1
|
Wang Z, Wu F, Yan J, Liang L, Chang F, Dong M, Diao J, Wu H. Ecdysterone Alleviates Atherosclerosis by Inhibiting NCF2 and Inhibiting Ferroptosis Mediated by the PI3K/Akt/Nrf2 Pathway. J Cell Mol Med 2025; 29:e70446. [PMID: 40045169 PMCID: PMC11882393 DOI: 10.1111/jcmm.70446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 01/14/2025] [Accepted: 02/12/2025] [Indexed: 03/09/2025] Open
Abstract
Ecdysterone (Ecd), an active ingredient in trianthema, has a strong anti-inflammatory effect. This study aimed to explore the potential mechanism by which Ecd improves atherosclerosis (AS). Here, we systematically investigated the mechanism of Ecd in human umbilical vein endothelial cells (HUVECs) treated with oxidised low-density lipoprotein (ox-LDL). In ox-LDL-treated HUVECs, Ecd promoted HUVEC viability as well as inhibited ferroptosis and the secretion of inflammatory factors (TNF-α, IL-6 and IL-1β). In addition, Ecd inhibited the expression of neutrophil cytoplasmic factor 2 (NCF2) and triggered the PI3K/AKT/Nrf2 signalling pathway, thereby alleviating the increase of ferroptosis in ox-LDL-treated HUVECs. More importantly, we constructed an AS mouse model by feeding ApoE-/- mice with a high-fat diet and found that Ecd treatment alleviated vasculopathy and arterial ferroptosis and inhibited the secretion of inflammatory factors in vivo, which could be reversed by overexpression of NCF2. Overall, this study showed that the protective effect of Ecd on AS is mainly achieved by inhibiting NCF2 and activating the PI3K/Akt/Nrf2 pathway to inhibit ferroptosis. Therefore, Ecd may be an effective drug to improve AS by inhibiting ferroptosis-induced inflammation.
Collapse
Affiliation(s)
- Zhenyu Wang
- Department of CardiologyShaanxi Provincial People's HospitalXi'anShaanxiChina
| | - Fengchao Wu
- Department of CardiologyShaanxi Provincial People's HospitalXi'anShaanxiChina
| | - Ju Yan
- Department of CardiologyShaanxi Provincial People's HospitalXi'anShaanxiChina
| | - Lei Liang
- Department of CardiologyShaanxi Provincial People's HospitalXi'anShaanxiChina
| | - Fengjun Chang
- Department of CardiologyShaanxi Provincial People's HospitalXi'anShaanxiChina
| | - Mengya Dong
- Department of CardiologyShaanxi Provincial People's HospitalXi'anShaanxiChina
| | - Jiayu Diao
- Department of CardiologyShaanxi Provincial People's HospitalXi'anShaanxiChina
| | - Haoyu Wu
- Department of CardiologyShaanxi Provincial People's HospitalXi'anShaanxiChina
| |
Collapse
|
2
|
Zhong C, Xu H, Chen J, Cai W, Zhou J, Peng H. Human Umbilical Cord Mesenchymal Stem Cells Prevent Steroid-Induced Avascular Necrosis of the Femoral Head by Modulating Cellular Autophagy. Biomedicines 2024; 12:2817. [PMID: 39767723 PMCID: PMC11673007 DOI: 10.3390/biomedicines12122817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/24/2024] [Accepted: 12/08/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Glucocorticoids (GCs) are critical regulatory molecules in the body, commonly utilized in clinical practice for their potent anti-inflammatory and immunosuppressive properties. However, prolonged, high-dose GC therapy is frequently associated with femoral head necrosis, a condition known as glucocorticoid-induced osteonecrosis of the femoral head (GC-ONFH). Emerging evidence suggests that enhanced autophagy may mitigate apoptosis, thereby protecting osteoblasts from GC-induced damage and delaying the progression of ONFH. This study aims to evaluate whether human umbilical cord mesenchymal stem cells (hUCMSCs) can alleviate GC-induced osteoblast injury through autophagy modulation. METHODS In vitro, osteoblasts were exposed to GCs for 48 h, followed by co-culture with hUCMSCs for an additional 12 h before further analysis. The osteoblasts were categorized into four experimental groups: (A) control group, (B) Dex group, (C) Dex + hUCMSC group, and (D) Dex + hUCMSC + 3-MA group. In vivo, rabbits were assigned to one of four groups: Con, MPS, core decompression (CD), and CD + hUCMSC (n = 12 per group), and subsequently subjected to CT imaging and HE staining. RESULTS In vitro results demonstrate that hUCMSC treatment mitigated GC-induced osteoblast apoptosis and preserved osteogenic activity through autophagy modulation. In vivo, infusion of hUCMSCs enhanced trabecular thickness in the femoral head and improved the femoral head microenvironment. CONCLUSIONS These findings suggest that hUCMSCs protect osteoblasts from GC-induced damage by regulating autophagy, offering new insights into the potential therapeutic use of hUCMSCs for treating ONFH via autophagy enhancement.
