Astaxanthin protects against osteoarthritis via Nrf2: a guardian of cartilage homeostasis

Kai Sun, Jiahui Luo, Xingzhi Jing, Jiachao Guo, Xudong Yao, Xiaoxia Hao, Yaping Ye, Shuang Liang, Jiamin Lin, Genchun Wang, Fengjing Guo, Kai Sun, Jiahui Luo, Xingzhi Jing, Jiachao Guo, Xudong Yao, Xiaoxia Hao, Yaping Ye, Shuang Liang, Jiamin Lin, Genchun Wang, Fengjing Guo

Abstract

Scope: Osteoarthritis (OA) is a progressive disease characterized by cartilage degradation. Astaxanthin (Ast), a natural compound with remarkable antioxidant activity and multiple medical applications due to its activation of Nrf2 signaling, has been studied for application to various degenerative diseases. Currently, however, little is known about its efficacy in treating OA. This study reports the effects of Ast on cartilage homeostasis in OA progression.

Methods: IL-1β, TNF-α, and tert-butyl hydroperoxide (TBHP) were used to impair cartilage homeostasis. Modulating effects of Ast on the Nrf2 signaling pathway, and damage-associated events including extracellular matrix (ECM) degradation, inflammation, oxidative stress, chondrocyte apoptosis, and in vivo cartilage degradation were examined.

Results: Ast attenuated ECM degradation of OA chondrocytes through the Nrf2 signaling, and ameliorated the IL-1β-induced inflammatory response and ECM degradation via blockade of MAPK signaling. Additionally, Ast alleviated TNF-α-induced ECM degradation and chondrocyte apoptosis by inhibiting the NF-κB signaling, suppressed TBHP-induced oxidative stress, and subsequently reduced chondrocyte apoptosis. In vitro results were finally corroborated in vivo by demonstrating that Ast attenuates the severity of cartilage destruction in a mouse model of OA.

Conclusions: Ast could protect against osteoarthritis via the Nrf2 signaling, suggesting Ast might be a potential therapeutic supplement for OA treatment.

Keywords: Nrf2; apoptosis; astaxanthin; cartilage homeostasis; osteoarthritis.

Conflict of interest statement

CONFLICTS OF INTEREST: The authors confirm that there are no conflicts of interest.

