Oxidative Stress and Inflammation in Osteoporosis: Molecular Mechanisms Involved and the Relationship with microRNAs
Abstract
:1. Introduction
2. Oxidative Stress and Osteoporosis: Principal Involved Molecular Mechanisms
3. Proinflammatory Mediators and Osteoporosis: Role of Immune Cells
4. miRNAs and Osteoporosis
5. Crosstalk among Oxidative Stress, Inflammation, and miRNAs in Osteoporosis
6. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Kenkre, J.S.; Bassett, J. The bone remodelling cycle. Ann. Clin. Biochem. 2018, 55, 308–327. [Google Scholar] [CrossRef] [PubMed]
- Robling, A.G.; Bonewald, L.F. The Osteocyte: New Insights. Annu. Rev. Physiol. 2020, 82, 485–506. [Google Scholar] [CrossRef] [PubMed]
- Zhou, B.; Peng, K.; Wang, G.; Chen, W.; Kang, Y. Polo Like Kinase 4 (PLK4) impairs human bone marrow mesenchymal stem cell (BMSC) viability and osteogenic differentiation. Biochem. Biophys. Res. Commun. 2021, 549, 221–228. [Google Scholar] [CrossRef] [PubMed]
- Bonnet, N.; Conway, S.J.; Ferrari, S.L. Regulation of beta catenin signaling and parathyroid hormone anabolic effects in bone by the matricellular protein periostin. Proc. Natl. Acad. Sci. USA 2012, 109, 15048–15053. [Google Scholar] [CrossRef] [PubMed]
- Orimo, H. The mechanism of mineralization and the role of alkaline phosphatase in health and disease. J. Nippon Med. Sch. 2010, 77, 4–12. [Google Scholar] [CrossRef]
- Licini, C.; Vitale-Brovarone, C.; Mattioli-Belmonte, M. Collagen and non-collagenous proteins molecular crosstalk in the pathophysiology of osteoporosis. Cytokine Growth Factor Rev. 2019, 49, 59–69. [Google Scholar] [CrossRef] [PubMed]
- Glass, D.A., 2nd; Bialek, P.; Ahn, J.D.; Starbuck, M.; Patel, M.S.; Clevers, H.; Taketo, M.M.; Long, F.; McMahon, A.P.; Lang, R.A.; et al. Canonical Wnt signaling in differentiated osteoblasts controls osteoclast differentiation. Dev. Cell 2005, 8, 751–764. [Google Scholar] [CrossRef] [PubMed]
- Boyce, B.F.; Yao, Z.; Xin, L. Osteoclasts have multiple roles in bone in addition to bone resorption. Crit. Rev. Eukaryot. Gene Expr. 2009, 19, 171–180. [Google Scholar] [CrossRef]
- Creecy, A.; Damrath, J.G.; Wallace, J.M. Control of Bone Matrix Properties by Osteocytes. Front. Endocrinol. 2021, 11, 578477. [Google Scholar] [CrossRef] [PubMed]
- Verborgt, O.; Gibson, G.J.; Schaffler, M.B. Loss of osteocyte integrity in association with microdamage and bone remodeling after fatigue in vivo. J. Bone Miner. Res. 2000, 15, 60–67. [Google Scholar] [CrossRef]
- Yang, J.; Shah, R.; Robling, A.G.; Templeton, E.; Yang, H.; Tracey, K.J.; Bidwell, J.P. HMGB1 is a bone-active cytokine. J. Cell. Physiol. 2008, 214, 730–739. [Google Scholar] [CrossRef]
- Yao, W.; Dai, W.; Jiang, J.X.; Lane, N.E. Glucocorticoids and osteocyte autophagy. Bone 2013, 54, 279–284. [Google Scholar] [CrossRef]
- Li, H.; Xiao, Z.; Quarles, L.D.; Li, W. Osteoporosis: Mechanism, Molecular Target and Current Status on Drug Development. Curr. Med. Chem. 2021, 28, 1489–1507. [Google Scholar] [CrossRef] [PubMed]
- Tatsumi, S.; Ishii, K.; Amizuka, N.; Li, M.; Kobayashi, T.; Kohno, K.; Ito, M.; Takeshita, S.; Ikeda, K. Targeted ablation of osteocytes induces osteoporosis with defective mechanotransduction. Cell Metab. 2007, 5, 464–475. [Google Scholar] [CrossRef] [PubMed]
- Khosla, S.; Oursler, M.J.; Monroe, D.G. Estrogen and the skeleton. Trends Endocrinol. Metab. 2012, 23, 576–581. [Google Scholar] [CrossRef] [PubMed]
- Almeida, M.; Laurent, M.R.; Dubois, V.; Claessens, F.; O’Brien, C.A.; Bouillon, R.; Vanderschueren, D.; Manolagas, S.C. Estrogens and Androgens in Skeletal Physiology and Pathophysiology. Physiol. Rev. 2017, 97, 135–187. [Google Scholar] [CrossRef]
- Emerton, K.B.; Hu, B.; Woo, A.A.; Sinofsky, A.; Hernandez, C.; Majeska, R.J.; Jepsen, K.J.; Schaffler, M.B. Osteocyte apoptosis and control of bone resorption following ovariectomy in mice. Bone 2010, 46, 577–583. [Google Scholar] [CrossRef]
- Wu, D.; Cline-Smith, A.; Shashkova, E.; Perla, A.; Katyal, A.; Aurora, R. T-Cell Mediated Inflammation in Postmenopausal Osteoporosis. Front. Immunol. 2021, 12, 687551. [Google Scholar] [CrossRef]
- Mohamad, N.V.; Ima-Nirwana, S.; Chin, K.Y. Are Oxidative Stress and Inflammation Mediators of Bone Loss Due to Estrogen Deficiency? A Review of Current Evidence. Endocr. Metab. Immune Disord. Drug Targets 2020, 20, 1478–1487. [Google Scholar] [CrossRef]
- Shahriarpour, Z.; Nasrabadi, B.; Hejri-Zarifi, S.; Shariati-Bafghi, S.E.; Yousefian-Sanny, M.; Karamati, M.; Rashidkhani, B. Oxidative balance score and risk of osteoporosis among postmenopausal Iranian women. Arch. Osteoporos. 2021, 16, 43. [Google Scholar] [CrossRef]
- Martín-Millán, M.; Castañeda Estrogens, S. Osteoarthritis and inflammation. Joint Bone Spine 2013, 80, 368–373. [Google Scholar] [CrossRef] [PubMed]
- Cenci, S.; Weitzmann, M.N.; Roggia, C.; Namba, N.; Novack, D.; Woodring, J.; Pacifici, R. Estrogen deficiency induces bone loss by enhancing T-cell production of TNF-alpha. J. Clin. Investig. 2000, 106, 1229–1237. [Google Scholar] [CrossRef]
- Zhou, X.; Yuan, W.; Xiong, X.; Zhang, Z.; Liu, J.; Zheng, Y.; Wang, J.; Liu, J. HO-1 in Bone Biology: Potential Therapeutic Strategies for Osteoporosis. Front. Cell Dev. Biol. 2021, 9, 791585. [Google Scholar] [CrossRef] [PubMed]
- Domazetovic, V.; Marcucci, G.; Iantomasi, T.; Brandi, M.L.; Vincenzini, M.T. Oxidative stress in bone remodeling: Role of antioxidants. Clin. Cases Miner. Bone Metab. 2017, 14, 209–216. [Google Scholar] [CrossRef] [PubMed]
- Agidigbi, T.S.; Kim, C. Reactive Oxygen Species in Osteoclast Differentiation and Possible Pharmaceutical Targets of ROS-Mediated Osteoclast Diseases. Int. J. Mol. Sci. 2019, 20, 3576. [Google Scholar] [CrossRef]
- Zha, L.; He, L.; Liang, Y.; Qin, H.; Yu, B.; Chang, L.; Xue, L. TNF-α contributes to postmenopausal osteoporosis by synergistically promoting RANKL-induced osteoclast formation. Biomed. Pharmacother. 2018, 102, 369–374. [Google Scholar] [CrossRef]
- Romagnoli, C.; Marcucci, G.; Favilli, F.; Zonefrati, R.; Mavilia, C.; Galli, G.; Tanini, A.; Iantomasi, T.; Brandi, M.L.; Vincenzini, M.T. Role of GSH/GSSG redox couple in osteogenic activity and osteoclastogenic markers of human osteoblast-like SaOS-2 cells. FEBS J. 2013, 280, 867–879. [Google Scholar] [CrossRef]
