Control of the Autophagy Pathway in Osteoarthritis: Key Regulators, Therapeutic Targets and Therapeutic Strategies
Abstract
:1. Background
2. Role of the Autophagic Process in Chondrogenic Differentiation
3. MicroRNAs Involvement in the Autophagy Process
4. Transcription Factors in OA
5. In Vitro and In Vivo Models for OA Studies
6. Therapeutic Targets in OA
6.1. Autophagy
6.2. Inflammation
6.3. Cell Senescence
- UBX0101(NCT04129944).
- Fisetin (NCT04210986).
6.4. microRNAs
7. Novel Therapeutic Strategies
8. Conclusions and Perspectives
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Lefebvre, V.; Angelozzi, M.; Haseeb, A. SOX9 in cartilage development and disease. Curr. Opin. Cell Biol. 2019, 61, 39–47. [Google Scholar] [CrossRef] [PubMed]
- Dong, X.; Zhao, B.; Iacob, R.E.; Zhu, J.; Koksal, A.C.; Lu, C.; Engen, J.R.; Springer, T.A. Force interacts with macromolecular structure in activation of TGF-β. Nature 2017, 542, 55–59. [Google Scholar] [CrossRef] [PubMed]
- Hügle, T.; Geurts, J.; Nüesch, C.; Müller-Gerbl, M.; Valderrabano, V. Aging and osteoarthritis: An inevitable encounter? J. Aging Res. 2012, 2012, 950192. [Google Scholar] [CrossRef] [PubMed]
- D’Adamo, S.; Cetrullo, S.; Minguzzi, M.; Silvestri, Y.; Borzì, R.M.; Flamigni, F. MicroRNAs and autophagy: Fine players in the control of chondrocyte homeostatic activities in osteoarthritis. Oxidative Med. Cell. Longev. 2017, 2017, 3720128. [Google Scholar] [CrossRef] [Green Version]
- Valenti, M.T.; Carbonare, L.D.; Mottes, M. Role of microRNAs in progenitor cell commitment and osteogenic differentiation in health and disease. Int. J. Mol. Med. 2018, 41, 2441–2449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cheng, N.-T.; Guo, A.; Meng, H. The protective role of autophagy in experimental osteoarthritis, and the therapeutic effects of Torin 1 on osteoarthritis by activating autophagy. BMC Musculoskelet. Disord. 2016, 17, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- O’Brien, M.; Philpott, H.T.; McDougall, J.J. Understanding osteoarthritis pain through animal models. Clin. Exp. Rheumatol. 2017, 35 (Suppl. 5), 47–52. [Google Scholar]
- Kang, X.; Yang, W.; Feng, D.; Jin, X.; Ma, Z.; Qian, Z.; Xie, T.; Li, H.; Liu, J.; Wang, R. Cartilage-specific autophagy deficiency promotes ER stress and impairs chondrogenesis in PERK-ATF4-CHOP–dependent manner. J. Bone Miner. Res. 2017, 32, 2128–2141. [Google Scholar] [CrossRef]
- Chang, N.C. Autophagy and stem cells: Self-eating for self-renewal. Front. Cell Dev. Biol. 2020, 8, 138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Horigome, Y.; Ida-Yonemochi, H.; Waguri, S.; Shibata, S.; Endo, N.; Komatsu, M. Loss of autophagy in chondrocytes causes severe growth retardation. Autophagy 2020, 16, 501–511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Luo, P.; Gao, F.; Niu, D.; Sun, X.; Song, Q.; Guo, C.; Liang, Y.; Sun, W. The role of autophagy in chondrocyte metabolism and osteoarthritis: A comprehensive research review. BioMed Res. Int. 2019, 2019, 5171602. [Google Scholar] [CrossRef] [PubMed]
- Kronenberg, H.M. Developmental regulation of the growth plate. Nature 2003, 423, 332–336. [Google Scholar] [CrossRef] [PubMed]
- Settembre, C.; Arteaga-Solis, E.; McKee, M.D.; de Pablo, R.; Al Awqati, Q.; Ballabio, A.; Karsenty, G. Proteoglycan desulfation determines the efficiency of chondrocyte autophagy and the extent of FGF signaling during endochondral ossification. Genes Dev. 2008, 22, 2645–2650. [Google Scholar] [CrossRef] [Green Version]
- Srinivas, V.; Bohensky, J.; Shapiro, I.M. Autophagy: A new phase in the maturation of growth plate chondrocytes is regulated by HIF, mTOR and AMP kinase. Cells Tissues Organs 2009, 189, 88–92. [Google Scholar] [CrossRef] [PubMed]