Figures

Figure 1
Figure 1
Ast did not affect cell viability and activated Nrf2 in mouse chondrocytes. (A) The cytotoxic effect of Ast (5, 10, 20, 40, and 80 μM) exposure for 24 and 48 h on chondrocytes was determined using a CCK8 assay. (B, C) Chondrocytes were treated with Ast (5, 10, and 20 μM) for 24 h. Expression levels of Cyclin D1, Nrf2, and Keap1 were determined by western blotting and quantified. (D, E) Nuclear translocation of Nrf2 was detected by western blotting and immunofluorescence after treatment of chondrocytes with Ast (10 μM) for 24 h, and the band density of Nrf2 in nucleus was quantified. The nuclear and cytoplasmic fractions used in the western blotting were obtained using a nuclear and cytoplasmic protein extraction kit (P0027, Beyotime, China). The data are presented as dot plots from three independent experiments. Significant differences among different groups are indicated as *p < 0.05 vs. control; **p<0.01 vs. control; ****p<0.0001 vs. control.
Figure 2
Figure 2
Effects of IL-1β, TNF-α, and TBHP on mouse chondrocytes. (A) The cytotoxic effect of IL-1β (1 and 5 ng/ml), TNF-α (1 and 5 ng/ml), and TBHP (50 and 100 μM) treatment on chondrocytes for 24 h was determined using a CCK8 assay. (B, C) After the indicated treatment for 24 h, the effects of IL-1β (5 ng/ml), TNF-α (5 ng/ml), and TBHP (100 μM) on the expression of MMP13, Collagen II, iNOS, and cleaved-caspase3 were determined by western blotting and quantified. The data are presented as dot plots from three independent experiments. Significant differences among different groups are indicated as *p < 0.05 vs. control; **p<0.01 vs. control; ***p<0.001 vs. control; ****p<0.0001 vs. control.
Figure 3
Figure 3
Effects of Ast on OA chondrocytes. Chondrocytes were pretreated with or without Ast (5, 10, and 20 μM) for 2 h and then exposed to IL-1β (5 ng/ml) for 24 h. Then, (AC) the expression of Collagen II, ADAMTS5, MMP3, and MMP13 was examined by RT-PCR and western blotting and quantified. Chondrocytes were treated with the vehicle or with increasing concentrations of Ast for 2 h followed by (5 ng/ml) TNF-α stimulation. (D, E) The expression of Collagen II, ADAMTS5, MMP3, and MMP13 were determined by western blotting and quantified. After the cells were treated with the vehicle or Ast (5, 10, and 20 μM) for 2 h, and stimulated with 100 μM TBHP, (F, G) the expression of Collagen II, ADAMTS5, and MMP13 was investigated by western blotting and quantified. The data are presented as dot plots from three independent experiments. Significant differences among different groups are indicated as **p<0.01 vs. control; ***p<0.001 vs. control; ****p<0.0001 vs. control. #p < 0.05 vs. IL-1β or TNF-α or TBHP group; ##p < 0.01 vs. IL-1β or TNF-α or TBHP group; ###p < 0.001 vs. IL-1β or TNF-α or TBHP group; ####p < 0.0001 vs. IL-1β or TNF-α or TBHP group.
Figure 4
Figure 4
Nrf2 signaling mediated protective effects of Ast on OA chondrocytes. Chondrocytes were cultured with vehicle or Ast (5, 10 and 20 μM) for 2 h, then stimulated with IL-1β (5 ng/ml), TNF-α (5 ng/ml) or TBHP (100 μM). (AC) Levels of Nrf2 signaling pathway proteins, including Nrf2, HO-1, and NQO1 were determined by western blotting and quantified. To knock down Nrf2 expression, chondrocytes were transfected with Nrf2 siRNA using Lipofectamine 3000. (D) Transfection efficiency was evaluated by detecting Nrf2 expression using western blotting. After 24h of