- Domazetovic, V.; Falsetti, I.; Ciuffi, S.; Iantomasi, T.; Marcucci, G.; Vincenzini, M.T.; Brandi, M.L. Effect of Oxidative Stress-Induced Apoptosis on Active FGF23 Levels in MLO-Y4 Cells: The Protective Role of 17-β-Estradiol. Int. J. Mo.l Sci. 2022, 23, 2103. [Google Scholar] [CrossRef]
- Lu, T.X.; Rothenberg, M.E. MicroRNA. J. Allergy Clin. Immunol. 2018, 141, 1202–1207. [Google Scholar] [CrossRef]
- Inukai, S.; Slack, F. MicroRNAs and the genetic network in aging. J. Mol. Biol. 2013, 425, 3601–3608. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sikora, M.; Marycz, K.; Smieszek, A. Small and Long Non-coding RNAs as Functional Regulators of Bone Homeostasis, Acting Alone or Cooperatively. Mol. Ther. Nucleic Acids 2020, 21, 792–803. [Google Scholar] [CrossRef] [PubMed]
- Feng, Q.; Zheng, S.; Zheng, J. The emerging role of microRNAs in bone remodeling and its therapeutic implications for osteoporosis. Biosci. Rep. 2018, 38, BSR20180453. [Google Scholar] [CrossRef] [PubMed]
- Jia, B.; Zhang, Z.; Qiu, X.; Chu, H.; Sun, X.; Zheng, X.; Zhao, J.; Li, Q. Analysis of the miRNA and mRNA involved in osteogenesis of adipose-derived mesenchymal stem cells. Exp. Ther. Med. 2018, 16, 1111–1120. [Google Scholar] [CrossRef] [PubMed]
- Li, Z.; Hassan, M.Q.; Jafferji, M.; Aqeilan, R.I.; Garzon, R.; Croce, C.M.; van Wijnen, A.J.; Stein, J.L.; Stein, G.S.; Lian, J.B. Biological functions of miR-29b contribute to positive regulation of osteoblast differentiation. J. Biol. Chem. 2009, 284, 15676–15684. [Google Scholar] [CrossRef]
- Vishnoi, A.; Rani, S. MiRNA Biogenesis and Regulation of Diseases: An Overview. In Methods in Molecular Biology; Springer: Berlin/Heidelberg, Germany, 2017; Volume 1509, pp. 1–10. [Google Scholar]
- Husain, A.; Jeffries, M.A. Epigenetics and Bone Remodeling. Curr. Osteoporos. Rep. 2017, 15, 450–458. [Google Scholar] [CrossRef]
- Ciuffi, S.; Marini, F.; Fossi, C.; Donati, S.; Giusti, F.; Botta, A.; Masi, L.; Isaia, G.; Marcocci, C.; Migliaccio, S.; et al. Circulating MicroRNAs as Biomarkers of Osteoporosis and Fragility Fractures. J. Clin. Endocrinol. Metab. 2022, 107, 2267–2285. [Google Scholar] [CrossRef]
- He, J.; Jiang, B.H. Interplay between Reactive oxygen Species and MicroRNAs in Cancer. Curr. Pharmacol. Rep. 2016, 2, 82–90. [Google Scholar] [CrossRef]
- Huffaker, T.B.; Hu, R.; Runtsch, M.C.; Bake, E.; Chen, X.; Zhao, J.; Round, J.L.; Baltimore, D.; O’Connell, R.M. Epistasis between microRNAs 155 and 146a during T cell-mediated antitumor immunity. Cell Rep. 2012, 2, 1697–1709. [Google Scholar] [CrossRef]
- Parascandolo, A.; Laukkanen, M.O. Carcinogenesis and Reactive Oxygen Species Signaling: Interaction of the NADPH Oxidase NOX1-5 and Superoxide Dismutase 1-3 Signal Transduction Pathways. Antioxid. Redox Signal. 2019, 30, 443–486. [Google Scholar] [CrossRef]
- Poljsak, B.; Šuput, D.; Milisav, I. Achieving the balance between ROS and antioxidants: When to use the synthetic antioxidants. Oxid. Med. Cell. Longev. 2013, 2013, 956792. [Google Scholar] [CrossRef]
- Catarzi, S.; Romagnoli, C.; Marcucci, G.; Favilli, F.; Iantomasi, T.; Vincenzini, M.T. Redox regulation of ERK1/2 activation induced by sphingosine 1-phosphate in fibroblasts: Involvement of NADPH oxidase and platelet-derived growth factor receptor. Biochim. Biophys. Acta 2011, 1810, 446–456. [Google Scholar] [CrossRef] [PubMed]
- Ray, P.D.; Huang, B.W.; Tsuji, Y. Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling. Cell. Signal. 2012, 24, 981–990. [Google Scholar] [CrossRef] [PubMed]
- Hong, S.E.; Lee, J.; Seo, D.H.; In Lee, H.; Ri Park, D.; Lee, G.R.; Jo, Y.J.; Kim, N.; Kwon, M.; Shon, H.; et al. Euphorbia factor L1 inhibits osteoclastogenesis by regulating cellular redox status and induces Fas-mediated apoptosis in osteoclast. Free Radic. Biol. Med. 2017, 112, 191–199. [Google Scholar] [CrossRef]
- Lee, N.K.; Choi, Y.G.; Baik, J.Y.; Han, S.Y.; Jeong, D.W.; Bae, Y.S.; Kim, N.; Lee, S.Y. A crucial role for reactive oxygen species in RANKL-induced osteoclast differentiation. Blood 2005, 106, 852–859. [Google Scholar] [CrossRef]
- Fatokun, A.A.; Stone, T.W.; Smith, R.A. Responses of differentiated MC3T3-E1 osteoblast-like cells to reactive oxygen species. Eur. J. Pharmacol. 2008, 587, 35–41. [Google Scholar] [CrossRef] [PubMed]
- Altindag, O.; Erel, O.; Soran, N.; Celik, H.; Selek, S. Total oxidative/anti-oxidative status and relation to bone mineral density in osteoporosis. Rheumatol. Int. 2008, 28, 317–321. [Google Scholar] [CrossRef] [PubMed]
- Kimball, J.S.; Johnson, J.P.; Carlson, D.A. Oxidative Stress and Osteoporosis. J. Bone Jt. Surg. 2021, 103, 1451–1461. [Google Scholar] [CrossRef]
- Wang, Y.F.; Chang, Y.Y.; Zhang, X.M.; Gao, M.T.; Zhang, Q.L.; Li, X.; Zhang, L.; Yao, W.F. Salidroside protects against osteoporosis in ovariectomized rats by inhibiting oxidative stress and promoting osteogenesis via Nrf2 activation. Phytomedicine 2022, 99, 154020. [Google Scholar] [CrossRef]
- Amarasekara, D.S.; Yun, H.; Kim, S.; Lee, N.; Kim, H.; Rho, J. Regulation of Osteoclast Differentiation by Cytokine Networks. Immune Netw. 2018, 18, e8. [Google Scholar] [CrossRef]
- Narimiya, T.; Kanzaki, H.; Yamaguchi, Y.; Wada, S.; Katsumata, Y.; Tanaka, K.; Tomonari, H. Nrf2 activation in osteoblasts suppresses osteoclastogenesis via inhibiting IL-6 expression. Bone Rep. 2019, 11, 100228. [Google Scholar] [CrossRef]
- Hyeon, S.; Lee, H.; Yang, Y.; Jeong, W. Nrf2 deficiency induces oxidative stress and promotes RANKL-induced osteoclast differentiation. Free Radic. Biol. Med. 2013, 65, 789–799. [Google Scholar] [CrossRef] [PubMed]
- Xu, L.; Shen, L.; Yu, X.; Li, P.; Wang, Q.; Li, C. Effects of irisin on osteoblast apoptosis and osteoporosis in postmenopausal osteoporosis rats through upregulating Nrf2 and inhibiting NLRP3 inflammasome. Exp. Ther. Med. 2020, 19, 1084–1090. [Google Scholar] [CrossRef] [PubMed]
- Zheng, D.; Cui, C.; Shao, C.; Wang, Y.; Ye, C.; Lv, G. Coenzyme Q10 inhibits RANKL-induced osteoclastogenesis by regulation of mitochondrial apoptosis and oxidative stress in RAW264.7 cells. J. Biochem. Mol. Toxicol. 2021, 35, e22778. [Google Scholar] [CrossRef] [PubMed]