- Kim, J.; Kundu, M.; Viollet, B.; Guan, K.-L. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat. Cell Biol. 2011, 13, 132–141. [Google Scholar] [CrossRef] [Green Version]
- Houck, S.A.; Ren, H.Y.; Madden, V.J.; Bonner, J.N.; Conlin, M.P.; Janovick, J.A.; Conn, P.M.; Cyr, D.M. Quality control autophagy degrades soluble ERAD-resistant conformers of the misfolded membrane protein GnRHR. Mol. Cell 2014, 54, 166–179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cecconi, F.; Levine, B. The role of autophagy in mammalian development: Cell makeover rather than cell death. Dev. Cell 2008, 15, 344–357. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ma, K.-G.; Shao, Z.-W.; Yang, S.-H.; Wang, J.; Wang, B.-C.; Xiong, L.-M.; Wu, Q.; Chen, S.-F. Autophagy is activated in compression-induced cell degeneration and is mediated by reactive oxygen species in nucleus pulposus cells exposed to compression. Osteoarthr. Cartil. 2013, 21, 2030–2038. [Google Scholar] [CrossRef] [Green Version]
- Fermor, B.; Gurumurthy, A.; Diekman, B.O. Hypoxia, RONS and energy metabolism in articular cartilage. Osteoarthr. Cartil. 2010, 18, 1167–1173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dalle Carbonare, L.; Mottes, M.; Cheri, S.; Deiana, M.; Zamboni, F.; Gabbiani, D.; Schena, F.; Salvagno, G.; Lippi, G. Valenti M Increased gene expression of RUNX2 and SOX9 in mesenchymal circulating progenitors is associated with autophagy during physical activity. Oxidative Med. Cell. Longev. 2019, 2019, 8426259. [Google Scholar] [CrossRef]
- Wallace, D.C. A mitochondrial bioenergetic etiology of disease. J. Clin. Investig. 2013, 123, 1405–1412. [Google Scholar] [CrossRef] [PubMed]
- Giorgi, C.; Marchi, S.; Simoes, I.C.; Ren, Z.; Morciano, G.; Perrone, M.; Patalas-Krawczyk, P.; Borchard, S.; Jędrak, P. Pierzynowska K Mitochondria and reactive oxygen species in aging and age-related diseases. Int. Rev. Cell Mol. Biol. 2018, 340, 209–344. [Google Scholar]
- Palikaras, K.; Lionaki, E.; Tavernarakis, N. Mechanisms of mitophagy in cellular homeostasis, physiology and pathology. Nat. Cell Biol. 2018, 20, 1013–1022. [Google Scholar] [CrossRef]
- Sun, K.; Jing, X.; Guo, J.; Yao, X.; Guo, F. Mitophagy in degenerative joint diseases. Autophagy 2020, 1–11. [Google Scholar] [CrossRef] [PubMed]
- Forni, M.F.; Peloggia, J.; Trudeau, K.; Shirihai, O.; Kowaltowski, A.J. Murine mesenchymal stem cell commitment to differentiation is regulated by mitochondrial dynamics. Stem Cells 2016, 34, 743–755. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Marycz, K.; Kornicka, K.; Grzesiak, J.; Śmieszek, A.; Szłapka, J. Macroautophagy and selective mitophagy ameliorate chondrogenic differentiation potential in adipose stem cells of equine metabolic syndrome: New findings in the field of progenitor cells differentiation. Oxidative Med. Cell. Longev. 2016, 2016, 3718468. [Google Scholar] [CrossRef]
- Caramés, B.; Taniguchi, N.; Otsuki, S.; Blanco, F.J.; Lotz, M. Autophagy is a protective mechanism in normal cartilage, and its aging-related loss is linked with cell death and osteoarthritis. Arthritis Rheum. 2010, 62, 791–801. [Google Scholar] [CrossRef] [Green Version]
- Yang, F.; Huang, R.; Ma, H.; Zhao, X.; Wang, G. miRNA-411 Regulates chondrocyte autophagy in osteoarthritis by targeting hypoxia-inducible factor 1 alpha (HIF-1α). Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2020, 26, e921155-1. [Google Scholar] [CrossRef]
- Li, H.; Li, Z.; Pi, Y.; Chen, Y.; Mei, L.; Luo, Y.; Xie, J.; Mao, X. MicroRNA-375 exacerbates knee osteoarthritis through repressing chondrocyte autophagy by targeting ATG2B. Aging 2020, 12, 7248. [Google Scholar] [CrossRef] [PubMed]
