, China
| | - Ting Lin
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
- Corresponding authors. Tel.: +86 592 2881105; fax: +86 592 2881105.
| | - Hu Zhou
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361102, China
- High Throughput Drug Screening Platform, Xiamen University, Xiamen 361102, China
- Corresponding authors. Tel.: +86 592 2881105; fax: +86 592 2881105.
| |
Collapse
|
31
|
Chen Z, Ding M, Cho E, Seong J, Lee S, Lee TH. 2-NPPA Mitigates Osteoclastogenesis via Reducing TRAF6-Mediated c-fos Expression. Front Pharmacol 2021; 11:599081. [PMID: 33574753 PMCID: PMC7870508 DOI: 10.3389/fphar.2020.599081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 12/21/2020] [Indexed: 11/23/2022] Open
Abstract
Excessive bone resorption leads to bone destruction in pathological bone diseases. Osteoporosis, which occurs when osteoclast-mediated bone resorption exceeds osteoblast-mediated bone synthesis, is regarded a global health challenge. Therefore, it is of great importance to identify agents that can regulate the activity of osteoclasts and prevent bone diseases mediated mainly by bone loss. We screened compounds for this purpose and found that 2-(2-chlorophenoxy)-N-[2-(4-propionyl-1piperazinyl) phenyl] acetamide (2-NPPA) exhibited a strong inhibitory effect on osteoclastogenesis. 2-NPPA suppressed the mRNA and protein expression of several osteoclast-specific markers and blocked the formation of mature osteoclasts, reducing the F-actin ring formation and bone resorption activity. In a cell signaling point of view, 2-NPPA exhibited a significant inhibitory effect on the phosphorylation of nuclear factor kappa-B (NF-κB) and c-fos expression in vitro and prevented ovariectomy-induced bone loss in vivo. These findings highlighted the potential of 2-NPPA as a drug for the treatment of bone loss-mediated disorders.
Collapse
Affiliation(s)
- Zhihao Chen
- Department of Molecular Medicine, Chonnam National University Graduate School, Gwangju, South Korea
| | - Mina Ding
- Department of Molecular Medicine, Chonnam National University Graduate School, Gwangju, South Korea
| | - Eunjin Cho
- Department of Oral Biochemistry, Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, South Korea
| | - Jihyoun Seong
- Department of Oral Biochemistry, Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, South Korea
| | - Sunwoo Lee
- Department of Chemistry, Chonnam National University, Gwangju, South Korea
| | - Tae-Hoon Lee
- Department of Molecular Medicine, Chonnam National University Graduate School, Gwangju, South Korea.,Department of Oral Biochemistry, Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, South Korea
| |
Collapse
|
32
|
Al‐Bari AA, Al Mamun A. Current advances in regulation of bone homeostasis. FASEB Bioadv 2020; 2:668-679. [PMID: 33205007 PMCID: PMC7655096 DOI: 10.1096/fba.2020-00058] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 08/18/2020] [Accepted: 08/21/2020] [Indexed: 02/06/2023] Open
Abstract
Bone homeostasis is securely controlled by the dynamic well-balanced actions among osteoclasts, osteoblasts and osteocytes. Osteoclasts are large multinucleated cells that degrade bone matrix and involve in the bone remodelling in conjunction with other bone cells, osteoblasts and osteocytes, the completely matured form of osteoblasts. Disruption of this controlling balance among these cells or any disparity in bone remodelling caused by a higher rate of resorption by osteoclasts over construction of bone by osteoblasts results in a reduction of bone matrix including bone mineral density (BMD) and bone marrow cells (BMCs). The dominating effect of osteoclasts results in advanced risk of bone crack and joint destruction in several diseases including osteoporosis and rheumatoid arthritis (RA). However, the boosted osteoblastic activity produces osteosclerotic phenotype and weakened its action primes to osteomalacia or rickets. On the other hand, senescent osteocytes predominately progress the senescence associated secretory phenotype (SASP) and may contribute to age related bone loss. Here, we discuss an advanced level work on newly identified cellular mechanisms controlling the remodelling of bone and crosstalk among bone cells as these relate to the therapeutic targeting of the skeleton.