- Liu, M.; Liu, S.; Zhang, Q.; Fang, Y.; Yu, Y.; Zhu, L.; Liu, Y.; Gong, W.; Zhao, L.; Qin, L.; et al. Curculigoside attenuates oxidative stress and osteoclastogenesis via modulating Nrf2/NF-κB signaling pathway in RAW264.7 cells. J. Ethnopharmacol. 2021, 275, 114129. [Google Scholar] [CrossRef]
- Kim, B.; Lee, K.Y.; Park, B. Icariin abrogates osteoclast formation through the regulation of the RANKL-mediated TRAF6/NF-κB/ERK signaling pathway in Raw264.7 cells. Phytomedicine 2018, 51, 181–190. [Google Scholar] [CrossRef] [PubMed]
- Ashtar, M.; Tenshin, H.; Teramachi, J.; Bat-Erdene, A.; Hiasa, M.; Oda, A.; Tanimoto, K.; Shimizu, S.; Higa, Y.; Harada, T.; et al. The Roles of ROS Generation in RANKL-Induced Osteoclastogenesis: Suppressive Effects of Febuxostat. Cancers 2020, 12, 929. [Google Scholar] [CrossRef]
- Thummuri, D.; Naidu, V.G.M.; Chaudhari, P. Carnosic acid attenuates RANKL-induced oxidative stress and osteoclastogenesis via induction of Nrf2 and suppression of NF-κB and MAPK signalling. J. Mol. Med. 2017, 95, 1065–1076. [Google Scholar] [CrossRef]
- Bai, X.C.; Lu, D.; Liu, A.L.; Zhang, Z.M.; Li, X.M.; Zou, Z.P.; Zeng, W.S.; Cheng, B.L.; Luo, S.Q. Reactive oxygen species stimulates receptor activator of NF-kappaB ligand expression in osteoblast. J. Biol. Chem. 2005, 280, 17497–17506. [Google Scholar] [CrossRef]
- Kanzaki, H.; Shinohara, F.; Kajiya, M.; Kodama, T. The Keap1/Nrf2 protein axis plays a role in osteoclast differentiation by regulating intracellular reactive oxygen species signaling. J. Biol. Chem. 2013, 288, 23009–23020. [Google Scholar] [CrossRef]
- Bai, X.C.; Lu, D.; Bai, J.; Zheng, H.; Ke, Z.Y.; Li, X.M.; Luo, S.Q. Oxidative stress inhibits osteoblastic differentiation of bone cells by ERK and NF-kappaB. Biochem. Biophys. Res. Commun. 2004, 314, 197–207. [Google Scholar] [CrossRef]
- Li, X.; Lin, H.; Zhang, X.; Jaspers, R.T.; Yu, Q.; Ji, Y.; Forouzanfar, T.; Wang, D.; Huang, S.; Wu, G. Notoginsenoside R1 attenuates oxidative stress-induced osteoblast dysfunction through JNK signalling pathway. J. Cell. Mol. Med. 2021, 25, 11278–11289. [Google Scholar] [CrossRef]
- Hu, X.; Li, B.; Wu, F.; Liu, X.; Liu, M.; Wang, C.; Shi, Y.; Ye, L. GPX7 Facilitates BMSCs Osteoblastogenesis via ER Stress and mTOR Pathway. J. Cell. Mol. Med. 2021, 25, 10454–10465. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Yang, X.; Zhang, J. Bridges between mitochondrial oxidative stress, ER stress and mTOR signaling in pancreatic β cells. Cell. Signal. 2016, 28, 1099–1104. [Google Scholar] [CrossRef]
- Almeida, M.; Han, L.; Martin-Millan, M.; O’Brien, C.A.; Manolagas, S.C. Oxidative stress antagonizes Wnt signaling in osteoblast precursors by diverting beta-catenin from T cell factor- to forkhead box O-mediated transcription. J. Biol. Chem. 2007, 282, 27298–27305. [Google Scholar] [CrossRef]
- Tian, L.M.; Xie, H.F.; Xiao, X.; Yang, T.; Hu, Y.H.; Wang, W.Z.; Liu, L.S.; Chen, X.; Li, J. Study on the roles of β-catenin in hydrogen peroxide-induced senescence in human skin fibroblasts. Exp. Dermatol. 2011, 20, 836–883. [Google Scholar] [CrossRef] [PubMed]
- Aaron, J.S.; Barfield, A.M.; Barton, S.; Dong, Y. Understanding Reactive Oxygen Species in Bone Regeneration: A Glance at Potential Therapeutics and Bioengineering Applications. Front. Bioeng. Biotechnol. 2022, 10, 836764. [Google Scholar]
- Takada, I.; Kouzmenko, A.P.; Kato, S. Wnt and γgamma signaling in osteoblastogenesis and adipogenesis. Nat. Rev. Rheumatol. 2009, 5, 442–447. [Google Scholar] [CrossRef]
- Li, L.; Wang, H.; Chen, X.; Li, X.; Wang, G.; Jie, Z.; Zhao, X.; Sun, X.; Huang, H.; Fan, S.; et al. Oxidative Stress-Induced Hypermethylation of KLF5 Promoter Mediated by DNMT3B Impairs Osteogenesis by Diminishing the Interaction with β-Catenin. Antioxid. Redox Signal. 2021, 35, 1–20. [Google Scholar] [CrossRef] [PubMed]
- Yang, Y.; Sun, Y.; Mao, W.W.; Zhang, H.; Ni, B.; Jiang, L. Oxidative stress induces downregulation of TP53INP2 and suppresses osteogenic differentiation of BMSCs during osteoporosis through the autophagy degradation pathway. Free Radic. Biol. Med. 2021, 166, 226–237. [Google Scholar] [CrossRef] [PubMed]
- Domazetovic, V.; Marcucci, G.; Falsetti, I.; Bilia, A.R.; Vincenzini, M.T.; Brandi, M.L.; Iantomasi, T. Blueberry Juice Antioxidants Protect Osteogenic Activity against Oxidative Stress and Improve Long-Term Activation of the Mineralization Process in Human Osteoblast-Like SaOS-2 Cells: Involvement of SIRT1. Antioxidants 2020, 9, 125. [Google Scholar] [CrossRef]
- Li, X.; Chen, Y.; Mao, Y.; Dai, P.; Sun, X.; Zhang, X.; Cheng, H.; Wang, Y.; Banda, I.; Wu, G.; et al. Curcumin Protects Osteoblasts From Oxidative Stress-Induced Dysfunction via GSK3β-Nrf2 Signaling Pathway. Front. Bioeng. Biotechnol. 2020, 8, 625. [Google Scholar] [CrossRef] [PubMed]
- Huang, L.; Su, W.; Wu, Z.; Zheng, L.; Lv, C. Glucosamine suppresses oxidative stress and induces protective autophagy in osteoblasts by blocking the ROS/Akt/mTOR signaling pathway. Cell Biol. Int. 2022, 46, 829–839. [Google Scholar] [CrossRef] [PubMed]
- Liu, M.; Wu, X.; Cui, Y.; Liu, P.; Xiao, B.; Zhang, X.; Zhang, J.; Sun, Z.; Song, M.; Shao, B.; et al. Mitophagy and apoptosis mediated by ROS participate in AlCl3-induced MC3T3-E1 cell dysfunction. Food Chem. Toxicol. 2021, 155, 112388. [Google Scholar] [CrossRef] [PubMed]
- Zhang, J.K.; Yang, L.; Meng, G.L.; Yuan, Z.; Fan, J.; Li, D.; Chen, J.Z.; Shi, T.Y.; Hu, H.M.; Wei, B.Y.; et al. Protection by salidroside against bone loss via inhibition of oxidative stress and bone-resorbing mediators. PLoS ONE 2013, 8, e57251. [Google Scholar] [CrossRef] [PubMed]
- Jie, J.; Li, W.; Wang, G.; Xu, X. FK506 ameliorates osteoporosis caused by osteoblast apoptosis via suppressing the activated CaN/NFAT pathway during oxidative stress. Inflamm. Res. 2021, 70, 789–797. [Google Scholar] [CrossRef]
- Guo, J.; Qin, W.; Xing, Q.; Gao, M.; Wei, F.; Song, Z.; Chen, L.; Lin, Y.; Gao, X.; Lin, Z. TRIM33 is essential for osteoblast proliferation and differentiation via BMP pathway. J. Cell. Physiol. 2017, 232, 3158–3169. [Google Scholar] [CrossRef]