transfection, chondrocytes were treated with vehicle or Ast (10 μM) for 2 h followed by stimulation with IL-1β (5 ng/ml) for 24 h. (E, F) The expression of Collagen II, ADAMTS5, MMP3, and MMP13 was measured by western blotting and quantified. The data are presented as dot plots from three independent experiments. Significant differences among different groups are indicated as *p<0.05 vs. control; **p<0.01 vs. control; ***p<0.001 vs. control; ****p<0.0001 vs. control. #p < 0.05 vs. IL-1β or TNF-α or TBHP group; ##p < 0.01 vs. IL-1β or TNF-α or TBHP group; ###p < 0.001 vs. IL-1β or TNF-α or TBHP group; ####p < 0.0001 vs. IL-1β or TNF-α or TBHP group. &p < 0.05 vs. (IL-1β or TNF-α or TBHP) + Ast group; &&p < 0.01 vs. (IL-1β or TNF-α or TBHP) + Ast group; &&&p < 0.001 vs. (IL-1β or TNF-α or TBHP) + Ast group.
Figure 5
Figure 5
Effects of Ast on IL-1β-induced inflammatory response. Chondrocytes were treated with Ast (5, 10, and 20 μM) for 2 h followed by stimulation with vehicle or IL-1β (5 ng/mL) for 24 h. (A) Expression of iNOS and COX-2 protein assessed by western blotting and quantified. (B) To detect altered phosphorylation within the MAPK signaling pathway, chondrocytes were serum-starved for 6 h followed by treatment with vehicle or Ast (5,10, and 20 μM) for 2 h. Cells were then stimulated with IL-1β (5 ng/mL) for 30 min. MAPK activation was examined using western blotting and quantified. (C) Phosphorylation of ERK was detected by western blotting after chondrocytes were pre-treated with vehicle or PD0325901 (a MEK inhibitor) for 2 h, followed by stimulation with IL-1β (5 ng/ml). (D) Chondrocytes were treated as indicated for 24 h. The expression of COX2, Collagen II, and MMP3 was determined using western blotting and quantified. The data are presented as dot plots from three independent experiments. Significant differences among different groups are indicated as *p<0.05 vs. control; **p<0.01 vs. control; ***p<0.001 vs. control; ****p<0.0001 vs. control. #p < 0.05 vs. IL-1β group; ##p < 0.01 vs. IL-1β group; ###p < 0.001 vs. IL-1β group; ####p < 0.0001 vs. IL-1β group.
Figure 6
Figure 6
Effects of Ast on TNF-α-induced apoptosis and ECM degradation. (A, B) Protein levels of BAX, Bcl-2, and cleaved caspase-3 were determined by western blotting, and apoptotic chondrocytes were stained with Annexin V-FITC/PI and examined by flow cytometry after treated as above for 24 h. Apoptosis rate was calculated and the data were expressed in (C). FL1 represents Annexin V-FITC and FL2 represents PI. To detect the activation of NF-κB signaling, chondrocytes were serum-starved for 6 h followed by treatment with the vehicle or Ast (5,10, and 20 μM) for 2 h. Cells were then stimulated with TNF-α (5 ng/ml) for 15 min. (D) Activation of the NF-κB signaling pathway was measured by western blotting and quantified. (E) Phosphorylation of p65 was detected by western blotting after the chondrocytes were pre-treated with the vehicle or QNZ (an inhibitor of the NF-κB pathway) for 2 h, followed by stimulation with TNF-α (5 ng/ml). (F, G) Chondrocytes were treated as indicated for 24 h. The expression of Collagen II, MMP3 and cleaved-caspase3 was determined using western blotting and quantified. The data are presented as dot plots from three independent experiments. Significant differences among different groups are indicated as *p<0.05 vs. control; **p<0.01 vs. control; ***p<0.001 vs. control. #p < 0.05 vs. TNF-α group; ##p < 0.01 vs. TNF-α group; ###p < 0.001 vs. TNF-α group.
