1
|
Tandon B, Aguilar Cosme JR, Xue R, Srirussamee K, Aguilar-Tadeo J, Ballestrem C, Blaker JJ, Cartmell SH. Co-stimulation with piezoelectric PVDF films and low intensity pulsed ultrasound enhances osteogenic differentiation. BIOMATERIALS ADVANCES 2025; 173:214283. [PMID: 40086006 DOI: 10.1016/j.bioadv.2025.214283] [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: 01/03/2025] [Revised: 03/06/2025] [Accepted: 03/08/2025] [Indexed: 03/16/2025]
Abstract
Bone tissue engineering has emerged as a promising approach to address the challenges of bone fracture repair and regeneration. The application of external stimuli (mechanical and electrical) can drive specific cellular responses and osteogenic differentiation, leading to the development of more effective treatments. Piezoelectric materials modulate cellular proliferation and osteogenic differentiation under both static (without mechanical stimulation) and dynamic (with mechanical stimulation) conditions, activating distinct gene expression pathways. In this work, we investigate the combinatorial effect of poly (vinylidene fluoride) (PVDF) poled and non-poled films, and low-intensity pulsed ultrasound (LIPUS) on early-stage osteogenic differentiation of mouse pre-osteoblasts. Static culture with PVDF poled films enhanced Runx2 and Col1α1 expression without impacting alkaline phosphatase (ALP) activity. Inhibition of ERK phosphorylation using U0126 in PVDF poled films resulted in a ~ 6-8-fold increase in ALP activity, suggesting the involvement of an alternative pathway in osteogenic differentiation. Dynamic culture with LIPUS generated an electric potential of approximately 500 mV across PVDF films and an electrical field of 0-10 mV mm-1. Co-stimulation led to a ~3-fold increase of ALP activity on stimulated PVDF compared to unstimulated films. This study underscores the potential of piezoelectric materials as non-invasive electrical stimulators to enhance the efficacy of ultrasound-based therapies for bone fracture repair.
Collapse
Affiliation(s)
- Biranche Tandon
- Department of Materials Science, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK
| | - Jose R Aguilar Cosme
- Department of Materials Science, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK; Henry Royce Institute, The University of Manchester, Manchester, UK
| | - Ruikang Xue
- Department of Materials Science, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK; Department of Mechanical Engineering, Faculty of Engineering Science, University College London, London WC1E 7JE, UK
| | - Kasama Srirussamee
- Department of Materials Science, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK; Department of Biomedical Engineering, School of Engineering, KMITL, Bangkok 10520, Thailand
| | - Julio Aguilar-Tadeo
- Department of Materials Science, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK
| | - Christoph Ballestrem
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK
| | - Jonny J Blaker
- Department of Materials Science, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK; Henry Royce Institute, The University of Manchester, Manchester, UK
| | - Sarah H Cartmell
- Department of Materials Science, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK; Henry Royce Institute, The University of Manchester, Manchester, UK.
| |
Collapse
|
2
|
Li X, Su K, Zhao L, Zhang H, Yang Q, Du P, Chen X, Pan H. Unraveling osteogenesis mechanisms of the empowered VitaFlux adaptive regeneration biomaterials for bone tissue engineering: Insights into the role of BBGs/BSBGs. Bioact Mater 2025; 49:271-290. [PMID: 40130079 PMCID: PMC11932761 DOI: 10.1016/j.bioactmat.2025.03.006] [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: 11/09/2024] [Revised: 02/07/2025] [Accepted: 03/06/2025] [Indexed: 03/26/2025] Open
Abstract
Bone tissue engineering materials are crucial for bone repair, but existing repair materials still face many challenges, including poor biocompatibility and bioactivity, slow self-repair processes, limited adaptability, inability to promote angiogenesis and so on. To address these issues, the development of third-generation bone repair materials, which are being designed to stimulate specific cellular responses at the molecular level, such as borate and borosilicate bioactive glasses (BBGs/BSBGs) that activate cells and genes, offers new potential for promoting bone tissue self-renewing. Their unique characteristic lies in a flow of life-giving energy, releasing beneficial ions such as boron, calcium and silicon to stimulate cell proliferation and differentiation, accelerating the regeneration of bones. Through this dynamic repair mechanism, these VitaFlux glasses operate like a "living system" within the body, not only speeding up the healing of damaged tissues but also interacting seamlessly with surrounding tissues during the repair process. In this review, we provide a comprehensive analysis of the current understanding of the osteogenesis mechanisms of BBGs/BSBGs, emphasizing their interactions with cells, including ion release and exchange, protein adsorption, and cell adhesion. We also examine key osteogenic signaling pathways related to the alkaline and ionic microenvironments of BBGs/BSBGs, such as the cell cycle, Wnt, MAPK, and BMP signaling pathways, along with macrophage polarization and angiogenesis. Additionally, strategies and future prospects for advancing BBGs/BSBGs research are discussed. Special attention is given to the NaBC1 and GPCR-mediated signaling pathways, which require further investigation.
Collapse
Affiliation(s)
- Xian Li
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Kun Su
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Limin Zhao
- Shenzhen Healthemes Biotechnology Co. Ltd, Shenzhen, 518102, PR China
- Geriatric Medicine Department and General Medicine Department, Shenzhen Longhua District Central Hospital, Shenzhen, 518000, PR China
| | - Hao Zhang
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Qiang Yang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, PR China
| | - Ping Du
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Xiaofeng Chen
- National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou, 510006, PR China
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, PR China
- School of Medicine, Foshan University, Foshan, 528000, PR China
| | - Haobo Pan
- Shenzhen Key Laboratory of Marine Biomedical Materials, CAS-HK Joint Lab of Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
- Shenzhen Healthemes Biotechnology Co. Ltd, Shenzhen, 518102, PR China
| |
Collapse
|
3
|
Sodré LI, Gall MEC, Elias MDB, de Oliveira LO, Lobo FATF, Carias RBV, Teodoro AJ. Osteogenic Effects of Bioactive Compounds Found in Fruits on Mesenchymal Stem Cells: A Review. Nutr Rev 2025; 83:675-691. [PMID: 39862385 DOI: 10.1093/nutrit/nuae209] [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] [Indexed: 01/27/2025] Open
Abstract
Phytochemicals, which are bioactive compounds contained in fruits, vegetables, and teas, have a positive effect on human health by having anti-inflammatory, antioxidant, and anticarcinogenic effects. Several studies have highlighted the ability of bioactive compounds to activate key cellular enzymes associated with important signaling pathways related to cell division and proliferation, as well as their role in inflammatory and immunological responses. Some phytochemicals are associated with increased proliferation, differentiation, and expression of markers related to osteogenesis, bone formation, and mineralization by activating various signaling pathways. The objective of this study was to clarify which bioactive compounds present in fruits have osteogenic effects on mesenchymal stem cells and the possible associated mechanisms. A literature search was conducted in the LILACS, MEDLINE, and PubMed databases for pertinent articles published between 2014 and 2024. This review included 34 articles that report the osteogenic effects of various bioactive compounds found in different fruits. All the articles reported that phytochemicals play a role in enhancing the regenerative properties of mesenchymal cells, such as proliferation, osteogenic differentiation, secretion of angiogenic factors, and extracellular matrix formation. This review highlights the potential of these phytochemicals in the prevention and treatment of bone diseases. However, more studies are recommended to identify and quantify the therapeutic dose of phytochemicals, investigate their mechanisms in humans, and ensure their safety and effectiveness for health, particularly for bone health.
Collapse
Affiliation(s)
- Lia Igel Sodré
- Graduate Program in Science of Nutrition, Fluminense Federal University, Niterói, RJ 24020-140, Brazil
| | - Maria Eduarda Cordebello Gall
- Graduate Program in Biotechnology, National Institute of Metrology Standardization and Industrial Quality, Xerém, RJ 25250-020, Brazil
| | - Monique de Barros Elias
- Graduate Program in Food and Nutrition Security, Fluminense Federal University/Faculty of Nutrition, Niterói, RJ 24020-140, Brazil
| | - Luana Oeby de Oliveira
- Programa de Pós-Graduação em Ciências Aplicadas a Produtos para a Saúde (PPG-CAPS)/Fluminense Federal University, Faculty of Nutrition, Niteroi, RJ 24020-140, Brazil
| | | | - Rosana Bizon Vieira Carias
- Regenerative Medicine Laboratory, Centro Universitário Arthur Sá Earp Neto, Petrópolis Medical School, Petrópolis, RJ 25680-120, Brazil
| | - Anderson Junger Teodoro
- Universidade Federal Fluminense (Fluminense Federal University), Nutrition and Dietetics Department, Food and Nutrition Integrated Center, Niterói, RJ CEP 24020-140, Brazil
| |
Collapse
|
4
|
Hung GY, Wang CY, Feng KC, Tu CS, Cheng IC, Mana-Ay H, Hsiao HY, Lai PL, Chen PY. Manipulating Mg/Ca ratios in MgO-CaO-SiO 2 bioactive glass for achieving accelerated osteogenic differentiation of human adipose-derived stem cells. BIOMATERIALS ADVANCES 2025; 169:214189. [PMID: 39826260 DOI: 10.1016/j.bioadv.2025.214189] [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: 10/02/2024] [Revised: 12/30/2024] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
Cell-containing biomaterial is a promising material for treating nonunion or critical bone defect. Human adipose-derived stem cells (hADSCs) are suitable for bone repair due to their abundance in the abdomen, thighs, and buttocks. However, the low osteogenic capacities of hADSCs hinder their extended development for bone regeneration application. The present goal explores a novel MgO-CaO-SiO2 bioactive glass with suitable Mg/Ca ratios to enhance the osteogenic differentiation and bioactivity of hADSCs. The synthetic bioglass can be expressed as xMgO-(2-x)CaO-SiO2 (abbreviated as Mg(x)Ca(2-x)Si2, x = 0, 0.25, 0.5, 0.75, and 1). The expression levels of osteoblast-related genes (i.e., BMP2, RUNX2, DLX5, COL1A1, BGLAP2, and SPP1) were evaluated by reverse transcription-quantitative PCR (RT-PCR). The proteins involved in the p38/Akt/ERK signaling pathways were analyzed with Western blots. The results indicated that the extractions from the Mg(x)Ca(2-x)Si2 bioglass promoted hADSCs proliferation. Among the Mg(x)Ca(2-x)Si2 bioglass with different Mg/Ca ratios, the bioglass with a low Mg/Ca ratio (x = 0.25) presented greater osteogenic differentiation of hADSCs by promoting the p38 signaling pathway. Interestingly, the bioglass with low Mg/Ca ratio (x = 0.25) further presented on osteogenic potential with greater osteointegration in rat femoral defect model. This work provides the optimal Mg/Ca ratio in Mg(x)Ca(2-x)Si2 bioglass to promote the osteogenic induction of hADSCs and bone regeneration.
Collapse
Affiliation(s)
- Guan-Yi Hung
- International Ph.D. Program in Innovative Technology of Biomedical Engineering and Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan
| | - Chi-Yun Wang
- International Ph.D. Program in Innovative Technology of Biomedical Engineering and Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan
| | - Kuei-Chih Feng
- International Ph.D. Program in Innovative Technology of Biomedical Engineering and Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan; Department of Mechanical Engineering, Ming Chi University of Technology, New Taipei City 24301, Taiwan
| | - Chi-Shun Tu
- International Ph.D. Program in Innovative Technology of Biomedical Engineering and Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan; Department of Physics, Fu Jen Catholic University, New Taipei City 24205, Taiwan
| | - I-Chien Cheng
- International Ph.D. Program in Innovative Technology of Biomedical Engineering and Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan
| | - Haidee Mana-Ay
- International Ph.D. Program in Innovative Technology of Biomedical Engineering and Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan; Department of Physics, Silliman University, Dumaguete City 6200, Philippines
| | - Hui-Yi Hsiao
- International Ph.D. Program in Innovative Technology of Biomedical Engineering and Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan; Department of Biomedical Sciences, Chang Gung University, Taoyuan City 33305, Taiwan; Center for Tissue Engineering, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan.
| | - Po-Liang Lai
- International Ph.D. Program in Innovative Technology of Biomedical Engineering and Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan; Bone and Joint Research Center, Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Taoyuan City 33305, Taiwan; College of Medicine, Chang Gung University, Taoyuan City 33305, Taiwan.
| | - Pin-Yi Chen
- International Ph.D. Program in Innovative Technology of Biomedical Engineering and Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan; Department of Mechanical Engineering, Chang Gung University, Taoyuan City 33302, Taiwan; Research Center for Intelligent Medical Devices, Ming Chi University of Technology, New Taipei City 24301, Taiwan.
| |
Collapse
|
5
|
Wu Q, Zhou J, Du D, Guo B, Wang H, Lv W. Mongolian medicine Sugemule-7 decoction prevents osteoporosis via Erk1/2 and p38 MAPK signaling pathways according to network pharmacology analysis. Int J Biol Macromol 2025; 292:139166. [PMID: 39743063 DOI: 10.1016/j.ijbiomac.2024.139166] [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: 06/17/2024] [Revised: 11/27/2024] [Accepted: 12/07/2024] [Indexed: 01/04/2025]
Abstract
Osteoporosis (OP) is a significant global public health concern that requires the development of safe and effective drugs for prevention and treatment. Sugemule-7 (SGML-7) decoction, a traditional Mongolian herbal prescription, has long been used for treating OP, but its components and mechanisms of action remain unclear. The study identified the main compounds of SGML-7 using UHPLC-Q Exactive MS and explored the multi-target mechanisms of SGML-7 in OP through network pharmacology and molecular docking. A retinoic acid (RA)-induced mouse OP model was utilized to confirm the therapeutic effects and potential mechanism of SGML-7. Additionally, mouse pre-osteoblastic (MC3T3-E1) cells treated with SGML-7 medicated serum were employed to delve deeper into the molecular mechanisms. The UHPLC-Q Exactive MS analysis, network pharmacology, and molecular docking suggested that the synergistic effect of multiple active compounds could be the main contributor to SGML-7 for its anti-OP activities. Moreover, MAPK1, JUN, ESR1, TP53, AKT1, NCOA1, FOS, and NR3C1 were identified as potential key targets, and the MAPK signaling pathway was among the signaling pathways possibly involved in the anti-OP activities of SGML-7. Consistent with these findings, experimental studies confirmed that SGML-7 prevented bone loss, enhanced bone quality in OP mice, and promoted osteoblastic activity and bone formation in MC3T3-E1 cells by modulating MAPK-associated targets. Taken together, SGML-7 shows promise as an effective and appealing anti-OP drug candidate.
Collapse
Affiliation(s)
- Qijin Wu
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing, Jiangsu 211198, China.
| | - Jing Zhou
- Department of Veterinary Medicine, College of Animal Science and Technology, Hebei North University, Zhangjiakou, Hebei 075131, China
| | - Donghua Du
- Department of Veterinary Medicine, College of Animal Science and Technology, Hebei North University, Zhangjiakou, Hebei 075131, China
| | - Bing Guo
- Department of Veterinary Medicine, College of Animal Science and Technology, Hebei North University, Zhangjiakou, Hebei 075131, China
| | - Haifeng Wang
- Department of Veterinary Medicine, College of Animal Science and Technology, Hebei North University, Zhangjiakou, Hebei 075131, China
| | - Wenting Lv
- Department of Veterinary Medicine, College of Animal Science and Technology, Hebei North University, Zhangjiakou, Hebei 075131, China.
| |
Collapse
|
6
|
Yao Z, Huang W, Yang Y, Zou L, Zhang Y, Zhang J, Luo G. Investigation of the osteogenic effects of ICA and ICSII on rat bone marrow mesenchymal stem cells. Sci Rep 2025; 15:3060. [PMID: 39856132 PMCID: PMC11760372 DOI: 10.1038/s41598-025-86501-1] [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: 01/11/2024] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
Icariin (ICA) serves as the primary biologically active compound in traditional Chinese medicine Epimedium, while Icariside II (ICSII) represents one of its gastrointestinal metabolites. Although ICA and ICSII have demonstrated osteogenic differentiation- promoting effects on BMSCs, there is limited literature comparing their effects and underlying mechanisms. This study aimed to compare the osteogenic effects of Icariin and Icariside II, along with their respective osteogenic mechanisms. In this study, we initially determined the optimal concentrations of Icariin (10-5 mol/L) and Icariside II (10-6 mol/L) for inducing rBMSCs osteogenic differentiation using CCK8, ALP activity assay, and flow apoptosis assay. Subsequently, we compared the vascularization and osteogenic capacity of the two groups through alizarin red staining assay, Western Blot, and RT-PCR. Subsequently, we assessed the phosphorylated and non-phosphorylated expression of JNK, ERK1/2, P38, and AKT at different time intervals. We observed their phosphorylated expression and the expression of angiogenic/osteogenic markers after blocking with their corresponding inhibitors. It was observed that both the Icariin and Icariside II groups promoted the expression of osteogenic/angiogenic markers RUNX2, OCN, OPN, VEGF, and ANG1. While there was no significant difference in their osteogenic abilities, ICSII exhibited a stronger promotion of angiogenic differentiation markers, VEGF, compared to ICA. Additionally, it was observed that both ICA and ICSII could activate ERK1/2 phosphorylation, thereby further promoting the osteogenic/angiogenic differentiation of rBMSCs through the activation of the MAPK/ERK1/2 signaling pathway.