- Zou, D.B.; Mou, Z.; Wu, W.; Liu, H. TRIM33 protects osteoblasts from oxidative stress-induced apoptosis in osteoporosis by inhibiting FOXO3a ubiquitylation and degradation. Aging Cell 2021, 20, e13367. [Google Scholar] [CrossRef] [PubMed]
- Wang, N.; Xu, P.; Wu, R.; Wang, X.; Wang, Y.; Shou, D.; Zhang, Y. Timosaponin BII improved osteoporosis caused by hyperglycemia through promoting autophagy of osteoblasts via suppressing the mTOR/NFκB signaling pathway. Free Radic. Biol. Med. 2021, 171, 112–123. [Google Scholar] [CrossRef] [PubMed]
- Schwartz, A.V. Efficacy of Osteoporosis Therapies in Diabetic Patients. Calcif. Tissue Int. 2017, 100, 165–173. [Google Scholar] [CrossRef] [PubMed]
- Costantini, S.; Conte, C. Bone health in diabetes and prediabetes. World J. Diabetes. 2019, 10, 421–445. [Google Scholar] [CrossRef]
- Zhao, Z.; Lu, Y.; Wang, H.; Gu, X.; Zhu, L.; Guo, H.; Li, N. ALK7 Inhibition Protects Osteoblast Cells Against High Glucoseinduced ROS Production via Nrf2/HO-1 Signaling Pathway. Curr. Mol. Med. 2022, 22, 354–364. [Google Scholar] [CrossRef] [PubMed]
- Conaway, H.H.; Henning, P.; Lie, A.; Tuckermann, J.; Lerner, U.H. Activation of dimeric glucocorticoid receptors in osteoclast progenitors potentiates RANKL induced mature osteoclast bone resorbing activity. Bone 2016, 93, 43–54. [Google Scholar] [CrossRef] [PubMed]
- Moutsatsou, P.; Kassi, E.; Papavassiliou, A.G. Glucocorticoid receptor signaling in bone cells. Trends Mol. Med. 2012, 18, 348–359. [Google Scholar] [CrossRef] [PubMed]
- Martin-Aragon, S.; Bermejo-Bescós, P.; Benedí, J.; Raposo, C.; Marques, F.; Kydonaki, E.K.; Gkiata, P.; Koutedakis, Y.; Ntina, G.; Carrillo, A.E.; et al. Neuroprotective Bovine Colostrum Attenuates Apoptosis in Dexamethasone-Treated MC3T3-E1 Osteoblastic Cells. Int. J. Mol. Sci. 2021, 22, 10195. [Google Scholar] [CrossRef] [PubMed]
- Huang, L.; Geng, D. Inhibition of MAGL activates the Keap1/Nrf2 pathway to attenuate glucocorticoid-induced osteonecrosis of the femoral head. Clin. Transl. Med. 2021, 11, e447. [Google Scholar]
- Zheng, Y.H.; Yang, J.J.; Tang, P.J.; Zhu, Y.; Chen, Z.; She, C.; Chen, G.; Cao, P.; Xu, X.Y. A novel Keap1 inhibitor iKeap1 activates Nrf2 signaling and ameliorates hydrogen peroxide-induced oxidative injury and apoptosis in osteoblasts. Cell Death Dis. 2021, 12, 679. [Google Scholar] [CrossRef]
- Kim, S.Y.; Cha, H.J.; Hwangbo, H.; Park, C.; Lee, H.; Song, K.S.; Shim, J.H.; Noh, J.S.; Kim, H.S.; Lee, B.J.; et al. Protection against Oxidative Stress-Induced Apoptosis by Fermented Sea Tangle (Laminaria japonica Aresch) in Osteoblastic MC3T3-E1 Cells through Activation of Nrf2 Signaling Pathway. Foods 2021, 10, 2807. [Google Scholar] [CrossRef]
- Vakili, S.; Zal, F.; Mostafavi-Pour, Z.; Savardashtaki, A.; Koohpeyma, F. Quercetin and vitamin E alleviate ovariectomy-induced osteoporosis by modulating autophagy and apoptosis in rat bone cells. J. Cell. Physiol. 2021, 236, 3495–3509. [Google Scholar] [CrossRef] [PubMed]
- Geng, Q.; Gao, H.; Yang, R.; Guo, K.; Miao, D. Pyrroloquinoline Quinone Prevents Estrogen Deficiency-Induced Osteoporosis by Inhibiting Oxidative Stress and Osteocyte Senescence. Int. J. Biol. Sci. 2019, 15, 58–68. [Google Scholar] [CrossRef] [PubMed]
- Chen, L.; Wang, G.; Wang, Q.; Liu, Q.; Sun, Q.; Chen, L. N-acetylcysteine prevents orchiectomy-induced osteoporosis by inhibiting oxidative stress and osteocyte senescence. Am. J. Transl. Res. 2019, 11, 4337–4347. [Google Scholar] [PubMed]
- Ru, J.Y.; Wang, Y.F. Osteocyte apoptosis: The roles and key molecular mechanisms in resorption-related bone diseases. Cell Death Dis. 2020, 11, 846. [Google Scholar] [CrossRef]
- Werner, S.L.; Sharma, R.; Woodruff, K.; Horn, D.; Harris, S.E.; Gorin, Y.; Lee, D.Y.; Hua, R.; Gu, S.; Fajardo, R.J.; et al. CSF-1 in Osteocytes Inhibits Nox4-mediated Oxidative Stress and Promotes Normal Bone Homeostasis. JBMR Plus 2019, 4, e10080. [Google Scholar] [CrossRef] [Green Version]
- Fontani, F.; Marcucci, G.; Iantomasi, T.; Brandi, M.L.; Vincenzini, M.T. Glutathione, N-acetylcysteine and lipoic acid down-regulate starvation-induced apoptosis, RANKL/OPG ratio and sclerostin in osteocytes: Involvement of JNK and ERK1/2 signalling. Calcif. Tissue Int. 2015, 96, 335–346. [Google Scholar] [CrossRef]
- Kar, R.; Riquelme, M.A.; Hua, R.; Jiang, J.X. Glucocorticoid-Induced Autophagy Protects Osteocytes Against Oxidative Stress Through Activation of MAPK/ERK Signaling. JBMR Plus 2018, 3, e10077. [Google Scholar] [CrossRef]
- Zeng, Y.; Liang, H.; Guo, Y.; Feng, Y.; Yao, Q. Adiponectin regulates osteocytic MLO-Y4 cell apoptosis in a high-glucose environment through the AMPK/FoxO3a signaling pathway. J. Cell. Physiol. 2021, 236, 7088–7096. [Google Scholar] [CrossRef]
- Yin, J.; Han, L.; Cong, W. Alpinumisoflavone rescues glucocorticoid-induced apoptosis of osteocytes via suppressing Nox2-dependent ROS generation. Pharmacol. Rep. 2018, 70, 270–276. [Google Scholar] [CrossRef]
- Pignolo, R.J.; Law, S.F.; Chandra, A. Bone Aging, Cellular Senescence, and Osteoporosis. JBMR Plus 2021, 5, e10488. [Google Scholar] [CrossRef]
- Ali, D.; Chen, L.; Kowal, J.M.; Okla, M.; Manikandan, M.; AlShehri, M.; AlMana, Y.; AlObaidan, R.; AlOtaibi, N.; Hamam, R.; et al. Resveratrol inhibits adipocyte differentiation and cellular senescence of human bone marrow stromal stem cells. Bone 2020, 133, 115252. [Google Scholar] [CrossRef]
- Kim, H.J.; Kim, W.J.; Shin, H.R.; Yoon, H.I.; Moon, J.I.; Lee, E.; Lim, J.M.; Cho, Y.D.; Lee, M.H.; Kim, H.G.; et al. ROS-induced PADI2 downregulation accelerates cellular senescence via the stimulation of SASP production and NFκB activation. Cell Mol. Life Sci. 2022, 79, 155. [Google Scholar] [CrossRef]
- Schoppa, A.M.; Chen, X.; Ramge, J.M.; Vikman, A.; Fischer, V.; Haffner-Luntzer, M.; Riegger, J.; Tuckermann, J.; Scharffetter-Kochanek, K.; Ignatius, A. Osteoblast lineage Sod2 deficiency leads to an osteoporosis-like phenotype in mice. Dis. Model. Mech. 2022, 15, dmm049392. [Google Scholar] [CrossRef]
- Martiniakova, M.; Babikova, M.; Mondockova, V.; Blahova, J.; Kovacova, V.; Omelka, R. The Role of Macronutrients, Micronutrients and Flavonoid Polyphenols in the Prevention and Treatment of Osteoporosis. Nutrients 2022, 14, 523. [Google Scholar] [CrossRef]