Collapse
Affiliation(s)
| | - Abdullah Al Mamun
- Department of Genetic Engineering and BiotechnologyShahjalal University of Science and TechnologySylhetBangladesh
| |
Collapse
|
33
|
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
|
34
|
Kim EN, Kwon J, Lee HS, Lee S, Lee D, Jeong GS. Inhibitory Effect of Cudratrixanthone U on RANKL-Induced Osteoclast Differentiation and Function in Macrophages and BMM Cells. Front Pharmacol 2020; 11:1048. [PMID: 32848726 PMCID: PMC7419678 DOI: 10.3389/fphar.2020.01048] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/26/2020] [Indexed: 12/19/2022] Open
Abstract
Cudratrixanthone U (CTU) is a prenylated xanthone compound isolated from Maclura tricuspidata Bureau (Moraceae). Prenylated xanthones have been reported to exhibit a variety of biological activities. However, the effects of prenylated xanthone on osteoclast differentiation and function are still unclear. Excessive bone resorption by osteoclasts is considered a major cause of diseases such as osteoporosis. Accordingly, suppression of excessive osteoclast formation and function is one of strategies for treating osteoclast related bone diseases. In this study, CTU inhibited osteoclast differentiation and function in RAW264.7 macrophages and BMM cells induced by receptor activator of nuclear factor-κB ligand (RANKL). CTU regulated the formation of TRAF6-TAK1 complex in RANKL-induced RAW264.7 macrophages and BMM cells. Osteoclast-specific genes including those encoding matrix metallopeptidase 9 (MMP-9), dendritic cell-specific transmembrane proteins (DC-STAMP), cathepsin K (CTSK) and chemokine CC motif ligand 4 (CCL4) play an important role in bone resorption and migration, and were effectively regulated by CTU. These results suggest that CTU is a potential therapeutic agent in osteoporosis.
Collapse
Affiliation(s)
- Eun-Nam Kim
- College of Pharmacy, Keimyung University, Daegu, South Korea
| | - Jaeyoung Kwon
- Natural Constituents Research Center, Korea Institute of Science and Technology (KIST), Gangneung, South Korea
| | - Hyun-Su Lee
- College of Pharmacy, Keimyung University, Daegu, South Korea
| | - Sooyeun Lee
- College of Pharmacy, Keimyung University, Daegu, South Korea
| | - Dongho Lee
- Department of Biosystems and Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, South Korea
| | - Gil-Saeng Jeong
- College of Pharmacy, Keimyung University, Daegu, South Korea
| |
Collapse
|
35
|
Combined administration of a small-molecule inhibitor of TRAF6 and Docetaxel reduces breast cancer skeletal metastasis and osteolysis. Cancer Lett 2020; 488:27-39. [PMID: 32474152 DOI: 10.1016/j.canlet.2020.05.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/08/2020] [Accepted: 05/18/2020] [Indexed: 12/20/2022]
Abstract
Tumour necrosis factor receptor-associated factor 6 (TRAF6) has been implicated in breast cancer and osteoclastic bone destruction. Here, we report that 6877002, a verified small-molecule inhibitor of TRAF6, reduced metastasis, osteolysis and osteoclastogenesis in models of osteotropic human and mouse breast cancer. First, we observed that TRAF6 is highly expressed in osteotropic breast cancer cells and its level of expression was higher in patients with bone metastasis. Pre-exposure of osteoclasts and osteoblasts to non-cytotoxic concentrations of 6877002 inhibited cytokine-induced NFκB activation and osteoclastogenesis, and reduced the ability of osteotropic human MDA-MB-231 and mouse 4T1 breast cancer cells to support bone cell activity. 6877002 inhibited human MDA-MB-231-induced osteolysis in the mouse calvaria organ system, and reduced soft tissue and bone metastases in immuno-competent mice following intra-cardiac injection of mouse 4T1-Luc2 cells. Of clinical relevance, combined administration of 6877002 with Docetaxel reduced metastasis and inhibited osteolytic bone damage in mice bearing 4T1-Luc2 cells. Thus, TRAF6 inhibitors such as 6877002 - alone or in combination with conventional chemotherapy - show promise for the treatment of metastatic breast cancer.
Collapse
|
36
|
Liu L, Jin R, Duan J, Yang L, Cai Z, Zhu W, Nie Y, He J, Xia C, Gong Q, Song B, Anderson JM, Ai H. Bioactive iron oxide nanoparticles suppress osteoclastogenesis and ovariectomy-induced bone loss through regulating the TRAF6-p62-CYLD signaling complex. Acta Biomater 2020; 103:281-292. [PMID: 31866569 DOI: 10.1016/j.actbio.2019.12.022] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 11/21/2019] [Accepted: 12/17/2019] [Indexed: 02/05/2023]
Abstract
Iron oxide nanoparticles (IONPs) have been widely used as contrast agents for magnetic resonance imaging (MRI) and other biomedical applications in both clinical and preclinical cases. In the present study, we show that two clinically used IONPs, ferumoxytol and ferucarbotran, have an intrinsic inhibitory effect on receptor activator NF-κB ligand (RANKL)-induced osteoclastogenesis of bone marrow-derived monocytes/macrophages (BMMs). IONPs significantly inhibited the formation of tartrate-resistant acid phosphatase (TRAP)-positive multinuclear osteoclasts and functional actin ring structures. More importantly, the inhibitory effect was also verified in vivo by its capacity to rescue the bone loss of ovariectomized (OVX) mice after intravenous injection with IONPs. Mechanistically, we found that IONPs trigger the upregulation of p62 which result in recruitment of CYLD and enhanced deubiquitination of TRAF6, a master controller of RANKL signaling. The downstream activation of NF-κB and MAPK signals was accordingly attenuated, ultimately leading to reduced expression of osteoclatogenesis-related genes. Taken together, clinically used IONPs can inhibit osteoclastogenesis through regulating TRAF6-p62-CYLD signaling complex, and they may be considered as alternative options for treatment of osteoporosis. STATEMENT OF SIGNIFICANCE: Nanoparticles have been developed as drug delivery systems for treatment of osteoporosis, mostly an age-related health problem with risk of fractures. In this work, we show that two clinically used iron oxide nanoparticles (IONPs) ferumoxytol and ferucarbotran themselves can significantly reduce the osteoporosis of ovariectomized (OVX) mice through inhibiting Osteoclastogenesis. We found that IONPs trigger the upregulation of p62 which result in recruitment of CYLD and enhanced deubiquitination of TRAF6, a master controller of RANKL signaling. The downstream activation of NF-κB and MAPK signals was accordingly attenuated, leading to reduced expression of osteoclatogenesis-related genes. Taken together, clinically used IONPs inhibit osteoclastogenesis through regulating TRAF6-p62-CYLD signaling complex, and they may be considered as alternative options for treatment of osteoporosis.