Collapse
Affiliation(s)
- Zhangshun Yao
- Kunming Medical University School and Hospital of Stomatology, Kunming, 650106, China
- Yunnan Key Laboratory of Stomatology, Kunming, 650106, China
| | - Weixiang Huang
- Kunming Medical University School and Hospital of Stomatology, Kunming, 650106, China
- Yunnan Key Laboratory of Stomatology, Kunming, 650106, China
| | - Yan Yang
- Kunming Medical University School and Hospital of Stomatology, Kunming, 650106, China
- Yunnan Key Laboratory of Stomatology, Kunming, 650106, China
| | - Leiyan Zou
- Kunming Medical University School and Hospital of Stomatology, Kunming, 650106, China
- Yunnan Key Laboratory of Stomatology, Kunming, 650106, China
| | - Yunpeng Zhang
- Kunming Medical University School and Hospital of Stomatology, Kunming, 650106, China
- Yunnan Key Laboratory of Stomatology, Kunming, 650106, China
| | - Jing Zhang
- Kunming Medical University School and Hospital of Stomatology, Kunming, 650106, China
- Yunnan Key Laboratory of Stomatology, Kunming, 650106, China
| | - Guangming Luo
- Kunming Medical University School and Hospital of Stomatology, Kunming, 650106, China.
- Yunnan Key Laboratory of Stomatology, Kunming, 650106, China.
| |
Collapse
|
7
|
Mouzoura P, Marazioti A, Gkartziou F, Metsiou DN, Antimisiaris SG. Potential of Liposomal FTY720 for Bone Regeneration: Proliferative, Osteoinductive, Chemoattractive, and Angiogenic Properties Compared to Free Bioactive Lipid. Int J Nanomedicine 2025; 20:239-265. [PMID: 39802384 PMCID: PMC11724662 DOI: 10.2147/ijn.s494512] [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: 10/09/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025] Open
Abstract
Introduction FTY720 bioactive lipid has proliferative, osteoinductive, chemo attractive, and angiogenic properties, being thus a potential exogenous administered agent for promotion of bone regeneration. Herein we developed FTY720-loaded liposomes as a potential delivery system that could retain and prolong the bioactivity of the bioactive lipid and at the same time reduce its cytotoxicity (at high doses). Methods FTY720 liposomes were prepared by thin-lipid hydration and microfluidic flow focusing, and evaluated for their ability to induce proliferation, osteoinduction, and chemoattraction in three cell types: MC3T3-E1 pre-osteoblast cells, L929 fibroblast cells, and ATDC5 chondrogenic cells. The angiogenic activity of free and liposomal FTY720 was investigated using a chick chorioallantoic membrane assay. NBD-FTY720 cellular uptake was quantitated using flow cytometry and morphologically assessed by confocal microscopy. Implicated cellular signaling mechanisms were investigated by quantifying phosphorylated MAPK and CREB proteins. Results FTY720 liposomes (~80-110 nm) with low polydispersity and ~100% loading were prepared using both methods. FTY720 demonstrated the ability to increase cell proliferation at 10-300nM doses but was cytotoxic at doses>400nM while the corresponding liposomal-FTY720 doses were non-cytotoxic, proving its reduced toxicity. In several cases (cells and doses), FTY720 liposomes demonstrated increased osteogenic differentiation of cells, proliferation, and migration compared to free FTY720, whereas both FTY720 forms demonstrated substantial angiogenic activity. Liposomal FTY720 cellular uptake was substantially higher than that of free FTY720 in some cases, a fact that may be connected to its higher bioactivity. Increased phosphorylated MAPK and CREB protein concentrations provided information about the potential cellular signaling mechanisms involved in FTY720-induced osteogenesis. Discussion The current results confirm the high potential of FTY720 bioactive lipid, especially in its liposomal form, that demonstrated substantial reduction of cytotoxicity and prolonged preservation of the lipids bioactivity (compared to the free lipid), for accelerated treatment of bone defects. Interestingly, the current studies prove the potential of FTY720, especially in its liposomal form, to promote reprogramming of L929 fibroblasts into osteoblasts, a novel finding deserving future exploitation.
Collapse
Affiliation(s)
- Panagiota Mouzoura
- Pharmaceutical Technology Laboratory, Department of Pharmacy, University of Patras, Rion, 26504, Greece
| | - Antonia Marazioti
- Pharmaceutical Technology Laboratory, Department of Pharmacy, University of Patras, Rion, 26504, Greece
- Laboratory of Basic Sciences, Department of Physiotherapy, University of the Peloponnese, Sparti, 23100, Greece
| | - Foteini Gkartziou
- Pharmaceutical Technology Laboratory, Department of Pharmacy, University of Patras, Rion, 26504, Greece
| | - Despoina-Nektaria Metsiou
- Pharmaceutical Technology Laboratory, Department of Pharmacy, University of Patras, Rion, 26504, Greece
| | - Sophia G Antimisiaris
- Pharmaceutical Technology Laboratory, Department of Pharmacy, University of Patras, Rion, 26504, Greece
- FORTH/ICE‑ΗΤ, Institute of Chemical Engineering Sciences, Platani, 26504, Greece
| |
Collapse
|
8
|
Su W, Zhang D, Wang Y, Lei L, Li H. G protein-coupled receptor 91 activations suppressed mineralization in Porphyromonas gingivalis-infected osteoblasts. Sci Rep 2024; 14:27606. [PMID: 39528607 PMCID: PMC11554824 DOI: 10.1038/s41598-024-78944-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Succinate receptor GPR91 is one of the G protein-coupled receptors (GPCRs) that interacts with various proteins to regulate diverse cellular functions such as cell morphology, apoptosis, and differentiation. In this study, we investigated whether the GPR91-mediated signaling pathway regulates mineralization in Porphyromonas gingivalis (P. gingivalis)-treated osteoblasts and to determine its potential role in osteoclast differentiation. Primary mouse osteoblasts from wild-type (WT) and GPR91 knockout (GPR91-/-) mice infected with P. gingivalis were used for in vitro experiments. The results showed that inhibition by 4C, a specific inhibitor, and GPR91 knockout promoted mineralization in P. gingivalis-infected osteoblasts. Surprisingly, GPR91 knockdown decreased the migration ability of osteoblasts. Moreover, compared with P. gingivalis-infected WT osteoblasts, GPR91-/- osteoblasts exhibited decreased RANKL production, and conditioned media (CM) from bacteria-infected GPR91-/- osteoblasts suppressed the formation of osteoclast precursors. Moreover, P. gingivalis mediated the role of GPR91 in osteoblast mineralization by activating the NF-κB pathway. These findings suggest that GPR91 activation reduces mineralization of P. gingivalis-infected osteoblasts and promotes osteoclastogenesis in macrophages. Therefore, targeting GPR91 may mitigate the loss of alveolar bone during bacterial infection.
Collapse
Affiliation(s)
- Wenqi Su
- Department of Periodontics, Nanjing Stomatological Hospital, Affiliated Hosptital of Medical School, Institute of Stomatology, Nanjing University, 30 Zhongyang Road, Nanjing, Jiangsu, 210008, People's Republic of China
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hosptital of Medical School, Institute of Stomatology, Nanjing University, Nanjing, People's Republic of China
| | - Dandan Zhang
- Department of Periodontics, Nanjing Stomatological Hospital, Affiliated Hosptital of Medical School, Institute of Stomatology, Nanjing University, 30 Zhongyang Road, Nanjing, Jiangsu, 210008, People's Republic of China
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hosptital of Medical School, Institute of Stomatology, Nanjing University, Nanjing, People's Republic of China
| | - Yujia Wang
- Department of Periodontics, Nanjing Stomatological Hospital, Affiliated Hosptital of Medical School, Institute of Stomatology, Nanjing University, 30 Zhongyang Road, Nanjing, Jiangsu, 210008, People's Republic of China
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hosptital of Medical School, Institute of Stomatology, Nanjing University, Nanjing, People's Republic of China
| | - Lang Lei
- Department of Orthodontics, Nanjing Stomatological Hospital, Affiliated Hosptital of Medical School, Institute of Stomatology, Nanjing University, Nanjing, People's Republic of China
| | - Houxuan Li
- Department of Periodontics, Nanjing Stomatological Hospital, Affiliated Hosptital of Medical School, Institute of Stomatology, Nanjing University, 30 Zhongyang Road, Nanjing, Jiangsu, 210008, People's Republic of China.
| |
Collapse
|
9
|
Guan Y, Zhang W, Mao Y, Li S. Nanoparticles and bone microenvironment: a comprehensive review for malignant bone tumor diagnosis and treatment. Mol Cancer 2024; 23:246. [PMID: 39487487 PMCID: PMC11529338 DOI: 10.1186/s12943-024-02161-1] [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: 09/23/2024] [Accepted: 10/17/2024] [Indexed: 11/04/2024] Open
Abstract
Malignant bone tumors, which are difficult to treat with current clinical strategies, originate from bone tissues and can be classified into primary and secondary types. Due to the specificity of the bone microenvironment, the results of traditional means of treating bone tumors are often unsatisfactory, so there is an urgent need to develop new treatments for malignant bone tumors. Recently, nanoparticle-based approaches have shown great potential in diagnosis and treatment. Nanoparticles (NPs) have gained significant attention due to their versatility, making them highly suitable for applications in bone tissue engineering, advanced imaging techniques, and targeted drug delivery. For diagnosis, NPs enhance imaging contrast and sensitivity by integrating targeting ligands, which significantly improve the specific recognition and localization of tumor cells for early detection. For treatment, NPs enable targeted drug delivery, increasing drug accumulation at tumor sites while reducing systemic toxicity. In conclusion, understanding bone microenvironment and using the unique properties of NPs holds great promise in improving disease management, enhancing treatment outcomes, and ultimately improving the quality of life for patients with malignant bone tumors. Further research and development will undoubtedly contribute to the advancement of personalized medicine in the field of bone oncology.
Collapse
Affiliation(s)
- Yujing Guan
- Second Ward of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, 110042, China
- The Liaoning Provincial Key Laboratory of Interdisciplinary Research on Gastrointestinal Tumor Combining Medicine with Engineering, Shenyang, Liaoning, 110042, China
- Institute of Cancer Medicine, Faculty of Medicine, Dalian University of Technology, Dalian, Liaoning, 116024, China
| | - Wei Zhang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province, 110016, P.R. China
| | - Yuling Mao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province, 110016, P.R. China.
| | - Shenglong Li
- Second Ward of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, 110042, China.
- The Liaoning Provincial Key Laboratory of Interdisciplinary Research on Gastrointestinal Tumor Combining Medicine with Engineering, Shenyang, Liaoning, 110042, China.
- Institute of Cancer Medicine, Faculty of Medicine, Dalian University of Technology, Dalian, Liaoning, 116024, China.
| |
Collapse
|
10
|
Wu Z, Li W, Jiang K, Lin Z, Qian C, Wu M, Xia Y, Li N, Zhang H, Xiao H, Bai J, Geng D. Regulation of bone homeostasis: signaling pathways and therapeutic targets. MedComm (Beijing) 2024; 5:e657. [PMID: 39049966 PMCID: PMC11266958 DOI: 10.1002/mco2.657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 06/22/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
As a highly dynamic tissue, bone is continuously rebuilt throughout life. Both bone formation by osteoblasts and bone resorption by osteoclasts constitute bone reconstruction homeostasis. The equilibrium of bone homeostasis is governed by many complicated signaling pathways that weave together to form an intricate network. These pathways coordinate the meticulous processes of bone formation and resorption, ensuring the structural integrity and dynamic vitality of the skeletal system. Dysregulation of the bone homeostatic regulatory signaling network contributes to the development and progression of many skeletal diseases. Significantly, imbalanced bone homeostasis further disrupts the signaling network and triggers a cascade reaction that exacerbates disease progression and engenders a deleterious cycle. Here, we summarize the influence of signaling pathways on bone homeostasis, elucidating the interplay and crosstalk among them. Additionally, we review the mechanisms underpinning bone homeostatic imbalances across diverse disease landscapes, highlighting current and prospective therapeutic targets and clinical drugs. We hope that this review will contribute to a holistic understanding of the signaling pathways and molecular mechanisms sustaining bone homeostasis, which are promising to contribute to further research on bone homeostasis and shed light on the development of targeted drugs.
Collapse
Affiliation(s)
- Zebin Wu
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Wenming Li
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Kunlong Jiang
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Zhixiang Lin
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Chen Qian
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Mingzhou Wu
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Yu Xia
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Ning Li
- Department of OrthopedicsCentre for Leading Medicine and Advanced Technologies of IHMDivision of Life Sciences and MedicineThe First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
| | - Hongtao Zhang
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Haixiang Xiao
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
- Department of OrthopedicsJingjiang People's HospitalSeventh Clinical Medical School of Yangzhou UniversityJingjiangJiangsu ProvinceChina
| | - Jiaxiang Bai
- Department of OrthopedicsCentre for Leading Medicine and Advanced Technologies of IHMDivision of Life Sciences and MedicineThe First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
| | - Dechun Geng
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| |
Collapse
|
11
|
Xu C, Wei Z, Dong X, Xing J, Meng X, Qiu Y, Zhou H, Zheng W, Xu Z, Huang S, Xia W, Lv L, Jiang H, Wang W, Zhao X, Liu Z, Akimoto Y, Zhao B, Wang S, Hu Z. A p38 MAP kinase inhibitor suppresses osteoclastogenesis and alleviates ovariectomy-induced bone loss through the inhibition of bone turnover. Biochem Pharmacol 2024; 226:116391. [PMID: 38914317 DOI: 10.1016/j.bcp.2024.116391] [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: 01/11/2024] [Revised: 05/26/2024] [Accepted: 06/21/2024] [Indexed: 06/26/2024]
Abstract
Inhibition of excessive osteoclastic activity is an efficient therapeutic strategy for many bone diseases induced by increased bone resorption, such as osteoporosis. BMS-582949, a clinical p38α inhibitor, is a promising drug in Phase II studies for treating rheumatoid arthritis. However, its function on bone resorption is largely unknown. In this study, we find that BMS-582949 represses RANKL-induced osteoclast differentiation in a dose-dependent manner. Moreover, BMS-582949 inhibits osteoclastic F-actin ring formation and osteoclast-specific gene expression. Mechanically, BMS-582949 treatment attenuates RANKL-mediated osteoclastogenesis through mitogen-activated protein kinases (MAPKs) and protein kinase B (AKT) signaling pathways without disturbing nuclear factor-κB (NF-κB) signaling. Interestingly, BMS-582949 impairs osteoclastic mitochondrial biogenesis and functions, such as oxidative phosphorylation (OXPHOS). Furthermore, BMS-582949 administration prevents bone loss in ovariectomized mouse mode by inhibiting both bone resorption and bone formation in vivo. Taken together, these findings indicate that BMS-582949 may be a potential and effective drug for the therapy of osteolytic diseases.