- Bellavia, D.; Caradonna, F.; Dimarco, E.; Costa, V.; Carina, V.; De Luca, A.; Raimondi, L.; Fini, M.; Gentile, C.; Giavaresi, G. Non-flavonoid polyphenols in osteoporosis: Preclinical evidence. Trends Endocrinol. Metab. 2021, 32, 515–529. [Google Scholar] [CrossRef]
- Saxena, Y.; Routh, S.; Mukhopadhaya, A. Immunoporosis: Role of Innate Immune Cells in Osteoporosis. Front. Immunol. 2021, 12, 687037. [Google Scholar] [CrossRef]
- Mundy, G.R. Osteoporosis and inflammation. Nutr. Rev. 2007, 65, S147–S151. [Google Scholar] [CrossRef]
- Fischer, V.; Haffner-Luntzer, M. Interaction between bone and immune cells: Implications for postmenopausal osteoporosis. Semin. Cell Dev. Biol. 2022, 123, 14–21. [Google Scholar] [CrossRef]
- Liu, H.; Wu, X.; Gang, N.; Wang, S.; Deng, W.; Zan, L.; Yu, S. Macrophage functional phenotype can be consecutively and reversibly shifted to adapt to microenvironmental changes. Int. J. Clin. Exp. Med. 2015, 8, 3044–3053. [Google Scholar]
- Dou, C.; Ding, N.; Zhao, C.; Hou, T.; Kang, F.; Cao, Z.; Liu, C.; Bai, Y.; Dai, Q.; Ma, Q.; et al. Estrogen Deficiency-Mediated M2 Macrophage Osteoclastogenesis Contributes to M1/M2 Ratio Alteration in Ovariectomized Osteoporotic Mice. J. Bone Miner. Res. 2018, 33, 899–908. [Google Scholar] [CrossRef]
- D’Amelio, P.; Grimaldi, A.; Pescarmona, G.P.; Tamone, C.; Roato, I.; Isaia, G. Spontaneous osteoclast formation from peripheral blood mononuclear cells in postmenopausal osteoporosis. FASEB J. 2005, 19, 410–412. [Google Scholar] [CrossRef]
- Salamanna, F.; Giardino, R.; Fini, M. Spontaneous osteoclastogenesis: Hypothesis for gender-unrelated osteoporosis screening and diagnosis. Med. Hypotheses 2017, 109, 70–72. [Google Scholar] [CrossRef]
- Salamanna, F.; Maglio, M.; Borsari, V.; Landini, M.P.; Fini, M. Blood factors as biomarkers in osteoporosis: Points from the COVID-19 era. Trends Endocrinol. Metab. 2021, 32, 672–679. [Google Scholar] [CrossRef]
- Cline-Smith, A.; Axelbaum, A.; Shashkova, E.; Chakraborty, M.; Sanford, J.; Panesar, P.; Peterson, M.; Cox, L.; Baldan, A.; Veis, D.; et al. Ovariectomy Activates Chronic Low-Grade Inflammation Mediated by Memory T Cells, Which Promotes Osteoporosis in Mice. J. Bone Miner. Res. 2020, 35, 1174–1187. [Google Scholar] [CrossRef]
- Narisawa, M.; Kubo, S.; Okada, Y.; Yamagata, K.; Nakayamada, S.; Sakata, K.; Yamaoka, K.; Tanaka, Y. Human dendritic cell-derived osteoclasts with high bone resorption capacity and T cell stimulation ability. Bone 2021, 142, 115616. [Google Scholar] [CrossRef]
- Alnaeeli, M.; Penninger, J.M.; Teng, Y.T. Immune interactions with CD4+ T cells promote the development of functional osteoclasts from murine CD11c+ dendritic cells. J. Immunol. 2006, 177, 3314–3326. [Google Scholar] [CrossRef] [Green Version]
- Yu, X.Y.; Li, X.S.; Li, Y.; Liu, T.; Wang, R.T. Neutrophil-lymphocyte ratio is associated with arterial stiffness in postmenopausal women with osteoporosis. Arch. Gerontol. Geriatr. 2015, 61, 76–80. [Google Scholar] [CrossRef]
- Iking-Konert, C.; Ostendorf, B.; Sander, O.; Jost, M.; Wagner, C.; Joosten, L.; Schneider, M.; Hänsch, G.M. Transdifferentiation of polymorphonuclear neutrophils to dendritic-like cells at the site of inflammation in rheumatoid arthritis: Evidence for activation by T cells. Ann. Rheum. Dis. 2005, 64, 1436–1442. [Google Scholar] [CrossRef]
- Ginaldi, L.; De Martinis, M.; Ciccarelli, F.; Saitta, S.; Imbesi, S.; Mannucci, C.; Gangemi, S. Increased levels of interleukin 31 (IL-31) in osteoporosis. BMC Immunol. 2015, 16, 60. [Google Scholar] [CrossRef]
- Kroner, J.; Kovtun, A.; Kemmler, J.; Messmann, J.J.; Strauss, G.; Seitz, S.; Schinke, T.; Amling, M.; Kotrba, J.; Froebel, J.; et al. Mast Cells Are Critical Regulators of Bone Fracture-Induced Inflammation and Osteoclast Formation and Activity. J. Bone Miner. Res. 2017, 32, 2431–2444. [Google Scholar] [CrossRef]
- Ragipoglu, D.; Dudeck, A.; Haffner-Luntzer, M.; Voss, M.; Kroner, J.; Ignatius, A.; Fischer, V. The Role of Mast Cells in Bone Metabolism and Bone Disorders. Front Immunol. 2020, 11, 163. [Google Scholar] [CrossRef]
- Fang, H.; Zhang, H.; Wang, Z.; Zhou, Z.; Li, Y.; Lu, L. Systemic immune-inflammation index acts as a novel diagnostic biomarker for postmenopausal osteoporosis and could predict the risk of osteoporotic fracture. J. Clin. Lab. Anal. 2020, 34, e23016. [Google Scholar] [CrossRef]
- Lei, Z.; Xiaoying, Z.; Xingguo, L. Ovariectomy-associated changes in bone mineral density and bone marrow haematopoiesis in rats. Int. J. Exp. Pathol. 2009, 90, 512–519. [Google Scholar] [CrossRef]
- Kireev, R.A.; Tresguerres, A.C.; Garcia, C.; Borras, C.; Ariznavarreta, C.; Vara, E.; Vina, J.; Tresguerres, J.A. Hormonal regulation of pro-inflammatory and lipid peroxidation processes in liver of old ovariectomized female rats. Biogerontology 2010, 11, 229–243. [Google Scholar] [CrossRef]
- Rodrigues, M.F.C.; Ferreira, F.C.; Silva-Magosso, N.S.; Barbosa, M.R.; Souza, M.V.C.; Domingos, M.M.; Canevazzi, G.H.R.; Stotzer, U.S.; Peviani, S.M.; de Lira, F.S.; et al. Effects of resistance training and estrogen replacement on adipose tissue inflammation in ovariectomized rats. Appl. Physiol. Nutr. Metab. 2017, 42, 605–612. [Google Scholar] [CrossRef]
- Zhai, X.; Yan, Z.; Zhao, J.; Chen, K.; Yang, Y.; Cai, M.; He, C.; Huang, C.; Li, B.; Yang, M.; et al. Muscone Ameliorates Ovariectomy-Induced Bone Loss and Receptor Activator of Nuclear Factor-κb Ligand-Induced Osteoclastogenesis by Suppressing TNF Receptor-Associated Factor 6-Mediated Signaling Pathways. Front. Pharmacol. 2020, 11, 348. [Google Scholar] [CrossRef] [Green Version]
- Iseme, R.A.; Mcevoy, M.; Kelly, B.; Agnew, L.; Walker, F.R.; Attia, J. Is osteoporosis an autoimmune mediated disorder? Bone Rep. 2017, 7, 121–131. [Google Scholar] [CrossRef]
- McDonald, M.M.; Khoo, W.H.; Ng, P.Y.; Xiao, Y.; Zamerli, J.; Thatcher, P.; Kyaw, W.; Pathmanandavel, K.; Grootveld, A.K.; Moran, I.; et al. Osteoclasts recycle via osteomorphs during RANKL-stimulated bone resorption. Cell 2021, 184, 1940. [Google Scholar] [CrossRef]