Collapse
Affiliation(s)
| | | | | | | | - Jianlin Zhou
- Department of Orthopedics Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China; (C.Z.); (J.C.); (H.X.); (W.C.)
| | - Hao Peng
- Department of Orthopedics Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China; (C.Z.); (J.C.); (H.X.); (W.C.)
| |
Collapse
|
3
|
Zhang X, Li Z, Xu X, Liu Z, Hao Y, Yang F, Li X, Zhang N, Hou Y, Zhang X. Huogu injection protects against SONFH by promoting osteogenic differentiation of BMSCs and preventing osteoblast apoptosis. Cell Tissue Res 2024; 395:63-79. [PMID: 38040999 PMCID: PMC10774174 DOI: 10.1007/s00441-023-03846-7] [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: 05/24/2023] [Accepted: 11/15/2023] [Indexed: 12/03/2023]
Abstract
To investigate the effect and mechanism of Huogu injection (HG) on steroid-induced osteonecrosis of the femoral head (SONFH), we established a SONFH model in rabbits using horse serum and dexamethasone (DEX) and applied HG locally at the hip joint. We evaluated the therapeutic efficacy at 4 weeks using scanning electron microscopy (SEM), micro-CT, and qualitative histology including H&E, Masson's trichrome, ALP, and TUNEL staining. In vitro, we induced osteogenic differentiation of bone marrow stromal cells (BMSCs) and performed analysis on days 14 and 21 of cell differentiation. The findings, in vivo, including SEM, micro-CT, and H&E staining, showed that HG significantly maintained bone quality and trabecular number. ALP staining indicated that HG promoted the proliferation of bone cells. Moreover, the results of Masson's trichrome staining demonstrated the essential role of HG in collagen synthesis. Additionally, TUNEL staining revealed that HG reduced apoptosis. ALP and ARS staining in vitro confirmed that HG enhanced osteogenic differentiation and mineralization, consistent with the WB and qRT-PCR analysis. Furthermore, Annexin V-FITC/PI staining verified that HG inhibited osteoblast apoptosis, in agreement with the WB and qRT-PCR analyses. Furthermore, combined with the UPLC analysis, we found that naringin enhanced the osteogenic differentiation and accelerated the deposition of calcium phosphate. Salvianolic acid B protected osteoblasts derived from BMSCs against GCs-mediated apoptosis. Thus, this study not only reveals the mechanism of HG in promoting osteogenesis and anti-apoptosis of osteoblasts but also identifies the active-related components in HG, by which we provide the evidence for the application of HG in SONFH.
Collapse
Affiliation(s)
- Xin Zhang
- Luoyang Orthopedic-Traumatological Hospital of Henan Province (Henan Provincial Orthopedic Hospital), Luoyang, 471002, Henan, China
| | - Ziyu Li
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, 150000, Heilongjiang, China
| | - Xilin Xu
- The Third Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150000, Heilongjiang, China
| | - Zhao Liu
- The First Affiliated Hospital of Zhejiang University of Chinese Medicine, Hangzhou, 310000, Zhejiang, China
| | - Yuanyuan Hao
- Shijiazhuang Yiling Pharmaceuticalco., ltd, Shijiazhuang, 050000, Hebei, China
| | - Fubiao Yang
- Heilongjiang University of Chinese Medicine, Harbin, 150000, Heilongjiang, China
| | - Xiaodong Li
- Heilongjiang University of Chinese Medicine, Harbin, 150000, Heilongjiang, China
| | - Ning Zhang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, 150000, Heilongjiang, China
| | - Yunlong Hou
- Shijiazhuang Yiling Pharmaceuticalco., ltd, Shijiazhuang, 050000, Hebei, China.