Collapse
Affiliation(s)
- Cheng Xu
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei 430056, China.
| | - Zhixin Wei
- Hubei Engineering Research Center for Protection and Utilization of Special Biological Resources in the Hanjiang River Basin, School of Life Sciences, Jianghan University, Wuhan, Hubei 430056, China
| | - Xiaoyu Dong
- Hubei Engineering Research Center for Protection and Utilization of Special Biological Resources in the Hanjiang River Basin, School of Life Sciences, Jianghan University, Wuhan, Hubei 430056, China
| | - Junqiao Xing
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei 430056, China
| | - Xiangrui Meng
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei 430056, China
| | - Yaxuan Qiu
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei 430056, China
| | - Huimei Zhou
- Hubei Engineering Research Center for Protection and Utilization of Special Biological Resources in the Hanjiang River Basin, School of Life Sciences, Jianghan University, Wuhan, Hubei 430056, China
| | - Wenrui Zheng
- Hubei Engineering Research Center for Protection and Utilization of Special Biological Resources in the Hanjiang River Basin, School of Life Sciences, Jianghan University, Wuhan, Hubei 430056, China
| | - Zhenyu Xu
- Hubei Engineering Research Center for Protection and Utilization of Special Biological Resources in the Hanjiang River Basin, School of Life Sciences, Jianghan University, Wuhan, Hubei 430056, China
| | - Shanhua Huang
- Hubei Engineering Research Center for Protection and Utilization of Special Biological Resources in the Hanjiang River Basin, School of Life Sciences, Jianghan University, Wuhan, Hubei 430056, China
| | - Wenwen Xia
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei 430056, China
| | - Longfei Lv
- Hubei Engineering Research Center for Protection and Utilization of Special Biological Resources in the Hanjiang River Basin, School of Life Sciences, Jianghan University, Wuhan, Hubei 430056, China
| | - Haochen Jiang
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei 430056, China
| | - Weihua Wang
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei 430056, China
| | - Xue Zhao
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei 430056, China
| | - Zixuan Liu
- Gogdel Cranleigh High School, Wuhan, Hubei 430312, China
| | | | - Baohong Zhao
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA; Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| | - Siyuan Wang
- Department of Medicinal Chemistry, College of Pharmacy, Shenzhen Technology University, Shenzhen, Guangdong 518118, China.
| | - Zhangfeng Hu
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei 430056, China; Hubei Engineering Research Center for Protection and Utilization of Special Biological Resources in the Hanjiang River Basin, School of Life Sciences, Jianghan University, Wuhan, Hubei 430056, China; Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, Jianghan University, Wuhan, Hubei 430056, China.
| |
Collapse
|
12
|
Yang X, Chen M, Wang S, Hu X, Zhou J, Yuan H, Zhu E, Wang B. Cortactin controls bone homeostasis through regulating the differentiation of osteoblasts and osteoclasts. Stem Cells 2024; 42:662-674. [PMID: 38655781 DOI: 10.1093/stmcls/sxae031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 04/10/2024] [Indexed: 04/26/2024]
Abstract
Cortactin (CTTN), a cytoskeletal protein and substrate of Src kinase, is implicated in tumor aggressiveness. However, its role in bone cell differentiation remains unknown. The current study revealed that CTTN was upregulated during osteoblast and adipocyte differentiation. Functional experiments demonstrated that CTTN promoted the in vitro differentiation of mesenchymal stem/progenitor cells into osteogenic and adipogenic lineages. Mechanistically, CTTN was able to stabilize the protein level of mechanistic target of rapamycin kinase (mTOR), leading to the activation of mTOR signaling. In-depth investigation revealed that CTTN could bind with casitas B lineage lymphoma-c (c-CBL) and counteract the function of c-CBL, a known E3 ubiquitin ligase responsible for the proteasomal degradation of mTOR. Silencing c-Cbl alleviated the impaired differentiation of osteoblasts and adipocytes caused by CTTN siRNA, while silencing mTOR mitigated the stimulation of osteoblast and adipocyte differentiation induced by CTTN overexpression. Notably, transplantation of CTTN-silenced bone marrow stromal cells (BMSCs) into the marrow of mice led to a reduction in trabecular bone mass, accompanied by a decrease in osteoblasts and an increase in osteoclasts. Furthermore, CTTN-silenced BMSCs expressed higher levels of receptor activator of nuclear factor κB ligand (RANKL) than control BMSCs did and promoted osteoclast differentiation when cocultured with bone marrow-derived osteoclast precursor cells. This study provides evidence that CTTN favors osteoblast differentiation by counteracting the c-CBL-induced degradation of mTOR and inhibits osteoclast differentiation by downregulating the expression of RANKL. It also suggests that maintaining an appropriate level of CTTN expression may be advantageous for maintaining bone homeostasis.
Collapse
Affiliation(s)
- Xiaoli Yang
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, People's Republic of China
| | - Meng Chen
- Department of hematology, the First Affiliated Hospital of Nanchang University, Nanchang 330006, People's Republic of China
| | - Shuang Wang
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, People's Republic of China
| | - Xingli Hu
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, People's Republic of China
| | - Jie Zhou
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, People's Republic of China
| | - Hairui Yuan
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, People's Republic of China
| | - Endong Zhu
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, People's Republic of China
| | - Baoli Wang
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, People's Republic of China
| |
Collapse
|
13
|
Sun Z, Liu K, Liang C, Wen L, Wu J, Liu X, Li X. Diosmetin as a promising natural therapeutic agent: In vivo, in vitro mechanisms, and clinical studies. Phytother Res 2024; 38:3660-3694. [PMID: 38748620 DOI: 10.1002/ptr.8214] [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: 11/08/2023] [Revised: 04/09/2024] [Accepted: 04/13/2024] [Indexed: 07/12/2024]
Abstract
Diosmetin, a natural occurring flavonoid, is primarily found in citrus fruits, beans, and other plants. Diosmetin demonstrates a variety of pharmacological activities, including anticancer, antioxidant, anti-inflammatory, antibacterial, metabolic regulation, cardiovascular function improvement, estrogenic effects, and others. The process of literature search was done using PubMed, Web of Science and ClinicalTrials databases with search terms containing Diosmetin, content, anticancer, anti-inflammatory, antioxidant, pharmacological activity, pharmacokinetics, in vivo, and in vitro. The aim of this review is to summarize the in vivo, in vitro and clinical studies of Diosmetin over the last decade, focusing on studies related to its anticancer, anti-inflammatory, and antioxidant activities. It is found that DIO has significant therapeutic effects on skin and cardiovascular system diseases, and its research in pharmacokinetics and toxicology is summarized. It provides the latest information for researchers and points out the limitations of current research and areas that should be strengthened in future research, so as to facilitate the relevant scientific research and clinical application of DIO.
Collapse
Affiliation(s)
- Zihao Sun
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kai Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chuipeng Liang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lin Wen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jijiao Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaolian Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaofang Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
14
|
Shang G, Lei L, Peng C. Bioinformatics Study on Mechanism of Postnatal Development of Craniofacial Bone. J Craniofac Surg 2024; 35:1368-1371. [PMID: 38847500 DOI: 10.1097/scs.0000000000010354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 05/04/2024] [Indexed: 07/24/2024] Open
Abstract
OBJECTIVE The postnatal development of craniofacial bone plays a crucial role in shaping the overall structure and functionality of the skull and face. Understanding the underlying mechanisms of this intricate process is essential for both clinical and research purposes. In this study, the authors conducted a bioinformatics analysis using the Gene Expression Omnibus database to investigate the molecular pathways and regulatory networks involved in the postnatal development of craniofacial bone. METHODS In this study, the online Gene Expression Omnibus microarray expression profiling data set GSE27976 was used to identify differentially expressed genes (DEGs) in different age groups. Protein-Protein Interaction network analyses, functional enrichment, and hub genes analysis were performed. The differences in immune infiltration and microenvironment among different types of cells were also analyzed. RESULTS In total, 523 DEGs, including 287 upregulated and 236 downregulated genes, were identified. GO and KEGG analysis showed that the DEGs were significantly enriched in multiple signaling pathways, such as skeletal system morphogenesis, osteoblast differentiation, and stem cell differentiation. Immune infiltration and microenvironment characteristics analysis showed that there were significant differences in fibroblasts, mesenchymal stem cell, osteoblast, stroma score, and microenvironment score between the two groups. Five hub genes, including IGF1, IL1B, ICAM1, MMP2 , and brain-derived neurotrophic factor, were filled out. CONCLUSION The findings of this study showed a significant shift in gene expression towards osteogenesis during the first 12 months after birth. These findings emphasize the critical role of the postnatal period in craniofacial bone development and provide valuable insights into the molecular mechanisms underlying this process.
Collapse
Affiliation(s)
- Guangling Shang
- Department of Liver Transplantation and Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University
| | - Liu Lei
- Department of Burns and Plastic Surgery, The Second Hospital of Shandong University, Shandong University
| | - Changliang Peng
- Department of Spine Surgery, The Second Hospital of Shandong University, Shandong University, Jinan, Shandong Province, People's Republic of China
| |
Collapse
|
15
|
Zhong Q, Wang D, Mai H, Chen R, Xu Y, Lei M, Xie J, Tang Z, Fu J, Chen Y, Wang J, Shi Z, Cheng H. Injectable thermo-responsive Poloxamer hydrogel/methacrylate gelatin microgels stimulates bone regeneration through biomimetic programmed release of SDF-1a and IGF-1. Int J Biol Macromol 2024; 271:132742. [PMID: 38821297 DOI: 10.1016/j.ijbiomac.2024.132742] [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: 02/20/2024] [Revised: 05/22/2024] [Accepted: 05/27/2024] [Indexed: 06/02/2024]
Abstract
Injectable hydrogels, offering adaptable drug delivery of growth factors (GFs), hold promise for treating bone defects. To optimize osteogenic efficacy, the release of GFs should mirror the natural bone healing. We developed an injectable thermo-responsive hydrogel/microgels platform for dual GF delivery for bone regeneration. Stromal cell-derived factor-1 alpha (SDF-1a) and the Methacrylate Gelatin (GelMA) microgels which encapsulated insulin-like growth factor-1 (IGF-1) loaded liposomes (Ls) were introduced into Poloxamer 407 (P407) hydrogel matrix. This system achieved the biomimetic release profile of SDF-1a and IGF-1, which covered the early stage from day 1 to 7 and the continuous stage from day 5 to 21, respectively. In vitro study confirmed the enhanced migration, osteogenic biomarker expression, and matrix mineralization of the bone marrow mesenchymal stem cells (BMSCs) co-cultivated with the dual GFs delivering hydrogel/microgels. Transcriptome sequencing revealed that the potential mechanism was associated with mitogen-activated protein kinase (MAPK) signaling activation and its downstream ribosomal protein S6 kinase 2 (RSK2) upregulation. In a critical-sized calvarial defect model in Sprague-Dawley (SD) rats, the injectable hydrogel/microgels system promoted significant bone regeneration. Collectively, our study suggested the current hydrogel/microgels system with the biomimetic release of SDF-1a and IGF-1 efficiently promoted bone regeneration, informing the future development of GF delivery systems intended for bone regeneration therapies.
Collapse
Affiliation(s)
- Qiang Zhong
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, 1838 # Guangzhou North Avenue, Guangzhou, Guangdong Province 510515, People's Republic of China
| | - Ding Wang
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, 1838 # Guangzhou North Avenue, Guangzhou, Guangdong Province 510515, People's Republic of China
| | - Huaming Mai
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, 1838 # Guangzhou North Avenue, Guangzhou, Guangdong Province 510515, People's Republic of China
| | - Rong Chen
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, 1838 # Guangzhou North Avenue, Guangzhou, Guangdong Province 510515, People's Republic of China
| | - Yixin Xu
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, 1838 # Guangzhou North Avenue, Guangzhou, Guangdong Province 510515, People's Republic of China
| | - Mingyuan Lei
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, 1838 # Guangzhou North Avenue, Guangzhou, Guangdong Province 510515, People's Republic of China
| | - Jiajun Xie
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, 1838 # Guangzhou North Avenue, Guangzhou, Guangdong Province 510515, People's Republic of China
| | - Zinan Tang
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, 1838 # Guangzhou North Avenue, Guangzhou, Guangdong Province 510515, People's Republic of China
| | - Jinlang Fu
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, 1838 # Guangzhou North Avenue, Guangzhou, Guangdong Province 510515, People's Republic of China
| | - Yuhang Chen
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, 1838 # Guangzhou North Avenue, Guangzhou, Guangdong Province 510515, People's Republic of China
| | - Jian Wang
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, 1838 # Guangzhou North Avenue, Guangzhou, Guangdong Province 510515, People's Republic of China.
| | - Zhanjun Shi
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, 1838 # Guangzhou North Avenue, Guangzhou, Guangdong Province 510515, People's Republic of China.
| | - Hao Cheng
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, 1838 # Guangzhou North Avenue, Guangzhou, Guangdong Province 510515, People's Republic of China.
| |
Collapse
|
16
|
Vyavahare S, Ahluwalia P, Gupta SK, Kolhe R, Hill WD, Hamrick M, Isales CM, Fulzele S. The Role of Aryl Hydrocarbon Receptor in Bone Biology. Int J Tryptophan Res 2024; 17:11786469241246674. [PMID: 38757095 PMCID: PMC11097734 DOI: 10.1177/11786469241246674] [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: 08/28/2023] [Accepted: 03/25/2024] [Indexed: 05/18/2024] Open
Abstract
Aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor, is crucial in maintaining the skeletal system. Our study focuses on encapsulating the role of AhR in bone biology and identifying novel signaling pathways in musculoskeletal pathologies using the GEO dataset. The GEO2R analysis identified 8 genes (CYP1C1, SULT6B1, CYB5A, EDN1, CXCR4B, CTGFA, TIPARP, and CXXC5A) involved in the AhR pathway, which play a pivotal role in bone remodeling. The AhR knockout in hematopoietic stem cells showed alteration in several novel bone-related transcriptomes (eg, Defb14, ZNF 51, and Chrm5). Gene Ontology Enrichment Analysis demonstrated 54 different biological processes associated with bone homeostasis. Mainly, these processes include bone morphogenesis, bone development, bone trabeculae formation, bone resorption, bone maturation, bone mineralization, and bone marrow development. Employing Functional Annotation and Clustering through DAVID, we further uncovered the involvement of the xenobiotic metabolic process, p450 pathway, oxidation-reduction, and nitric oxide biosynthesis process in the AhR signaling pathway. The conflicting evidence of current research of AhR signaling on bone (positive and negative effects) homeostasis may be due to variations in ligand binding affinity, binding sites, half-life, chemical structure, and other unknown factors. In summary, our study provides a comprehensive understanding of the underlying mechanisms of the AhR pathway in bone biology.