- Meng, J.; Zhang, X.; Guo, X.; Cheng, W.; Qi, X.; Huang, J.; Lin, W. Briarane-type diterpenoids suppress osteoclastogenisis by regulation of Nrf2 and MAPK/NF-kB signaling pathway. Bioorg. Chem. 2021, 112, 104976. [Google Scholar] [CrossRef]
- Ruscitti, P.; Cipriani, P.; Carubbi, F.; Liakouli, V.; Zazzeroni, F.; Di Benedetto, P.; Berardicurti, O.; Alesse, E.; Giacomelli, R. The role of IL-1β in the bone loss during rheumatic diseases. Mediat. Inflamm. 2015, 2015, 782382. [Google Scholar] [CrossRef]
- Mansoori, M.N.; Shukla, P.; Kakaji, M.; Tyagi, A.M.; Srivastava, K.; Shukla, M.; Dixit, M.; Kureel, J.; Gupta, S.; Singh, D. IL-18BP is decreased in osteoporotic women: Prevents Inflammasome mediated IL-18 activation and reduces Th17 differentiation. Sci. Rep. 2016, 6, 33680. [Google Scholar] [CrossRef] [PubMed]
- Adami, G. Regulation of bone mass in inflammatory diseases. Best Pract. Res. Clin. Endocrinol. Metab. 2022, 36, 101611. [Google Scholar] [CrossRef]
- Mao, C.Y.; Wang, Y.G.; Zhang, X.; Zheng, X.Y.; Tang, T.T.; Lu, E.Y. Double-edged-sword effect of IL-1β on the osteogenesis of periodontal ligament stem cells via crosstalk between the NF-κB, MAPK and BMP/Smad signaling pathways. Cell Death Dis. 2016, 7, e2296. [Google Scholar] [CrossRef]
- Huang, R.L.; Yuan, Y.; Tu, J.; Zou, G.M.; Li, Q. Opposing TNF-α/IL-1β- and BMP-2-activated MAPK signaling pathways converge on Runx2 to regulate BMP-2-induced osteoblastic differentiation. Cell Death Dis. 2014, 5, e1187. [Google Scholar] [CrossRef] [PubMed]
- Palmqvist, P.; Persson, E.; Conaway, H.H.; Lerner, U.H. IL-6, leukemia inhibitory factor, and oncostatin M stimulate bone resorption and regulate the expression of receptor activator of NF-kappa B ligand, osteoprotegerin, and receptor activator of NF-kappa B in mouse calvariae. J. Immunol. 2002, 169, 3353–3362. [Google Scholar] [CrossRef]
- Malysheva, K.; de Rooij, K.; Lowik, C.W.; Baeten, D.L.; Rose-John, S.; Stoika, R.; Korchynskyi, O. Interleukin 6/Wnt interactions in rheumatoid arthritis: Interleukin 6 inhibits Wnt signaling in synovial fibroblasts and osteoblasts. Croat Med. J. 2016, 57, 89–98. [Google Scholar] [CrossRef] [PubMed]
- Adami, G.; Orsolini, G.; Adami, S.; Viapiana, O.; Idolazzi, L.; Gatti, D.; Rossini, M. Effects of TNF Inhibitors on Parathyroid Hormone and Wnt Signaling Antagonists in Rheumatoid Arthritis. Calcif. Tissue Int. 2016, 99, 360–364. [Google Scholar] [CrossRef]
- Xu, L.; Zhang, L.; Wang, Z.; Li, C.; Li, S.; Li, L.; Fan, Q.; Zheng, L. Melatonin Suppresses Estrogen Deficiency-Induced Osteoporosis and Promotes Osteoblastogenesis by Inactivating the NLRP3 Inflammasome. Calcif. Tissue Int. 2018, 103, 400–410. [Google Scholar] [CrossRef]
- Yin, W.; Liu, S.; Dong, M.; Liu, Q.; Shi, C.; Bai, H.; Wang, Q.; Yang, X.; Niu, W.; Wang, L. A New NLRP3 Inflammasome Inhibitor, Dioscin, Promotes Osteogenesis. Small 2020, 16, e1905977. [Google Scholar] [CrossRef]
- An, Y.; Zhang, H.; Wang, C.; Jiao, F.; Xu, H.; Wang, X.; Luan, W.; Ma, F.; Ni, L.; Tang, X.; et al. Activation of ROS/MAPKs/NF-κB/NLRP3 and inhibition of efferocytosis in osteoclast-mediated diabetic osteoporosis. FASEB J. 2019, 33, 12515–12527. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.; Wang, Q.; Su, H.; Cheng, J. Exosomes from adipose derived mesenchymal stem cells alleviate diabetic osteoporosis in rats through suppressing NLRP3 inflammasome activation in osteoclasts. J. Biosci. Bioeng. 2021, 131, 671–678. [Google Scholar] [CrossRef] [PubMed]
- Alippe, Y.; Wang, C.; Ricci, B.; Xiao, J.; Qu, C.; Zou, W.; Novack, D.V.; Abu-Amer, Y.; Civitelli, R.; Mbalaviele, G. Bone matrix components activate the NLRP3 inflammasome and promote osteoclast differentiation. Sci. Rep. 2017, 7, 6630. [Google Scholar] [CrossRef] [PubMed]
- Wang, L.; Chen, K.; Wan, X.; Wang, F.; Guo, Z.; Mo, Z. NLRP3 inflammasome activation in mesenchymal stem cells inhibits osteogenic differentiation and enhances adipogenic differentiation. Biochem. Biophys. Res. Commun. 2017, 484, 871–877. [Google Scholar] [CrossRef]
- Shi, J.; Zhao, Y.; Wang, K.; Shi, X.; Wang, Y.; Huang, H.; Zhuang, Y.; Cai, T.; Wang, F.; Shao, F. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature 2015, 526, 660–665. [Google Scholar] [CrossRef]
- Liu, S.; Du, J.; Li, D.; Yang, P.; Kou, Y.; Li, C.; Zhou, Q.; Lu, Y.; Hasegawa, T.; Li, M. Oxidative stress induced pyroptosis leads to osteogenic dysfunction of MG63 cells. J. Mol. Histol. 2020, 51, 221–232. [Google Scholar] [CrossRef]
- Zemanova, N.; Omelka, R.; Mondockova, V.; Kovacova, V.; Martiniakova, M. Roles of Gut Microbiome in Bone Homeostasis and Its Relationship with Bone-Related Diseases. Biology 2022, 11, 1402. [Google Scholar] [CrossRef]
- D’Amelio, P.; Sassi, F. Gut Microbiota, Immune System, and Bone. Calcif. Tissue Int. 2018, 102, 415–425. [Google Scholar] [CrossRef]
- Martiniakova, M.; Babikova, M.; Omelka, R. Pharmacological agents and natural compounds: Available treatments for osteoporosis. J. Physiol. Pharmacol. 2020, 71, 307–320. [Google Scholar] [CrossRef]
- Weilner, S.; Skalicky, S.; Salzer, B.; Keider, V.; Wagner, M.; Hildner, F.; Gabriel, C.; Dovjak, P.; Pietschmann, P.; Grillari-Voglauer, R.; et al. Differentially circulating miRNAs after recent osteoporotic fractures can influence osteogenic differentiation. Bone 2015, 79, 43–51. [Google Scholar] [CrossRef]
- Li, Z.; Xue, H.; Tan, G.; Xu, Z. Effects of miRNAs, lncRNAs and circRNAs on osteoporosis as regulatory factors of bone homeostasis (Review). Mol. Med. Rep. 2021, 24, 788. [Google Scholar] [CrossRef]
- Hu, G.; Drescher, K.M.; Chen, X.M. Exosomal miRNAs: Biological Properties and Therapeutic Potential. Front. Genet. 2012, 3, 56. [Google Scholar] [CrossRef]
- Salloum-Asfar, S.; Satheesh, N.J.; Abdulla, S.A. Circulating miRNAs, Small but Promising Biomarkers for Autism Spectrum Disorder. Front. Mol. Neurosci. 2019, 12, 253. [Google Scholar] [CrossRef]
- Liu, J.; Dang, L.; Wu, X.; Li, D.; Ren, Q.; Lu, A.; Zhang, G. microRNA-Mediated Regulation of Bone Remodeling: A Brief Review. JBMR Plus 2019, 3, e10213. [Google Scholar] [CrossRef]
- Wang, H.; Sun, Z.; Wang, Y.; Hu, Z.; Zhou, H.; Zhang, L.; Hong, B.; Zhang, S.; Cao, X. miR-33-5p, a novel mechano-sensitive microRNA promotes osteoblast differentiation by targeting Hmga2. Sci. Rep. 2016, 6, 23170. [Google Scholar] [CrossRef]