Figure 7
Figure 7
Effects of Ast on TBHP-induced oxidative stress, and chondrocyte apoptosis. Chondrocytes were treated with Ast (10 μM) for 2 h, then stimulated with TBHP (100 μM) for 24 h. The samples with the same concentration of chondrocytes were used to detect intracellular ROS. Intracellular ROS was stained by DCFH-DA followed by fluorescence spectrophotometry and flow cytometric analysis. (A) Green fluorescence indicates that the intracellular ROS production. (B) Representative data of flow cytometric measurement of ROS production. (C) Dot plots graphs show the mean fluorescence intensity (MFI) of ROS in chondrocytes. (D, E) Mitochondrial membrane potential (MMP) was detected by fluorescence and flow cytometric analysis after chondrocytes were incubated with JC-1. Red fluorescence represents JC-1 aggregates in healthy mitochondria, while green fluorescence is emitted by JC-1 monomers, representing MMP dissipation. Merged images exhibit co-localization of JC-1 aggregates and monomers. FL1 represents JC-1 green and FL2 represents JC-1 red. (F) Dot plots graphs represent flow cytometric analysis of MMP levels, represented as the ratio of red/green MFI. (H) To measure chondrocyte apoptosis, cells were stained with Annexin V-FITC/PI and apoptotic cells were counted by flow cytometry. FL1 represents Annexin V-FITC and FL2 represents PI. Apoptosis rates were evaluated, and the results are expressed as dot plots graphs (I). (G) Chondrocytes were treated with vehicle or Ast (5, 10, and 20 μM) for 2 h, then stimulated with TBHP (100 μM) for 24 h. Protein levels of BAX, Bcl-2, and cleaved-caspase3 were examined by western blotting and quantified. (J) Expression of cleaved-caspase 3 protein in chondrocytes under treatment with scrambled siRNA (NC), TBHP (100 μM), N-acetylcysteine (NAC)+TBHP, Nrf2-siRNA+TBHP, respectively. The data are presented as dot plots from three independent experiments. Significant differences among different groups are indicated as *p<0.05 vs. control; **p<0.01 vs. control; ***p<0.001 vs. control. #p < 0.05 vs. TBHP group; ##p < 0.01 vs. TBHP group.
Figure 8
Figure 8
Effects of Ast on cartilage degradation in vivo. Mice received intra-articular injections of Ast (20 mg/kg) or vehicle for 8 weeks after DMM surgery. (A) Histological analysis of cartilage degradation was evaluated by safranin O staining. Representative safranin O staining of cartilage (scale bar: 200 μm and 50 μm). (B) Osteoarthritis Research Society International (OARSI) scores of three groups. (C) Nrf2 expression in cartilage was detected by immunofluorescence staining (scale bar: 50 μm). (D) Dot plots graphs represent rates of nuclear Nrf2 in total chondrocytes from each cartilage section. The data are presented as dot plots from three independent experiments. Significant differences among different groups are indicated as **P < 0.01; ***P<0.001; ****P < 0.0001.
Figure 9
Figure 9
Schematic diagram of Ast-mediated protective effects on cartilage homeostasis. Ast attenuated ECM degradation of OA chondrocytes through the Nrf2 signaling, and ameliorated the IL-1β-induced inflammatory response and ECM degradation via blockade of MAPK signaling. In addition, Ast alleviated TNF-α-induced ECM degradation and chondrocyte apoptosis by inhibiting the NF-kB signaling, suppressed TBHP-induced oxidative stress, and subsequently reduced chondrocyte apoptosis.