Collapse
Affiliation(s)
- Sagar Vyavahare
- Department of Medicine, Augusta University, Augusta, GA, USA
| | | | | | - Ravindra Kolhe
- Department of Pathology, Augusta University, Augusta, GA, USA
| | - William D Hill
- Department of Pathology, Medical University of South Carolina, Charleston, SC, USA
| | - Mark Hamrick
- Department of Cell Biology and Anatomy, Augusta University, Augusta, GA, USA
- Center for Healthy Aging, Augusta University, Augusta, GA, USA
| | - Carlos M Isales
- Department of Medicine, Augusta University, Augusta, GA, USA
- Center for Healthy Aging, Augusta University, Augusta, GA, USA
| | - Sadanand Fulzele
- Department of Medicine, Augusta University, Augusta, GA, USA
- Department of Cell Biology and Anatomy, Augusta University, Augusta, GA, USA
- Center for Healthy Aging, Augusta University, Augusta, GA, USA
| |
Collapse
|
17
|
Park EJ, Truong VL, Jeong WS, Min WK. Brain-Derived Neurotrophic Factor (BDNF) Enhances Osteogenesis and May Improve Bone Microarchitecture in an Ovariectomized Rat Model. Cells 2024; 13:518. [PMID: 38534361 PMCID: PMC10969057 DOI: 10.3390/cells13060518] [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: 01/08/2024] [Revised: 03/09/2024] [Accepted: 03/14/2024] [Indexed: 03/28/2024] Open
Abstract
BACKGROUND Brain-derived neurotrophic factor (BDNF) has gained attention as a therapeutic agent due to its potential biological activities, including osteogenesis. However, the molecular mechanisms involved in the osteogenic activity of BDNF have not been fully understood. This study aimed to investigate the action of BDNF on the osteoblast differentiation in bone marrow stromal cells, and its influence on signaling pathways. In addition, to evaluate the clinical efficacy, an in vivo animal study was performed. METHODS Preosteoblast cells (MC3T3-E1), bone marrow-derived stromal cells (ST2), and a direct 2D co-culture system were treated with BDNF. The effect of BDNF on cell proliferation was determined using the CCK-8 assay. Osteoblast differentiation was assessed based on alkaline phosphatase (ALP) activity and staining and the protein expression of multiple osteoblast markers. Calcium accumulation was examined by Alizarin red S staining. For the animal study, we used ovariectomized Sprague-Dawley rats and divided them into BDNF and normal saline injection groups. MicroCT, hematoxylin and eosin (H&E), and tartrate-resistant acid phosphatase (TRAP) stain were performed for analysis. RESULTS BDNF significantly increased ALP activity, calcium deposition, and the expression of osteoblast differentiation-related proteins, such as ALP, osteopontin, etc., in both ST-2 and the MC3T3-E1 and ST-2 co-culture systems. Moreover, the effect of BDNF on osteogenic differentiation was diminished by blocking tropomyosin receptor kinase B, as well as inhibiting c-Jun N-terminal kinase and p38 MAPK signals. Although the animal study results including bone density and histology showed increased osteoblastic and decreased osteoclastic activity, only a portion of parameters reached statistical significance. CONCLUSIONS Our study results showed that BDNF affects osteoblast differentiation through TrkB receptor, and JNK and p38 MAPK signal pathways. Although not statistically significant, the trend of such effects was observed in the animal experiment.
Collapse
Affiliation(s)
- Eugene J. Park
- Department of Orthopedic Surgery, Kyungpook National University Hospital, College of Medicine, Kyungpook National University, Daegu 41566, Republic of Korea;
| | - Van-Long Truong
- Food and Bio-Industry Research Institute, School of Food Science & Biotechnology, College of Agriculture and Life Sciences, Kyungpook National University, Daegu 41566, Republic of Korea;
| | - Woo-Sik Jeong
- Food and Bio-Industry Research Institute, School of Food Science & Biotechnology, College of Agriculture and Life Sciences, Kyungpook National University, Daegu 41566, Republic of Korea;
| | - Woo-Kie Min
- Department of Orthopedic Surgery, Kyungpook National University Hospital, College of Medicine, Kyungpook National University, Daegu 41566, Republic of Korea;
| |
Collapse
|
18
|
Wang TH, Watanabe K, Hamada N, Tani-Ishii N. Role of MAPKs in TGF-β1-induced maturation and mineralization in human osteoblast-like cells. J Oral Biosci 2024; 66:61-67. [PMID: 38110177 DOI: 10.1016/j.job.2023.12.003] [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: 10/06/2023] [Revised: 12/14/2023] [Accepted: 12/14/2023] [Indexed: 12/20/2023]
Abstract
OBJECTIVES Our study aimed to clarify the role of mitogen-activated protein kinases (MAPKs) in transforming growth factor (TGF)-β1-stimulated mineralization in the human osteoblast-like MG63 cells. METHODS The viability of MG63 cells under TGF-β1 stimulation was assessed by MTS assay. Western blotting determined TGF-β1-mediated activation of extracellular signal-related protein kinase (ERK), p38, and c-Jun amino-terminal kinase (JNK). Mineralization-related gene expression was examined by quantitative real-time PCR, and mineral deposition levels were evaluated by alizarin red S staining. RESULTS TGF-β1 had no effect on MG63 cell proliferation. Activation of p38 was observed at 3 h post TGF-β1 stimulation. Moreover, JNK phosphorylation was upregulated by TGF-β1 from 1 to 6 h post stimulation, but had no activation on ERK phosphorylation throughout the experimental period. Treatment with JNK inhibitor diminished the alizarin red S-stained area in a dose-dependent manner. Mineral deposition was unaffected by MEK inhibitor, whereas p38 inhibitor increased the red-stained area. Gene expression levels of ALP and BSP were significantly decreased under treatment with JNK inhibitor and p38 inhibitor. The MEK inhibitor had no effect on the TGF-β1-mediated upregulation of ALP and BSP. Although all three inhibitors suppressed expression of COL I, none were found to stimulate expression of OCN. CONCLUSIONS Human osteoblast-like MG63 cells maturation and mineralization are induced through JNK activation of MAPK signaling in response to TGF-β1.
Collapse
Affiliation(s)
- Ting-Hsuan Wang
- Department of Pulp Biology and Endodontics, Graduate School of Dentistry, Kanagawa Dental University, 82 Inaoka-cho, Yokosuka, 238-8580, Japan
| | - Kiyoko Watanabe
- Department of Liberal Arts Education, Kanagawa Dental University, 82 Inaoka-cho, Yokosuka, 238-8580, Japan
| | - Nobushiro Hamada
- Department of Oral Microbiology, Graduate School of Dentistry, Kanagawa Dental University, 82 Inaoka-cho, Yokosuka, 238-8580, Japan
| | - Nobuyuki Tani-Ishii
- Department of Pulp Biology and Endodontics, Graduate School of Dentistry, Kanagawa Dental University, 82 Inaoka-cho, Yokosuka, 238-8580, Japan.
| |
Collapse
|
19
|
Zhang YY, Xie N, Sun XD, Nice EC, Liou YC, Huang C, Zhu H, Shen Z. Insights and implications of sexual dimorphism in osteoporosis. Bone Res 2024; 12:8. [PMID: 38368422 PMCID: PMC10874461 DOI: 10.1038/s41413-023-00306-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 11/04/2023] [Accepted: 11/27/2023] [Indexed: 02/19/2024] Open
Abstract
Osteoporosis, a metabolic bone disease characterized by low bone mineral density and deterioration of bone microarchitecture, has led to a high risk of fatal osteoporotic fractures worldwide. Accumulating evidence has revealed that sexual dimorphism is a notable feature of osteoporosis, with sex-specific differences in epidemiology and pathogenesis. Specifically, females are more susceptible than males to osteoporosis, while males are more prone to disability or death from the disease. To date, sex chromosome abnormalities and steroid hormones have been proven to contribute greatly to sexual dimorphism in osteoporosis by regulating the functions of bone cells. Understanding the sex-specific differences in osteoporosis and its related complications is essential for improving treatment strategies tailored to women and men. This literature review focuses on the mechanisms underlying sexual dimorphism in osteoporosis, mainly in a population of aging patients, chronic glucocorticoid administration, and diabetes. Moreover, we highlight the implications of sexual dimorphism for developing therapeutics and preventive strategies and screening approaches tailored to women and men. Additionally, the challenges in translating bench research to bedside treatments and future directions to overcome these obstacles will be discussed.
Collapse
Affiliation(s)
- Yuan-Yuan Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Na Xie
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Xiao-Dong Sun
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Yih-Cherng Liou
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, 117543, Republic of Singapore
| | - Canhua Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Huili Zhu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, Department of Reproductive Medicine, West China Second University Hospital of Sichuan University, Chengdu, China.
| | - Zhisen Shen
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China.
| |
Collapse
|
20
|
Zhao D, Saiding Q, Li Y, Tang Y, Cui W. Bone Organoids: Recent Advances and Future Challenges. Adv Healthc Mater 2024; 13:e2302088. [PMID: 38079529 DOI: 10.1002/adhm.202302088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 11/23/2023] [Indexed: 12/21/2023]
Abstract
Bone defects stemming from tumorous growths, traumatic events, and diverse conditions present a profound conundrum in clinical practice and research. While bone has the inherent ability to regenerate, substantial bone anomalies require bone regeneration techniques. Bone organoids represent a new concept in this field, involving the 3D self-assembly of bone-associated stem cells guided in vitro with or without extracellular matrix material, resulting in a tissue that mimics the structural, functional, and genetic properties of native bone tissue. Within the scientific panorama, bone organoids ascend to an esteemed status, securing significant experimental endorsement. Through a synthesis of current literature and pioneering studies, this review offers a comprehensive survey of the bone organoid paradigm, delves into the quintessential architecture and ontogeny of bone, and highlights the latest progress in bone organoid fabrication. Further, existing challenges and prospective directions for future research are identified, advocating for interdisciplinary collaboration to fully harness the potential of this burgeoning domain. Conclusively, as bone organoid technology continues to mature, its implications for both clinical and research landscapes are poised to be profound.
Collapse
Affiliation(s)
- Ding Zhao
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Qimanguli Saiding
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Yihan Li
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Yunkai Tang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| |
Collapse
|
21
|
Xiao Y, Xie X, Chen Z, Yin G, Kong W, Zhou J. Advances in the roles of ATF4 in osteoporosis. Biomed Pharmacother 2023; 169:115864. [PMID: 37948991 DOI: 10.1016/j.biopha.2023.115864] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/01/2023] [Accepted: 11/07/2023] [Indexed: 11/12/2023] Open
Abstract
Osteoporosis (OP) is characterized by reduced bone mass, decreased strength, and enhanced bone fragility fracture risk. Activating transcription factor 4 (ATF4) plays a role in cell differentiation, proliferation, apoptosis, redox balance, amino acid uptake, and glycolipid metabolism. ATF4 induces the differentiation of bone marrow mesenchymal stem cells (BM-MSCs) into osteoblasts, increases osteoblast activity, and inhibits osteoclast formation, promoting bone formation and remodeling. In addition, ATF4 mediates the energy metabolism in osteoblasts and promotes angiogenesis. ATF4 is also involved in the mediation of adipogenesis. ATF4 can selectively accumulate in osteoblasts. ATF4 can directly interact with RUNT-related transcription factor 2 (RUNX2) and up-regulate the expression of osteocalcin (OCN) and osterix (Osx). Several upstream factors, such as Wnt/β-catenin and BMP2/Smad signaling pathways, have been involved in ATF4-mediated osteoblast differentiation. ATF4 promotes osteoclastogenesis by mediating the receptor activator of nuclear factor κ-B (NF-κB) ligand (RANKL) signaling. Several agents, such as parathyroid (PTH), melatonin, and natural compounds, have been reported to regulate ATF4 expression and mediate bone metabolism. In this review, we comprehensively discuss the biological activities of ATF4 in maintaining bone homeostasis and inhibiting OP development. ATF4 has become a therapeutic target for OP treatment.
Collapse
Affiliation(s)
- Yaosheng Xiao
- Department of Orthopaetics, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Xunlu Xie
- Department of Pathology, Ganzhou People's Hospital, Ganzhou 341000, China
| | - Zhixi Chen
- Department of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Guoqiang Yin
- Ganzhou Hospital Affiliated to Nanchang University, Ganzhou 341000, China
| | - Weihao Kong
- Department of Joint Surgery, Ganzhou People's Hospital, Ganzhou 341000, China
| | - Jianguo Zhou
- Department of Joint Surgery, Ganzhou People's Hospital, Ganzhou 341000, China.
| |
Collapse
|
22
|
Sugimoto A, Iwata K, Kurogoushi R, Tanaka M, Nakashima Y, Yamakawa Y, Oishi A, Yoshizaki K, Fukumoto S, Yamamoto A, Ishimaru N, Iwamoto T. C-terminus of PIEZO1 governs Ca 2+ influx and intracellular ERK1/2 signaling pathway in mechanotransduction. Biochem Biophys Res Commun 2023; 682:39-45. [PMID: 37801988 DOI: 10.1016/j.bbrc.2023.09.080] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 09/25/2023] [Indexed: 10/08/2023]
Abstract
Cells sense and respond to extracellular mechanical stress through mechanotransduction receptors and ion channels, which regulate cellular behaviors such as cell proliferation and differentiation. Among them, PIEZO1, piezo-type mechanosensitive ion channel component 1, has recently been highlighted as a mechanosensitive ion channel in various cell types including mesenchymal stem cells. We previously reported that PIEZO1 is essential for ERK1/2 phosphorylation and osteoblast differentiation in bone marrow-derived mesenchymal stem cells (BMSCs), induced by hydrostatic pressure loading and treatment with the PIEZO1-specific activator Yoda1. However, the molecular mechanism underlying how PIEZO1 induces mechanotransduction remains unclear. In this study, we investigated that the role of the C-terminus in regulating extracellular Ca2+ influx and activating the ERK1/2 signaling pathway. We observed the activation of Fluo-4 AM in the Yoda1-stimulated human BMSC line UE7T-13, but not in a calcium-depleted cell culture medium. Similarly, Western blotting analysis revealed that Yoda1 treatment induced ERK1/2 phosphorylation, but this induction was not observed in calcium-depleted cell culture medium. To investigate the functional role of the C-terminus of PIEZO1, we generated HEK293 cells stably expressing the full-length mouse PIEZO1 (PIEZO1-FL) and a deletion-type PIEZO1 lacking the C-terminal intracellular region containing the R-Ras-binding domain (PIEZO1-ΔR-Ras). We found that Yoda1 treatment predominantly activated Flou-4 AM and ERK1/2 in PIEZO1-FL-trasfected cells but neither in PIEZO1-ΔR-Ras-transfected cells nor control cells. Our results indicate that the C-terminus of PIEZO1, which contains the R-Ras binding domain, plays an essential role in Ca2+ influx and activation of the ERK1/2 signaling pathway, suggesting that this domain is crucial for the mechanotransduction of osteoblastic differentiation in BMSCs.