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine, Shijiazhuang, 050000, Hebei, China.
| | - Xiaofeng Zhang
- Heilongjiang University of Chinese Medicine, Harbin, 150000, Heilongjiang, China.
| |
Collapse
|
4
|
Tocotrienol as a Protecting Agent against Glucocorticoid-Induced Osteoporosis: A Mini Review of Potential Mechanisms. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27185862. [PMID: 36144598 PMCID: PMC9506150 DOI: 10.3390/molecules27185862] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/03/2022] [Accepted: 09/08/2022] [Indexed: 11/18/2022]
Abstract
Glucocorticoid-induced osteogenic dysfunction is the main pathologyical mechanism underlying the development of glucocorticoid-induced osteoporosis. Glucocorticoids promote adipogenic differentiation and osteoblast apoptosis through various pathways. Various ongoing studies are exploring the potential of natural products in preventing glucocorticoid-induced osteoporosis. Preclinical studies have consistently shown the bone protective effects of tocotrienol through its antioxidant and anabolic effects. This review aims to summarise the potential mechanisms of tocotrienol in preventing glucocorticoid-induced osteoporosis based on existing in vivo and in vitro evidence. The current literature showed that tocotrienol prevents oxidative damage on osteoblasts exposed to high levels of glucocorticoids. Tocotrienol reduces lipid peroxidation and increases oxidative stress enzyme activities. The reduction in oxidative stress protects the osteoblasts and preserves the bone microstructure and biomechanical strength of glucocorticoid-treated animals. In other animal models, tocotrienol has been shown to activate the Wnt/β-catenin pathway and lower the RANKL/OPG ratio, which are the targets of glucocorticoids. In conclusion, tocotrienol enhances osteogenic differentiation and bone formation in glucocorticoid-treated osteoblasts while improving structural integrity in glucocorticoid-treated rats. This is achieved by preventing oxidative stress and osteoblast apoptosis. However, these preclinical results should be validated in a randomised controlled trial.
Collapse
|
5
|
Yan CP, Wang XK, Jiang K, Yin C, Xiang C, Wang Y, Pu C, Chen L, Li YL. β-Ecdysterone Enhanced Bone Regeneration Through the BMP-2/SMAD/RUNX2/Osterix Signaling Pathway. Front Cell Dev Biol 2022; 10:883228. [PMID: 35669516 PMCID: PMC9164109 DOI: 10.3389/fcell.2022.883228] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 05/03/2022] [Indexed: 11/19/2022] Open
Abstract
Bone defects are a global public health problem. However, the available methods for inducing bone regeneration are limited. The application of traditional Chinese herbs for bone regeneration has gained popularity in recent years. β-ecdysterone is a plant sterol similar to estrogen, that promotes protein synthesis in cells; however, its function in bone regeneration remains unclear. In this study, we investigated the function of β-ecdysterone on osteoblast differentiation and bone regeneration in vitro and in vivo. MC3T3-E1 cells were used to test the function of β-ecdysterone on osteoblast differentiation and bone regeneration in vitro. The results of the Cell Counting Kit-8 assay suggested that the proliferation of MC3T3-E1 cells was promoted by β-ecdysterone. Furthermore, β-ecdysterone influenced the expression of osteogenesis-related genes, and the bone regeneration capacity of MC3T3-E1 cells was detected by polymerase chain reaction, the alkaline phosphatase (ALP) test, and the alizarin red test. β-ecdysterone could upregulate the expression of osteoblastic-related genes, and promoted ALP activity and the formation of calcium nodules. We also determined that β-ecdysterone increased the mRNA and protein levels of components of the BMP-2/Smad/Runx2/Osterix pathway. DNA sequencing further confirmed these target effects. β-ecdysterone promoted bone formation by enhancing gene expression of the BMP-2/Smad/Runx2/Osterix signaling pathway and by enrichment biological processes. For in vivo experiments, a femoral condyle defect model was constructed by drilling a bone defect measuring 3 mm in diameter and 4 mm in depth in the femoral condyle of 8-week-old Sprague Dawley male rats. This model was used to further assess the bone regenerative functions of β-ecdysterone. The results of micro-computed tomography showed that β-ecdysterone could accelerate bone regeneration, exhibiting higher bone volume, bone surface, and bone mineral density at each observation time point. Immunohistochemistry confirmed that the β-ecdysterone also increased the expression of collagen, osteocalcin, and bone morphogenetic protein-2 in the experiment group at 4 and 8 weeks. In conclusion, β-ecdysterone is a new bone regeneration regulator that can stimulate MC3T3-E1 cell proliferation and induce bone regeneration through the BMP-2/Smad/Runx2/Osterix pathway. This newly discovered function of β-ecdysterone has revealed a new direction of osteogenic differentiation and has provided novel therapeutic strategies for treating bone defects.
