Effects of Autophagy on Chondrocyte Apoptosis in Osteoarthritis: An Investigation Based on lncRNA/Hedgehog Signaling Pathway Expression
-
摘要:
目的 探究骨关节炎中lncRNA /Hedgehog信号通路介导的自噬对软骨细胞功能的影响。 方法 在OA软骨细胞(SW1353)建立LPS诱导的软骨细胞炎症模型,并通过胶原II免疫荧光染色和甲苯胺蓝染色进行鉴定,分为Normal组、LPS组、LPS/lncRNA HHIP-AS1 抑制剂组和LPS/Scr组,RT-qPCR检测lncRNA HHIP- as1和HHIP在软骨细胞中的表达,蛋白印迹HHIP、Gli1、Gli2、LC3B-I ab、p62表达,TUNEL染色和流式细胞术(FC)检测细胞凋亡,免疫荧光法(IFA)检测自噬LC3B表达。 结果 用LPS处理SW1细胞,与正常软骨细胞相比,LPS诱导后,软骨细胞体积增大,胞浆内空泡增多,细胞核体积增大,部分细胞形态不规则,数量相对减少,甲苯胺蓝染色和II型胶原免疫组化染色减少,LPS刺激会诱导细胞死亡和自噬,lncRNA HHIP- as1和HHIP上调(P < 0.05),Hedgehog信号通路HHIP、Gli1和Gli2的关键分子持续上调(P < 0.05),LPS处理的软骨细胞有明显的凋亡(P < 0.05),LC3B(绿色)积累,LC3B生物合成和加工升高(LC3B I和II水平升高),p62降解升高(P < 0.05),lncRNA HHIP-AS1抑制剂降低LPS诱导的OA软骨细胞凋亡(细胞凋亡率降低)和自噬(降低了LPS处理的软骨细胞的自噬率,LC3B生物合成和加工减少(LC3B I和II水平降低),p62降解减弱),差异有统计学意义(P < 0.05)。 结论 LncRNA HHIP-AS1可能通过调控Hedgehog信号通路抑制LPS诱导的OA软骨细胞凋亡和自噬。 -
关键词:
- 骨关节炎 /
- lncRNA HHIP-AS1 /
- Hedgehog /
- 自噬 /
- 软骨细胞
Abstract:Objective To investigate the effects of lncRNA/Hedgehog signaling pathway-mediated autophagy on chondrocyte function in osteoarthritis (OA). Methods Established an LPS-induced inflammatory chondrocyte model in OA chondrocytes (SW1353), and identified it through collagen II immunofluorescence staining and toluidine blue staining, dividing the groups into Normal, LPS, LPS/lncRNA HHIP-AS1 inhibitor, and LPS/Scr groups. RT-qPCR was used to detect lncRNA HHIP-AS1 and HHIP expression in chondrocytes, Western blot was used to assess HHIP, Gli1, Gli2, LC3B-I/II, and p62 protein expression, TUNEL staining and flow cytometry (FC) were used to detect cell apoptosis, and immunofluorescence assay (IFA) was used to detect autophagy LC3B expression. Results When SW1 cells were treated with LPS, compared with normal chondrocytes, after LPS induction, the volume of chondrocytes increased, the number of vacuoli in the cytoplasm increased, the volume of the nucleus increased, the morphology of some cells was irregular, and the number relatively decreased. Toluidine blue staining and type II collagen immunohistochemical staining decreased. LPS stimulation would induce cell death and autophagy. lncRNA HIP-AS1 and HHIP were upregulated (P < 0.05), the key molecules of the Hedgehog signaling pathway HHIP, Gli1 and Gli2 were continuously upregulated (P < 0.05), chondrocytes treated with LPS showed obvious apoptosis (P < 0.05), and LC3B (green) accumulated. The biosynthesis and processing of LC3B increased (the levels of LC3B I and II increased), the degradation of p62 increased (P < 0.05), and the lncRNA HIP-AS1 inhibitor reduced LPS-induced apoptosis of OA chondrocytes (decreased apoptosis rate) and autophagy (decreased autophagy rate of chondrocytes treated with LPS). The biosynthesis and processing of LC3B decreased (the levels of LC3B I and II decreased), and the degradation of p62 weakened), and the difference was statistically significant (P < 0.05). Conclusion The lncRNA HHIP-AS1 may inhibit LPS-induced OA chondrocyte apoptosis and autophagy by regulating the Hedgehog signaling pathway. -
