1
|
Guo M, Liu N, Guo Z. MiR-221-5p/Smad3 axis in osteoclastogenesis and its function: Potential therapeutic target for osteoporosis. Steroids 2022; 185:109063. [PMID: 35700796 DOI: 10.1016/j.steroids.2022.109063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 05/16/2022] [Accepted: 06/07/2022] [Indexed: 10/18/2022]
Abstract
OBJECTIVE To probe the role of miR-221-5p in osteoclastogenesis and the underlying mechanism. METHODS Serum from patients with postmenopausal osteoporosis and healthy controls was collected for determination of miR-221-5p expression. For in vitro experiment, RAW264.7 macrophages, in which the expression of miR-221-5p and/or Smad3 was altered, were induced by RANKL to differentiate into osteoclasts. For in vivo experiment, ovariectomy was performed to construct osteoporosis mouse models, followed by tail vein injection of miR-221-5p agomir. qRT-PCR and/or western blot were applied to measure the expression of miR-221-5p, Smad3, and osteoclastogenesis-related genes (NFATc1 and TRAF6). TRAP staining was utilized for assessment of osteoclast formation, MTT assay for assessment of osteoclast viability, and H&E staining for observation of histomorphological changes. The targeting relationship between miR-221-5p and Smad3 was verified by dual-luciferase reporter gene assay. RESULTS Compared with healthy controls, patients with postmenopausal osteoporosis had decreased miR-221-5p expression and lower lumbar vertebra bone mineral density. MiR-221-5p expression was decreased and Smad3 level was increased during osteoclastogenesis. The osteoclastogenesis was suppressed by miR-221-5p and promoted by Smad3, as evidenced by diminished number and viability of osteoclasts following overexpression of miR-221-5p or knockdown of Smad3. MiR-221-5p negatively mediated Smad3 expression. Smad3 suppression nullified the pro-osteoclastogenesis effect of miR-221-5p inhibition. Consistent results were observed in osteoporosis mouse models. CONCLUSION MiR-221-5p may alleviate postmenopausal osteoporosis through suppressing osteoclastogenesis via Smad3, which provides new ideas for molecule-targeted therapy of osteoporosis.
Collapse
Affiliation(s)
- Min Guo
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China.
| | - Na Liu
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Zhanjun Guo
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| |
Collapse
|
2
|
He D, Gao J, Zheng L, Liu S, Ye L, Lai H, Pan B, Pan W, Lou C, Chen Z, Fan S. TGF‑β inhibitor RepSox suppresses osteosarcoma via the JNK/Smad3 signaling pathway. Int J Oncol 2021; 59:84. [PMID: 34533199 PMCID: PMC8460063 DOI: 10.3892/ijo.2021.5264] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 07/29/2021] [Indexed: 02/06/2023] Open
Abstract
Osteosarcoma (OS) is the most common malignant bone tumor and the long-term survival rates remain unsatisfactory. Transforming growth factor-β (TGF-β) has been revealed to play a crucial role in OS progression, and RepSox is an effective TGF-β inhibitor. In the present study, the effect of RepSox on the proliferation of the OS cell lines (HOS and 143B) was detected. The results revealed that RepSox effectively inhibited the proliferation of OS cells by inducing S-phase arrest and apoptosis. Moreover, the inhibitory effect of RepSox on cell migration and invasion was confirmed by wound-healing and Transwell assays. Furthermore, western blotting revealed that the protein levels of molecules associated with the epithelial-mesenchymal transition (EMT) phenotype, including E-cadherin, N-cadherin, Vimentin, matrix metalloproteinase (MMP)-2 and MMP-9, were reduced by RepSox treatment. Concurrently, it was also revealed that the JNK and Smad3 signaling pathway was inhibited. Our in vivo findings using a xenograft model also revealed that RepSox markedly inhibited the growth of tumors. In general, our data demonstrated that RepSox suppressed OS proliferation, EMT and promoted apoptosis by inhibiting the JNK/Smad3 signaling pathway. Thus, RepSox may be a potential anti-OS drug.