Collapse
Affiliation(s)
- Cai-Ping Yan
- Department of Orthopaedics, Affiliated Hospital of North Sichuan Medical College, Nanchong, China.,Laboratory of Biological Tissue Engineering and Digital Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xing-Kuan Wang
- Department of Orthopaedics, Affiliated Hospital of North Sichuan Medical College, Nanchong, China.,Laboratory of Biological Tissue Engineering and Digital Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Ke Jiang
- Department of Orthopaedics, Affiliated Hospital of North Sichuan Medical College, Nanchong, China.,Laboratory of Biological Tissue Engineering and Digital Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Chong Yin
- Laboratory for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, NPU-UAB Joint Laboratory for Bone Metabolism, Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Chao Xiang
- Department of Orthopaedics, Affiliated Hospital of North Sichuan Medical College, Nanchong, China.,Laboratory of Biological Tissue Engineering and Digital Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yong Wang
- Department of Orthopaedics, Affiliated Hospital of North Sichuan Medical College, Nanchong, China.,Laboratory of Biological Tissue Engineering and Digital Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Chaoyu Pu
- Department of Orthopaedics, Affiliated Hospital of North Sichuan Medical College, Nanchong, China.,Laboratory of Biological Tissue Engineering and Digital Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Lu Chen
- Department of Orthopaedics, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yu-Ling Li
- Department of Orthopaedics, Affiliated Hospital of North Sichuan Medical College, Nanchong, China.,Laboratory of Biological Tissue Engineering and Digital Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| |
Collapse
|
6
|
Gado M, Baschant U, Hofbauer LC, Henneicke H. Bad to the Bone: The Effects of Therapeutic Glucocorticoids on Osteoblasts and Osteocytes. Front Endocrinol (Lausanne) 2022; 13:835720. [PMID: 35432217 PMCID: PMC9008133 DOI: 10.3389/fendo.2022.835720] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/10/2022] [Indexed: 02/06/2023] Open
Abstract
Despite the continued development of specialized immunosuppressive therapies in the form of monoclonal antibodies, glucocorticoids remain a mainstay in the treatment of rheumatological and auto-inflammatory disorders. Therapeutic glucocorticoids are unmatched in the breadth of their immunosuppressive properties and deliver their anti-inflammatory effects at unparalleled speed. However, long-term exposure to therapeutic doses of glucocorticoids decreases bone mass and increases the risk of fractures - particularly in the spine - thus limiting their clinical use. Due to the abundant expression of glucocorticoid receptors across all skeletal cell populations and their respective progenitors, therapeutic glucocorticoids affect skeletal quality through a plethora of cellular targets and molecular mechanisms. However, recent evidence from rodent studies, supported by clinical data, highlights the considerable role of cells of the osteoblast lineage in the pathogenesis of glucocorticoid-induced osteoporosis: it is now appreciated that cells of the osteoblast lineage are key targets of therapeutic glucocorticoids and have an outsized role in mediating their undesirable skeletal effects. As part of this article, we review the molecular mechanisms underpinning the detrimental effects of supraphysiological levels of glucocorticoids on cells of the osteoblast lineage including osteocytes and highlight the clinical implications of recent discoveries in the field.
Collapse
Affiliation(s)
- Manuel Gado
- Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, Dresden, Germany
| | - Ulrike Baschant
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Center for Healthy Aging, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Lorenz C. Hofbauer
- Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, Dresden, Germany
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Center for Healthy Aging, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Holger Henneicke
- Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, Dresden, Germany
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Center for Healthy Aging, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- *Correspondence: Holger Henneicke,
| |
Collapse
|
7
|
Todorova V, Ivanov K, Ivanova S. Comparison between the Biological Active Compounds in Plants with Adaptogenic Properties ( Rhaponticum carthamoides, Lepidium meyenii, Eleutherococcus senticosus and Panax ginseng). PLANTS (BASEL, SWITZERLAND) 2021; 11:64. [PMID: 35009068 PMCID: PMC8747685 DOI: 10.3390/plants11010064] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/22/2021] [Accepted: 12/24/2021] [Indexed: 06/01/2023]
Abstract
BACKGROUND In the 1960s, research into plant adaptogens began. Plants with adaptogenic properties have rich phytochemical compositions and have been used by humanity since ancient times. However, it is not still clear whether the adaptogenic properties are because of specific compounds or because of the whole plant extracts. The aim of this review is to compare the bioactive compounds in the different parts of these plants. METHODS The search strategy was based on studies related to the isolation of bioactive compounds from Rhaponticum carthamoides, Lepidium meyenii, Eleutherococcus senticosus, and Panax ginseng. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines were followed. RESULTS This review includes data from 259 articles. The phytochemicals isolated from Rhaponticum carthamoides, Lepidium meyenii, Eleutherococcus senticosus, and Panax ginseng were described and classified in several categories. CONCLUSIONS Plant species have always played an important role in drug discovery because their effectiveness is based on the hundreds of years of experience with folk medicine in different nations. In our view, there is great potential in the near future for some of the phytochemicals found in these plants species to become pharmaceutical agents.