Collapse
Affiliation(s)
- Asuna Sugimoto
- Department of Pediatric Dentistry / Special Needs Dentistry, Division of Oral Health Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, 113-8549, Japan; Department of Pediatric Dentistry, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, 770-8504, Japan
| | - Kokoro Iwata
- Department of Pediatric Dentistry / Special Needs Dentistry, Division of Oral Health Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, 113-8549, Japan
| | - Rika Kurogoushi
- Department of Pediatric Dentistry / Special Needs Dentistry, Division of Oral Health Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, 113-8549, Japan
| | - Manami Tanaka
- Department of Pediatric Dentistry / Special Needs Dentistry, Division of Oral Health Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, 113-8549, Japan
| | - Yumiko Nakashima
- Department of Pediatric Dentistry / Special Needs Dentistry, Division of Oral Health Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, 113-8549, Japan
| | - Yoshihito Yamakawa
- Department of Pediatric Dentistry, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, 770-8504, Japan
| | - Atsushi Oishi
- Department of Pediatric Dentistry / Special Needs Dentistry, Division of Oral Health Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, 113-8549, Japan
| | - Keigo Yoshizaki
- Orthodontics and Dentofacial Orthopedics Section, Division of Oral Health, Growth and Development, Kyushu University Faculty of Dental Science, Fukuoka, 812-8582, Japan
| | - Satoshi Fukumoto
- Pediatric Dentistry Section, Division of Oral Health, Growth and Development, Kyushu University Faculty of Dental Science, Fukuoka, 812-8582, Japan
| | - Akihito Yamamoto
- Department of Tissue Regeneration, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, 770-8504, Japan
| | - Naozumi Ishimaru
- Department of Oral Molecular Pathology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, 770-8504, Japan
| | - Tsutomu Iwamoto
- Department of Pediatric Dentistry / Special Needs Dentistry, Division of Oral Health Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, 113-8549, Japan.
| |
Collapse
|
23
|
He XYX, Zhao WL, Yao LP, Sun P, Cheng G, Liu YL, Yu Y, Liu Y, Wang TJ, Zhang QY, Qin LP, Zhang QL. Orcinol glucoside targeted p38 as an agonist to promote osteogenesis and protect glucocorticoid-induced osteoporosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 119:154953. [PMID: 37573809 DOI: 10.1016/j.phymed.2023.154953] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 07/01/2023] [Accepted: 07/06/2023] [Indexed: 08/15/2023]
Abstract
BACKGROUND Glucocorticoids (GC)-induced osteoporosis (GIOP) is the most common cause of secondary osteoporosis, which leads to an increased risk of fracture in patients. The inhibition of the osteoblast effect is one of the main pathological characteristics of GIOP, but without effective drugs on treatment. PURPOSE The aim of this study was to investigate the potential effects of orcinol glucoside (OG) on osteoblast cells and GIOP mice, as well as the mechanism of the underlying molecular target protein of OG both in vitro osteoblast cell and in vivo GIOP mice model. METHODS GIOP mice were used to determine the effect of OG on bone density and bone formation. Then, a cellular thermal shift assay coupled with mass spectrometry (CETSA-MS) method was used to identify the target of OG. Surface plasmon resonance (SPR), enzyme activity assay, molecular docking, and molecular dynamics were used to detect the affinity, activity, and binding site between OG and its target, respectively. Finally, the anti-osteoporosis effect of OG through the target signal pathway was investigated in vitro osteoblast cell and in vivo GIOP mice model. RESULTS OG treatment increased bone mineral density (BMD) in GIOP mice and effectively promoted osteoblast proliferation, osteogenic differentiation, and mineralization in vitro. The CETSA-MS result showed that the target of OG acting on the osteoblast is the p38 protein. SPR, molecular docking assay and enzyme activity assay showed that OG could direct bind to the p38 protein and is a p38 agonist. The cellular study found that OG could promote p38 phosphorylation and upregulate the proteins expression of its downstream osteogenic (Runx2, Osx, Collagen Ⅰ, Dlx5). Meanwhile, it could also inhibit the nuclear transport of GR by increasing the phosphorylation site at GR226 in osteoblast cell. In vivo GIOP mice experiment further confirmed that OG could prevent bone loss in the GIOP mice model through promoting p38 activity as well as its downstream proteins expression and activity. CONCLUSIONS This study has established that OG could promote osteoblast activity and revise the bone loss in GIOP mice by direct binding to the p38 protein and is a p38 agonist to improve its downstream signaling, which has great potential in GIOP treatment for targeting p38. This is the first report to identify OG anti-osteoporosis targets using a label-free strategy (CETSA-MS).
Collapse
Affiliation(s)
- Xin-Yun-Xi He
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Wan-Lu Zhao
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Li-Ping Yao
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Peng Sun
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Gang Cheng
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Yu-Ling Liu
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Yang Yu
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Yan Liu
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Teng-Jian Wang
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Qiao-Yan Zhang
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China.
| | - Lu-Ping Qin
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China.
| | - Quan-Long Zhang
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China.
| |
Collapse
|
24
|
Abstract
Osteoarthritis (OA) is mainly caused by ageing, strain, trauma, and congenital joint abnormalities, resulting in articular cartilage degeneration. During the pathogenesis of OA, the changes in subchondral bone (SB) are not only secondary manifestations of OA, but also an active part of the disease, and are closely associated with the severity of OA. In different stages of OA, there were microstructural changes in SB. Osteocytes, osteoblasts, and osteoclasts in SB are important in the pathogenesis of OA. The signal transduction mechanism in SB is necessary to maintain the balance of a stable phenotype, extracellular matrix (ECM) synthesis, and bone remodelling between articular cartilage and SB. An imbalance in signal transduction can lead to reduced cartilage quality and SB thickening, which leads to the progression of OA. By understanding changes in SB in OA, researchers are exploring drugs that can regulate these changes, which will help to provide new ideas for the treatment of OA.
Collapse
Affiliation(s)
- Pan Luo
- Department of Joint Surgery, HongHui Hospital, Xi’an Jiaotong University, Xi'an, China
| | - Qi-ling Yuan
- Department of Joint Surgery, HongHui Hospital, Xi’an Jiaotong University, Xi'an, China
| | - Mingyi Yang
- Department of Joint Surgery, HongHui Hospital, Xi’an Jiaotong University, Xi'an, China
| | - Xianjie Wan
- Department of Joint Surgery, HongHui Hospital, Xi’an Jiaotong University, Xi'an, China
| | - Peng Xu
- Department of Joint Surgery, HongHui Hospital, Xi’an Jiaotong University, Xi'an, China
| |
Collapse
|
25
|
Song X, Dong C, Man X. Phosphorylated MAPK11 promotes the progression of clear cell renal cell carcinoma by maintaining RUNX2 protein abundance. J Cell Mol Med 2023; 27:2583-2593. [PMID: 37525479 PMCID: PMC10468653 DOI: 10.1111/jcmm.17870] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 06/20/2023] [Accepted: 07/16/2023] [Indexed: 08/02/2023] Open
Abstract
Previous studies have demonstrated that mitogen-activated protein kinase 11 (MAPK11) functions as an important point of integration in signalling transduction pathways and controlling endocellular processes, including viability of cells, differentiation, proliferation and apoptosis, through the sequence phosphorylation of the substrate protein Ser/Thr kinase protein cascade. Though MAPK 11 plays an important role in various tumours, especially in the invasive and metastatic processes, its expression and molecular mechanism in clear cell renal cell carcinoma (ccRCC) remain unclear. Runt-associated transcription factor 2 (RUNX2), a main transcription factor for osteoblast differentiation and chondrocyte maturation, has high expression in a number of tumours. In this study, the mRNA and protein levels of targeted genes in ccRCC tissues and adjacent tissues are analysed using the Cancer Genome Atlas (TCGA) database and western blotting. The ccRCC cell proliferation was measured with colony formation and EdU assay, and cell migration was examined through transwell assay. The interactive behaviour between proteins was detected with immunoprecipitation. Half-life period of RUNX2 protein was measured with cycloheximide chase assay. The results of the study indicated overexpression of MAPK11 and RUNX2 in ccRCC tissues and cell lines. MAPK11 and RUNX2 promoted the ccRCC cell proliferation and migration. Additionally, physical interaction took place between RUNX2 and P-MAPK11, which functioned to sustain the stability of RUNX2 protein. The high expression of RUNX2 could neutralize the functional degradation in MAPK11. And the outcomes of the study suggest that the P-MAPK11/RUNX2 axis may be used as a potential therapeutic target of ccRCC.
Collapse
Affiliation(s)
- Xiandong Song
- Department of UrologyThe First Hospital of China Medical UniversityShenyangLiaoningChina
| | - Changming Dong
- Department of UrologyThe First Hospital of China Medical UniversityShenyangLiaoningChina
| | - Xiaojun Man
- Department of UrologyThe First Hospital of China Medical UniversityShenyangLiaoningChina
| |
Collapse
|
26
|
Agriesti F, Landini F, Tamma M, Pacelli C, Mazzoccoli C, Calice G, Ruggieri V, Capitanio G, Mori G, Piccoli C, Capitanio N. Bioenergetic profile and redox tone modulate in vitro osteogenesis of human dental pulp stem cells: new perspectives for bone regeneration and repair. Stem Cell Res Ther 2023; 14:215. [PMID: 37608350 PMCID: PMC10463344 DOI: 10.1186/s13287-023-03447-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 08/10/2023] [Indexed: 08/24/2023] Open
Abstract
BACKGROUND Redox signaling and energy metabolism are known to be involved in controlling the balance between self-renewal and proliferation/differentiation of stem cells. In this study we investigated metabolic and redox changes occurring during in vitro human dental pulp stem cells (hDPSCs) osteoblastic (OB) differentiation and tested on them the impact of the reactive oxygen species (ROS) signaling. METHODS hDPSCs were isolated from dental pulp and subjected to alkaline phosphatase and alizarin red staining, q-RT-PCR, and western blotting analysis of differentiation markers to assess achievement of osteogenic/odontogenic differentiation. Moreover, a combination of metabolic flux analysis and confocal cyto-imaging was used to profile the metabolic phenotype and to evaluate the redox tone of hDPSCs. RESULTS In differentiating hDPSCs we observed the down-regulation of the mitochondrial respiratory chain complexes expression since the early phase of the process, confirmed by metabolic flux analysis, and a reduction of the basal intracellular peroxide level in its later phase. In addition, dampened glycolysis was observed, thereby indicating a lower energy-generating phenotype in differentiating hDPSCs. Treatment with the ROS scavenger Trolox, applied in the early-middle phases of the process, markedly delayed OB differentiation of hDPSCs assessed as ALP activity, Runx2 expression, mineralization capacity, expression of stemness and osteoblast marker genes (Nanog, Lin28, Dspp, Ocn) and activation of ERK1/2. In addition, the antioxidant partly prevented the inhibitory effect on cell metabolism observed following osteogenic induction. CONCLUSIONS Altogether these results provided evidence that redox signaling, likely mediated by peroxide species, influenced the stepwise osteogenic expansion/differentiation of hDPSCs and contributed to shape its accompanying metabolic phenotype changes thus improving their efficiency in bone regeneration and repair.
Collapse
Affiliation(s)
- Francesca Agriesti
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy
| | - Francesca Landini
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Mirko Tamma
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Consiglia Pacelli
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Carmela Mazzoccoli
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy
| | - Giovanni Calice
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy
| | - Vitalba Ruggieri
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy
- Clinical Pathology Unit, “Madonna delle Grazie’’ Hospital, Matera, Italy
| | - Giuseppe Capitanio
- Department of Translational Biomedicine and Neuroscience “DiBraiN”, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Giorgio Mori
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Claudia Piccoli
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Nazzareno Capitanio
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| |
Collapse
|
27
|
Yallowitz AR, Shim JH, Xu R, Greenblatt MB. An angiogenic approach to osteoanabolic therapy targeting the SHN3-SLIT3 pathway. Bone 2023; 172:116761. [PMID: 37030497 PMCID: PMC10198948 DOI: 10.1016/j.bone.2023.116761] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/30/2023] [Accepted: 04/05/2023] [Indexed: 04/10/2023]
Abstract
Often, disorders of impaired bone formation involve not only a cell intrinsic defect in the ability of osteoblasts to form bone, but moreover a broader dysfunction of the skeletal microenvironment that limits osteoblast activity. Developing approaches to osteoanabolic therapy that not only augment osteoblast activity but moreover correct this microenvironmental dysfunction may enable both more effective osteoanabolic therapies and also addressing a broader set of indications where vasculopathy or other forms microenvironment dysfunction feature prominently. We here review evidence that SHN3 acts as a suppressor of not only the cell intrinsic bone formation activity of osteoblasts, but moreover of the creation of a local osteoanabolic microenvironment. Mice lacking Schnurri3 (SHN3, HIVEP3) display a very robust increase in bone formation, that is due to de-repression of ERK pathway signaling in osteoblasts. In addition to loss of SHN3 augmenting the differentiation and bone formation activity of osteoblasts, loss of SHN3 increases secretion of SLIT3 by osteoblasts, which in a skeletal context acts as an angiogenic factor. Through this angiogenic activity, SLIT3 creates an osteoanabolic microenvironment, and accordingly treatment with SLIT3 can increase bone formation and enhance fracture healing. These features both validate vascular endothelial cells as a therapeutic target for disorders of low bone mass alongside the traditionally targeted osteoblasts and osteoclasts and indicate that targeting the SHN3/SLIT3 pathway provides a new mechanism to induce therapeutic osteoanabolic responses.
Collapse
Affiliation(s)
- Alisha R Yallowitz
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY 10065, United States of America
| | - Jae-Hyuck Shim
- Department of Medicine, Division of Rheumatology, University of Massachusetts Chan Medical School, USA; Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Ren Xu
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cells, State Key Laboratory of Cellular Stress Biology, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361005, China; Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Matthew B Greenblatt
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY 10065, United States of America; Research Division, Hospital for Special Surgery, New York, NY 10065, United States of America.
| |
Collapse
|
28
|
Wang X, Zhu X, Wang D, Li X, Wang J, Yin G, Huang Z, Pu X. Identification of a Specific Phage as Growth Factor Alternative Promoting the Recruitment and Differentiation of MSCs in Bone Tissue Regeneration. ACS Biomater Sci Eng 2023; 9:2426-2437. [PMID: 37023478 DOI: 10.1021/acsbiomaterials.2c01538] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Abstract
Inefficient use and loss of exogenously implanted mesenchymal stem cells (MSCs) are major concerns in MSCs-based bone tissue engineering. It is a promising approach to overcome the above issues by recruiting and regulation of endogenous MSCs. However, there are few substances that can recruit MSCs effectively and specifically to the site of bone injury. In this study, we identified a phage clone (termed P11) with specific affinity for MSCs through phage display biopanning, and further investigated the effects of P11 on the cytological behavior of MSCs and macrophages. The results showed that P11 could bind MSCs specifically and promote the proliferation and migration of MSCs. Meanwhile, P11 could polarize macrophages to the M1 phenotype and significantly changed their morphology, which further enhanced the chemotaxis of MSCs. Additionally, RNA-seq results revealed that P11 could promote the secretion of osteogenesis-related markers in MSCs through the TPL2-MEK-ERK signaling pathway. Altogether, P11 has great potential to be used as growth factor alternatives in bone tissue engineering, with the advantages of cheaper and stable activity. Our study also advances the understanding of the effects of phages on macrophages and MSCs, and provides a new idea for the development in the field of phage-based tissue engineering.
Collapse
Affiliation(s)
- Xingming Wang
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Xiupeng Zhu
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, People's Republic of China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, People's Republic of China
| | - Danni Wang
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Xiaoxu Li
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Juan Wang
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Guangfu Yin
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Zhongbing Huang
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| | - Ximing Pu
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, People's Republic of China
| |
Collapse
|
29
|
Tantalum as Trabecular Metal for Endosseous Implantable Applications. Biomimetics (Basel) 2023; 8:biomimetics8010049. [PMID: 36810380 PMCID: PMC9944482 DOI: 10.3390/biomimetics8010049] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
During the last 20 years, tantalum has known ever wider applications for the production of endosseous implantable devices in the orthopedic and dental fields. Its excellent performances are due to its capacity to stimulate new bone formation, thus improving implant integration and stable fixation. Tantalum's mechanical features can be mainly adjusted by controlling its porosity thanks to a number of versatile fabrication techniques, which allow obtaining an elastic modulus similar to that of bone tissue, thus limiting the stress-shielding effect. The present paper aims at reviewing the characteristics of tantalum as a solid and porous (trabecular) metal, with specific regard to biocompatibility and bioactivity. Principal fabrication methods and major applications are described. Moreover, the osteogenic features of porous tantalum are presented to testify its regenerative potential. It can be concluded that tantalum, especially as a porous metal, clearly possesses many advantageous characteristics for endosseous applications but it presently lacks the consolidated clinical experience of other metals such as titanium.
Collapse
|
30
|
Wang M, Liu M, Zheng J, Xiong L, Wang P. Exendin-4 regulates the MAPK and WNT signaling pathways to alleviate the osteogenic inhibition of periodontal ligament stem cells in a high glucose environment. Open Med (Wars) 2023; 18:20230692. [PMID: 37034502 PMCID: PMC10080709 DOI: 10.1515/med-2023-0692] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 03/01/2023] [Accepted: 03/14/2023] [Indexed: 04/11/2023] Open
Abstract
Diabetes mellitus (DM) increases the destruction of periodontal tissue and impairs osteogenesis differentiation. Exendin-4 (Ex-4), a glucagon-like peptide-1 (GLP-1) analogue, can be used for treating DM and promotes bone regeneration. The aim of this study was to explore the effect and mechanism of Ex-4 on improving the osteogenesis of periodontal ligament stem cells (PDLSCs) in a high glucose environment. Alkaline phosphatase staining and alizarin red staining were used to detect the osteogenic differentiation of PDLSCs. The results showed that 10 nM Ex-4 could reduce the osteogenesis inhibition of PDLSCs induced by high glucose. RT-PCR and western blot results showed that Ex-4 increased the osteogenesis-related gene expression of ALP, Runx2, and Osx, and upregulated the phosphorylation of P38, JNK, and ERK1/2; the peak effect was observed in the range 0.5-1.0 h. Mitogen-activated protein kinase (MAPK) inhibitors PD98059, SB203580, and SP600125 blocked the effects of Ex-4 on MAPK activation and decreased the expression of ALP, Runx2, and Osx in PDLSCs. Moreover, after Ex-4 treatment, the total β-catenin, p-GSK3β, LEF, and Runx2 protein levels increased under normal or high glucose environments. In conclusion, our results indicated that Ex-4 regulates the MAPK and WNT signaling pathways to alleviate the osteogenic inhibition of PDLSCs in a high glucose environment.