- Feng, L.; Feng, C.; Wang, C.X.; Xu, D.Y.; Chen, J.J.; Huang, J.F.; Tan, P.L.; Shen, J.M. Circulating microRNA let‑7e is decreased in knee osteoarthritis, accompanied by elevated apoptosis and reduced autophagy. Int. J. Mol. Med. 2020, 45, 1464–1476. [Google Scholar] [CrossRef]
- Yu, Y.; Zhao, J. Modulated autophagy by microRNAs in osteoarthritis chondrocytes. BioMed Res. Int. 2019, 2019, 1484152. [Google Scholar] [CrossRef] [PubMed]
- Lambert, S.A.; Jolma, A.; Campitelli, L.F.; Das, P.K.; Yin, Y.; Albu, M.; Chen, X.; Taipale, J.; Hughes, T.R.; Weirauch, M.T. The human transcription factors. Cell 2018, 172, 650–665. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Accili, D.; Arden, K.C. FoxOs at the crossroads of cellular metabolism, differentiation, and transformation. Cell 2004, 117, 421–426. [Google Scholar] [CrossRef] [Green Version]
- Roeder, R.G. The role of general initiation factors in transcription by RNA polymerase II. Trends Biochem. Sci. 1996, 21, 327–335. [Google Scholar] [CrossRef]
- Maston, G.A.; Evans, S.K.; Green, M.R. Transcriptional regulatory elements in the human genome. Annu. Rev. Genom. Hum. Genet. 2006, 7, 29–59. [Google Scholar] [CrossRef] [Green Version]
- Mullen, A.C.; Orlando, D.A.; Newman, J.J.; Lovén, J.; Kumar, R.M.; Bilodeau, S.; Reddy, J.; Guenther, M.G.; DeKoter, R.P.; Young, R.A. Master transcription factors determine cell-type-specific responses to TGF-β signaling. Cell 2011, 147, 565–576. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, L.-F.; Williams, S.A.; Mu, Y.; Nakano, H.; Duerr, J.M.; Buckbinder, L.; Greene, W.C. NF-κB RelA phosphorylation regulates RelA acetylation. Mol. Cell. Biol. 2005, 25, 7966–7975. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lefebvre, V.; Dvir-Ginzberg, M. SOX9 and the many facets of its regulation in the chondrocyte lineage. Connect. Tissue Res. 2017, 58, 2–14. [Google Scholar] [CrossRef]
- Csukasi, F.; Duran, I.; Zhang, W.; Martin, J.H.; Barad, M.; Bamshad, M.; Weis, M.A.; Eyre, D.; Krakow, D.; Cohn, D.H. Dominant-negative SOX9 mutations in campomelic dysplasia. Hum. Mutat. 2019, 40, 2344–2352. [Google Scholar] [CrossRef] [PubMed]
- Zhong, L.; Huang, X.; Karperien, M.; Post, J.N. Correlation between gene expression and osteoarthritis progression in human. Int. J. Mol. Sci. 2016, 17, 1126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nakamura, Y.; He, X.; Kato, H.; Wakitani, S.; Kobayashi, T.; Watanabe, S.; Iida, A.; Tahara, H.; Warman, M.L.; Watanapokasin, R. Sox9 is upstream of microRNA-140 in cartilage. Appl. Biochem. Biotechnol. 2012, 166, 64–71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Martinez-Sanchez, A.; Murphy, C.L. miR-1247 functions by targeting cartilage transcription factor SOX9. J. Biol. Chem. 2013, 288, 30802–30814. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, H.; Wang, Y.; Yang, G.; Yu, H.; Zhou, Z.; Tang, M. MicroRNA‑30a regulates chondrogenic differentiation of human bone marrow‑derived mesenchymal stem cells through targeting Sox9. Exp. Ther. Med. 2019, 18, 4689–4697. [Google Scholar] [CrossRef]
- Yu, X.-M.; Meng, H.-Y.; Yuan, X.-L.; Wang, Y.; Guo, Q.-Y.; Peng, J.; Wang, A.-Y.; Lu, S.-B. MicroRNAs’ involvement in osteoarthritis and the prospects for treatments. Evid. Based Complementary Altern. Med. 2015, 2015, 236179. [Google Scholar] [CrossRef] [Green Version]
- Neefjes, M.; van Caam, A.P.; van der Kraan, P.M. Transcription Factors in Cartilage Homeostasis and Osteoarthritis. Biology 2020, 9, 290. [Google Scholar] [CrossRef] [PubMed]