Collapse
Affiliation(s)
- Li Liu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Rongrong Jin
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China.
| | - Jimei Duan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Li Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Zhongyuan Cai
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Wencheng Zhu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China; Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yu Nie
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Jing He
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Chunchao Xia
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiyong Gong
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - James M Anderson
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA; Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Hua Ai
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China; Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
37
|
Al-Bari MAA, Hossain S, Mia U, Al Mamun MA. Therapeutic and Mechanistic Approaches of Tridax Procumbens Flavonoids for the Treatment of Osteoporosis. Curr Drug Targets 2020; 21:1687-1702. [PMID: 32682372 DOI: 10.2174/1389450121666200719012116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 11/22/2022]
Abstract
Homeostasis of bone is closely regulated by the balanced activities between the bone resorbing activity of osteoclast cells and bone-forming ability of osteoblast cells. Multinucleated osteoclasts degrade bone matrix and involve in the dynamic bone remodelling in coordination with osteoblasts. Disruption of this regulatory balance between these cells or any imbalance in bone remodelling caused by a higher rate of resorption over construction of bone results in a decrease of bone matrix including bone mineral density (BMD). These osteoclast-dominant effects result in a higher risk of bone crack and joint demolition in several bone-related diseases, including osteoporosis and rheumatoid arthritis (RA). Tridax procumbens is a very interesting perennial plant and its secondary metabolites called here T. procumbens flavonoids (TPFs) are well-known phytochemical agents owing to various therapeutic practices such as anti-inflammatory, anti-anaemic and anti-diabetic actions. This review designed to focus the systematic convention concerning the medicinal property and mechanism of actions of TPFs for the management of bone-related diseases. Based on the current literature, the review offers evidence-based information of TPFs for basic researchers and clinicians for the prevention and treatment of bone related diseases, including osteoporosis. It also emphasizes the medical significance for more research to comprehend the cellular signalling pathways of TPFs for the regulation of bone remodelling and discusses the possible promising ethnobotanical resource that can convey the preclinical and clinical clues to develop the next generation therapeutic agents for the treatment of bonerelated disorders.
Collapse
Affiliation(s)
| | - Showna Hossain
- Department of Pharmacy, University of Rajshahi, Rajshahi-6205, Bangladesh
| | - Ujjal Mia
- Department of Pharmacy, University of Rajshahi, Rajshahi-6205, Bangladesh
| | - Md Abdullah Al Mamun
- Department of Genetic Engineering and Biotechnology, Shahjalal University of Science and Technology, Sylhet-3114, Bangladesh
| |
Collapse
|
38
|
Jin H, Wang Q, Chen K, Xu K, Pan H, Chu F, Ye Z, Wang Z, Tickner J, Qiu H, Wang C, Kenny J, Xu H, Wang T, Xu J. Astilbin prevents bone loss in ovariectomized mice through the inhibition of RANKL-induced osteoclastogenesis. J Cell Mol Med 2019; 23:8355-8368. [PMID: 31603626 PMCID: PMC6850941 DOI: 10.1111/jcmm.14713] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 07/27/2019] [Accepted: 08/07/2019] [Indexed: 01/01/2023] Open
Abstract
Osteoporosis is the most common osteolytic disease characterized by excessive osteoclast formation and resultant bone loss, which afflicts millions of patients around the world. Astilbin, a traditional herb, is known to have anti-inflammatory, antioxidant and antihepatic properties, but its role in osteoporosis treatment has not yet been confirmed. In our study, astilbin was found to have an inhibitory effect on the RANKL-induced formation and function of OCs in a dose-dependent manner without cytotoxicity. These effects were attributed to its ability to suppress the activity of two transcription factors (NFATc1 and c-Fos) indispensable for osteoclast formation, followed by inhibition of the expression of bone resorption-related genes and proteins (Acp5/TRAcP, CTSK, V-ATPase-d2 and integrin β3). Furthermore, we examined the underlying mechanisms and found that astilbin repressed osteoclastogenesis by blocking Ca2+ oscillations and the NF-κB and MAPK pathways. In addition, the therapeutic effect of MA on preventing bone loss in vivo was further confirmed in an ovariectomized mouse model. Therefore, considering its ability to inhibit RANKL-mediated osteoclastogenesis and the underlying mechanisms, astilbin might be a potential candidate for treating osteolytic bone diseases.