Collapse
Affiliation(s)
- Velislava Todorova
- Department of Pharmacognosy and Pharmaceutical Chemistry, Faculty of Pharmacy, Medical University-Plovdiv, 4002 Plovdiv, Bulgaria; (K.I.); (S.I.)
| | | | | |
Collapse
|
8
|
Tang Y, Mo Y, Xin D, Xiong Z, Zeng L, Luo G, Cao Y. Regulation of osteoblast autophagy based on PI3K/AKT/mTOR signaling pathway study on the effect of β-ecdysterone on fracture healing. J Orthop Surg Res 2021; 16:719. [PMID: 34924000 PMCID: PMC8684673 DOI: 10.1186/s13018-021-02862-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 11/24/2021] [Indexed: 11/24/2022] Open
Abstract
Objectives To investigate the effects of β-ecdysterone on fracture healing and the underlying mechanism. Methods MTT assay was used to detect the cell viability. AO/PI and flow cytometry assays were used to determine the apoptotic rate. The expression level of RunX2, ATG7 and LC3 was evaluated by qRT-PCR and Western blot assays. X-ray and HE staining were conducted on the fractured femur. Immunohistochemical assay was used to detect the expression level of Beclin-1 and immunofluorescence assay was used to measure the expression level of LC3 in the fractured femurs. Western blot was utilized to determine the expression level of PI3K, p-AKT1, AKT1, p-mTOR, mTOR, p-p70S6K, and p70S6K. Results The ALP activity and the expression of RunX2 in fractured osteoblasts were significantly elevated, the apoptotic rate was suppressed by rapamycin, 60, and 80 μM β-ecdysterone. The state of autophagy both in fractured osteoblasts and femurs was facilitated by rapamycin and β-ecdysterone. Compared to control, Garrett score was significantly promoted in rapamycin and β-ecdysterone groups, accompanied by ameliorated pathological state. Lastly, the PI3K/AKT/mTOR pathway both in fractured osteoblasts and femurs was inhibited by rapamycin and β-ecdysterone. Conclusion β-ecdysterone might facilitate fracture healing by activating autophagy through suppressing PI3K/AKT/mTOR signal pathway.
Collapse
Affiliation(s)
- Yanghua Tang
- Department of Orthopedics, Hospital of Traditional Chinese Medicine of Xiaoshan District, No. 156, Yucai Road, Xiaoshan District, Hangzhou City, Zhejiang Province, China
| | - Yafeng Mo
- Department of Orthopedics, Hospital of Traditional Chinese Medicine of Xiaoshan District, No. 156, Yucai Road, Xiaoshan District, Hangzhou City, Zhejiang Province, China
| | - Dawei Xin
- Department of Orthopedics, Hospital of Traditional Chinese Medicine of Xiaoshan District, No. 156, Yucai Road, Xiaoshan District, Hangzhou City, Zhejiang Province, China
| | - Zhenfei Xiong
- Department of Orthopedics, Hospital of Traditional Chinese Medicine of Xiaoshan District, No. 156, Yucai Road, Xiaoshan District, Hangzhou City, Zhejiang Province, China
| | - Linru Zeng
- Department of Orthopedics, Hospital of Traditional Chinese Medicine of Xiaoshan District, No. 156, Yucai Road, Xiaoshan District, Hangzhou City, Zhejiang Province, China
| | - Gan Luo
- Department of Orthopedics, Hospital of Traditional Chinese Medicine of Xiaoshan District, No. 156, Yucai Road, Xiaoshan District, Hangzhou City, Zhejiang Province, China
| | - Yanguang Cao
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, No. 318, ChaoWang Road, Gongshu District, Hangzhou City, Zhejiang Province, China.
| |
Collapse
|
9
|
Li P, Mao W, Zhang S, Zhang L, Chen Z, Lu Z. MicroRNA-22 contributes to dexamethasone-induced osteoblast differentiation inhibition and dysfunction through targeting caveolin-3 expression in osteoblastic cells. Exp Ther Med 2021; 21:336. [PMID: 33732309 PMCID: PMC7903452 DOI: 10.3892/etm.2021.9767] [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: 05/30/2020] [Accepted: 10/30/2020] [Indexed: 12/14/2022] Open
Abstract
Osteoporosis is a common complication of long-term use of glucocorticoids (GCs) characterized by the loss of bone mass and damage of the microarchitecture as well as osteoblast dysfunction. Previous studies have demonstrated that microRNA-22 (miR-22) is the negative modulator of osteogenesis that may target caveolin-3 (CAV3), which has been reported to enhance bone formation and inhibit the progression of osteoporosis as well as apoptosis. The present study aimed to investigate whether miR-22 may be involved in dexamethasone (DEX)-induced inhibition of osteoblast differentiation and dysfunction by regulating CAV3 expression. Reverse transcription-quantitative PCR (RT-qPCR) was performed to measure the expression of miR-22 and western blotting was performed to determine protein levels. The results demonstrated that miR-22 expression was upregulated in DEX-treated osteoblastic cells compared with the control group. In addition, miR-22 mimic aggravated, whereas miR-22 inhibitor mitigated DEX-induced damage in osteoblastic cells compared with the control groups. Additionally, CAV3 was identified as the target of miR-22 in osteoblasts using RT-qPCR, western blotting and dual-luciferase reporter gene assay analysis. The results also demonstrated that silencing of CAV3 blocked the beneficial effects of miR-22 inhibitor against DEX-induced cell damage and apoptosis in osteoblasts, as evidenced by the increased expression levels of cleaved caspase-3, Bax and alkaline phosphatase activity as well as decreased cell viability and Bcl-2 levels. Collectively, these results indicate a novel molecular mechanism by which miR-22 contributes to DEX-induced osteoblast dysfunction and apoptosis via the miR-22/CAV3 pathway.