- Tang, X.; Lin, J.; Wang, G.; Lu, J. MicroRNA-433-3p promotes osteoblast differentiation through targeting DKK1 expression. PLoS ONE 2017, 12, e0179860. [Google Scholar] [CrossRef]
- Li, J.; He, X.; Wei, W.; Zhou, X. MicroRNA-194 promotes osteoblast differentiation via downregulating STAT1. Biochem. Biophys. Res. Commun. 2015, 460, 482–788. [Google Scholar] [CrossRef]
- Hu, R.; Liu, W.; Li, H.; Yang, L.; Chen, C.; Xia, Z.Y.; Guo, L.J.; Xie, H.; Zhou, H.D.; Wu, X.P.; et al. A Runx2/miR-3960/miR-2861 regulatory feedback loop during mouse osteoblast differentiation. J. Biol. Chem. 2011, 286, 12328–12339. [Google Scholar] [CrossRef]
- Krzeszinski, J.Y.; Wei, W.; Huynh, H.; Jin, Z.; Wang, X.; Chang, T.C.; Xie, X.J.; He, L.; Mangala, L.S.; Lopez-Berestein, G.; et al. Retraction Note: miR-34a blocks osteoporosis and bone metastasis by inhibiting osteoclastogenesis and Tgif2. Nature 2020, 582, 134. [Google Scholar] [CrossRef]
- Lee, Y.; Kim, H.J.; Park, C.K.; Kim, Y.G.; Lee, H.J.; Kim, J.Y.; Kim, H.H. MicroRNA-124 regulates osteoclast differentiation. Bone 2013, 56, 383–389. [Google Scholar] [CrossRef]
- Nakasa, T.; Shibuya, H.; Nagata, Y.; Niimoto, T.; Ochi, M. The inhibitory effect of microRNA-146a expression on bone destruction in collagen-induced arthritis. Arthritis Rheum. 2011, 63, 1582–1590. [Google Scholar] [CrossRef]
- Wang, H.; Xie, Z.; Hou, T.; Li, Z.; Huang, K.; Gong, J.; Zhou, W.; Tang, K.; Xu, J.; Dong, S. MiR-125b Regulates the Osteogenic Differentiation of Human Mesenchymal Stem Cells by Targeting BMPR1b. Cell Physiol. Biochem. 2017, 41, 530–542. [Google Scholar] [CrossRef]
- Du, F.; Wu, H.; Zhou, Z.; Liu, Y.U. microRNA-375 inhibits osteogenic differentiation by targeting runt-related transcription factor 2. Exp. Ther. Med. 2015, 10, 207–212. [Google Scholar] [CrossRef]
- Zhang, W.; Wu, Y.; Shiozaki, Y.; Sugimoto, Y.; Takigawa, T.; Tanaka, M.; Matsukawa, A.; Ozaki, T. miRNA-133a-5p Inhibits the Expression of Osteoblast Differentiation-Associated Markers by Targeting the 3’ UTR of RUNX2. DNA Cell Biol. 2018, 37, 199–209. [Google Scholar] [CrossRef]
- Sugatani, T.; Vacher, J.; Hruska, K.A. A microRNA expression signature of osteoclastogenesis. Blood 2011, 117, 3648–3657. [Google Scholar] [CrossRef]
- Franceschetti, T.; Kessler, C.B.; Lee, S.K.; Delany, A.M. miR-29 promotes murine osteoclastogenesis by regulating osteoclast commitment and migration. J. Cell. Biochem. 2013, 288, 33347–33360. [Google Scholar] [CrossRef]
- Ke, K.; Sul, O.J.; Rajasekaran, M.; Choi, H.S. MicroRNA-183 increases osteoclastogenesis by repressing heme oxygenase-1. Bone 2015, 81, 237–246. [Google Scholar] [CrossRef]
- Zhang, Y.; Xie, R.L.; Croce, C.M.; Stein, J.L.; Lian, J.B.; van Wijnen, A.J.; Stein, G.S. A program of microRNAs controls osteogenic lineage progression by targeting transcription factor Runx2. Proc. Natl. Acad. Sci. USA 2011, 108, 9863–9868. [Google Scholar] [CrossRef]
- Schaap-Oziemla, A.M.; Raymaker, R.A.; Bergevoet, S.M.; Gilissen, C.; Jansen, B.J.; Adema, G.J.; Kögler, G.; le Sage, C.; Agami, R.; van der Reijden, B.A.; et al. MicroRNA hsa-miR-135b regulates mineralization in osteogenic differentiation of human unrestricted somatic stem cells. Stem Cells Dev. 2010, 19, 877–885. [Google Scholar] [CrossRef]
- Chen, C.; Cheng, P.; Xie, H.; Zhou, H.D.; Wu, X.P.; Liao, E.Y.; Luo, X.H. MiR-503 regulates osteoclastogenesis via targeting RANK. J. Bone Miner. Res. 2014, 29, 338–347. [Google Scholar] [CrossRef]
- Mandourah, A.Y.; Ranganath, L.; Barraclough, R.; Vinjamuri, S.; Hof, R.V.; Hamill, S.; Czanner, G.; Dera, A.A.; Wang, D.; Barraclough, D.L. Circulating microRNAs as potential diagnostic biomarkers for osteoporosis. Sci. Rep. 2018, 8, 8421. [Google Scholar] [CrossRef]
- Olivieri, F.; Prattichizzo, F.; Giuliani, A.; Matacchione, G.; Rippo, M.R.; Sabbatinelli, J.; Bonafè, M. miR-21 and miR-146a: The microRNAs of inflammaging and age-related diseases. Ageing Res. Rev. 2021, 70, 101374. [Google Scholar] [CrossRef]
- Boldin, M.P.; Baltimore, D. MicroRNAs, new effectors and regulators of NF-κB. Immunol. Rev. 2012, 246, 205–220. [Google Scholar] [CrossRef]
- Yang, Y.K.; Ogando, C.R.; Wang See, C.; Chang, T.Y.; Barabino, G.A. Changes in phenotype and differentiation potential of human mesenchymal stem cells aging in vitro. Stem Cell Res. Ther. 2018, 9, 131. [Google Scholar] [CrossRef]
- Liao, L.; Su, X.; Yang, X.; Hu, C.; Li, B.; Lv, Y.; Shuai, Y.; Jing, H.; Deng, Z.; Jin, Y. TNF-α Inhibits FoxO1 by Upregulating miR-705 to Aggravate Oxidative Damage in Bone Marrow-Derived Mesenchymal Stem Cells during Osteoporosis. Stem Cells 2016, 34, 1054–1067. [Google Scholar] [CrossRef]
- Miller, C.H.; Smith, S.M.; Elguindy, M.; Zhang, T.; Xiang, J.Z.; Hu, X.; Ivashkiv, L.B.; Zhao, B. RBP-J-Regulated miR-182 Promotes TNF-α-Induced Osteoclastogenesis. J. Immunol. 2016, 196, 4977–4986. [Google Scholar] [CrossRef]
- Shen, G.; Ren, H.; Shang, Q.; Zhang, Z.; Zhao, W.; Yu, X.; Tang, J.; Yang, Z.; Liang, D.; Jiang, X. miR-128 plays a critical role in murine osteoclastogenesis and estrogen deficiency-induced bone loss. Theranostics 2020, 10, 4334–4348. [Google Scholar] [CrossRef]
- Wu, L.; Zhang, G.; Guo, C.; Zhao, X.; Shen, D.; Yang, N. MiR-128-3p mediates TNF-α-induced inflammatory responses by regulating Sirt1 expression in bone marrow mesenchymal stem cells. Biochem. Biophys. Res. Commun. 2020, 521, 98–105. [Google Scholar] [CrossRef]
- Fariyike, B.; Singleton, Q.; Hunter, M.; Hill, W.D.; Isales, C.M.; Hamrick, M.W.; Fulzele, S. Role of MicroRNA-141 in the Aging Musculoskeletal System: A Current Overview. Mech. Ageing Dev. 2019, 178, 9–15. [Google Scholar] [CrossRef]
- Magenta, A.; Cencioni, C.; Fasanaro, P.; Zaccagnini, G.; Greco, S.; Sarra-Ferraris, G.; Antonini, A.; Martelli, F.; Capogrossi, M.C. miR-200c is upregulated by oxidative stress and induces endothelial cell apoptosis and senescence via ZEB1 inhibition. Cell Death Differ. 2011, 18, 1628–1639. [Google Scholar] [CrossRef]
- Lam, W.Y.; Yeung, A.C.; Ngai, K.L.; Li, M.S.; To, K.F.; Tsui, S.K.; Chan, P.K. Effect of avian influenza A H5N1 infection on the expression of microRNA-141 in human respiratory epithelial cells. BMC Microbiol. 2013, 13, 104. [Google Scholar] [CrossRef]