Collapse
Affiliation(s)
- Dengwei He
- Department of Orthopedics, Affiliated Lishui Hospital of Zhejiang University, Lishui, Zhejiang 323000, P.R. China
| | - Jiawei Gao
- Department of Orthopedics, Affiliated Lishui Hospital of Zhejiang University, Lishui, Zhejiang 323000, P.R. China
| | - Lin Zheng
- Department of Orthopedics, Affiliated Lishui Hospital of Zhejiang University, Lishui, Zhejiang 323000, P.R. China
| | - Shijie Liu
- Department of Orthopedics, Affiliated Lishui Hospital of Zhejiang University, Lishui, Zhejiang 323000, P.R. China
| | - Lin Ye
- Department of Orthopedics, Affiliated Lishui Hospital of Zhejiang University, Lishui, Zhejiang 323000, P.R. China
| | - Hehuan Lai
- Department of Orthopedics, Affiliated Lishui Hospital of Zhejiang University, Lishui, Zhejiang 323000, P.R. China
| | - Bin Pan
- Department of Orthopedics, Affiliated Lishui Hospital of Zhejiang University, Lishui, Zhejiang 323000, P.R. China
| | - Wenzheng Pan
- Department of Orthopedics, Affiliated Lishui Hospital of Zhejiang University, Lishui, Zhejiang 323000, P.R. China
| | - Chao Lou
- Department of Orthopedics, Affiliated Lishui Hospital of Zhejiang University, Lishui, Zhejiang 323000, P.R. China
| | - Zhenzhong Chen
- Department of Orthopedics, Affiliated Lishui Hospital of Zhejiang University, Lishui, Zhejiang 323000, P.R. China
| | - Shunwu Fan
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, P.R. China
| |
Collapse
|
3
|
Guo Y, Zhu H, Li X, Ma C, Li Y, Sun T, Wang Y, Wang C, Guan W, Liu C. RepSox effectively promotes the induced differentiation of sheep fibroblasts into adipocytes via the inhibition of the TGF‑β1/Smad pathway. Int J Mol Med 2021; 48:148. [PMID: 34132357 PMCID: PMC8208630 DOI: 10.3892/ijmm.2021.4981] [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: 08/24/2020] [Accepted: 05/10/2021] [Indexed: 01/22/2023] Open
Abstract
Previous reports have demonstrated that RepSox can function as a replacement for cMyc and Sox2 in the reprogramming of cells into induced pluripotent stem cells (iPSCs), as well as increasing the levels of bone morphogenetic protein (BMP)-3 and inducing the phosphorylation of Smad1 in mouse embryonic stem cells. In the present study, it was demonstrated that RepSox caused the visible morphological transformation of sheep fibroblasts; however, no significant alterations in cell proliferation, apoptosis or chromosome aberrations were observed. Moreover, RepSox increased the plasticity of long-term cryopreserved sheep fibroblasts, and further promoted differentiation into adipocytes. RepSox treatment led to a notable decrease in the expression of components of the transforming growth factor (TGF)-β signaling pathway, particularly Smad2/3 phosphorylation. RepSox also activated the BMP pathway, promoted the reprogramming of cells from fibroblasts into adipocytes and induced mesenchymal-epithelial transition. It is worth noting that RepSox notably increased the expression of octamer-binding transcription factor 4 and L-Myc, whereas Sox2 and Nanog expression were not detected. The results of high-throughput RNA sequencing revealed that the levels of differentially expressed genes (DEGs) involved in various metabolic processes were markedly upregulated in the RepSox-treated fibroblasts, while the DEGs in the majority of signaling pathways were markedly downregulated. On the whole, the present study demonstrates that RepSox can promote the plasticity of sheep fibroblasts and facilitates the differentiation of adipocytes via increasing BMP expression and inhibiting the activation of the TGF-β signaling pathway.