Collapse
Affiliation(s)
- Peng Li
- Department of Orthopedics, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Weiwei Mao
- Clinical Skill Center of Yinchuan First People's Hospital, Yinchuan, Ningxia 750001, P.R. China
| | - Shuai Zhang
- Department of Orthopedics, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Liang Zhang
- Department of Orthopedics, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Zhirong Chen
- Department of Orthopedics, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Zhidong Lu
- Department of Orthopedics, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| |
Collapse
|
10
|
Li P, Mao WW, Zhang S, Zhang L, Chen ZR, Lu ZD. Sodium hydrosulfide alleviates dexamethasone-induced cell senescence and dysfunction through targeting the miR-22/sirt1 pathway in osteoblastic MC3T3-E1 cells. Exp Ther Med 2021; 21:238. [PMID: 33603846 PMCID: PMC7851607 DOI: 10.3892/etm.2021.9669] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 09/15/2020] [Indexed: 01/30/2023] Open
Abstract
Glucocorticoid-induced osteoporosis is characterized by osteoblastic cell and microarchitecture dysfunction, as well as a loss of bone mass. Cell senescence contributes to the pathological process of osteoporosis and sodium hydrosulfide (NaHS) regulates the potent protective effects through delaying cell senescence. The aim of the present study was to investigate whether senescence could contribute to dexamethasone (Dex)-induced osteoblast impairment and to examine the effect of NaHS on Dex-induced cell senescence and damage. It was found that the levels of the senescence-associated markers, p53 and p21, were markedly increased in osteoblasts exposed to Dex. A p53 inhibitor reversed Dex-induced osteoblast injury, a process that was mitigated by NaHS administration through alleviating osteoblastic cell senescence. MicroRNA (miR)-22 blocked the impact of NaHS on Dex-induced osteoblast damage and senescence through targeting the regulation of Sirtuin 1 (sirt1) expression, as shown by the decreased cell viability and alkaline phosphatase activity, as well as an increased expression of p53 and p21. It was revealed that the sirt1 gene was the target of miR-22 in osteoblastic MC3T3-E1 cells through combining the results of dual luciferase reporter assays and reverse transcription-quantitative PCR, as well as western blot analyses. Silencing of sirt1 abolished the protective effect of NaHS against Dex-associated osteoblast senescence and injury. Taken together, the present study showed that NaHS prevents Dex-induced cell senescence and damage through targeting the miR-22/sirt1 pathway in osteoblastic MC3T3-E1 cells.