- Fisch, K.M.; Gamini, R.; Alvarez-Garcia, O.; Akagi, R.; Saito, M.; Muramatsu, Y.; Sasho, T.; Koziol, J.A.; Su, A.I.; Lotz, M.K. Identification of transcription factors responsible for dysregulated networks in human osteoarthritis cartilage by global gene expression analysis. Osteoarthr. Cartil. 2018, 26, 1531–1538. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kobayashi, H.; Hirata, M.; Saito, T.; Itoh, S.; Chung, U.-I.; Kawaguchi, H. Transcriptional induction of ADAMTS5 protein by nuclear factor-κB (NF-κB) family member RelA/p65 in chondrocytes during osteoarthritis development. J. Biol. Chem. 2013, 288, 28620–28629. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Takahata, Y.; Nakamura, E.; Hata, K.; Wakabayashi, M.; Murakami, T.; Wakamori, K.; Yoshikawa, H.; Matsuda, A.; Fukui, N.; Nishimura, R. Sox4 is involved in osteoarthritic cartilage deterioration through induction of ADAMTS4 and ADAMTS5. FASEB J. 2019, 33, 619–630. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Z.; Kang, Y.; Zhang, H.; Duan, X.; Liu, J.; Li, X..; Liao, W. Expression of microRNAs during chondrogenesis of human adipose-derived stem cells. Osteoarthr. Cartil. 2012, 20, 1638–1646. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hu, G.; Codina, M.; Fisher, S. Multiple enhancers associated with ACAN suggest highly redundant transcriptional regulation in cartilage. Matrix Biol. 2012, 31, 328–337. [Google Scholar] [CrossRef] [Green Version]
- Settembre, C.; Di Malta, C.; Polito, V.A.; Arencibia, M.G.; Vetrini, F.; Erdin, S.; Erdin, S.U.; Huynh, T.; Medina, D.; Colella, P. TFEB links autophagy to lysosomal biogenesis. Science 2011, 332, 1429–1433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zheng, G.; Zhan, Y.; Li, X.; Pan, Z.; Zheng, F.; Zhang, Z.; Zhou, Y.; Wu, Y.; Wang, X.; Gao, W. TFEB, a potential therapeutic target for osteoarthritis via autophagy regulation. Cell Death Dis. 2018, 9, 1–15. [Google Scholar] [CrossRef] [Green Version]
- Chauhan, S.; Goodwin, J.G.; Chauhan, S.; Manyam, G.; Wang, J.; Kamat, A.M.; Boyd, D.D. ZKSCAN3 is a master transcriptional repressor of autophagy. Mol. Cell 2013, 50, 16–28. [Google Scholar] [CrossRef] [Green Version]
- Hu, H.; Ji, Q.; Song, M.; Ren, J.; Liu, Z.; Wang, Z.; Liu, X.; Yan, K.; Hu, J.; Jing, Y. ZKSCAN3 counteracts cellular senescence by stabilizing heterochromatin. Nucleic Acids Res. 2020, 48, 6001–6018. [Google Scholar] [CrossRef] [PubMed]
- Brunet, A.; Sweeney, L.B.; Sturgill, J.F.; Chua, K.F.; Greer, P.L.; Lin, Y.; Tran, H.; Ross, S.E.; Mostoslavsky, R.; Cohen, H.Y. Stress-dependent regulation of FOXO transcription factors by the SIRT1 deacetylase. Science 2004, 303, 2011–2015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pietrocola, F.; Izzo, V.; Niso-Santano, M.; Vacchelli, E.; Galluzzi, L.; Maiuri, M.C.; Kroemer, G. Regulation of autophagy by stress-responsive transcription factors. In Seminars in Cancer Biology; Elsevier: Amsterdam, The Netherlands, 2013. [Google Scholar]
- Akasaki, Y.; Alvarez-Garcia, O.; Saito, M.; Caramés, B.; Iwamoto, Y.; Lotz, M.K. FoxO transcription factors support oxidative stress resistance in human chondrocytes. Arthritis Rheumatol. 2014, 66, 3349–3358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Matsuzaki, T.; Alvarez-Garcia, O.; Mokuda, S.; Nagira, K.; Olmer, M.; Gamini, R.; Miyata, K.; Akasaki, Y.; Su, A.I.; Asahara, H.; et al. FoxO transcription factors modulate autophagy and proteoglycan 4 in cartilage homeostasis and osteoarthritis. Sci. Transl. Med. 2018, 10, eaan0746. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xie, J.; Lin, J.; Wei, M.; Teng, Y.; He, Q.; Yang, G.; Yang, X. Sustained Akt signaling in articular chondrocytes causes osteoarthritis via oxidative stress-induced senescence in mice. Bone Res. 2019, 7, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, K.M.; Park, S.J.; Jung, S.H.; Kim, E.J.; Jogeswar, G.; Ajita, J.; Rhee, Y.; Kim, C.H.; Lim, S.K. miR-182 is a negative regulator of osteoblast proliferation, differentiation, and skeletogenesis through targeting FoxO1. J. Bone Miner. Res. 2012, 27, 1669–1679. [Google Scholar] [CrossRef]