Collapse
Affiliation(s)
- Min Wang
- Department of Stomatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Min Liu
- Department of Stomatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jiawen Zheng
- Department of Stomatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Li Xiong
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ping Wang
- Department of Stomatology, The First Affiliated Hospital of Chongqing Medical University, Youyi
Road 1, Chongqing, 400016, China
| |
Collapse
|
31
|
Feng Y, Han Z, Jiang W, Shen H, Yu Y, Zhou N, Huang X. Promotion of osteogenesis in BMSC under hypoxia by ATF4 via the PERK-eIF2α signaling pathway. In Vitro Cell Dev Biol Anim 2022; 58:886-897. [PMID: 36378269 DOI: 10.1007/s11626-022-00732-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 10/25/2022] [Indexed: 11/16/2022]
Abstract
Mandibular distraction osteogenesis (MDO) is an endogenous tissue engineering technology in which bone marrow mesenchymal stem cells (BMSC) play a key role in MDO-related osteogenesis. Activating transcription factor 4 (ATF4) is involved in osteogenesis through activation of PERK (Protein kinase R-like endoplasmic reticulum kinase) in endoplasmic reticulum stress (ERS) condition under hypoxia. However, the specific role of ATF4 in MDO with BMSC remains unknown. The aim of this study was to explore the effects of ATF4 in MDO with BMSC under hypoxia. Briefly, canine BMSCs were cultured in a hypoxic chamber, and effects of hypoxia were evaluated using cell migration assay and Alizarin Red S staining. Expression levels of protein kinase R-like endoplasmic reticulum kinase, eukaryotic translation initiation factor 2α, ATF4, osteocalcin, and bone sialoprotein were evaluated using quantitative polymerase chain reaction and western blotting. BMSCs were transduced with the ATF4-small interfering RNA lentivirus. The effects were evaluated using all the aforementioned experiments. The results showed that hypoxia promoted migration, osteoblast differentiation, and ATF4 expression in BMSC. ATF4 knockdown in BMSC significantly inhibited migration and osteoblast differentiation abilities, while hypoxia reversed these effects to some extent. In addition, the molecular mechanism partly depended on the ERS signaling pathway, with ATF4 as the key factor. In summary, we presented a novel mechanism of ATF4-mediated regulation of BMSC under hypoxia.
Collapse
Affiliation(s)
- Yuan Feng
- Guangxi Medical University, Nanning, People's Republic of China
- Department of Oral and Maxillofacial Surgery, College of Stomatology, Hospital of Stomatology, Guangxi Medical University, 10 Shuangyong Road, Nanning, Guangxi, 530021, People's Republic of China
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, People's Republic of China
| | - Zhiqi Han
- Guangxi Medical University, Nanning, People's Republic of China
- Department of Oral and Maxillofacial Surgery, College of Stomatology, Hospital of Stomatology, Guangxi Medical University, 10 Shuangyong Road, Nanning, Guangxi, 530021, People's Republic of China
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, People's Republic of China
| | - Weidong Jiang
- Guangxi Medical University, Nanning, People's Republic of China
- Department of Oral and Maxillofacial Surgery, College of Stomatology, Hospital of Stomatology, Guangxi Medical University, 10 Shuangyong Road, Nanning, Guangxi, 530021, People's Republic of China
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, People's Republic of China
| | - Huijuan Shen
- Guangxi Medical University, Nanning, People's Republic of China
- Department of Oral and Maxillofacial Surgery, College of Stomatology, Hospital of Stomatology, Guangxi Medical University, 10 Shuangyong Road, Nanning, Guangxi, 530021, People's Republic of China
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, People's Republic of China
| | - Yangyang Yu
- Guangxi Medical University, Nanning, People's Republic of China
- Department of Oral and Maxillofacial Surgery, College of Stomatology, Hospital of Stomatology, Guangxi Medical University, 10 Shuangyong Road, Nanning, Guangxi, 530021, People's Republic of China
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, People's Republic of China
| | - Nuo Zhou
- Guangxi Medical University, Nanning, People's Republic of China.
- Department of Oral and Maxillofacial Surgery, College of Stomatology, Hospital of Stomatology, Guangxi Medical University, 10 Shuangyong Road, Nanning, Guangxi, 530021, People's Republic of China.
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, People's Republic of China.
| | - Xuanping Huang
- Guangxi Medical University, Nanning, People's Republic of China.
- Department of Oral and Maxillofacial Surgery, College of Stomatology, Hospital of Stomatology, Guangxi Medical University, 10 Shuangyong Road, Nanning, Guangxi, 530021, People's Republic of China.
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Clinical Research Center for Craniofacial Deformity, Nanning, People's Republic of China.
| |
Collapse
|
32
|
Betaine promotes osteogenic differentiation in immortalized human dental pulp-derived cells. BDJ Open 2022; 8:31. [PMID: 36207319 PMCID: PMC9546879 DOI: 10.1038/s41405-022-00123-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 09/07/2022] [Accepted: 09/12/2022] [Indexed: 11/05/2022] Open
Abstract
Objectives This study aimed to evaluate the effect of betaine (BET) on immortalized human dental pulp stem cell (ihDP) osteogenic differentiation. Materials and methods hDPs were immortalized using SV40 T-antigen transfection. Characterization, multilineage differentiation, proliferation, cell cycle, colony-forming unit, and cellular senescence were evaluated (n = 4). The effect of BET on ihDP response was assessed (n = 4). Osteogenic differentiation was detected using ALP, ARS staining, and RT-qPCR (n = 4). To investigate the involvement of calcium signaling, the cells were pretreated with either 8-(NN-diethylamino)octyl-3,4,5-trimethoxybenzoate (TMB-8) or thapsigargin before BET treatment (n = 6). Results ihDPs retained similar phenotypic characteristics presented in hDPs but exhibited an increase in cell proliferation and extended culture to passage 25. An increased proportion of cells in S and G2/M phases without senescence was observed in ihDPs. BET (50 mM) treatment significantly increased mineral deposition at 14 days and upregulated ALP, MSX2, BMP2, and RUNX2 expression. TMB-8 pretreatment reduced the effect of BET-induced ihDP osteogenic differentiation, whereas thapsigargin promoted osteogenic differentiation in ihDPs synergistically with BET. Conclusion ihDPs showed superior proliferation ability and a longer life span, which could serve as a promising cell for regenerative dentistry. BET promoted odonto/osteogenic differentiation via intracellular calcium regulation.
Collapse
|
33
|
Jiang S, Yin C, Dang K, Zhang W, Huai Y, Qian A. Comprehensive ceRNA network for MACF1 regulates osteoblast proliferation. BMC Genomics 2022; 23:695. [PMID: 36207684 PMCID: PMC9541005 DOI: 10.1186/s12864-022-08910-0] [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: 04/20/2022] [Accepted: 09/26/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Previous studies have shown that microtubule actin crosslinking factor 1 (MACF1) can regulate osteoblast proliferation and differentiation through non-coding RNA (ncRNA) in bone-forming osteoblasts. However, the role of MACF1 in targeting the competing endogenous RNA (ceRNA) network to regulate osteoblast differentiation remains poorly understood. Here, we profiled messenger RNA (mRNA), microRNA (miRNA), and long ncRNA (lncRNA) expression in MACF1 knockdown MC3TC‑E1 pre‑osteoblast cells. RESULTS In total, 547 lncRNAs, 107 miRNAs, and 376 mRNAs were differentially expressed. Significantly altered lncRNAs, miRNAs, and mRNAs were primarily found on chromosome 2. A lncRNA-miRNA-mRNA network was constructed using a bioinformatics computational approach. The network indicated that mir-7063 and mir-7646 were the most potent ncRNA regulators and mef2c was the most potent target gene. Pathway enrichment analysis showed that the fluid shear stress and atherosclerosis, p53 signaling, and focal adhesion pathways were highly enriched and contributed to osteoblast proliferation. Importantly, the fluid shear stress and atherosclerosis pathway was co-regulated by lncRNAs and miRNAs. In this pathway, Dusp1 was regulated by AK079370, while Arhgef2 was regulated by mir-5101. Furthermore, Map3k5 was regulated by AK154638 and mir-466q simultaneously. AK003142 and mir-3082-5p as well as Ak141402 and mir-446 m-3p were identified as interacting pairs that regulate target genes. CONCLUSION This study revealed the global expression profile of ceRNAs involved in the differentiation of MC3TC‑E1 osteoblasts induced by MACF1 deletion. These results indicate that loss of MACF1 activates a comprehensive ceRNA network to regulate osteoblast proliferation.
Collapse
Affiliation(s)
- Shanfeng Jiang
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
| | - Chong Yin
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
- Department of Clinical Laboratory, Academician (expert) workstation, Lab of epigenetics and RNA therapy, Affiliated Hospital of North Sichuan Medical College, 637000, Nanchong, China
| | - Kai Dang
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
| | - Wenjuan Zhang
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
| | - Ying Huai
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
| | - Airong Qian
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China.
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China.
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China.
| |
Collapse
|
34
|
Yuan H, Ikegame M, Fukuhara Y, Takemoto F, Yu Y, Teramachi J, Weng Y, Guo J, Yamada D, Takarada T, Li Y, Okamura H, Zhang B. Vestigial-Like 3 Plays an Important Role in Osteoblast Differentiation by Regulating the Expression of Osteogenic Transcription Factors and BMP Signaling. Calcif Tissue Int 2022; 111:331-344. [PMID: 35750933 DOI: 10.1007/s00223-022-00997-7] [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: 02/14/2022] [Accepted: 06/01/2022] [Indexed: 11/02/2022]
Abstract
Our previous gene profiling analysis showed that the transcription cofactor vestigial-like 3 (VGLL3) gene expression was upregulated by mechanical tension in the mouse cranial suture, coinciding with accelerated osteoblast differentiation. Therefore, we hypothesized that VGLL3 plays a significant role in osteogenic differentiation. To clarify the function of VGLL3 in osteoblasts, we examined its expression characteristics in mouse bone tissue and the osteoblastic cell line MC3T3-E1. We further examined the effects of Vgll3 knockdown on osteoblast differentiation and bone morphogenetic protein (BMP) signaling. In the mouse cranial suture, where membranous ossification occurs, VGLL3 was immunohistochemically detected mostly in the nucleus of osteoblasts, preosteoblasts, and fibroblastic cells. VGLL3 expression in MC3T3-E1 cells was transient and peaked at a relatively early stage of differentiation. RNA sequencing revealed that downregulated genes in Vgll3-knockdown cells were enriched in gene ontology terms associated with osteoblast differentiation. Interestingly, most of the upregulated genes were related to cell division. Targeted Vgll3 knockdown markedly suppressed the expression of major osteogenic transcription factors (Runx2, Sp7/osterix, and Dlx5) and osteoblast differentiation. It also attenuated BMP signaling; moreover, exogenous BMP2 partially restore osteogenic transcription factors' expression in Vgll3-knockdown cells. Furthermore, overexpression of Vgll3 increased the expression of osteogenic transcription factors. These results suggest that VGLL3 plays a critical role in promoting osteoblast differentiation and that part of the process is mediated by BMP signaling. Further elucidation of VGLL3 function will increase our understanding of osteogenesis and skeletal disease etiology.
Collapse
Affiliation(s)
- Haoze Yuan
- Institute of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, 246 Xuefu Road, Nangang, Harbin, 150001, Heilongjiang, People's Republic of China
- Department of Oral Morphology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
| | - Mika Ikegame
- Department of Oral Morphology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan.
| | - Yoko Fukuhara
- Department of Oral Morphology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
| | - Fumiko Takemoto
- Department of Oral Morphology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
- Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yaqiong Yu
- Department of Oral Morphology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
- Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, P.R. China
| | - Jumpei Teramachi
- Department of Oral Function & Anatomy, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yao Weng
- Department of Oral Morphology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
| | - Jiajie Guo
- Department of Oral Morphology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
- Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, P.R. China
| | - Daisuke Yamada
- Department of Regenerative Science, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University , Okayama, Japan
| | - Takeshi Takarada
- Department of Regenerative Science, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University , Okayama, Japan
| | - Ying Li
- Institute of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, 246 Xuefu Road, Nangang, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Hirohiko Okamura
- Department of Oral Morphology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
| | - Bin Zhang
- Institute of Hard Tissue Development and Regeneration, The Second Affiliated Hospital of Harbin Medical University, Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, 246 Xuefu Road, Nangang, Harbin, 150001, Heilongjiang, People's Republic of China.
- Heilongjiang Academy of Medical Sciences, Harbin, 150001, Heilongjiang, P.R. China.
| |
Collapse
|
35
|
Munmun F, Mohiuddin OA, Hoang VT, Burow ME, Bunnell BA, Sola VM, Carpentieri AR, Witt-Enderby PA. The role of MEK1/2 and MEK5 in melatonin-mediated actions on osteoblastogenesis, osteoclastogenesis, bone microarchitecture, biomechanics, and bone formation. J Pineal Res 2022; 73:e12814. [PMID: 35674448 DOI: 10.1111/jpi.12814] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 04/30/2022] [Accepted: 05/11/2022] [Indexed: 12/11/2022]
Abstract
Melatonin, the primary hormone involved in circadian entrainment, plays a significant role in bone physiology. This study aimed to assess the role of MEK1/2 and MEK5 in melatonin-mediated actions in mouse and human mesenchymal stem cells (MSCs) and on bone using small-molecule inhibitors and CRISPR/Cas9 knockout approaches. Consistent with in vitro studies performed in mMSCs and hMSCs, nightly (25 mg/kg, i.p., 45 days) injections with PD184352 (MEK1/2 inhibitor) or Bix02189 (MEK5 inhibitor) or SC-1-151 (MEK1/2/5 inhibitor) demonstrated that MEK1/2 and MEK5 were the primary drivers underlying melatonin's actions on bone density, microarchitecture (i.e., trabecular number, separation, and connectivity density), and bone mechanical properties (i.e., ultimate stress) through increases in osteogenic (RUNX2, BMP-2, FRA-1, OPG) expression and decreases in PPARγ. Furthermore, CRISPR/Cas9 knockout of MEK1 or MEK5 in mMSCs seeded on PLGA scaffolds and placed into critical-size calvarial defects in Balb(c) mice (male and female) revealed that treatment with melatonin (15 mg/L; p.o., nightly, 90 days) mediates sex-specific actions of MEK1 and MEK5 in new bone formation. This study is the first to demonstrate a role for MEK1/2 and MEK5 in modulating melatonin-mediated actions on bone formation in vivo and in a sex-specific manner.