Collapse
Affiliation(s)
- Haiming Jin
- Key Laboratory of Orthopaedics of Zhejiang ProvinceThe Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
- School of Biomedical SciencesThe University of Western AustraliaPerthWAAustralia
| | - Qingqing Wang
- Key Laboratory of Orthopaedics of Zhejiang ProvinceThe Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
- School of Biomedical SciencesThe University of Western AustraliaPerthWAAustralia
| | - Kai Chen
- School of Biomedical SciencesThe University of Western AustraliaPerthWAAustralia
| | - Ke Xu
- Key Laboratory of Orthopaedics of Zhejiang ProvinceThe Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Hao Pan
- Key Laboratory of Orthopaedics of Zhejiang ProvinceThe Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Feifan Chu
- Key Laboratory of Orthopaedics of Zhejiang ProvinceThe Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Zhen Ye
- Key Laboratory of Orthopaedics of Zhejiang ProvinceThe Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Ziyi Wang
- School of Biomedical SciencesThe University of Western AustraliaPerthWAAustralia
| | - Jennifer Tickner
- School of Biomedical SciencesThe University of Western AustraliaPerthWAAustralia
| | - Heng Qiu
- School of Biomedical SciencesThe University of Western AustraliaPerthWAAustralia
| | - Chao Wang
- School of Biomedical SciencesThe University of Western AustraliaPerthWAAustralia
| | - Jacob Kenny
- School of Biomedical SciencesThe University of Western AustraliaPerthWAAustralia
| | - Huazi Xu
- Key Laboratory of Orthopaedics of Zhejiang ProvinceThe Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Te Wang
- Key Laboratory of Orthopaedics of Zhejiang ProvinceThe Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Jiake Xu
- Key Laboratory of Orthopaedics of Zhejiang ProvinceThe Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
- School of Biomedical SciencesThe University of Western AustraliaPerthWAAustralia
| |
Collapse
|
39
|
Wang Q, Yao L, Xu K, Jin H, Chen K, Wang Z, Liu Q, Cao Z, kenny J, Liu Y, Tickner J, Xu H, Xu J. Madecassoside inhibits estrogen deficiency-induced osteoporosis by suppressing RANKL-induced osteoclastogenesis. J Cell Mol Med 2019; 23:380-394. [PMID: 30338925 PMCID: PMC6307845 DOI: 10.1111/jcmm.13942] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 09/10/2018] [Accepted: 09/11/2018] [Indexed: 12/31/2022] Open
Abstract
Osteoporosis is the most common osteolytic disease characterized by excessive osteoclast formation and resultant bone loss, which afflicts millions of patients around the world. Madecassoside (MA), isolated from Centella asiatica, was reported to have anti-inflammatory and antioxidant activities, but its role in osteoporosis treatment has not yet been confirmed. In our study, MA was found to have an inhibitory effect on the RANKL-induced formation and function of OCs in a dose-dependent manner without cytotoxicity. These effects were attributed to its ability to suppress the activity of two transcription factors (NFATc1 and c-Fos) indispensable for osteoclast formation, followed by inhibition of the expression of bone resorption-related genes and proteins (Acp5/TRAcP, CTSK, ATP6V0D2/V-ATPase-d2, and integrin β3). Furthermore, we examined the underlying mechanisms and found that MA represses osteoclastogenesis by blocking Ca2+ oscillations and the NF-κB and MAPK pathways. In addition, the therapeutic effect of MA on preventing bone loss in vivo was further confirmed in an ovariectomized mouse model. Therefore, considering its ability to inhibit RANKL-mediated osteoclastogenesis and the underlying mechanisms, MA might be a potential candidate for treating osteolytic bone diseases.