- Huang, P.-Y.; Wu, J.-G.; Gu, J.; Zhang, T.-Q.; Li, L.-F.; Wang, S.-Q.; Wang, M. Bioinformatics analysis of miRNA and mRNA expression profiles to reveal the key miRNAs and genes in osteoarthritis. J. Orthop. Surg. Res. 2021, 16, 1–9. [Google Scholar] [CrossRef]
- Duan, R.; Xie, H.; Liu, Z.-Z. The Role of Autophagy in Osteoarthritis. Front. Cell Dev. Biol. 2020, 8, 1437. [Google Scholar] [CrossRef] [PubMed]
- McCorry, M.C.; Puetzer, J.L.; Bonassar, L.J. Characterization of mesenchymal stem cells and fibrochondrocytes in three-dimensional co-culture: Analysis of cell shape, matrix production, and mechanical performance. Stem Cell Res. Ther. 2016, 7, 1–10. [Google Scholar] [CrossRef] [Green Version]
- Samvelyan, H.J.; Hughes, D.; Stevens, C.; Staines, K.A. Models of osteoarthritis: Relevance and new insights. Calcif. Tissue Int. 2020, 1–14. [Google Scholar] [CrossRef] [Green Version]
- Guilak, F.; Alexopoulos, L.G.; Upton, M.L.; Youn, I.; Choi, J.B.; Cao, L.; Setton, L.A.; Haider, M.A. The pericellular matrix as a transducer of biomechanical and biochemical signals in articular cartilage. Ann. N. Y. Acad. Sci. 2006, 1068, 498–512. [Google Scholar] [CrossRef]
- Cope, P.; Ourradi, K.; Li, Y.; Sharif, M. Models of osteoarthritis: The good, the bad and the promising. Osteoarthr. Cartil. 2019, 27, 230–239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- van der Kraan, P. Relevance of zebrafish as an OA research model. Osteoarthr. Cartil. 2013, 21, 261–262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brunt, L.H.; Roddy, K.A.; Rayfield, E.J.; Hammond, C.L. Building finite element models to investigate zebrafish jaw biomechanics. J. Vis. Exp. 2016, 2016, e54811. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lawrence, E.A.; Aggleton, J.A.; van Loon, J.J.; Godivier, J.; Harniman, R.L.; Pei, J.; Nowlan, N.; Hammond, C.L. Exposure to hypergravity during zebrafish development alters cartilage material properties and strain distribution. Bone Jt. Res. 2021, 10, 137–148. [Google Scholar] [CrossRef] [PubMed]
- Weiner, A.M.; Scampoli, N.L.; Steeman, T.J.; Dooley, C.M.; Busch-Nentwich, E.M.; Kelsh, R.N.; Calcaterra, N.B. Dicer1 is required for pigment cell and craniofacial development in zebrafish. Biochim. Biophys. Acta (BBA)-Gene Regul. Mech. 2019, 1862, 472–485. [Google Scholar] [CrossRef] [PubMed]
- Sorial, A.K.; Hofer, I.M.; Tselepi, M.; Cheung, K.; Parker, E.; Deehan, D.J.; Rice, S.J.; Loughlin, J. Multi-tissue epigenetic analysis of the osteoarthritis susceptibility locus mapping to the plectin gene PLEC. Osteoarthr. Cartil. 2020, 28, 1448–1458. [Google Scholar] [CrossRef]
- Almarza, D.; Cucchiarini, M.; Loughlin, J. Genome editing for human osteoarthritis–a perspective. Osteoarthr. Cartil. 2017, 25, 1195–1198. [Google Scholar] [CrossRef] [PubMed]
- Moss, J.J.; Hammond, C.L.; Lane, J.D. Zebrafish as a model to study autophagy and its role in skeletal development and disease. Histochem. Cell Biol. 2020, 1–16. [Google Scholar] [CrossRef] [PubMed]
- Butterfield, N.C.; Curry, K.F.; Steinberg, J.; Dewhurst, H.; Komla-Ebri, D.; Mannan, N.S.; Adoum, A.-T.; Leitch, V.D.; Logan, J.G.; Waung, J.A. Accelerating functional gene discovery in osteoarthritis. Nat. Commun. 2021, 12, 1–18. [Google Scholar] [CrossRef]
- Brunt, L.; Kague, E.; Hammond, C. Developmental Insights into Osteoarthritis Increase the Applicability of New Animal Models. J. Musculoskelet. Disord. Treat. 2016, 2, 17. [Google Scholar]