Collapse
Affiliation(s)
- Peng Li
- Department of Orthopedics, General Hospital of Ningxia Medical University, Xingqing, Yinchuan, Ningxia 750004, P.R. China
| | - Wei-Wei Mao
- Clinical Skill Center of Yinchuan First People's Hospital, Yinchuan, Ningxia 750001, P.R. China
| | - Shuai Zhang
- Department of Orthopedics, General Hospital of Ningxia Medical University, Xingqing, Yinchuan, Ningxia 750004, P.R. China
| | - Liang Zhang
- Department of Orthopedics, General Hospital of Ningxia Medical University, Xingqing, Yinchuan, Ningxia 750004, P.R. China
| | - Zhi-Rong Chen
- Department of Orthopedics, General Hospital of Ningxia Medical University, Xingqing, Yinchuan, Ningxia 750004, P.R. China
| | - Zhi-Dong Lu
- Department of Orthopedics, General Hospital of Ningxia Medical University, Xingqing, Yinchuan, Ningxia 750004, P.R. China
| |
Collapse
|
11
|
Tang Y, Mo Y, Xin D, Zeng L, Yue Z, Xu C. β-ecdysterone alleviates osteoarthritis by activating autophagy in chondrocytes through regulating PI3K/AKT/mTOR signal pathway. Am J Transl Res 2020; 12:7174-7186. [PMID: 33312358 PMCID: PMC7724317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 10/09/2020] [Indexed: 06/12/2023]
Abstract
PURPOSE To investigate the therapeutic effects of β-ecdysterone on osteoarthritis (OA) and the underlying mechanism. METHODS OA model was established on rats by injecting MIA. ELSA was used to determine the concentration of IL-1β, IL-6, NO and TNF-α in the chondrocytes and cartilage tissues. Immunofluorescence assay was used to determine the expression of collagen II in the chondrocytes. The survival rate of chondrocytes was evaluated by MTT assay. The apoptosis of chondrocytes was checked by AO/PI staining and flow cytometry assay. The expression level of Atg7, PI3K and caspase-3 was evaluated by qRT-PCR. Western Blot was used determine the expression of PI3K, p-AKT1, AKT1, p-mTOR, mTOR, p70S6K, p-p70S6K, LC3I, LC3II and caspase-3. HE staining was used to check the pathological state of cartilage tissues. RESULTS Chondrocytes were tolerable to rapamycin, 3-methyladenine and β-ecdysterone at the concentration of 10 mM, 100 nM and 40 μM, respectively. The apoptosis of chondrocytes was inhibited by rapamycin and β-ecdysterone, and induced by 3-methyladenine. PI3K, p-AKT1, p-mTOR, p-p70S6K and caspase-3 were down-regulated by rapamycin and β-ecdysterone, and up-regulated by 3-methyladenine in both the chondrocytes and the cartilage tissues. The expression of Atg7 and LC3II/LC3I were regulated in a opposite way. The inflammation state was improved by rapamycin and β-ecdysterone both the chondrocytes and the cartilage tissues. HE staining results showed that the pathological state of cartilage tissues was alleviated by β-ecdysterone. CONCLUSION β-ecdysterone might alleviate osteoarthritis by activating autophagy in chondrocytes through regulating PI3K/AKT/mTOR signal pathway.
Collapse
Affiliation(s)
- Yanghua Tang
- Department of Orthopedics, Xiaoshan Hospital of Traditional Chinese MedicineNo. 156 Yucai Road, Xiaoshan District, Hangzhou, Zhejiang Province, China
| | - Yafeng Mo
- Department of Orthopedics, Third Clinical College of Zhejiang Chinese Medical UniversityNo. 548 Binwen Road, Binjiang District, Hangzhou, Zhejiang Province, China
| | - Dawei Xin
- Department of Orthopedics, Xiaoshan Hospital of Traditional Chinese MedicineNo. 156 Yucai Road, Xiaoshan District, Hangzhou, Zhejiang Province, China
| | - Linru Zeng
- Department of Orthopedics, Xiaoshan Hospital of Traditional Chinese MedicineNo. 156 Yucai Road, Xiaoshan District, Hangzhou, Zhejiang Province, China
| | - Zhenshuang Yue
- Department of Orthopedics, Xiaoshan Hospital of Traditional Chinese MedicineNo. 156 Yucai Road, Xiaoshan District, Hangzhou, Zhejiang Province, China
| | - Canda Xu
- Department of Orthopedics, Xiaoshan Hospital of Traditional Chinese MedicineNo. 156 Yucai Road, Xiaoshan District, Hangzhou, Zhejiang Province, China
| |
Collapse
|
12
|
Zhang L, Haddouti EM, Welle K, Burger C, Wirtz DC, Schildberg FA, Kabir K. The Effects of Biomaterial Implant Wear Debris on Osteoblasts. Front Cell Dev Biol 2020; 8:352. [PMID: 32582688 PMCID: PMC7283386 DOI: 10.3389/fcell.2020.00352] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/21/2020] [Indexed: 12/14/2022] Open
Abstract
Aseptic loosening subsequent to periprosthetic osteolysis is the leading cause for the revision of arthroplasty failure. The biological response of macrophages to wear debris has been well established, however, the equilibrium of bone remodeling is not only dictated by osteoclastic bone resorption but also by osteoblast-mediated bone formation. Increasing evidence shows that wear debris significantly impair osteoblastic physiology and subsequent bone formation. In the present review, we update the current state of knowledge regarding the effect of biomaterial implant wear debris on osteoblasts. The interaction of osteoblasts with osteoclasts and macrophages under wear debris challenge, and potential treatment options targeting osteoblasts are also presented.