References

    1. Morales-Ivorra I, Romera-Baures M, Roman-Viñas B, Serra-Majem L. Osteoarthritis and the Mediterranean Diet: A Systematic Review. Nutrients. 2018; 10:10. 10.3390/nu10081030
    1. Glyn-Jones S, Palmer AJ, Agricola R, Price AJ, Vincent TL, Weinans H, Carr AJ. Osteoarthritis. Lancet. 2015; 386:376–87. 10.1016/S0140-6736(14)60802-3
    1. Hunter DJ, Bierma-Zeinstra S. Osteoarthritis. Lancet. 2019; 393:1745–59. 10.1016/S0140-6736(19)30417-9
    1. Lotz MK, Caramés B. Autophagy and cartilage homeostasis mechanisms in joint health, aging and OA. Nat Rev Rheumatol. 2011; 7:579–87. 10.1038/nrrheum.2011.109
    1. Hosseinzadeh A, Kamrava SK, Joghataei MT, Darabi R, Shakeri-Zadeh A, Shahriari M, Reiter RJ, Ghaznavi H, Mehrzadi S. Apoptosis signaling pathways in osteoarthritis and possible protective role of melatonin. J Pineal Res. 2016; 61:411–25. 10.1111/jpi.12362
    1. Kapoor M, Martel-Pelletier J, Lajeunesse D, Pelletier JP, Fahmi H. Role of proinflammatory cytokines in the pathophysiology of osteoarthritis. Nat Rev Rheumatol. 2011; 7:33–42. 10.1038/nrrheum.2010.196
    1. Giorgio M, Trinei M, Migliaccio E, Pelicci PG. Hydrogen peroxide: a metabolic by-product or a common mediator of ageing signals? Nat Rev Mol Cell Biol. 2007; 8:722–28. 10.1038/nrm2240
    1. Bolduc JA, Collins JA, Loeser RF. Reactive oxygen species, aging and articular cartilage homeostasis. Free Radic Biol Med. 2019; 132:73–82. 10.1016/j.freeradbiomed.2018.08.038
    1. Cuadrado A, Rojo AI, Wells G, Hayes JD, Cousin SP, Rumsey WL, Attucks OC, Franklin S, Levonen AL, Kensler TW, Dinkova-Kostova AT. Therapeutic targeting of the NRF2 and KEAP1 partnership in chronic diseases. Nat Rev Drug Discov. 2019; 18:295–317. 10.1038/s41573-018-0008-x
    1. Kumar H, Kim IS, More SV, Kim BW, Choi DK. Natural product-derived pharmacological modulators of Nrf2/ARE pathway for chronic diseases. Nat Prod Rep. 2014; 31:109–39. 10.1039/C3NP70065H
    1. Khan NM, Ahmad I, Haqqi TM. Nrf2/ARE pathway attenuates oxidative and apoptotic response in human osteoarthritis chondrocytes by activating ERK1/2/ELK1-P70S6K-P90RSK signaling axis. Free Radic Biol Med. 2018; 116:159–71. 10.1016/j.freeradbiomed.2018.01.013
    1. Khan NM, Haseeb A, Ansari MY, Devarapalli P, Haynie S, Haqqi TM. Wogonin, a plant derived small molecule, exerts potent anti-inflammatory and chondroprotective effects through the activation of ROS/ERK/Nrf2 signaling pathways in human Osteoarthritis chondrocytes. Free Radic Biol Med. 2017; 106:288–301. 10.1016/j.freeradbiomed.2017.02.041
    1. Guerin M, Huntley ME, Olaizola M. Haematococcus astaxanthin: applications for human health and nutrition. Trends Biotechnol. 2003; 21:210–16. 10.1016/S0167-7799(03)00078-7
    1. Fakhri S, Abbaszadeh F, Dargahi L, Jorjani M. Astaxanthin: A mechanistic review on its biological activities and health benefits. Pharmacol Res. 2018; 136:1–20. 10.1016/j.phrs.2018.08.012
    1. Farruggia C, Kim MB, Bae M, Lee Y, Pham TX, Yang Y, Han MJ, Park YK, Lee JY. Astaxanthin exerts anti-inflammatory and antioxidant effects in macrophages in NRF2-dependent and independent manners. J Nutr Biochem. 2018; 62:202–09. 10.1016/j.jnutbio.2018.09.005
    1. Xue XL, Han XD, Li Y, Chu XF, Miao WM, Zhang JL, Fan SJ. Astaxanthin attenuates total body irradiation-induced hematopoietic system injury in mice via inhibition of oxidative stress and apoptosis. Stem Cell Res Ther. 2017; 8:7. 10.1186/s13287-016-0464-3
    1. Huang LJ, Chen WP. Astaxanthin ameliorates cartilage damage in experimental osteoarthritis. Mod Rheumatol. 2015; 25:768–71. 10.3109/14397595.2015.1008724