- Le, L.T.; Swingler, T.E.; Crowe, N.; Vincent, T.L.; Barter, M.J.; Donell, S.T.; Delany, A.M.; Dalmay, T.; Young, D.A.; Clark, I.M. The microRNA-29 family in cartilage homeostasis and osteoarthritis. J. Mol. Med. 2016, 94, 583–596. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cheng, Y.; Ren, X.; Hait, W.N.; Yang, J.-M. Therapeutic targeting of autophagy in disease: Biology and pharmacology. Pharmacol. Rev. 2013, 65, 1162–1197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cooper, C.; Chapurlat, R.; Al-Daghri, N.; Herrero-Beaumont, G.; Bruyère, O.; Rannou, F.; Roth, R.; Uebelhart, D.; Reginster, J.-Y. Safety of oral non-selective non-steroidal anti-inflammatory drugs in osteoarthritis: What does the literature say? Drugs Aging 2019, 36, 15–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Takayama, K.; Kawakami, Y.; Kobayashi, M.; Greco, N.; Cummins, J.H.; Matsushita, T.; Kuroda, R.; Kurosaka, M.; Fu, F.H.; Huard, J. Local intra-articular injection of rapamycin delays articular cartilage degeneration in a murine model of osteoarthritis. Arthritis Res. Ther. 2014, 16, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ouyang, J.; Jiang, H.; Fang, H.; Cui, W.; Cai, D. Isoimperatorin ameliorates osteoarthritis by downregulating the mammalian target of rapamycin C1 signaling pathway. Mol. Med. Rep. 2017, 16, 9636–9644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sun, M.M.-G.; Beier, F.; Pest, M.A. Recent developments in emerging therapeutic targets of osteoarthritis. Curr. Opin. Rheumatol. 2017, 29, 96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shi, Y.; Hu, X.; Cheng, J.; Zhang, X.; Zhao, F.; Shi, W.; Ren, B.; Yu, H.; Yang, P.; Li, Z. A small molecule promotes cartilage extracellular matrix generation and inhibits osteoarthritis development. Nat. Commun. 2019, 10, 1–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, T.; Zhang, L.; Joo, D.; Sun, S.-C. NF-κB signaling in inflammation. Signal Transduct. Target. Ther. 2017, 2, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yi, H.; Zhang, W.; Cui, Z.-M.; Cui, S.-Y.; Fan, J.-B.; Zhu, X.-H.; Liu, W. Resveratrol alleviates the interleukin-1β-induced chondrocytes injury through the NF-κB signaling pathway. J. Orthop. Surg. Res. 2020, 15, 1–9. [Google Scholar] [CrossRef]
- Ruan, G.; Xu, J.; Wang, K.; Wu, J.; Zhu, Q.; Ren, J.; Bian, F.; Chang, B.; Bai, X.; Han, W. Associations between knee structural measures, circulating inflammatory factors and MMP13 in patients with knee osteoarthritis. Osteoarthr. Cartil. 2018, 26, 1063–1069. [Google Scholar] [CrossRef] [Green Version]
- Mehana, E.-S.E.; Khafaga, A.F.; El-Blehi, S.S. The role of matrix metalloproteinases in osteoarthritis pathogenesis: An updated review. Life Sci. 2019, 234, 116786. [Google Scholar] [CrossRef] [PubMed]
- Yang, C.-Y.; Chanalaris, A.; Troeberg, L. ADAMTS and ADAM metalloproteinases in osteoarthritis–looking beyond the ‘usual suspects’. Osteoarthr. Cartil. 2017, 25, 1000–1009. [Google Scholar] [CrossRef] [Green Version]
- Wang, M.; Sampson, E.R.; Jin, H.; Li, J.; Ke, Q.H.; Im, H.-J.; Chen, D. MMP13 is a critical target gene during the progression of osteoarthritis. Arthritis Res. Ther. 2013, 15, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhao, L.; Huang, J.; Fan, Y.; Li, J.; You, T.; He, S.; Xiao, G.; Chen, D. Exploration of CRISPR/Cas9-based gene editing as therapy for osteoarthritis. Ann. Rheum. Dis. 2019, 78, 676–682. [Google Scholar] [CrossRef] [PubMed]
- Fields, G.B. The rebirth of matrix metalloproteinase inhibitors: Moving beyond the dogma. Cells 2019, 8, 984. [Google Scholar] [CrossRef] [Green Version]