Collapse
Affiliation(s)
- Li Zhang
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - El-Mustapha Haddouti
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Kristian Welle
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Christof Burger
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Dieter C Wirtz
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Frank A Schildberg
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Koroush Kabir
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
13
|
Biological Factors, Metals, and Biomaterials Regulating Osteogenesis through Autophagy. Int J Mol Sci 2020; 21:ijms21082789. [PMID: 32316424 PMCID: PMC7215394 DOI: 10.3390/ijms21082789] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/10/2020] [Accepted: 04/13/2020] [Indexed: 01/18/2023] Open
Abstract
Bone loss raises great concern in numerous situations, such as ageing and many diseases and in both orthopedic and dentistry fields of application, with an extensive impact on health care. Therefore, it is crucial to understand the mechanisms and the determinants that can regulate osteogenesis and ensure bone balance. Autophagy is a well conserved lysosomal degradation pathway, which is known to be highly active during differentiation and development. This review provides a revision of the literature on all the exogen factors that can modulate osteogenesis through autophagy regulation. Metal ion exposition, mechanical stimuli, and biological factors, including hormones, nutrients, and metabolic conditions, were taken into consideration for their ability to tune osteogenic differentiation through autophagy. In addition, an exhaustive overview of biomaterials, both for orthopedic and dentistry applications, enhancing osteogenesis by modulation of the autophagic process is provided as well. Already investigated conditions regulating bone regeneration via autophagy need to be better understood for finely tailoring innovative therapeutic treatments and designing novel biomaterials.
Collapse
|
14
|
Zheng X, Yu Y, Shao B, Gan N, Chen L, Yang D. Osthole improves therapy for osteoporosis through increasing autophagy of mesenchymal stem cells. Exp Anim 2019; 68:453-463. [PMID: 31155553 PMCID: PMC6842796 DOI: 10.1538/expanim.18-0178] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 04/11/2019] [Indexed: 12/27/2022] Open
Abstract
Osteoporosis is a common skeletal disorder resulting in elevated fracture risk. Improvement of osteogenic differentiation is thought to be the top priority in osteoporosis treatment projects. Significant characteristics of bone marrow mesenchymal stem cells (BMMSCs), especially attractive ability to differentiate into osteoblasts, have made them alternatives for osteoporosis treatment. However, therapeutic effect with BMMSCs remains to be improved. Here, osthole, a bioactive simple coumarin derivative extracted from many medicinal plants, was introduced to pre-stimulate BMMSCs and then applied in osteoporosis therapy. The results showed that osthole-treated-BMMSCs (OBMMSCs) brought a better outcome than BMMSCs alone in estrogen deficiency-induced osteoporosis model. And elevated autophagy level was suggested to be the underlying mechanism of the ability of osthole to promote osteoblast differentiation, which is indicated by the upregulation of protein and mRNA expression level of autophagy-associated genes, Beclin1 and LC3. We concluded from these experiments that OBMMSCs are more effective than BMMSCs in osteoporosis treatment maybe through upregulation level of autophagy level induced by osthole.
Collapse
Affiliation(s)
- Xuedan Zheng
- Department of Endodontics, Stomatological Hospital of Chongqing Medical University, No. 426 Songshi Bei Road, Yubei, 401147 Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, No. 426 Songshi Bei Road, Yubei, 401147 Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, No. 426 Songshi Bei Road, Yubei, 401147 Chongqing, China
| | - Yang Yu
- Department of Endodontics, Stomatological Hospital of Chongqing Medical University, No. 426 Songshi Bei Road, Yubei, 401147 Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, No. 426 Songshi Bei Road, Yubei, 401147 Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, No. 426 Songshi Bei Road, Yubei, 401147 Chongqing, China
| | - Binyi Shao
- Department of Endodontics, Stomatological Hospital of Chongqing Medical University, No. 426 Songshi Bei Road, Yubei, 401147 Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, No. 426 Songshi Bei Road, Yubei, 401147 Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, No. 426 Songshi Bei Road, Yubei, 401147 Chongqing, China
| | - Ning Gan
- Department of Endodontics, Stomatological Hospital of Chongqing Medical University, No. 426 Songshi Bei Road, Yubei, 401147 Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, No. 426 Songshi Bei Road, Yubei, 401147 Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, No. 426 Songshi Bei Road, Yubei, 401147 Chongqing, China
| | - Liang Chen
- Department of Endodontics, Stomatological Hospital of Chongqing Medical University, No. 426 Songshi Bei Road, Yubei, 401147 Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, No. 426 Songshi Bei Road, Yubei, 401147 Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, No. 426 Songshi Bei Road, Yubei, 401147 Chongqing, China
| | - Deqin Yang
- Department of Endodontics, Stomatological Hospital of Chongqing Medical University, No. 426 Songshi Bei Road, Yubei, 401147 Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, No. 426 Songshi Bei Road, Yubei, 401147 Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, No. 426 Songshi Bei Road, Yubei, 401147 Chongqing, China
| |
Collapse
|