Collapse
Affiliation(s)
- Qingqing Wang
- Department of OrthopaedicsThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityZhejiangChina
- School of Biomedical SciencesThe University of Western AustraliaPerthWestern AustraliaAustralia
| | - Lingya Yao
- Department of OrthopaedicsThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityZhejiangChina
| | - Ke Xu
- Department of OrthopaedicsThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityZhejiangChina
| | - Haiming Jin
- Department of OrthopaedicsThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityZhejiangChina
- School of Biomedical SciencesThe University of Western AustraliaPerthWestern AustraliaAustralia
| | - Kai Chen
- School of Biomedical SciencesThe University of Western AustraliaPerthWestern AustraliaAustralia
| | - Ziyi Wang
- School of Biomedical SciencesThe University of Western AustraliaPerthWestern AustraliaAustralia
| | - Qian Liu
- Research Centre for Regenerative Medicine and Guangxi Key Laboratory of Regenerative MedicineGuangxi Medical UniversityGuangxiChina
| | - Zhen Cao
- Department of Biomedical Materials ScienceThird Military Medical UniversityChongqingChina
| | - Jacob kenny
- School of Biomedical SciencesThe University of Western AustraliaPerthWestern AustraliaAustralia
| | - Yuhao Liu
- School of Biomedical SciencesThe University of Western AustraliaPerthWestern AustraliaAustralia
- The Lab of Orthopaedics and Traumatology of Lingnan Medical Research CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Jennifer Tickner
- School of Biomedical SciencesThe University of Western AustraliaPerthWestern AustraliaAustralia
| | - Huazi Xu
- Department of OrthopaedicsThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityZhejiangChina
| | - Jiake Xu
- Department of OrthopaedicsThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityZhejiangChina
- School of Biomedical SciencesThe University of Western AustraliaPerthWestern AustraliaAustralia
| |
Collapse
|
40
|
Kim B, Lee KY, Park B. Icariin abrogates osteoclast formation through the regulation of the RANKL-mediated TRAF6/NF-κB/ERK signaling pathway in Raw264.7 cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 51:181-190. [PMID: 30466615 DOI: 10.1016/j.phymed.2018.06.020] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 04/13/2018] [Accepted: 06/18/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Icariin is pharmacologically active prenylated flavonoid glycoside that has various biologic effects such as antioxidant, anticancer, and anti-inflammatory activities. In addition, icariin has been used in Chinese medicine for thousands of years to treat osteoporosis and it is still being used today. However, direct mechanism of icariin in the treatment of bone disease is not understood. PURPOSE The purpose of this study is to investigate whether icariin influences RANKL-induced osteoclast formation in murine macrophages. METHODS Osteoclastogenesis was determined by TRAP staining and activity assay. Inhibition of signaling pathways and marker protein expression were evaluated by western blot analysis. The NF-κB (p65) nuclear localization was detected by immunofluorescence assay, and NF-κB/DNA-binding activity was detected by electrophoretic mobility shift assay (EMSA). RESULTS In our study, icariin inhibited the differentiation of pre-osteoclast cells into osteoclasts and suppressed expression of various genes involved in osteoclast formation and bone resorption. Also, icariin blocked the osteoclastogenesis induced by MCF7 and MDA-MB-231 breast cancer cells through inhibition of NF-κB activation. We found that icariin inhibited RANKL-stimulated TRAF-6 expression, and subsequently suppressed the phosphorylation of ERK, but icariin did not show an effect on p38, JNK, and Akt activation. CONCLUSION These results indicate that icariin is likely to be a candidate for bone-related disease treatment and that icariin provides insights into the molecular mechanisms that influence RANKL-induced osteoclast differentiation.
Collapse
Affiliation(s)
- Buyun Kim
- College of Pharmacy, Keimyung University, 1095 Dalgubeoldaero, Dalseo-Gu, Daegu 704-701, Republic of Korea
| | - Ki Yong Lee
- College of Pharmacy, Korea University, Sejong Campus 2511 Sejong-ro, Sejong City 339-770, Republic of Korea.
| | - Byoungduck Park
- College of Pharmacy, Keimyung University, 1095 Dalgubeoldaero, Dalseo-Gu, Daegu 704-701, Republic of Korea.
| |
Collapse
|
41
|
Kim H, Lee S, Lee SW. TRAF6 Distinctly Regulates Hematopoietic Stem and Progenitors at Different Periods of Development in Mice. Mol Cells 2018; 41:753-761. [PMID: 30037215 PMCID: PMC6125416 DOI: 10.14348/molcells.2018.0191] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 06/14/2018] [Accepted: 06/19/2018] [Indexed: 12/27/2022] Open
Abstract
Tumor necrosis factor receptor-associated factor 6 (TRAF6) is identified as a signaling adaptor protein that regulates bone metabolism, immunity, and the development of several tissues. Therefore, its functions are closely associated with multiple diseases. TRAF6 is also involved in the regulation of hematopoiesis under steady-state conditions, but the role of TRAF6 in modulating hematopoietic stem and progenitor cells (HSPCs) during the developmental stages remains unknown. Here, we report that the deletion of TRAF6 in hematopoietic lineage cells resulted in the upregulation of HSPCs in the fetal liver at the prenatal period. However, in the early postnatal period, deletion of TRAF6 drastically diminished HSPCs in the bone marrow (BM), with severe defects in BM development and extramedullary hematopoiesis in the spleen being identified. In the analysis of adult HSPCs in a BM reconstitution setting, TRAF6 played no significant role in HSPC homeostasis, albeit it affected the development of T cells. Taken together, our results suggest that the role of TRAF6 in regulating HSPCs is altered in a spatial and temporal manner during the developmental course of mice.