- Santamaria, S. ADAMTS-5: A difficult teenager turning 20. Int. J. Exp. Pathol. 2020, 101, 4–20. [Google Scholar] [CrossRef] [PubMed]
- Kang, C.; Elledge, S.J. How autophagy both activates and inhibits cellular senescence. Autophagy 2016, 12, 898–899. [Google Scholar] [CrossRef]
- Di Micco, R.; Krizhanovsky, V.; Baker, D.; d’Adda di Fagagna, F. Cellular senescence in ageing: From mechanisms to therapeutic opportunities. Nat. Rev. Mol. Cell Biol. 2020, 22, 75–95. [Google Scholar] [CrossRef]
- Zhu, Y.; Doornebal, E.J.; Pirtskhalava, T.; Giorgadze, N.; Wentworth, M.; Fuhrmann-Stroissnigg, H.; Niedernhofer, L.J.; Robbins, P.D.; Tchkonia, T.; Kirkland, J.L. New agents that target senescent cells: The flavone, fisetin, and the BCL-XL inhibitors, A1331852 and A1155463. Aging 2017, 9, 955. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kirkland, J.L.; Tchkonia, T.; Zhu, Y.; Niedernhofer, L.J.; Robbins, P.D. The clinical potential of senolytic drugs. J. Am. Geriatr. Soc. 2017, 65, 2297–2301. [Google Scholar] [CrossRef] [PubMed]
- Lewinska, A.; Adamczyk-Grochala, J.; Bloniarz, D.; Olszowka, J.; Kulpa-Greszta, M.; Litwinienko, G.; Tomaszewska, A.; Wnuk, M.; Pazik, R. AMPK-mediated senolytic and senostatic activity of quercetin surface functionalized Fe3O4 nanoparticles during oxidant-induced senescence in human fibroblasts. Redox Biol. 2020, 28, 101337. [Google Scholar] [CrossRef] [PubMed]
- Deng, Z.; Li, Y.; Liu, H.; Xiao, S.; Li, L.; Tian, J.; Cheng, C.; Zhang, G.; Zhang, F. The role of sirtuin 1 and its activator, resveratrol in osteoarthritis. Biosci. Rep. 2019, 39, BSR20190189. [Google Scholar] [CrossRef] [Green Version]
- Wang, X.-B.; Zhao, F.-C.; Yi, L.-H.; Tang, J.-L.; Zhu, Z.-Y.; Pang, Y.; Chen, Y.-S.; Li, D.-Y.; Guo, K.-J.; Zheng, X. MicroRNA-21-5p as a novel therapeutic target for osteoarthritis. Rheumatology 2019, 58, 1485–1497. [Google Scholar] [CrossRef]
- Geng, Y.; Chen, J.; Alahdal, M.; Chang, C.; Duan, L.; Zhu, W.; Mou, L.; Xiong, J.; Wang, M.; Wang, D. Intra-articular injection of hUC-MSCs expressing miR-140-5p induces cartilage self-repairing in the rat osteoarthritis. J. Bone Miner. Metab. 2020, 38, 277–288. [Google Scholar] [CrossRef] [PubMed]
- Liu, X.; Shortt, C.; Zhang, F.; Bater, M.Q.; Cowman, M.K.; Kirsch, T. Extracellular Vesicles Released From Articular Chondrocytes Play a Major Role in Cell–Cell Communication. J. Orthop. Res.® 2020, 38, 731–739. [Google Scholar] [CrossRef] [PubMed]
- Tao, S.-C.; Yuan, T.; Zhang, Y.-L.; Yin, W.-J.; Guo, S.-C.; Zhang, C.-Q. Exosomes derived from miR-140-5p-overexpressing human synovial mesenchymal stem cells enhance cartilage tissue regeneration and prevent osteoarthritis of the knee in a rat model. Theranostics 2017, 7, 180. [Google Scholar] [CrossRef] [PubMed]
- Mao, G.; Zhang, Z.; Hu, S.; Zhang, Z.; Chang, Z.; Huang, Z.; Liao, W.; Kang, Y. Exosomes derived from miR-92a-3p-overexpressing human mesenchymal stem cells enhance chondrogenesis and suppress cartilage degradation via targeting WNT5A. Stem Cell Res. Ther. 2018, 9, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Qiu, B.; Xu, X.; Yi, P.; Hao, Y. Curcumin reinforces MSC-derived exosomes in attenuating osteoarthritis via modulating the miR-124/NF-kB and miR-143/ROCK1/TLR9 signalling pathways. J. Cell. Mol. Med. 2020, 24, 10855–10865. [Google Scholar] [CrossRef]
- Liu, X.; Yang, Y.; Li, Y.; Niu, X.; Zhao, B.; Wang, Y.; Bao, C.; Xie, Z.; Lin, Q.; Zhu, L. Integration of stem cell-derived exosomes with in situ hydrogel glue as a promising tissue patch for articular cartilage regeneration. Nanoscale 2017, 9, 4430–4438. [Google Scholar] [CrossRef]
- Chen, P.; Zheng, L.; Wang, Y.; Tao, M.; Xie, Z.; Xia, C.; Gu, C.; Chen, J.; Qiu, P.; Mei, S. Desktop-stereolithography 3D printing of a radially oriented extracellular matrix/mesenchymal stem cell exosome bioink for osteochondral defect regeneration. Theranostics 2019, 9, 2439. [Google Scholar] [CrossRef]
- Tanikella, A.S.; Hardy, M.J.; Frahs, S.M.; Cormier, A.G.; Gibbons, K.D.; Fitzpatrick, C.K.; Oxford, J.T. Emerging gene-editing modalities for osteoarthritis. Int. J. Mol. Sci. 2020, 21, 6046. [Google Scholar] [CrossRef]
- Seidl, C.; Fulga, T.; Murphy, C. CRISPR-Cas9 targeting of MMP13 in human chondrocytes leads to significantly reduced levels of the metalloproteinase and enhanced type II collagen accumulation. Osteoarthr. Cartil. 2019, 27, 140–147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Karlsen, T.A.; Pernas, P.F.; Staerk, J.; Caglayan, S.; Brinchmann, J. Generation of IL1β-resistant chondrocytes using CRISPR-CAS genome editing. Osteoarthr. Cartil. 2016, 24, S325. [Google Scholar] [CrossRef]
- Lambert, L.J.; Challa, A.K.; Niu, A.; Zhou, L.; Tucholski, J.; Johnson, M.S.; Nagy, T.R.; Eberhardt, A.W.; Estep, P.N.; Kesterson, R.A. Increased trabecular bone and improved biomechanics in an osteocalcin-null rat model created by CRISPR/Cas9 technology. Dis. Models Mech. 2016, 9, 1169–1179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huang, Y.; Askew, E.B.; Knudson, C.B.; Knudson, W. CRISPR/Cas9 knockout of HAS2 in rat chondrosarcoma chondrocytes demonstrates the requirement of hyaluronan for aggrecan retention. Matrix Biol. 2016, 56, 74–94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Adkar, S.S.; Brunger, J.M.; Willard, V.P.; Wu, C.-L.; Gersbach, C.A.; Guilak, F. Genome engineering for personalized arthritis therapeutics. Trends in molecular medicine 2017, 23, 917–931. [Google Scholar] [CrossRef] [PubMed]
- Uzieliene, I.; Kalvaityte, U.; Bernotiene, E.; Mobasheri, A. Non-viral Gene Therapy for Osteoarthritis. Front. Bioeng. Biotechnol. 2020, 8, 8. [Google Scholar] [CrossRef]
- Mancuso, P.; Raman, S.; Glynn, A.; Barry, F.; Murphy, J.M. Mesenchymal stem cell therapy for osteoarthritis: The critical role of the cell secretome. Front. Bioeng. Biotechnol. 2019, 7, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Senolytic Drug | Targeted Molecules/Pathway |
---|---|
FISETIN | ⊥ P13/AKT/mTOR ⊥ Bcl2-xL |
QUERCETIN | ⊥ P13/AKT/mTOR ⊥ Bcl2-w |
NAVITOCLAX | ⊥ Bcl2, Bcl-xL, Bcl-w |
FOXO4-DRI | ⊗ FOXO4-p53 interaction, no p53 in the nucleus |
USP7 inhibitor | MDM2 ubiquitination |
UBX0101 | MDM2, p32 |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 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 (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Valenti, M.T.; Dalle Carbonare, L.; Zipeto, D.; Mottes, M. Control of the Autophagy Pathway in Osteoarthritis: Key Regulators, Therapeutic Targets and Therapeutic Strategies. Int. J. Mol. Sci. 2021, 22, 2700. https://doi.org/10.3390/ijms22052700
Valenti MT, Dalle Carbonare L, Zipeto D, Mottes M. Control of the Autophagy Pathway in Osteoarthritis: Key Regulators, Therapeutic Targets and Therapeutic Strategies. International Journal of Molecular Sciences. 2021; 22(5):2700. https://doi.org/10.3390/ijms22052700
Chicago/Turabian StyleValenti, Maria Teresa, Luca Dalle Carbonare, Donato Zipeto, and Monica Mottes. 2021. "Control of the Autophagy Pathway in Osteoarthritis: Key Regulators, Therapeutic Targets and Therapeutic Strategies" International Journal of Molecular Sciences 22, no. 5: 2700. https://doi.org/10.3390/ijms22052700
APA StyleValenti, M. T., Dalle Carbonare, L., Zipeto, D., & Mottes, M. (2021). Control of the Autophagy Pathway in Osteoarthritis: Key Regulators, Therapeutic Targets and Therapeutic Strategies. International Journal of Molecular Sciences, 22(5), 2700. https://doi.org/10.3390/ijms22052700