    1. Chen WP, Xiong Y, Shi YX, Hu PF, Bao JP, Wu LD. Astaxanthin reduces matrix metalloproteinase expression in human chondrocytes. Int Immunopharmacol. 2014; 19:174–77. 10.1016/j.intimp.2013.12.007
    1. Higuera-Ciapara I, Félix-Valenzuela L, Goycoolea FM. Astaxanthin: a review of its chemistry and applications. Crit Rev Food Sci Nutr. 2006; 46:185–96. 10.1080/10408690590957188
    1. Tu C, Huang X, Xiao Y, Song M, Ma Y, Yan J, You H, Wu H, Schisandrin A. Schisandrin A Inhibits the IL-1β-Induced Inflammation and Cartilage Degradation via Suppression of MAPK and NF-κB Signal Pathways in Rat Chondrocytes. Front Pharmacol. 2019; 10:41. 10.3389/fphar.2019.00041
    1. Ma CH, Wu CH, Jou IM, Tu YK, Hung CH, Hsieh PL, Tsai KL. PKR activation causes inflammation and MMP-13 secretion in human degenerated articular chondrocytes. Redox Biol. 2018; 14:72–81. 10.1016/j.redox.2017.08.011
    1. Young IC, Chuang ST, Hsu CH, Sun YJ, Lin FH. C-phycocyanin alleviates osteoarthritic injury in chondrocytes stimulated with H2O2 and compressive stress. Int J Biol Macromol. 2016; 93:852–59. 10.1016/j.ijbiomac.2016.09.051
    1. Iwasa K, Hayashi S, Fujishiro T, Kanzaki N, Hashimoto S, Sakata S, Chinzei N, Nishiyama T, Kuroda R, Kurosaka M. PTEN regulates matrix synthesis in adult human chondrocytes under oxidative stress. J Orthop Res. 2014; 32:231–37. 10.1002/jor.22506
    1. Xie X, Chen Q, Tao J. Astaxanthin Promotes Nrf2/ARE Signaling to Inhibit HG-Induced Renal Fibrosis in GMCs. Mar Drugs. 2018; 16:16. 10.3390/md16040117
    1. Robinson WH, Lepus CM, Wang Q, Raghu H, Mao R, Lindstrom TM, Sokolove J. Low-grade inflammation as a key mediator of the pathogenesis of osteoarthritis. Nat Rev Rheumatol. 2016; 12:580–92. 10.1038/nrrheum.2016.136
    1. Lee SW, Rho JH, Lee SY, Kim JH, Cheong JH, Kim HY, Jeong NY, Chung WT, Yoo YH. Leptin protects rat articular chondrocytes from cytotoxicity induced by TNF-α in the presence of cyclohexamide. Osteoarthritis Cartilage. 2015; 23:2269–78. 10.1016/j.joca.2015.06.005
    1. Yan L, Zhou L, Xie D, Du W, Chen F, Yuan Q, Tong P, Shan L, Efferth T. Chondroprotective effects of platelet lysate towards monoiodoacetate-induced arthritis by suppression of TNF-α-induced activation of NF-ĸB pathway in chondrocytes. Aging (Albany NY). 2019; 11:2797–811. 10.18632/aging.101952
    1. Liu ZG. Molecular mechanism of TNF signaling and beyond. Cell Res. 2005; 15:24–27. 10.1038/sj.cr.7290259
    1. Lee SJ, Bai SK, Lee KS, Namkoong S, Na HJ, Ha KS, Han JA, Yim SV, Chang K, Kwon YG, Lee SK, Kim YM. Astaxanthin inhibits nitric oxide production and inflammatory gene expression by suppressing I(kappa)B kinase-dependent NF-kappaB activation. Mol Cells. 2003; 16:97–105.
    1. Kim SH, Kim H. Inhibitory Effect of Astaxanthin on Oxidative Stress-Induced Mitochondrial Dysfunction-A Mini-Review. Nutrients. 2018; 10:10. 10.3390/nu10091137
    1. Mobasheri A, Rayman MP, Gualillo O, Sellam J, van der Kraan P, Fearon U. The role of metabolism in the pathogenesis of osteoarthritis. Nat Rev Rheumatol. 2017; 13:302–11. 10.1038/nrrheum.2017.50
    1. El-Agamy SE, Abdel-Aziz AK, Wahdan S, Esmat A, Azab SS. Astaxanthin Ameliorates Doxorubicin-Induced Cognitive Impairment (Chemobrain) in Experimental Rat Model: Impact on Oxidative, Inflammatory, and Apoptotic Machineries. Mol Neurobiol. 2018; 55:5727–40. 10.1007/s12035-017-0797-7
    1. Kim B, Farruggia C, Ku CS, Pham TX, Yang Y, Bae M, Wegner CJ, Farrell NJ, Harness E, Park YK, Koo SI, Lee JY. Astaxanthin inhibits inflammation and fibrosis in the liver and adipose tissue of mouse models of diet-induced obesity and nonalcoholic steatohepatitis. J Nutr Biochem. 2017; 43:27–35. 10.1016/j.jnutbio.2016.01.006