Collapse
Affiliation(s)
- Hyekang Kim
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang 37673,
Korea
| | - Seungwon Lee
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang 37673,
Korea
| | - Seung-Woo Lee
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang 37673,
Korea
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673,
Korea
| |
Collapse
|
42
|
Song D, Cao Z, Tickner J, Qiu H, Wang C, Chen K, Wang Z, Guo C, Dong S, Xu J. Poria cocos polysaccharide attenuates RANKL-induced osteoclastogenesis by suppressing NFATc1 activity and phosphorylation of ERK and STAT3. Arch Biochem Biophys 2018; 647:76-83. [PMID: 29678628 DOI: 10.1016/j.abb.2018.04.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 03/23/2018] [Accepted: 04/16/2018] [Indexed: 11/17/2022]
Abstract
Pathological fractures caused by osteolytic lesions seriously threaten the health of patients. Osteoclasts play important roles in bone resorption whose hyperfunction are closely related to osteolytic lesions. Studies on osteoclast differentiation and function assist in the prevention of excessive bone loss associated diseases. We screened a variety of natural compounds with anti-inflammatory effect and found that poria cocos polysaccharide (PCP) inhibited RANKL-induced osteoclast formation and bone resorption via TRAcP staining, immunofluorescence, RT-PCR and western blot. PCP down-regulated phosphorylation of STAT3, P38, ERK and JNK, and thus repressed the expression of NFAcT1 and c-Fos during RANKL-induced osteoclastogenesis. Besides, the expression of bone resorption related genes such as TRAcP and CTSK was suppressed by PCP. The results suggest that PCP can be invoked as a candidate for the treatment of osteolytic diseases by inhibiting osteoclastogenesis.
Collapse
Affiliation(s)
- Dezhi Song
- Department of Microbiology, Guangxi Medical University, Nanning, 530021, Guangxi, China; Research Centre for Regenerative Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China; Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China; School of Biomedical Sciences, University of Western Australia, Perth, WA, 6009, Australia
| | - Zhen Cao
- Department of Biomedical Materials Science, Third Military Medical University, Chongqing, 400038, China; Department of Anatomy, Third Military Medical University, Chongqing, 400038, China; School of Biomedical Sciences, University of Western Australia, Perth, WA, 6009, Australia
| | - Jennifer Tickner
- School of Biomedical Sciences, University of Western Australia, Perth, WA, 6009, Australia
| | - Heng Qiu
- School of Biomedical Sciences, University of Western Australia, Perth, WA, 6009, Australia
| | - Chao Wang
- School of Biomedical Sciences, University of Western Australia, Perth, WA, 6009, Australia
| | - Kai Chen
- School of Biomedical Sciences, University of Western Australia, Perth, WA, 6009, Australia
| | - Ziyi Wang
- School of Biomedical Sciences, University of Western Australia, Perth, WA, 6009, Australia
| | - Chunyu Guo
- Department of Neurosurgery, Nanning Second People's Hospital, Nanning, 530031, Guangxi, China
| | - Shiwu Dong
- Department of Biomedical Materials Science, Third Military Medical University, Chongqing, 400038, China.
| | - Jiake Xu
- School of Biomedical Sciences, University of Western Australia, Perth, WA, 6009, Australia.
| |
Collapse
|
43
|
Hodge CD, Spyracopoulos L, Glover JNM. Ubc13: the Lys63 ubiquitin chain building machine. Oncotarget 2018; 7:64471-64504. [PMID: 27486774 PMCID: PMC5325457 DOI: 10.18632/oncotarget.10948] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 07/19/2016] [Indexed: 12/25/2022] Open
Abstract
Ubc13 is an ubiquitin E2 conjugating enzyme that participates with many different E3 ligases to form lysine 63-linked (Lys63) ubiquitin chains that are critical to signaling in inflammatory and DNA damage response pathways. Recent studies have suggested Ubc13 as a potential therapeutic target for intervention in various human diseases including several different cancers, alleviation of anti-cancer drug resistance, chronic inflammation, and viral infections. Understanding a potential therapeutic target from different angles is important to assess its usefulness and potential pitfalls. Here we present a global review of Ubc13 from its structure, function, and cellular activities, to its natural and chemical inhibition. The aim of this article is to review the literature that directly implicates Ubc13 in a biological function, and to integrate structural and mechanistic insights into the larger role of this critical E2 enzyme. We discuss observations of multiple Ubc13 structures that suggest a novel mechanism for activation of Ubc13 that involves conformational change of the active site loop.