Key words:
- Osteoarthritis /
- lncRNA HIP-AS1 /
- Hedgehog /
- Autophagy /
- Chondrocyte
-
表 1 lncRNA HHIP-AS1和HHIP在LPS诱导的小鼠OA软骨细胞中表达($\bar x \pm s $)
Table 1. Expression of lncRNA HHIP-AS1 and HHIP in LPS-induced mouse OA chondrocytes ($\bar x \pm s $)
分组 lncRNA HHIP-AS1 HHIP mRNA Normal组(n=9) 1.070±0.370 0.890±0.320 LPS组(n=9) 2.650±0.360 2.250±0.390 t 9.180 8.080 P <0.001* <0.001* *P < 0.05。 表 2 lncRNA HHIP-AS1对Hedgehog信号通路的影响($\bar x \pm s $)
Table 2. Effects of lncRNA HHIP-AS1 on the Hedgehog signaling pathway($\bar x \pm s $)
分组 HHIP/GAPDH Gli1/GAPDH Gli2/GAPDH Normal组(n=9) 0.530±0.170 0.390±0.120 0.490±0.110 LPS组(n=9) 1.350±0.260Δ 1.010±0.140Δ 1.140±0.130Δ LPS/HHIP-AS1 inhibitor(n=9) 0.270±0.050 0.400±0.100 0.620±0.150 LPS/Scr(n=9) 1.110±0.160# 1.000±0.150# 1.060±0.190# F 98.520 45.140 32.75 P <0.001* <0.001* <0.001* *P < 0.05;与Normal组比较,ΔP < 0.05;与LPS组比较,#P < 0.05。 表 3 lncRNA HHIP-AS1/Hedgehog信号通路对LPS诱导的OA软骨细胞凋亡的影响($\bar x \pm s $)
Table 3. Impact of lncRNA HHIP-AS1/Hedgehog signaling pathway on apoptosis of LPS-induced OA chondrocytes ($\bar x \pm s $)
分组 TUNEL染色
阳性细胞(%)凋亡率(%) Normal组(n=9) 5.080±1.010 7.450±1.110 LPS组(n=9) 19.120±1.960Δ 22.210±2.120Δ LPS/HHIP-AS1 inhibitor(n=9) 4.210±1.040 6.410±2.010 LPS/Scr(n=9) 20.130±1.660# 23.340±2.110# F 289.120 245.090 P <0.001* <0.001* 与Normal组比较,ΔP < 0.05;与LPS组比较,#P < 0.05;*P < 0.05。 表 4 lncRNA HHIP-AS1/Hedgehog信号通路对LPS诱导的OA软骨细胞自噬的影响($\bar x \pm s $)
Table 4. Effects of lncRNA HHIP-AS1/Hedgehog signaling pathway on autophagy of LPS-induced OA chondrocytes ($\bar x \pm s $)
分组 LC3B/GAPDH p62/GAPDH Normal组(n=9) 0.380±0.090 1.440±0.150 LPS组(n=9) 1.320±0.110Δ 0.310±0.120Δ LPS/HHIP-AS1 inhibitor(n=9) 0.510±0.130 1.410±0.110 LPS/Scr(n=9) 1.430±0.160# 0.390±0.140# F 294.83 198.48 P 0.001* 0.001* 与Normal组比较,ΔP < 0.05;与LPS组比较,#P < 0.05;*P < 0.05。 -
[1] Abramoff B,Caldera F E. Osteoarthritis: Pathology,diagnosis,and treatment options[J]. Med Clin North Am,2020,104(2):293-311. doi: 10.1016/j.mcna.2019.10.007 [2] Xia B,Di Chen,Zhang J,et al. Osteoarthritis pathogenesis: A review of molecular mechanisms[J]. Calcif Tissue Int,2014,95(6):495-505. doi: 10.1007/s00223-014-9917-9 [3] Yao Q,Wu X,Tao C,et al. Osteoarthritis: Pathogenic signaling pathways and therapeutic targets[J]. Signal Transduct Target Ther,2023,8(1):56. doi: 10.1038/s41392-023-01330-w [4] Yao Z T,Yang Y M,Sun M M,et al. New insights into the interplay between long non-coding RNAs and RNA-binding proteins in cancer[J]. Cancer Commun (Lond),2022,42(2):117-140. doi: 10.1002/cac2.12254 [5] Gu Y,Liu X,Liao L,et al. Relationship between lipid metabolism and Hedgehog signaling pathway[J]. J Steroid