Collapse
Affiliation(s)
- Fahima Munmun
- Division of Pharmaceutical Sciences, Duquesne University School of Pharmacy, Pittsburgh, Pennsylvania, USA
| | - Omair A Mohiuddin
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Van T Hoang
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Matthew E Burow
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Bruce A Bunnell
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Veronica M Sola
- Department of Oral Biology, Faculty of Odontology, National University of Cordoba, Cordoba, Argentina
| | - Agata R Carpentieri
- Faculty of Odontology, National University of Cordoba and National Council for Scientific and Technical Research (CONICET); Institute for Health Sciences Research (INICSA), Cordoba, Argentina
| | - Paula A Witt-Enderby
- Division of Pharmaceutical Sciences, Duquesne University School of Pharmacy, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
36
|
Chu X, He C, Sang B, Yang C, Yin C, Ji M, Qian A, Tian Y. Transfer RNAs-derived small RNAs and their application potential in multiple diseases. Front Cell Dev Biol 2022; 10:954431. [PMID: 36072340 PMCID: PMC9441921 DOI: 10.3389/fcell.2022.954431] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 07/11/2022] [Indexed: 11/17/2022] Open
Abstract
The role of tRNAs is best known as adapter components of translational machinery. According to the central dogma of molecular biology, DNA is transcribed to RNA and in turn is translated into proteins, in which tRNA outstands by its role of the cellular courier. Recent studies have led to the revision of the canonical function of transfer RNAs (tRNAs), which indicates that tRNAs also serve as a source for short non-coding RNAs called tRNA-derived small RNAs (tsRNAs). tsRNAs play key roles in cellular processes by modulating complicated regulatory networks beyond translation and are widely involved in multiple diseases. Herein, the biogenesis and classification of tsRNAs were firstly clarified. tsRNAs are generated from pre-tRNAs or mature tRNAs and are classified into tRNA-derived fragments (tRFs) and tRNA halves (tiRNA). The tRFs include five types according to the incision loci: tRF-1, tRF-2, tRF-3, tRF-5 and i-tRF which contain 3′ tiRNA and 5′ tiRNA. The functions of tsRNAs and their regulation mechanisms involved in disease processes are systematically summarized as well. The mechanisms can elaborate on the specific regulation of tsRNAs. In conclusion, the current research suggests that tsRNAs are promising targets for modulating pathological processes, such as breast cancer, ischemic stroke, respiratory syncytial virus, osteoporosis and so on, and maintain vital clinical implications in diagnosis and therapeutics of various diseases.
Collapse
Affiliation(s)
- Xiaohua Chu
- Lab for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, SN, China
| | - Chenyang He
- Department of Oncology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Bo Sang
- Lab for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, SN, China
| | - Chaofei Yang
- Lab for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, SN, China
| | - Chong Yin
- Department of Clinical Laboratory, Academician (expert) Workstation, Lab of Epigenetics and RNA Therapy, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Mili Ji
- Lab for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, SN, China
| | - Airong Qian
- Lab for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, SN, China
- *Correspondence: Airong Qian, ; Ye Tian,
| | - Ye Tian
- Lab for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, SN, China
- *Correspondence: Airong Qian, ; Ye Tian,
| |
Collapse
|
37
|
Kim JM, Yang YS, Hong J, Chaugule S, Chun H, van der Meulen MCH, Xu R, Greenblatt MB, Shim JH. Biphasic regulation of osteoblast development via the ERK MAPK-mTOR pathway. eLife 2022; 11:78069. [PMID: 35975983 PMCID: PMC9417416 DOI: 10.7554/elife.78069] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 08/16/2022] [Indexed: 11/13/2022] Open
Abstract
Emerging evidence supports that osteogenic differentiation of skeletal progenitors is a key determinant of overall bone formation and bone mass. Despite extensive studies showing the function of mitogen-activated protein kinases (MAPKs) in osteoblast differentiation, none of these studies show in vivo evidence of a role for MAPKs in osteoblast maturation subsequent to lineage commitment. Here, we describe how the extracellular signal-regulated kinase (ERK) pathway in osteoblasts controls bone formation by suppressing the mechanistic target of rapamycin (mTOR) pathway. We also show that, while ERK inhibition blocks the differentiation of osteogenic precursors when initiated at an early stage, ERK inhibition surprisingly promotes the later stages of osteoblast differentiation. Accordingly, inhibition of the ERK pathway using a small compound inhibitor or conditional deletion of the MAP2Ks Map2k1 (MEK1) and Map2k2 (MEK2), in mature osteoblasts and osteocytes, markedly increased bone formation due to augmented osteoblast differentiation. Mice with inducible deletion of the ERK pathway in mature osteoblasts also displayed similar phenotypes, demonstrating that this phenotype reflects continuous postnatal inhibition of late-stage osteoblast maturation. Mechanistically, ERK inhibition increases mitochondrial function and SGK1 phosphorylation via mTOR2 activation, which leads to osteoblast differentiation and production of angiogenic and osteogenic factors to promote bone formation. This phenotype was partially reversed by inhibiting mTOR. Our study uncovers a surprising dichotomy of ERK pathway functions in osteoblasts, whereby ERK activation promotes the early differentiation of osteoblast precursors, but inhibits the subsequent differentiation of committed osteoblasts via mTOR-mediated regulation of mitochondrial function and SGK1.
Collapse
Affiliation(s)
- Jung-Min Kim
- Department of Medicine, University of Massachusetts Medical School, Worcester, United States
| | - Yeon-Suk Yang
- Department of Medicine, University of Massachusetts Medical School, Worcester, United States
| | - Jaehyoung Hong
- Department of Mathematical Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Sachin Chaugule
- Department of Medicine, University of Massachusetts Medical School, Worcester, United States
| | - Hyonho Chun
- Department of Mathematical Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Marjolein C H van der Meulen
- Meinig School of Biomedical Engineering and Sibley School of Mechanical & Aerospace Engineering, Cornell University, Ithaca, United States.,Research Division, Hospital for Special Surgery, New York, United States
| | - Ren Xu
- State Key Laboratory of Cellular Stress Biology, School of Medicine, Xiamen University, Fujian, China.,Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Matthew B Greenblatt
- Research Division, Hospital for Special Surgery, New York, United States.,Department of Pathology and Laboratory Medicine, Weill Cornell, New York, United States
| | - Jae-Hyuck Shim
- Department of Medicine, University of Massachusetts Medical School, Worcester, United States.,Horae Gene Therapy Center, Worcester, United States.,Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, Worcester, United States
| |
Collapse
|
38
|
Pathogenic Variants of the PHEX Gene. ENDOCRINES 2022. [DOI: 10.3390/endocrines3030040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Twenty-five years ago, a pathogenic variant of the phosphate-regulating endopeptidase homolog X-linked (PHEX) gene was identified as the cause of X-linked hypophosphatemic rickets (XLH). Subsequently, the overproduction of fibroblast growth factor 23 (FGF23) due to PHEX defects has been found to be associated with XLH pathophysiology. However, the mechanism by which PHEX deficiency contributes to the upregulation of FGF23 and the function of PHEX itself remain unclear. To date, over 700 pathogenic variants have been identified in patients with XLH, and functional assays and genotype–phenotype correlation analyses based on pathogenic variant data derived from XLH patients have been reported. Genetic testing for XLH is useful for the diagnosis. Not only have single-nucleotide variants causing missense, nonsense, and splicing variants and small deletion/insertion variants causing frameshift/non-frameshift alterations been observed, but also gross deletion/duplication variants causing copy number variants have been reported as pathogenic variants in PHEX. With the development of new technologies including next generation sequencing, it is expected that an increasing number of pathogenic variants will be identified. This chapter aimed to summarize the genotype of PHEX and related analyses and discusses the pathophysiology of PHEX defects to seek clues on unsolved questions.
Collapse
|
39
|
Effects of Triterpene Soyasapogenol B from Arachis hypogaea (Peanut) on Differentiation, Mineralization, Autophagy, and Necroptosis in Pre-Osteoblasts. Int J Mol Sci 2022; 23:ijms23158297. [PMID: 35955423 PMCID: PMC9368047 DOI: 10.3390/ijms23158297] [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: 07/04/2022] [Revised: 07/23/2022] [Accepted: 07/25/2022] [Indexed: 12/10/2022] Open
Abstract
Triterpenes are a diverse group of natural compounds found in plants. Soyasapogenol B (SoyB) from Arachis hypogaea (peanut) has various pharmacological properties. This study aimed to elucidate the pharmacological properties and mechanisms of SoyB in bone-forming cells. In the present study, 1–20 μM of SoyB showed no cell proliferation effects, whereas 30–100 μM of SoyB increased cell proliferation in MC3T3-E1 cells. Next, osteoblast differentiation was analyzed, and it was found that SoyB enhanced ALP staining and activity and bone mineralization. SoyB also induced RUNX2 expression in the nucleus with the increased phosphorylation of Smad1/5/8 and JNK2 during osteoblast differentiation. In addition, SoyB-mediated osteoblast differentiation was not associated with autophagy and necroptosis. Furthermore, SoyB increased the rate of cell migration and adhesion with the upregulation of MMP13 levels during osteoblast differentiation. The findings of this study provide new evidence that SoyB possesses biological effects in bone-forming cells and suggest a potentially beneficial role for peanut-based foods.
Collapse
|
40
|
Zhou J, Yang J, Dong Y, Shi Y, Zhu E, Yuan H, Li X, Wang B. Oncostatin M receptor regulates osteoblast differentiation via extracellular signal-regulated kinase/autophagy signaling. Stem Cell Res Ther 2022; 13:278. [PMID: 35765036 PMCID: PMC9241272 DOI: 10.1186/s13287-022-02958-1] [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: 04/18/2022] [Accepted: 06/12/2022] [Indexed: 11/10/2022] Open
Abstract
Background Oncostatin M receptor (OSMR), as one of the receptors for oncostatin M (OSM), has previously been shown to mediate the stimulatory role of OSM in osteoclastogenesis and bone resorption. However, it remains to be clarified whether and how OSMR affects the differentiation of osteoblasts. Methods The expression level of OSMR during osteoblast and adipocyte differentiation was examined. The role of OSMR in the differentiation was investigated using in vitro gain-of-function and loss-of-function experiments. The mechanisms by which OSMR regulates bone cell differentiation were explored. Finally, in vivo function of OSMR in cell fate determination and bone homeostasis was studied after transplantation of OSMR-silenced bone marrow stromal cells (BMSCs) to the marrow of ovariectomized mice. Results OSMR was regulated during osteogenic and adipogenic differentiation of marrow stromal progenitor cells and increased in the metaphysis of ovariectomized mice. OSMR suppressed osteogenic differentiation and stimulated adipogenic differentiation of progenitor cells. Mechanistic investigations showed that OSMR inhibited extracellular signal-regulated kinase (ERK) and autophagy signaling. The downregulation of autophagy, which was mediated by ERK inhibition, suppressed osteogenic differentiation of progenitor cells. Additionally, inactivation of ERK/autophagy signaling attenuated the stimulation of osteogenic differentiation induced by Osmr siRNA. Furthermore, transplantation of BMSCs in which OSMR was silenced to the marrow of mice promoted osteoblast differentiation, attenuated fat accumulation and osteoclast differentiation, and thereby relieved the osteopenic phenotype in the ovariectomized mice. Conclusions Our study has for the first time established the direct role of OSMR in regulating osteogenic differentiation of marrow stromal progenitor cells through ERK-mediated autophagy signaling. OSMR thus contributes to bone homeostasis through dual regulation of osteoblasts and osteoclasts. It also suggests that OSMR may be a potential target for the treatment of metabolic disorders such as osteoporosis. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02958-1.
Collapse
Affiliation(s)
- Jie Zhou
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital and Institute of Endocrinology, Tianjin Medical University, 6 Huan-Rui-Bei Road, Tianjin, 300134, China
| | - Junying Yang
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital and Institute of Endocrinology, Tianjin Medical University, 6 Huan-Rui-Bei Road, Tianjin, 300134, China.,College of Basic Medical Sciences, Tianjin Medical University, 22 Qi-Xiang-Tai Road, Tianjin, 300070, China
| | - Yuan Dong
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital and Institute of Endocrinology, Tianjin Medical University, 6 Huan-Rui-Bei Road, Tianjin, 300134, China.,College of Basic Medical Sciences, Tianjin Medical University, 22 Qi-Xiang-Tai Road, Tianjin, 300070, China
| | - Yaru Shi
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital and Institute of Endocrinology, Tianjin Medical University, 6 Huan-Rui-Bei Road, Tianjin, 300134, China
| | - Endong Zhu
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital and Institute of Endocrinology, Tianjin Medical University, 6 Huan-Rui-Bei Road, Tianjin, 300134, China
| | - Hairui Yuan
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital and Institute of Endocrinology, Tianjin Medical University, 6 Huan-Rui-Bei Road, Tianjin, 300134, China
| | - Xiaoxia Li
- College of Basic Medical Sciences, Tianjin Medical University, 22 Qi-Xiang-Tai Road, Tianjin, 300070, China
| | - Baoli Wang
- NHC Key Lab of Hormones and Development, Tianjin Key Lab of Metabolic Diseases, Chu Hsien-I Memorial Hospital and Institute of Endocrinology, Tianjin Medical University, 6 Huan-Rui-Bei Road, Tianjin, 300134, China.
| |
Collapse
|
41
|
Cerqueira A, García-Arnáez I, Romero-Gavilán F, Azkargorta M, Elortza F, Martín de Llanos JJ, Carda C, Gurruchaga M, Goñi I, Suay J. Complex effects of Mg-biomaterials on the osteoblast cell machinery: A proteomic study. BIOMATERIALS ADVANCES 2022; 137:212826. [PMID: 35929259 DOI: 10.1016/j.bioadv.2022.212826] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/08/2022] [Accepted: 04/20/2022] [Indexed: 12/16/2022]
Abstract
The cell-biomaterial interface is highly complex; thousands of molecules and many processes participate in its formation. Growing demand for improved biomaterials has highlighted the need to understand the structure and functions of this interface. Proteomic methods offer a viable alternative to the traditional in vitro techniques for analyzing such systems. Magnesium is a promoter of cell adhesion and osteogenesis. Here, we used the LC-MS/MS to compare the protein expression profiles of human osteoblasts (HOb) exposed to sol-gel coatings without (MT) and with Mg (MT1.5Mg) for 1, 3, and 7 days. PANTHER, DAVID, and IPA databases were employed for protein identification and data analysis. Confocal microscopy and gene expression analysis were used for further characterization. Exposure to MT1.5Mg increased the HOb cell area and the expression of SP7, RUNX2, IBP3, COL3A1, MXRA8, and FBN1 genes. Proteomic analysis showed that MT1.5Mg affected the early osteoblast maturation (PI3/AKT, mTOR, ERK/MAPK), insulin metabolism, cell adhesion (integrin, FAK, actin cytoskeleton regulation) and oxidative stress pathways. Thus, the effects of Mg on cell adhesion and osteogenesis are rather complex, affecting several pathways rather than single processes. Our analysis also confirms the potential of proteomics in biomaterial characterization, showing a good correlation with in vitro results.