Collapse
Affiliation(s)
- Curtis D Hodge
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Leo Spyracopoulos
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - J N Mark Glover
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
44
|
|
45
|
Marino S, Bishop RT, Logan JG, Mollat P, Idris AI. Pharmacological evidence for the bone-autonomous contribution of the NFκB/β-catenin axis to breast cancer related osteolysis. Cancer Lett 2017; 410:180-190. [DOI: 10.1016/j.canlet.2017.09.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 09/18/2017] [Accepted: 09/21/2017] [Indexed: 01/21/2023]
|
46
|
Park MY, Kim HS, Lee M, Park B, Lee HY, Cho EB, Seong JY, Bae YS. FAM19A5, a brain-specific chemokine, inhibits RANKL-induced osteoclast formation through formyl peptide receptor 2. Sci Rep 2017; 7:15575. [PMID: 29138422 PMCID: PMC5686125 DOI: 10.1038/s41598-017-15586-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 10/30/2017] [Indexed: 01/01/2023] Open
Abstract
Osteoclasts can be differentiated from bone marrow-derived macrophages (BMDM). They play a key role in bone resorption. Identifying novel molecules that can regulate osteoclastogenesis has been an important issue. In this study, we found that FAM19A5, a neurokine or brain-specific chemokine, strongly stimulated mouse BMDM, resulting in chemotactic migration and inhibition of RANKL-induced osteoclastogenesis. Expression levels of osteoclast-related genes such as RANK, TRAF6, OSCAR, TRAP, Blimp1, c-fos, and NFATc1 were markedly decreased by FAM19A5. However, negative regulators of osteoclastogenesis such as MafB and IRF-8 were upregulated by FAM19A5. FAM19A5 also downregulated expression levels of RANKL-induced fusogenic genes such as OC-STAMP, DC-STAMP, and Atp6v0d2. FAM19A5-induced inhibitory effect on osteoclastogenesis was significantly reversed by a formyl peptide receptor (FPR) 2 antagonist WRW4 or by FPR2-deficiency, suggesting a crucial role of FPR2 in the regulation of osteoclastogenesis. Collectively, our results suggest that FAM19A5 and its target receptor FPR2 can act as novel endogenous ligand/receptor to negatively regulate osteoclastogenesis. They might be regarded as potential targets to control osteoclast formation and bone disorders.
Collapse
Affiliation(s)
- Min Young Park
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hyung Sik Kim
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Republic of Korea
| | - Mingyu Lee
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Byunghyun Park
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Republic of Korea
| | - Ha Young Lee
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Republic of Korea
| | - Eun Bee Cho
- Graduate School of Medicine, Korea University, Seoul, Republic of Korea
| | - Jae Young Seong
- Graduate School of Medicine, Korea University, Seoul, Republic of Korea
| | - Yoe-Sik Bae
- Department of Biological Sciences, Sungkyunkwan University, Suwon, Republic of Korea.
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea.
| |
Collapse
|
47
|
NUMBL Interacts with TAK1, TRAF6 and NEMO to Negatively Regulate NF-κB Signaling During Osteoclastogenesis. Sci Rep 2017; 7:12600. [PMID: 28974699 PMCID: PMC5626749 DOI: 10.1038/s41598-017-12707-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/13/2017] [Indexed: 11/09/2022] Open
Abstract
NF-κB signaling is essential for osteoclast differentiation and skeletal homeostasis. We have reported recently that NUMB-like (NUMBL) protein modulates osteoclastogenesis by down regulating NF-κB activation. Herein, we decipher the mechanism underlying this phenomenon. We found that whereas NUMBL mRNA expression decreases upon stimulation of wild type (WT) bone marrow macrophages (BMMs) with RANKL, TAK1 deficiency in these cells leads to increased NUMBL and decreased TRAF6 and NEMO expression. These changes were restored upon WT-TAK1 expression, but not with catalytically inactive TAK1-K63W, suggesting that TAK1 enzymatic activity is required for these events. Forced expression of NUMBL inhibits osteoclast differentiation and function as evident by reduction in all hallmarks of osteoclastogenesis. Conversely, NUMBL-null BMMs, show increased osteoclast differentiation and mRNA expression of osteoclast marker genes. Post-translationally, K48-linked poly-ubiquitination of NUMBL is diminished in TAK1-null BMMs compared to elevated K48-poly-ubiquitination in WT cells, indicating increased stability of NUMBL in TAK1-null conditions. Further, our studies show that NUMBL directly interacts with TRAF6 and NEMO, and induces their K48-poly-ubiquitination mediated proteasomal degradation. Collectively, our data suggest that NUMBL and TAK1 are reciprocally regulated and that NUMBL acts as an endogenous regulator of NF-κB signaling and osteoclastogenesis by targeting the TAK1-TRAF6-NEMO axis.
Collapse
|
48
|
Salvianolic Acid A Inhibits OX-LDL Effects on Exacerbating Choroidal Neovascularization via Downregulating CYLD. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:6210694. [PMID: 29081889 PMCID: PMC5610829 DOI: 10.1155/2017/6210694] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 02/28/2017] [Accepted: 03/08/2017] [Indexed: 11/17/2022]
Abstract
Backgrounds Age-related macular degeneration is closely related to lipid oxidation, while relationship between OX-LDL and choroidal neovascularization is unclear. Recently, cylindromatosis is proved to regulate angiogenesis. However, its role in CNV progression remained unclear. Salvianolic acid A is widely used in vascular diseases. We investigated the relationship between OX-LDL and CNV and explore antineovascularization mechanism of Sal A. Methods C57BL6/J mice were randomized into four groups and injected with PBS or OX-LDL, together with Sal A for one week. CNV was induced by laser; CNV severity was analyzed by fundus fluorescein angiography, H&E staining, and choroid flat mount after 1 week. In in vitro experiments, ARPE-19 and HUVECs were cultured with OX-LDL (with or without Sal A) for 48 hours. Angiogenic proteins, cell junction integrity, and tube formation were measured. CYLD siRNA and specific inhibitors were used to explore mechanisms of CYLD in promoting OX-LDL-induced CNV progression. Results OX-LDL promoted laser-induced CNV volume by increasing VEGF, PDGF, and CYLD levels. Sal A antagonized OX-LDL effects and restrained CNV progression by decreasing VEGF/PDGF/CYLD, increasing antiangiostatin levels, and promoting P62-CYLD-TRAF6 interaction. Conclusions We demonstrated oxidation damage exacerbates CNV progression, and Sal A could be a clinical therapeutic reagent to exudative AMD.