Collapse
Affiliation(s)
- Yu Guo
- Department of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, Anhui 233000, P.R. China
| | - Huan Zhu
- Department of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, Anhui 233000, P.R. China
| | - Xiangchen Li
- Institute of Beijing Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China
| | - Caiyun Ma
- Department of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, Anhui 233000, P.R. China
| | - Yanan Li
- Institute of Beijing Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China
| | - Tingting Sun
- Department of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, Anhui 233000, P.R. China
| | - Yuanyuan Wang
- Department of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, Anhui 233000, P.R. China
| | - Chunjing Wang
- Department of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, Anhui 233000, P.R. China
| | - Weijun Guan
- Institute of Beijing Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China
| | - Changqing Liu
- Department of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, Anhui 233000, P.R. China
| |
Collapse
|
4
|
Huang C, Zheng Y, Bai J, Shi C, Shi X, Shan H, Zhou X. Hepatocyte growth factor overexpression promotes osteoclastogenesis and exacerbates bone loss in CIA mice. J Orthop Translat 2020; 27:9-16. [PMID: 33344167 PMCID: PMC7732867 DOI: 10.1016/j.jot.2020.10.011] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 09/11/2020] [Accepted: 10/28/2020] [Indexed: 01/20/2023] Open
Abstract
Background Hepatocyte growth factor (HGF) is a multifunctional growth factor that promotes various biological processes. However, the effect of HGF on bone metabolism in rheumatoid arthritis (RA) remains unknown. Here, we investigated the role of HGF in regulating osteoclastogenesis and bone resorption in RA. Methods The expression of HGF in RA patients and collagen-induced arthritis (CIA) mice was examined. The role of HGF on osteoclastogenesis was analysed by osteoclastogenesis and bone resorption assays. The effect of HGF inhibition was evaluated in a CIA mice model. The mechanism of HGF in regulating osteoclastogenesis and bone resorption was explored by a series of in vitro studies. Results HGF was overexpressed in CIA and RA. HGF stimulated osteoclastogenesis in vitro. SU11274, a selective small molecule blocker of c-Met, impeded the effect of HGF on osteoclastogenesis and bone resorption. HGF regulated osteoclastogenesis by JNK and AKT-GSK-3β-NFATc1 signallings. SU11274 protected CIA mice from pathological bone loss. Conclusions These data strongly suggest that the highly expressed HGF in the joint tissues contributes to bone loss in RA. Inhibition of HGF/c-Met could effectively alleviate pathological bone loss and inflammatory symptoms in CIA mice. HGF/c-Met may be used as a new target for the treatment of bone loss in RA.
Collapse
Affiliation(s)
- Chaoming Huang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, China.,Department of Orthopedics, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, Jiangsu, 223800, China
| | - Yufan Zheng
- Institute of Neuroscience, Soochow University, Suzhou, 215000, China
| | - Jinyu Bai
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, China
| | - Ce Shi
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, China.,Department of Orthopedics, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, Jiangsu, 223800, China
| | - Xin Shi
- Institute of Neuroscience, Soochow University, Suzhou, 215000, China
| | - Huajian Shan
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, China
| | - Xiaozhong Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215004, China
| |
Collapse
|
5
|
Kim EN, Kim YG, Lee JH, Min BS, Jeong GS. 6,7,4'-Trihydroxyflavone inhibits osteoclast formation and bone resorption in vitro and in vivo. Phytother Res 2019; 33:2948-2959. [PMID: 31478281 DOI: 10.1002/ptr.6468] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 07/05/2019] [Accepted: 07/09/2019] [Indexed: 12/30/2022]
Abstract
The balance between the osteoblasts and the osteoclasts is important for the maintenance of the skeleton of the human body. The osteoclasts absorb bone after differentiated into polymorphonuclear cells by the fusion of monocytes/macrophages. We have found that 6,7,4'-Trihydroxyflavone (THF), a compound from the heartwood of Dalbergia Odorifera inhibits receptor activator of NF-κB ligand (RANKL)-induced osteoclast differentiation, actin ring formation, and bone resorption in RAW 264.7 cells and bone marrow macrophage. THF significantly inhibited the c-Jun-N-terminal kinase signaling pathway without affecting extracellular signal-regulated kinase, p38, and AKT signaling. Moreover, THF inhibited the expression of c-Fos, nuclear factor-activated T cells cytoplasm 1, cathepsin K, and c-src by RANKL. We used a lipopolysaccharide (LPS)-induced bone loss model in mice. Consequently, bone volume per tissue volume, trabecular number's reduction was recovered in THF-treated mice, and trabecular separation's augmentation was also attenuated by THF administration. In summary, THF inhibits RANKL-induced osteoclast differentiation by MAPK signaling pathway and inhibits bone resorption by destroying the actin ring in mature osteoclasts. THF also prevented LPS-induced bone loss in a mice model. Thus, THF may be useful in the treatment of bone diseases associated with excessive osteoclast differentiation and bone resorption.