Biochem Mol Biol,2021,209(1):105825. doi: 10.1016/j.jsbmb.2021.105825 [6] Yin X H,Wang X Y,Liu S C,et al. lncRNA HHIP-AS1/HHIP modulates osteogenic differentiation of BM-MSCs by regulating Hedgehog signaling pathway[J]. Aging (Albany NY),2022,14(21):8839-8855. doi: 10.18632/aging.204381 [7] Deng C,Zhu H,Li J,et al. Bioactive scaffolds for regeneration of cartilage and subchondral bone interface[J]. Theranostics,2018,8(7):1940-1955. doi: 10.7150/thno.23674 [8] Guo Q,Xu J,Shi Q,et al. PDLIM2 protects articular chondrocytes from lipopolysaccharide-induced apoptosis,degeneration and inflammatory injury through down-regulation of nuclear factor (NF)-kappaB signaling[J]. Int Immunopharmacol,2020,88(1):106883. doi: 10.1016/j.intimp.2020.106883 [9] Steele N G,Biffi G,Kemp S B,et al. Inhibition of hedgehog signaling alters fibroblast composition in pancreatic cancer[J]. Clin Cancer Res,2021,27(7):2023-2037. doi: 10.1158/1078-0432.CCR-20-3715 [10] Cai C,Min S,Yan B,et al. MiR-27a promotes the autophagy and apoptosis of IL-1β treated-articular chondrocytes in osteoarthritis through PI3K/AKT/mTOR signaling[J]. Aging (Albany NY),2019,11(16):6371-6384. doi: 10.18632/aging.102194 [11] Motta F,Barone E,Sica A,et al. Inflammaging and osteoarthritis[J]. Clin Rev Allergy Immunol,2023,64(2):222-238. [12] Chen G,Liu T,Yu B,et al. CircRNA-UBE2G1 regulates LPS-induced osteoarthritis through miR-373/HIF-1a axis[J]. Cell Cycle,2020,19(13):1696-1705. doi: 10.1080/15384101.2020.1772545 [13] Hodgkinson T,Kelly D C,Curtin C M,et al. Mechanosignalling in cartilage: An emerging target for the treatment of osteoarthritis[J]. Nat Rev Rheumatol,2022,18(2):67-84. doi: 10.1038/s41584-021-00724-w [14] Zhang M,Hu W,Cai C,et al. Advanced application of stimuli-responsive drug delivery system for inflammatory arthritis treatment[J]. Mater Today Bio,2022,14(1):100223. doi: 10.1016/j.mtbio.2022.100223 [15] Jhun J,Cho K H,Lee D H,et al. Oral administration of lactobacillus rhamnosus ameliorates the progression of osteoarthritis by inhibiting joint pain and inflammation[J]. Cells,2021,10(5):1057. doi: 10.3390/cells10051057 [16] Liu W B,Li G S,Shen P,et al. Long non-coding RNA HOTAIRM1-1 silencing in cartilage tissue induces osteoarthritis through microRNA-125b[J]. Exp Ther Med,2021,22(3):933. doi: 10.3892/etm.2021.10365 [17] Qin Q,Yang H,Zhang C,lncRNA HHIP-AS1 promotes the osteogenic differentiation potential and inhibits the migration ability of periodontal ligament stem cells[J]. Stem Cells Int,2021,2021(1): 5595580. [18] Zhou H,Zhang L,Chen Y,et al. Research progress on the hedgehog signalling pathway in regulating bone formation and homeostasis[J]. Cell Prolif,2022,55(1):e13162. doi: 10.1111/cpr.13162 [19] Zeng X,Ju D. Hedgehog signaling pathway and autophagy in cancer[J]. Int J Mol Sci,2018,19(8):2279. doi: 10.3390/ijms19082279 -
下载: