Icariin alleviates osteoarthritis by inhibiting NLRP3-mediated pyroptosis

Yan Zu, Yue Mu, Qiang Li, Shu-Ting Zhang, Hong-Juan Yan, Yan Zu, Yue Mu, Qiang Li, Shu-Ting Zhang, Hong-Juan Yan

Abstract

Background: Osteoarthritis (OA) is the common chronic degenerative joint bone disease that is mainly featured by joint stiffness and cartilage degradation. Icariin (ICA), an extract from Epimedium, has been preliminarily proven to show anti-osteoporotic and anti-inflammatory effects in OA. However, the underlying mechanisms of ICA on chondrocytes need to be elucidated.

Methods: LPS-treated chondrocytes and monosodium iodoacetate (MIA)-treated Wistar rats were used as models of OA in vitro and in vivo, respectively. LDH and MTT assays were performed to detect cytotoxicity and cell viability. The expression levels of NLRP3, IL-1β, IL-18, MMP-1, MMP-13, and collagen II were detected by qRT-PCR and Western blotting. The release levels of IL-1β and IL-18 were detected by ELISA assay. Caspase-1 activity was assessed by flow cytometry. Immunofluorescence and immunohistochemistry were used to examine the level of NLRP3 in chondrocytes and rat cartilage, respectively. The progression of OA was monitored with hematoxylin-eosin (H&E) staining and safranin O/fast green staining.

Results: ICA could suppress LPS-induced inflammation and reduction of collagen formation in chondrocytes. Furthermore, ICA could inhibit NLRP3 inflammasome-mediated caspase-1 signaling pathway to alleviate pyroptosis induced by LPS. Overexpression of NLRP3 reversed the above changes caused by ICA. It was further confirmed in the rat OA model that ICA alleviated OA by inhibiting NLRP3-mediated pyroptosis.

Conclusion: ICA inhibited OA via repressing NLRP3/caspase-1 signaling-mediated pyroptosis in models of OA in vitro and in vivo, suggesting that ICA might be a promising compound in the treatment of OA.

Keywords: Caspase-1 signaling; Icariin; NLRP3 inflammasome; Osteoarthritis; Pyroptosis.

Conflict of interest statement

The authors declare that they have no conflict of interest.

Figures

Fig. 1
Fig. 1
Effects of LPS on chondrocytes inflammation and collagen formation. a The mRNA levels of NRLP3, IL-1β, and IL-18 were analyzed by qRT-PCR in rat chondrocytes treated with LPS. b The release levels of IL-1β and IL-18 in the cell supernatant of LPS treated rat chondrocytes was checked by ELISA assay. c The expression level of collagen formation and pyroptosis-related protein was analyzed by Western blotting in rat chondrocytes treated with LPS. d Quantitative analysis of protein band gray in c. GAPDH was used as internal control. All the results were shown as mean ± SD (n = 3), which were three separate experiments performed in triplicate. *p < 0.05 and **p < 0.01
Fig. 2
Fig. 2
Effects of ICA on chondrocytes injury and pyroptosis. a The cell activity was detected by MTT assay in chondrocytes treated with various concentrations of ICA for 24 h. b Leakage of LDH was assessed by ELISA assay in chondrocytes treated with LPS or ICA. The mRNA levels of IL-1β (c) and IL-18 (d) were analyzed by qRT-PCR in chondrocytes treated with LPS or ICA. The release level of IL-1β (e) and IL-18 (f) were analyzed by ELISA assay in chondrocytes treated with LPS or ICA. g The protein level of collagen formation and pyroptosis-related protein was analyzed by Western blotting in rat chondrocytes treated with LPS or ICA. h Quantitative analysis of protein band gray in g. GAPDH was used as internal control. All the results were shown as mean ± SD (n = 3), which were three separate experiments performed in triplicate. *p < 0.05 and **p < 0.01
Fig. 3
Fig. 3
ICA exerted negative effects on LPS-induced NLRP3 expression and pyroptosis. a Immunofluorescent was used to detect NLRP3 in chondrocytes treated with LPS or ICA. b Flow cytometry analysis was used to measure caspase-1 activity in chondrocytes treated with LPS or ICA. c Quantitative statistics of doublepositive for caspase-1 and PI stain. d The protein level of pyroptosis-related protein was analyzed by Western blotting in chondrocytes treated with LPS or ICA. e Quantitative analysis of protein band gray in d. GAPDH was used as internal control. All the results were shown as mean ± SD (n = 3), which were three separate experiments performed in triplicate. *p < 0.05 and **p < 0.01
Fig. 4
Fig. 4
ICA suppressed chondrocytes injury and pyroptosis through NLRP3. a The mRNA level of NLRP3 was validated by qRT-PCR in chondrocytes. b The protein level was assessed by Western blotting in chondrocytes. GAPDH was used as internal control. c Leakage of LDH was assessed by ELISA assay in chondrocytes. The release level of IL-1β (d) and IL-18 (e) were analyzed by ELISA assay in chondrocytes. f The protein level of pyroptosis-related protein was analyzed by Western blotting in rat chondrocytes. g Quantitative analysis of protein band gray in e.GAPDH was used as internal control. All the results were shown as mean ± SD (n = 3), which were three separate experiments performed in triplicate. *p < 0.05 and **p < 0.01
Fig. 5
Fig. 5
ICA protected rat against osteoarthritis. a H&E staining and safranin O/fast green staining were used to evaluate cartilage histopathology in OA rats, and immunohitochemical analysis was used to detect NLRP3 in OA rat cartilage tissue. b Statistical analysis of NLRP3 positive rate. All the results were shown as mean ± SD (n = 3), which were three separate experiments performed in triplicate. *p < 0.05 and **p < 0.01
Fig. 6
Fig. 6
ICA attenuated pyroptosis and promoted collagen formation in OA rats. The mRNA levels of NLRP3 (a), IL-1β (b), and IL-18 (c) were examined with qRT-PCR in OA rat cartilage tissue. d The expression level of collagen formation and pyroptosis-related protein was analyzed by Western blotting in OA rat cartilage tissue. e Quantitative analysis of protein band gray in d. GAPDH was used as internal control. All the results were shown as mean ± SD (n = 3), which were three separate experiments performed in triplicate. *p < 0.05 and **p < 0.01

References

    1. Guermazi A, et al. Why radiography should no longer be considered a surrogate outcome measure for longitudinal assessment of cartilage in knee osteoarthritis. Arthritis Res Ther. 2011;13(6):247. doi: 10.1186/ar3488.
    1. Singh R, Akhtar N, Haqqi TM. Green tea polyphenol epigallocatechin-3-gallate: inflammation and arthritis. [corrected] Life Sci. 2010;86(25):907–918. doi: 10.1016/j.lfs.2010.04.013.
    1. Zheng Jiapeng, Li Qiang. Methylene blue regulates inflammatory response in osteoarthritis by noncoding long chain RNA CILinc02. Journal of Cellular Biochemistry. 2018;120(3):3331–3338. doi: 10.1002/jcb.27602.
    1. Favero Marta, Belluzzi Elisa, Trisolino Giovanni, Goldring Mary B, Goldring Steven R, Cigolotti Augusto, Pozzuoli Assunta, Ruggieri Pietro, Ramonda Roberta, Grigolo Brunella, Punzi Leonardo, Olivotto Eleonora. Inflammatory molecules produced by meniscus and synovium in early and end‐stage osteoarthritis: a coculture study. Journal of Cellular Physiology. 2018;234(7):11176–11187. doi: 10.1002/jcp.27766.
    1. He XF, et al. Investigation for effects of iNOS on biological function of chondrocytes in rats with post-traumatic osteoarthritis. Eur Rev Med Pharmacol Sci. 2018;22(21):7140–7147.
    1. Patel L, et al. Tenascin-C induces inflammatory mediators and matrix degradation in osteoarthritic cartilage. BMC Musculoskelet Disord. 2011;12:164. doi: 10.1186/1471-2474-12-164.
    1. Lu YC, Evans CH, Grodzinsky AJ. Effects of short-term glucocorticoid treatment on changes in cartilage matrix degradation and chondrocyte gene expression induced by mechanical injury and inflammatory cytokines. Arthritis Res Ther. 2011;13(5):R142. doi: 10.1186/ar3456.
    1. Miao EA, Rajan JV, Aderem A. Caspase-1-induced pyroptotic cell death. Immunological Rev. 2011;243(1):206–214. doi: 10.1111/j.1600-065X.2011.01044.x.
    1. Zhao LR, et al. NLRP1 and NLRP3 inflammasomes mediate LPS/ATP-induced pyroptosis in knee osteoarthritis. Mol Med Reports. 2018;17:4.
    1. Jin C, et al. NLRP3 inflammasome plays a critical role in the pathogenesis of hydroxyapatite-associated arthropathy. Proc Natl Acad Sci U S A. 2011;108(36):14867–14872. doi: 10.1073/pnas.1111101108.
    1. Zhou J, et al. Differential miRNAomics of the synovial membrane in knee osteoarthritis induced by bilateral anterior cruciate ligament transection in rats. Mol Med Rep. 2018;18(4):4051–4057.
    1. Zhang W, et al. Effect of Chinese traditional herb Epimedium grandiflorum C. Morren and its extract Icariin on osteoarthritis via suppressing NF-kappaB pathway. Indian J Exp Biol. 2013;51(4):313–321.
    1. Mi B, et al. Icariin activates autophagy via down-regulation of the NF-κB signaling-mediated apoptosis in chondrocytes. Front Pharmacol. 2018;9:605. doi: 10.3389/fphar.2018.00605.
    1. Wang Z, et al. Effects of icariin on the regulation of the OPG-RANKL-RANK system are mediated through the MAPK pathways in IL-1β-stimulated human SW1353 chondrosarcoma cells. Int J Mol Med. 2014;34(6):1720–6. doi: 10.3892/ijmm.2014.1952.
    1. Zeng L, et al. Icariin inhibits MMP-1, MMP-3 and MMP-13 expression through MAPK pathways in IL-1β-stimulated SW1353 chondrosarcoma cells. Mol Med Rep. 2017;15(5):2853–2858. doi: 10.3892/mmr.2017.6312.
    1. Vincourt JB, et al. Establishment of a reliable method for direct proteome characterization of human articular cartilage. Mol Cell Proteomics. 2006;5(10):1984–1995. doi: 10.1074/mcp.T600007-MCP200.
    1. Parra-Torres NM, Cázares-Raga FE, Kouri JB. Proteomic analysis of rat cartilage: the identification of differentially expressed proteins in the early stages of osteoarthritis. Proteome Sci. 2014;12(1):55. doi: 10.1186/s12953-014-0055-0.
    1. Qu Y, et al. Morin exhibits anti-inflammatory effects on IL-1β-stimulated human osteoarthritis chondrocytes by activating the Nrf2 signaling pathway. Cell Physiol Biochem. 2018;51(4):1830–1838. doi: 10.1159/000495684.
    1. Scotece M, et al. Oleocanthal inhibits catabolic and inflammatory mediators in LPS-activated human primary osteoarthritis (OA) chondrocytes through MAPKs/NF-κB pathways. Cell Physiol Biochem. 2018;49(6):2414–2426. doi: 10.1159/000493840.
    1. Chin KY, Ima-Nirwana S. The role of vitamin E in preventing and treating osteoarthritis - a review of the current evidence. Front Pharmacol. 2018;9:946. doi: 10.3389/fphar.2018.00946.
    1. Zhong G, et al. Artemisinin ameliorates osteoarthritis by inhibiting the Wnt/β-catenin signaling pathway. Cell Physiol Biochem. 2018;51(6):2575–2590. doi: 10.1159/000495926.
    1. Li M, et al. Icariin ameliorates palmitate-induced insulin resistance through reducing thioredoxin-interacting protein (TXNIP) and suppressing ER stress in C2C12 myotubes. Front Pharmacol. 2018;9:1180. doi: 10.3389/fphar.2018.01180.
    1. Zhang Y, Kong WN, Chai XQ. Compound of icariin, astragalus, and puerarin mitigates iron overload in the cerebral cortex of Alzheimer’s disease mice. Neural Regen Res. 2018;13(4):731–736. doi: 10.4103/1673-5374.230303.
    1. Chen WF, et al. Neuroprotective properties of icariin in MPTP-induced mouse model of Parkinson’s disease: involvement of PI3K/Akt and MEK/ERK signaling pathways. Phytomedicine. 2017;25:93–99. doi: 10.1016/j.phymed.2016.12.017.
    1. Jin J, et al. An outline for the pharmacological effect of icariin in the nervous system. Eur J Pharmacol. 2018;842:20–32. doi: 10.1016/j.ejphar.2018.10.006.
    1. McAllister MJ, et al. NLRP3 as a potentially novel biomarker for the management of osteoarthritis. Osteoarthritis Cartilage. 2018;26(5):612–619. doi: 10.1016/j.joca.2018.02.901.
    1. Sun Y, et al. Curcumin prevents osteoarthritis by inhibiting the activation of inflammasome NLRP3. J Interferon Cytokine Res. 2017;37(10):449–455. doi: 10.1089/jir.2017.0069.
    1. Shi J, et al. Estradiol inhibits NLRP3 inflammasome in fibroblast-like synoviocytes activated by lipopolysaccharide and adenosine triphosphate. Int J Rheum Dis. 2017.
    1. Man Si Ming, Karki Rajendra, Kanneganti Thirumala-Devi. Molecular mechanisms and functions of pyroptosis, inflammatory caspases and inflammasomes in infectious diseases. Immunological Reviews. 2017;277(1):61–75. doi: 10.1111/imr.12534.
    1. Feng S, Fox D, Man SM. Mechanisms of Gasdermin family members in inflammasome signaling and cell death. J Mol Biol. 2018;430(18):3068–3080. doi: 10.1016/j.jmb.2018.07.002.
    1. McKenzie BA, et al. Caspase-1 inhibition prevents glial inflammasome activation and pyroptosis in models of multiple sclerosis. Proc Natl Acad Sci U S A. 2018;115(26):E6065–E6074. doi: 10.1073/pnas.1722041115.
    1. Chien H, Dix RD. Evidence for multiple cell death pathways during development of experimental cytomegalovirus retinitis in mice with retrovirus-induced immunosuppression: apoptosis, necroptosis, and pyroptosis. J Virol. 2012;86(20):10961–10978. doi: 10.1128/JVI.01275-12.
    1. Mamik MK, Power C. Inflammasomes in neurological diseases: emerging pathogenic and therapeutic concepts. Brain. 2017;140(9):2273–2285. doi: 10.1093/brain/awx133.
    1. Tan CC, et al. NLRP1 inflammasome is activated in patients with medial temporal lobe epilepsy and contributes to neuronal pyroptosis in amygdala kindling-induced rat model. J Neuroinflammation. 2015;12:18. doi: 10.1186/s12974-014-0233-0.
    1. Lacey CA, et al. Caspases-1 and caspase-11 mediate pyroptosis, inflammation, and control of. Infect Immun. 2018.
    1. Joosten LA, et al. Inflammatory arthritis in caspase 1 gene-deficient mice: contribution of proteinase 3 to caspase 1-independent production of bioactive interleukin-1beta. Arthritis Rheum. 2009;60(12):3651–3662. doi: 10.1002/art.25006.

Source: PubMed

3
Suscribir