Collapse
|
49
|
Uev1A facilitates osteosarcoma differentiation by promoting Smurf1-mediated Smad1 ubiquitination and degradation. Cell Death Dis 2017; 8:e2974. [PMID: 28771228 PMCID: PMC5596555 DOI: 10.1038/cddis.2017.366] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 06/06/2017] [Accepted: 07/02/2017] [Indexed: 02/07/2023]
Abstract
Malignant bone tumor osteosarcoma (OS) displays high metastasis incidence and poor prognosis. Its stem cell properties could serve to explain tumor recurrence and resistance to conventional treatments. In this study, we identified UEV1A as a novel suppressor of OS. Elevated UEV1A diminishes stem cell properties of OS cells and drives them to terminal differentiation. Importantly, UEV1A-overexpressed OS cells delay proliferation and are more sensitive to chemotherapeutic agents than control cells. Uev1A appears to be involved in the BMP signaling pathway in which it collaborates with a ubiquitin E3 ligase Smurf1 to promote Smad1 degradation in a Ubc13-independent manner. Indeed, Smad1 is identified as a dominant downstream effector of Uev1A, which unravels the mechanism underlying Uev1A-orchestrated tumor suppression in OS. The above findings identify UEV1A as a potential OS tumor suppression gene, and shed lights to future OS diagnosis and treatment.
Collapse
|
50
|
Biswas S, Zimman A, Gao D, Byzova TV, Podrez EA. TLR2 Plays a Key Role in Platelet Hyperreactivity and Accelerated Thrombosis Associated With Hyperlipidemia. Circ Res 2017; 121:951-962. [PMID: 28775078 DOI: 10.1161/circresaha.117.311069] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 07/27/2017] [Accepted: 08/03/2017] [Indexed: 01/19/2023]
Abstract
RATIONALE Platelet hyperreactivity, which is common in many pathological conditions, is associated with increased atherothrombotic risk. The mechanisms leading to platelet hyperreactivity are complex and not yet fully understood. OBJECTIVE Platelet hyperreactivity and accelerated thrombosis, specifically in dyslipidemia, have been mechanistically linked to the accumulation in the circulation of a specific group of oxidized phospholipids (oxPCCD36) that are ligands for the platelet pattern recognition receptor CD36. In the current article, we tested whether the platelet innate immune system contributes to responses to oxPCCD36 and accelerated thrombosis observed in hyperlipidemia. METHODS AND RESULTS Using in vitro approaches, as well as platelets from mice with genetic deletion of MyD88 (myeloid differentiation factor 88) or TLRs (Toll-like receptors), we demonstrate that TLR2 and TLR6 are required for the activation of human and murine platelets by oxPCCD36. oxPCCD36 induce formation of CD36/TLR2/TLR6 complex in platelets and activate downstream signaling via TIRAP (Toll-interleukin 1 receptor domain containing adaptor protein)-MyD88-IRAK (interleukin-1 receptor-associated kinase)1/4-TRAF6 (TNF receptor-associated factor 6), leading to integrin activation via the SFK (Src family kinase)-Syk (spleen tyrosine kinase)-PLCγ2 (phospholipase Cγ2) pathway. Intravital thrombosis studies using ApoE-/- mice with genetic deficiency of TLR2 or TLR6 have demonstrated that oxPCCD36 contribute to accelerated thrombosis specifically in the setting of hyperlipidemia. CONCLUSIONS Our studies reveal that TLR2 plays a key role in platelet hyperreactivity and the prothrombotic state in the setting of hyperlipidemia by sensing a wide range of endogenous lipid peroxidation ligands and activating innate immune signaling cascade in platelets.
Collapse
Affiliation(s)
- Sudipta Biswas
- From the Department of Molecular Cardiology, Cleveland Clinic, OH
| | - Alejandro Zimman
- From the Department of Molecular Cardiology, Cleveland Clinic, OH
| | - Detao Gao
- From the Department of Molecular Cardiology, Cleveland Clinic, OH
| | - Tatiana V Byzova
- From the Department of Molecular Cardiology, Cleveland Clinic, OH
| | - Eugene A Podrez
- From the Department of Molecular Cardiology, Cleveland Clinic, OH.
| |
Collapse
|