Collapse
Affiliation(s)
- Eun-Nam Kim
- College of Pharmacy, Keimyung University, Daegu, Republic of Korea
| | - Yu Gyeong Kim
- College of Pharmacy, Keimyung University, Daegu, Republic of Korea
| | - Jeong-Hyung Lee
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, Republic of Korea
| | - Byung Sun Min
- College of Pharmacy, Catholic University of Daegu, Gyeongsan, Republic of Korea
| | - Gil-Saeng Jeong
- College of Pharmacy, Keimyung University, Daegu, Republic of Korea
| |
Collapse
|
6
|
Bottini A, Wu DJ, Ai R, Le Roux M, Bartok B, Bombardieri M, Doody KM, Zhang V, Sacchetti C, Zoccheddu M, Lonic A, Li X, Boyle DL, Hammaker D, Meng TC, Liu L, Corr M, Stanford SM, Lewis M, Wang W, Firestein GS, Khew-Goodall Y, Pitzalis C, Bottini N. PTPN14 phosphatase and YAP promote TGFβ signalling in rheumatoid synoviocytes. Ann Rheum Dis 2019; 78:600-609. [PMID: 30808624 DOI: 10.1136/annrheumdis-2018-213799] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 01/28/2019] [Accepted: 01/30/2019] [Indexed: 11/04/2022]
Abstract
OBJECTIVE We aimed to understand the role of the tyrosine phosphatase PTPN14-which in cancer cells modulates the Hippo pathway by retaining YAP in the cytosol-in fibroblast-like synoviocytes (FLS) from patients with rheumatoid arthritis (RA). METHODS Gene/protein expression levels were measured by quantitative PCR and/or Western blotting. Gene knockdown in RA FLS was achieved using antisense oligonucleotides. The interaction between PTPN14 and YAP was assessed by immunoprecipitation. The cellular localisation of YAP and SMAD3 was examined via immunofluorescence. SMAD reporter studies were carried out in HEK293T cells. The RA FLS/cartilage coimplantation and passive K/BxN models were used to examine the role of YAP in arthritis. RESULTS RA FLS displayed overexpression of PTPN14 when compared with FLS from patients with osteoarthritis (OA). PTPN14 knockdown in RA FLS impaired TGFβ-dependent expression of MMP13 and potentiation of TNF signalling. In RA FLS, PTPN14 formed a complex with YAP. Expression of PTPN14 or nuclear YAP-but not of a non-YAP-interacting PTPN14 mutant-enhanced SMAD reporter activity. YAP promoted TGFβ-dependent SMAD3 nuclear localisation in RA FLS. Differences in epigenetic marks within Hippo pathway genes, including YAP, were found between RA FLS and OA FLS. Inhibition of YAP reduced RA FLS pathogenic behaviour and ameliorated arthritis severity. CONCLUSION In RA FLS, PTPN14 and YAP promote nuclear localisation of SMAD3. YAP enhances a range of RA FLS pathogenic behaviours which, together with epigenetic evidence, points to the Hippo pathway as an important regulator of RA FLS behaviour.
Collapse
Affiliation(s)
- Angel Bottini
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA.,Division of Cellular Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Dennis J Wu
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA
| | - Rizi Ai
- Dept. of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA
| | - Michelle Le Roux
- Division of Cellular Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Beatrix Bartok
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA
| | - Michele Bombardieri
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Karen M Doody
- Division of Cellular Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Vida Zhang
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA.,Division of Cellular Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Cristiano Sacchetti
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA.,Division of Cellular Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Martina Zoccheddu
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA
| | - Ana Lonic
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia
| | - Xiaochun Li
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia
| | - David L Boyle
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA
| | - Deepa Hammaker
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA
| | - Tzu-Ching Meng
- Institute for Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Lin Liu
- Dept. of Family Medicine and Public Health, University of California San Diego, La Jolla, CA, USA
| | - Maripat Corr
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA
| | - Stephanie M Stanford
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA.,Division of Cellular Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Myles Lewis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Wei Wang
- Dept. of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA.,Dept. of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Gary S Firestein
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA
| | - Yeesim Khew-Goodall
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia
| | - Costantino Pitzalis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Nunzio Bottini
- Dept. of Medicine, University of California San Diego, La Jolla, California, USA .,Division of Cellular Biology, La Jolla Institute for Immunology, La Jolla, CA, USA
| |
Collapse
|