- Bergo, M.O.; Gavino, B.; Ross, J.; Schmidt, W.K.; Hong, C.; Kendall, L.V.; Mohr, A.; Meta, M.; Genant, H.; Jiang, Y.; et al. Zmpste24 deficiency in mice causes spontaneous bone fractures, muscle weakness, and a prelamin A processing defect. Proc. Natl. Acad. Sci. USA 2002, 99, 13049–13054. [Google Scholar] [CrossRef]
- Chen, H.; Li, S.; Yin, H.; Hua, Z.; Shao, Y.; Wei, J.; Wang, J. MYC-mediated miR-320a affects receptor activator of nuclear factor κB ligand (RANKL)-induced osteoclast formation by regulating phosphatase and tensin homolog (PTEN). Bioengineered 2021, 12, 12677–12687. [Google Scholar] [CrossRef]
- Chalhoub, N.; Baker, S.J. PTEN and the PI3-kinase pathway in cancer. Annu. Rev. Pathol. 2009, 4, 127–150. [Google Scholar] [CrossRef]
- Kong, Y.; Nie, Z.K.; Li, F.; Guo, H.M.; Yang, X.L.; Ding, S.F. MiR-320a was highly expressed in postmenopausal osteoporosis and acts as a negative regulator in MC3T3E1 cells by reducing MAP9 and inhibiting PI3K/AKT signaling pathway. Exp. Mol. Pathol. 2019, 110, 104282. [Google Scholar] [CrossRef]
- De-Ugarte, L.; Balcells, S.; Nogues, X.; Grinberg, D.; Diez-Perez, A.; Garcia-Giralt, N. Pro-osteoporotic miR-320a impairs osteoblast function and induces oxidative stress. PLoS ONE 2018, 13, e0208131. [Google Scholar] [CrossRef]
- Gao, Z.; Chen, Z.; Xiong, Z.; Liu, X. Ferroptosis—A new target of osteoporosis. Exp. Gerontol. 2022, 165, 111836. [Google Scholar] [CrossRef]
- Wang, X.; Cao, Z.; Liu, L.; Liu, G.; Chen, B.; Zhu, K.; Xu, Y.; Xu, Y. Osteoporotic bone loss from excess iron accumulation is driven by NOX4-triggered ferroptosis in osteoblasts. Free Radic. Biol. Med. 2023. [Google Scholar] [CrossRef]
- Luo, Y.; Huang, Q.; He, B.; Liu, Y.; Huang, S.; Xiao, J. Regulation of ferroptosis by non-coding RNAs in the development and treatment of cancer (Review). Oncol. Rep. 2021, 45, 29–48. [Google Scholar] [CrossRef]
- Liu, H.; Wang, Y.W.; Chen, W.D.; Dong, H.H.; Xu, Y.J. Iron accumulation regulates osteoblast apoptosis through lncRNA XIST/miR-758-3p/caspase 3 axis leading to osteoporosis. IUBMB Life 2021, 73, 432–443. [Google Scholar] [CrossRef]
- Feng, Y.; He, P.Y.; Kong, W.D.; Cen, W.J.; Wang, P.L.; Liu, C.; Zhang, W.; Li, S.S.; Jiang, J.W. Apoptosis-promoting properties of miR-3074-5p in MC3T3-E1 cells under iron overload conditions. Cell. Mol. Biol. Lett. 2021, 26, 37. [Google Scholar] [CrossRef]
- Zhang, S.; Wu, W.; Jiao, G.; Li, C.; Liu, H. MiR-455-3p activates Nrf2/ARE signaling via HDAC2 and protects osteoblasts from oxidative stress. Int. J. Biol. Macromol. 2018, 107, 2094–2101. [Google Scholar] [CrossRef]
- Yan, T.B.; Li, C.; Jiao, G.J.; Wu, W.L.; Liu, H.C. TIMP-1 suppressed by miR-138 participates in endoplasmic reticulum stress-induced osteoblast apoptosis in osteoporosis. Free Radic. Res. 2018, 52, 223–231. [Google Scholar] [CrossRef]
- Zhang, W.; Cui, S.Y.; Yi, H.; Zhu, X.H.; Liu, W.; Xu, Y.J. MiR-708 inhibits MC3T3-E1 cells against H2O2-induced apoptosis through targeting PTEN. J. Orthop. Surg. Res. 2020, 15, 255. [Google Scholar] [CrossRef]
- Lu, X.Z.; Yang, Z.H.; Zhang, H.J.; Zhu, L.L.; Mao, X.L.; Yuan, Y. MiR-214 protects MC3T3-E1 osteoblasts against H2O2-induced apoptosis by suppressing oxidative stress and targeting ATF4. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 4762–4770. [Google Scholar] [PubMed]
miRNA | miRNA Target | Effect | Species | Ref. |
---|---|---|---|---|
miRNA-705 (+) | FOXO1 (−) | Oxidative damage | Osteoporotic mouse BMSCs | Liao et al. [172] |
miRNA-182 (+) | FOXO3 (−) Maml1 (−) | TNFα-induced osteoclastogenesis | Murine BMSCs | Miller et al. [173] |
miRNA-128 (+) | Sirt-1 (−) NF-kB (+) | Osteoclasogenesis | Mouse bone-marrow-derived macrophages | Shen et al. [174] |
miRNA-128-3p (+) | Sirt-1 (−) | Exacerbation of TNFα inflammation | Human BMSCs | Wu et al. [175] |
miRNA-141 (+) | ZMPSTE24 (−) | Alteration bone formation | Mice | Bergo et al. [179] |
miRNA-320 (+) | (PTEN) (−) PI3K/AKT (−) Osteoblastic genes and genes involved in redox homeostasis (−) | Osteoclastogenesis Oxidative damage Oxidative stress and reduced osteoblastic differentiation and functionality | RAW 264.7 cells MC3T3-E1 cells Human primary osteoblasts | Chen et al. [180] Kong et al. [182] De-Ugarte et al. [183] |
miRNA-138 (+) | TIMP-1 (−) | Apoptosis osteoblasts | MC3T3-E1 cells | Yan et al. [190] |
miRNA-455-3p (−) | HDAC2 (+) | Inhibition of Nfr2 Oxidative stress | MC3T3-E1 cells | Zhang et al. [189] |
miRNA-708 (−) | PTEN (+) | Apoptosis osteoblast Oxidative stress | MC3T3-E1 cells | Zhang et al. [191] |
miRNA-214 (−) | (ATF4) (+) | Apoptosis osteoblast Oxidative stress | MC3T3-E1 cells | Lu et al. [192] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Iantomasi, T.; Romagnoli, C.; Palmini, G.; Donati, S.; Falsetti, I.; Miglietta, F.; Aurilia, C.; Marini, F.; Giusti, F.; Brandi, M.L. Oxidative Stress and Inflammation in Osteoporosis: Molecular Mechanisms Involved and the Relationship with microRNAs. Int. J. Mol. Sci. 2023, 24, 3772. https://doi.org/10.3390/ijms24043772
Iantomasi T, Romagnoli C, Palmini G, Donati S, Falsetti I, Miglietta F, Aurilia C, Marini F, Giusti F, Brandi ML. Oxidative Stress and Inflammation in Osteoporosis: Molecular Mechanisms Involved and the Relationship with microRNAs. International Journal of Molecular Sciences. 2023; 24(4):3772. https://doi.org/10.3390/ijms24043772
Chicago/Turabian StyleIantomasi, Teresa, Cecilia Romagnoli, Gaia Palmini, Simone Donati, Irene Falsetti, Francesca Miglietta, Cinzia Aurilia, Francesca Marini, Francesca Giusti, and Maria Luisa Brandi. 2023. "Oxidative Stress and Inflammation in Osteoporosis: Molecular Mechanisms Involved and the Relationship with microRNAs" International Journal of Molecular Sciences 24, no. 4: 3772. https://doi.org/10.3390/ijms24043772
APA StyleIantomasi, T., Romagnoli, C., Palmini, G., Donati, S., Falsetti, I., Miglietta, F., Aurilia, C., Marini, F., Giusti, F., & Brandi, M. L. (2023). Oxidative Stress and Inflammation in Osteoporosis: Molecular Mechanisms Involved and the Relationship with microRNAs. International Journal of Molecular Sciences, 24(4), 3772. https://doi.org/10.3390/ijms24043772