Collapse
Affiliation(s)
- Andreia Cerqueira
- Department of Industrial Systems Engineering and Design, Universitat Jaume I, Av. Vicent Sos Baynat s/n, 12071 Castellón de la Plana, Spain
| | - Iñaki García-Arnáez
- Department of Science and Technology of Polymers, University of the Basque Country, P. M. de Lardizábal, 3, 20018 San Sebastián, Spain
| | - Francisco Romero-Gavilán
- Department of Industrial Systems Engineering and Design, Universitat Jaume I, Av. Vicent Sos Baynat s/n, 12071 Castellón de la Plana, Spain.
| | - Mikel Azkargorta
- Proteomics Platform, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), CIBERehd, ProteoRed-ISCIII, Bizkaia Science and Technology Park, 48160 Derio, Spain
| | - Félix Elortza
- Proteomics Platform, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), CIBERehd, ProteoRed-ISCIII, Bizkaia Science and Technology Park, 48160 Derio, Spain
| | - José Javier Martín de Llanos
- Department of Pathology Medicine and Odontology, Medicine Faculty, University of Valencia, Av Blasco Ibáñez, 13, 46010, Valencia, Spain; Research Institute of the University Clinical Hospital of Valencia (INCLIVA), C. de Menéndez y Pelayo, 4, 46010, Valencia, Spain
| | - Carmen Carda
- Department of Pathology Medicine and Odontology, Medicine Faculty, University of Valencia, Av Blasco Ibáñez, 13, 46010, Valencia, Spain; Research Institute of the University Clinical Hospital of Valencia (INCLIVA), C. de Menéndez y Pelayo, 4, 46010, Valencia, Spain
| | - Mariló Gurruchaga
- Department of Science and Technology of Polymers, University of the Basque Country, P. M. de Lardizábal, 3, 20018 San Sebastián, Spain
| | - Isabel Goñi
- Department of Science and Technology of Polymers, University of the Basque Country, P. M. de Lardizábal, 3, 20018 San Sebastián, Spain
| | - Julio Suay
- Department of Industrial Systems Engineering and Design, Universitat Jaume I, Av. Vicent Sos Baynat s/n, 12071 Castellón de la Plana, Spain
| |
Collapse
|
42
|
Park KR, Kwon YJ, Jeong YH, Hong JT, Yun HM. Thelephoric acid, p-terphenyl, induces bone-forming activities in pre-osteoblasts. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
43
|
Kim JH, Kim K, Kim I, Seong S, Koh JT, Kim N. The ATF3-OPG Axis Contributes to Bone Formation by Regulating the Differentiation of Osteoclasts, Osteoblasts, and Adipocytes. Int J Mol Sci 2022; 23:ijms23073500. [PMID: 35408860 PMCID: PMC8998270 DOI: 10.3390/ijms23073500] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 01/25/2023] Open
Abstract
Activating transcription factor 3 (ATF3) has been identified as a negative regulator of osteoblast differentiation in in vitro study. However, it was not associated with osteoblast differentiation in in vivo study. To provide an understanding of the discrepancy between the in vivo and in vitro findings regarding the function of ATF3 in osteoblasts, we investigated the unidentified roles of ATF3 in osteoblast biology. ATF3 enhanced osteoprotegerin (OPG) production, not only in osteoblast precursor cells, but also during osteoblast differentiation and osteoblastic adipocyte differentiation. In addition, ATF3 increased nodule formation in immature osteoblasts and decreased osteoblast-dependent osteoclast formation, as well as the transdifferentiation of osteoblasts to adipocytes. However, all these effects were reversed by the OPG neutralizing antibody. Taken together, these results suggest that ATF3 contributes to bone homeostasis by regulating the differentiation of various cell types in the bone microenvironment, including osteoblasts, osteoclasts, and adipocytes via inducing OPG production.
Collapse
Affiliation(s)
- Jung Ha Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469, Korea; (J.H.K.); (K.K.); (I.K.); (S.S.)
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju 61186, Korea;
| | - Kabsun Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469, Korea; (J.H.K.); (K.K.); (I.K.); (S.S.)
| | - Inyoung Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469, Korea; (J.H.K.); (K.K.); (I.K.); (S.S.)
| | - Semun Seong
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469, Korea; (J.H.K.); (K.K.); (I.K.); (S.S.)
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju 61186, Korea;
| | - Jeong-Tae Koh
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju 61186, Korea;
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju 61186, Korea
| | - Nacksung Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469, Korea; (J.H.K.); (K.K.); (I.K.); (S.S.)
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju 61186, Korea;
- Correspondence: ; Tel.: +82-61-379-2835
| |
Collapse
|
44
|
Greenblatt MB, Shim JH, Bok S, Kim JM. The Extracellular Signal-Regulated Kinase Mitogen-Activated Protein Kinase Pathway in Osteoblasts. J Bone Metab 2022; 29:1-15. [PMID: 35325978 PMCID: PMC8948490 DOI: 10.11005/jbm.2022.29.1.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 02/17/2022] [Indexed: 12/01/2022] Open
Abstract
Extracellular signal-regulated kinases (ERKs) are evolutionarily ancient signal transducers of the mitogen-activated protein kinase (MAPK) family that have long been linked to the regulation of osteoblast differentiation and bone formation. Here, we review the physiological functions, biochemistry, upstream activators, and downstream substrates of the ERK pathway. ERK is activated in skeletal progenitors and regulates osteoblast differentiation and skeletal mineralization, with ERK serving as a key regulator of Runt-related transcription factor 2, a critical transcription factor for osteoblast differentiation. However, new evidence highlights context-dependent changes in ERK MAPK pathway wiring and function, indicating a broader set of physiological roles associated with changes in ERK pathway components or substrates. Consistent with this importance, several human skeletal dysplasias are associated with dysregulation of the ERK MAPK pathway, including neurofibromatosis type 1 and Noonan syndrome. The continually broadening array of drugs targeting the ERK pathway for the treatment of cancer and other disorders makes it increasingly important to understand how interference with this pathway impacts bone metabolism, highlighting the importance of mouse studies to model the role of the ERK MAPK pathway in bone formation.
Collapse
Affiliation(s)
- Matthew B. Greenblatt
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical, New York, NY,
USA
- Research Division, Hospital for Special Surgery, New York, NY,
USA
| | - Jae-Hyuck Shim
- Division of Rheumatology, Department of Medicine, UMass Chan Medical School, Worcester, MA,
USA
- Horae Gene Therapy Center, and Li Weibo Institute for Rare Diseases Research, UMass Chan Medical School, Worcester, MA,
USA
| | - Seoyeon Bok
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical, New York, NY,
USA
| | - Jung-Min Kim
- Division of Rheumatology, Department of Medicine, UMass Chan Medical School, Worcester, MA,
USA
| |
Collapse
|
45
|
Xia K, Yu LY, Huang XQ, Zhao ZH, Liu J. Epigenetic regulation by long noncoding RNAs in osteo-/adipogenic differentiation of mesenchymal stromal cells and degenerative bone diseases. World J Stem Cells 2022; 14:92-103. [PMID: 35126830 PMCID: PMC8788182 DOI: 10.4252/wjsc.v14.i1.92] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 07/07/2021] [Accepted: 01/05/2022] [Indexed: 02/06/2023] Open
Abstract
Bone is a complex tissue that undergoes constant remodeling to maintain homeostasis, which requires coordinated multilineage differentiation and proper proliferation of mesenchymal stromal cells (MSCs). Mounting evidence indicates that a disturbance of bone homeostasis can trigger degenerative bone diseases, including osteoporosis and osteoarthritis. In addition to conventional genetic modifications, epigenetic modifications (i.e., DNA methylation, histone modifications, and the expression of noncoding RNAs) are considered to be contributing factors that affect bone homeostasis. Long noncoding RNAs (lncRNAs) were previously regarded as ‘transcriptional noise’ with no biological functions. However, substantial evidence suggests that lncRNAs have roles in the epigenetic regulation of biological processes in MSCs and related diseases. In this review, we summarized the interactions between lncRNAs and epigenetic modifiers associated with osteo-/adipogenic differentiation of MSCs and the pathogenesis of degenerative bone diseases and highlighted promising lncRNA-based diagnostic and therapeutic targets for bone diseases.
Collapse
Affiliation(s)
- Kai Xia
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Li-Yuan Yu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Xin-Qi Huang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Zhi-He Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Jun Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
46
|
Han X, Deng F, Zhu R, Li K, Yang S, Jin L, Ma Z, Ning C, Shi X, Li Y. Osteoimmune reaction caused by novel silicocarnotite bioceramic promoting osteogenesis through MAPK pathway. Biomater Sci 2022; 10:2877-2891. [DOI: 10.1039/d2bm00125j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The host immune response to implant is a key factor in determining the fate of bone grafts, which is thought to be a regulator of tissue regeneration. Figuring out the...
Collapse
|
47
|
Huang G, Pan ST, Qiu JX. The osteogenic effects of porous Tantalum and Titanium alloy scaffolds with different unit cell structure. Colloids Surf B Biointerfaces 2021; 210:112229. [PMID: 34875470 DOI: 10.1016/j.colsurfb.2021.112229] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/30/2021] [Accepted: 11/15/2021] [Indexed: 12/28/2022]
Abstract
Porous scaffolds have long been regarded as optimal substitute for bone tissue repairing. In order to explore the influence of unit cell structure and inherent material characteristics on the porous scaffolds in terms of mechanical and biological performance, selective laser melting (SLM) technology was used to fabricate porous tantalum (Ta) and titanium alloy (Ti6Al4V) with diamond (Di) or rhombic dodecahedron (Do) unit cell structure. The mechanical strength of all the porous scaffolds could match that of trabecular bone, while the biological performance of each scaffold was diverse from each other. Moreover, the ILK/ERK1/2/Runx2 signaling pathway had been verified to be involved in the osteogenic differentiation of rat bone mesenchymal stem cells (rBMSCs) cultured on those porous scaffolds. Unit cell structure and material characteristics of the porous Ta and Ti6Al4V scaffolds can synergistically modulate this axis and further impact on the osteogenic effects. Our results hence illustrate that porous Ta scaffold with diamond unit cell structure possesses excellent osteogenic effects and moderate mechanical strength and porous Ti6Al4V scaffold with rhombic dodecahedron unit cell structure has the highest mechanical strength and moderate osteogenic effects. Both porous Ta and Ti6Al4V can be applied in different settings requiring either better biological performance or higher mechanical demand.
Collapse
Affiliation(s)
- Gan Huang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Shu-Ting Pan
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Jia-Xuan Qiu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China.
| |
Collapse
|
48
|
Zhang X, Jiang Y, Mao J, Ren X, Ji Y, Mao Y, Chen Y, Sun X, Pan Y, Ma J, Huang S. Hydroxytyrosol prevents periodontitis-induced bone loss by regulating mitochondrial function and mitogen-activated protein kinase signaling of bone cells. Free Radic Biol Med 2021; 176:298-311. [PMID: 34610362 DOI: 10.1016/j.freeradbiomed.2021.09.027] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/24/2021] [Accepted: 09/30/2021] [Indexed: 01/10/2023]
Abstract
Reactive oxygen species (ROS) overproduction promotes the alveolar bone loss during the development of periodontitis. Mitochondria are the principal source of ROS. Hydroxytyrosol (HT), a natural phenolic compound present in olive oil, is well known for its antioxidant and mitochondrial-protective prosperities. Nonetheless, the impact of HT on periodontitis and its related mechanisms underlying bone cell behavior remains unknown. Osteoclasts differentiated from RAW264.7 model and oxidative stress (OS) induced pre-osteoblast MC3T3-E1 cell injury model were treated with and without HT. Cell viability, apoptosis, differentiation, mitochondrial function along with mitogen-activated protein kinase (MAPK) signaling pathway were investigated. Meanwhile, the effect and related mechanisms of HT on bone loss in mice with periodontitis were also detected. HT inhibited osteoclast differentiation and prevented OS induced pre-osteoblast cells injury via regulating mitochondrial function as well as ERK and JNK signaling pathways. Moreover, HT attenuated the alveolar bone loss, increased bone forming activity, inhibited the osteoclasts differentiation and decreased the level of OS in mice with periodontitis. Our findings, for the first time, revealed a novel function of HT in bone remodeling of periodontitis, and highlighted its therapeutical potential for the prevention/treatment of periodontitis.
Collapse
Affiliation(s)
- Xiaorong Zhang
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China; Department of Endodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Yun Jiang
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China; Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Jiajie Mao
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China; Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Xuekun Ren
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China; Department of Endodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Yinghui Ji
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China; Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Yixin Mao
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China; Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Yang Chen
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China; Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Xiaoyu Sun
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China; Department of Periodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Yihuai Pan
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China; Department of Endodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.
| | - Jianfeng Ma
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China; Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.
| | - Shengbin Huang
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China; Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
49
|
Molecular insights for an anti-osteoporotic properties of Litsea glutinosa on Saos-2 cells: An in-vitro approach. J Ayurveda Integr Med 2021; 13:100501. [PMID: 34799209 PMCID: PMC8728066 DOI: 10.1016/j.jaim.2021.07.017] [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: 02/01/2021] [Revised: 07/22/2021] [Accepted: 07/23/2021] [Indexed: 01/26/2023] Open
Abstract
Osteoporosis is a skeletal disease that is identified by the deterioration of micro-architecture of bone tissue, leading to enhanced bone brittleness and a consequential increase in fracture threat. There are many treatments available for osteoporosis such as bisphosphonate therapy, hormonal replacement therapy, herbal therapy etc. For decades, there are several herbs that are attributed to have anti-osteoporotic effects however the candidate genes involved in it remained unknown. In line with this, the present study is focused to elucidate the anti-osteoporotic property of Litsea glutinosa (LG). To understand the proliferative effect and identify involved players, gene expression was studied on the Saos-2 osteocytes in-vitro. The expression profile of candidate genes involved in different signaling pathways such as Egr-2, RUNX2, MAPK3, NFATc1, CREB, ERβ, along with proliferation and apoptotic markers in osteoporosis were selected for the study. The gene expression profile demonstrated a significant up-regulation of Egr-2, RUNX2, MAPK3, CREB, EBβ in the range of 1.5–2.2 folds, whereas NFATc1 was found to be down-regulated up to 0.4 times compared to control when treated with 250 μg/mL of LG. Besides this, anti-apoptosis effect of LG was also supported by flow cytometry results which also proved that LG induces proliferation and inhibits apoptosis, suggesting the proliferative role of LG. In conclusion, the present study gathers the potency of LG extract for its proliferative and anti-apoptotic effect on Saos-2 osteocytes and opens a new avenue for detailing the mechanistic actions of it on mitigating the pathophysiology of osteoporosis.
Collapse
|
50
|
Xie B, Zeng Z, Liao S, Zhou C, Wu L, Xu D. Kaempferol Ameliorates the Inhibitory Activity of Dexamethasone in the Osteogenesis of MC3T3-E1 Cells by JNK and p38-MAPK Pathways. Front Pharmacol 2021; 12:739326. [PMID: 34675808 PMCID: PMC8524096 DOI: 10.3389/fphar.2021.739326] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 09/21/2021] [Indexed: 11/23/2022] Open
Abstract
Kaempferol has been reported to exhibit beneficial effect on the osteogenic differentiation in mesenchymal stem cells (MSC) and osteoblasts. In our previous study, dexamethasone (DEX) demonstrated inhibitory effect on MC3T3-E1 cells differentiation. In this study, we mainly explored the protective effect of kaempferol on the inhibitory activity of DEX in the osteogenesis of MC3T3-E1 cells. We found that kaempferol ameliorated the proliferation inhibition, cell cycle arrest, and cell apoptosis and increased the activity of alkaline phosphatase (ALP) and the mineralization in DEX-treated MC3T3-E1 cells. Kaempferol also significantly enhanced the expression of osterix (Osx) and runt-related transcription factor 2 (Runx2) in MC3T3-E1 cells treated with DEX. In addition, kaempferol attenuated DEX-induced reduction of cyclin D1 and Bcl-2 expression and elevation of p53 and Bax expression. Kaempferol also activated JNK and p38-MAPK pathways in DEX-treated MC3T3-E1 cells. Furthermore, kaempferol improved bone mineralization in DEX-induced bone damage in a zebrafish larvae model. These data suggested that kaempferol ameliorated the inhibitory activity of DEX in the osteogenesis of MC3T3-E1 cells by activating JNK and p38-MAPK signaling pathways. Kaempferol exhibited great potentials in developing new drugs for treating glucocorticoid-induced osteoporosis.
Collapse
Affiliation(s)
- Baocheng Xie
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Public Service Platform of South China Sea for R&D Marine Biomedicine Resources, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.,Department of Pharmacy, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, China
| | - Zhanwei Zeng
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Public Service Platform of South China Sea for R&D Marine Biomedicine Resources, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.,Key Laboratory of Traditional Chinese Medicine and New Pharmacy Development, Guangdong Medical University, Dongguan, China
| | - Shiyi Liao
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Public Service Platform of South China Sea for R&D Marine Biomedicine Resources, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.,Key Laboratory of Traditional Chinese Medicine and New Pharmacy Development, Guangdong Medical University, Dongguan, China
| | - Chenhui Zhou
- School of Nursing, Guangdong Medical University, Dongguan, China
| | - Longhuo Wu
- College of Pharmacy, Gannan Medical University, Ganzhou, China
| | - Daohua Xu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Public Service Platform of South China Sea for R&D Marine Biomedicine Resources, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.,Key Laboratory of Traditional Chinese Medicine and New Pharmacy Development, Guangdong Medical University, Dongguan, China
| |
Collapse
|