    1. Li X, Lin J, Ding X, Xuan J, Hu Z, Wu D, Zhu X, Feng Z, Ni W, Wu A. The protective effect of sinapic acid in osteoarthritis: in vitro and in vivo studies. J Cell Mol Med. 2019; 23:1940–50. 10.1111/jcmm.14096
    1. Cai D, Yin S, Yang J, Jiang Q, Cao W. Histone deacetylase inhibition activates Nrf2 and protects against osteoarthritis. Arthritis Res Ther. 2015; 17:269. 10.1186/s13075-015-0774-3
    1. Sellam J, Berenbaum F. The role of synovitis in pathophysiology and clinical symptoms of osteoarthritis. Nat Rev Rheumatol. 2010; 6:625–35. 10.1038/nrrheum.2010.159
    1. Leonidou A, Lepetsos P, Mintzas M, Kenanidis E, Macheras G, Tzetis M, Potoupnis M, Tsiridis E. Inducible nitric oxide synthase as a target for osteoarthritis treatment. Expert Opin Ther Targets. 2018; 22:299–318. 10.1080/14728222.2018.1448062
    1. Li MY, Sun L, Niu XT, Chen XM, Tian JX, Kong YD, Wang GQ. Astaxanthin protects lipopolysaccharide-induced inflammatory response in Channa argus through inhibiting NF-κB and MAPKs signaling pathways. Fish Shellfish Immunol. 2019; 86:280–86. 10.1016/j.fsi.2018.11.011
    1. Kim YH, Koh HK, Kim DS. Down-regulation of IL-6 production by astaxanthin via ERK-, MSK-, and NF-κB-mediated signals in activated microglia. Int Immunopharmacol. 2010; 10:1560–72. 10.1016/j.intimp.2010.09.007
    1. Kühn K, D’Lima DD, Hashimoto S, Lotz M. Cell death in cartilage. Osteoarthritis Cartilage. 2004; 12:1–16. 10.1016/j.joca.2003.09.015
    1. Stanic I, Facchini A, Borzì RM, Vitellozzi R, Stefanelli C, Goldring MB, Guarnieri C, Facchini A, Flamigni F. Polyamine depletion inhibits apoptosis following blocking of survival pathways in human chondrocytes stimulated by tumor necrosis factor-alpha. J Cell Physiol. 2006; 206:138–46. 10.1002/jcp.20446
    1. Pan T, Chen R, Wu D, Cai N, Shi X, Li B, Pan J. Alpha-Mangostin suppresses interleukin-1β-induced apoptosis in rat chondrocytes by inhibiting the NF-κB signaling pathway and delays the progression of osteoarthritis in a rat model. Int Immunopharmacol. 2017; 52:156–62. 10.1016/j.intimp.2017.08.021
    1. Sies H, Berndt C, Jones DP. Oxidative Stress. Annu Rev Biochem. 2017; 86:715–48. 10.1146/annurev-biochem-061516-045037
    1. Wang JY, Lee YJ, Chou MC, Chang R, Chiu CH, Liang YJ, Wu LS. Astaxanthin protects steroidogenesis from hydrogen peroxide-induced oxidative stress in mouse Leydig cells. Mar Drugs. 2015; 13:1375–88. 10.3390/md13031375
    1. Song X, Wang B, Lin S, Jing L, Mao C, Xu P, Lv C, Liu W, Zuo J. Astaxanthin inhibits apoptosis in alveolar epithelial cells type II in vivo and in vitro through the ROS-dependent mitochondrial signalling pathway. J Cell Mol Med. 2014; 18:2198–212. 10.1111/jcmm.12347
    1. Liang S, Lv ZT, Zhang JM, Wang YT, Dong YH, Wang ZG, Chen K, Cheng P, Yang Q, Guo FJ, Lu WW, Zhu WT, Chen AM. Necrostatin-1 Attenuates Trauma-Induced Mouse Osteoarthritis and IL-1β Induced Apoptosis via HMGB1/TLR4/SDF-1 in Primary Mouse Chondrocytes. Front Pharmacol. 2018; 9:1378. 10.3389/fphar.2018.01378
    1. Jing X, Du T, Chen K, Guo J, Xiang W, Yao X, Sun K, Ye Y, Guo F. Icariin protects against iron overload-induced bone loss via suppressing oxidative stress. J Cell Physiol. 2019; 234:10123–10137. 10.1002/jcp.27678
    1. Ma HL, Blanchet TJ, Peluso D, Hopkins B, Morris EA, Glasson SS. Osteoarthritis severity is sex dependent in a surgical mouse model. Osteoarthritis Cartilage. 2007; 15:695–700. 10.1016/j.joca.2006.11.005
    1. Glasson SS, Chambers MG, Van Den Berg WB, Little CB. The OARSI histopathology initiative -recommendations for histological assessments of osteoarthritis in the mouse. Osteoarthritis Cartilage. 2010. (Suppl 3); 18:S17–23. 10.1016/j.joca.2010.05.025

Source: PubMed

3
Se inscrever