LncRNA MALAT1 promotes osteogenic differentiation through the miR‐217/AKT3 axis: a possible strategy to alleviate osteoporosis

Author(s):  
Chenyang Song ◽  
Yu Guo ◽  
Fenyong Chen ◽  
Wenge Liu
2021 ◽  
Author(s):  
Yuan Zhou ◽  
Zhuo Xu ◽  
Yuanyi Wang ◽  
Qiang Song ◽  
Ruofeng Yin

Abstract Background: Accumulating evidence demonstrates that long non-coding RNAs (lncRNAs) are associated with the development of osteoporosis. This study aimed to investigate the effects of metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) on osteogenic differentiation and cell apoptosis in osteoporosis. Methods: hindlimb unloading (HU) was performed to establish osteoporosis model in vivo. MicroCT was applied for pathological analysis. Microgravity (MG) was used to construct osteoporosis in vitro. The mRNA and miRNA expression was determined using RT-qPCR. Protein expression was determined using western blot. The binding sites between miR-485-5p and MALAT1/Wnt family member 7B (WNT7B) was predicted by bioinformatics analysis and verified by luciferase and RNA pull-down assays. Cellular functions were determined by ALP staining, Alizarin red staining, and flow cytometry assays. Results: MALAT1 expression was downregulated in HU mice and MG treated MC3T3-E1 cells. However, overexpression of MALAT1 upregulated the expression of Bmp4, Col1a1, Spp1, and enhanced ALP activity. Additionally, overexpression of MALAT1 inhibited apoptosis, decreased Bax and caspase-3 levels, and increased Bcl-2 level. Moreover, MALAT1 overexpression improved bone phenotype in vivo. MALAT1 functioned as a ceRNA to upregulate WNT7B. Overexpression of miR-485-5p rescued the promotion of osteogenic differentiation and the inhibition of apoptosis induced by MALAT1. Knockdown of WNT7B abolished the facilitation of osteogenic differentiation and the suppression of apoptosis induced by downregulation of miR-485-5p. Conclusion: In conclusion, MALAT1 promoted osteogenic differentiation and inhibited cell apoptosis through miR-485-5p/WNT7B axis, which suggested that MALAT1 is a potential target to alleviate osteoporosis.


Author(s):  
Zewu Zhu ◽  
Fang Huang ◽  
Weiping Xia ◽  
Huimin Zeng ◽  
Meng Gao ◽  
...  

BackgroundThe current belief is that Randall’s plaques (RP) constitute a nidus for the formation of idiopathic calcium oxalate stones, but the upstream events in RP formation remain unclear. The present study aimed to investigate whether RP formation shares similarities with biomineralization and to illustrate the potential role played by the lncRNA MALAT1 in osteogenic differentiation of human renal interstitial fibroblasts (hRIFs).Materials and MethodsBiomineralization and MALAT1 expression were assessed in RP, and hRIFs were isolated and induced under osteogenic conditions for further experiments. The transcription initiation and termination sites in MALAT1 were identified by 5′ and 3′ RACE. RNA immunoprecipitation assays and luciferase assays were used to validate the interactions among MALAT1, Runx2 and miRNAs.ResultsUpregulated expression of osteogenic markers and MALAT1 was observed in RP and hRIFs induced with osteogenic medium. Biomineralization in RP and calcium phosphate (CaP) deposits in induced hRIFs were further verified by electron microscopy. Furthermore, overexpression of MALAT1 promoted the osteogenic phenotype of hRIFs, while treatment with a miR-320a-5p mimic and knockdown of Runx2 significantly suppressed the osteogenic phenotype. Further analysis showed that MALAT1 functioned as a competing endogenous RNA to sponge miR-320a-5p, leading to upregulation of Runx2 and thus promoting osteogenic differentiation of hRIFs.ConclusionEctopic calcification and MALAT1 partially contributed to the formation of RP, in which MALAT1 might promote Runx2 expression to regulate osteogenic differentiation of hRIFs by sponging miRNA-320a-5p. The current study sheds new light on the lncRNA-directed mechanism of RP formation via a process driven by osteogenic-like cells.


2020 ◽  
Vol 10 (5) ◽  
pp. 730-736
Author(s):  
Qingyun Pan ◽  
Biao Dong ◽  
Yong He ◽  
Xiaohui Wang

Osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) is controlled by elaborate genetic programs. lncRNA MALAT1 plays an important role in many diseases. However, the role of lncRNA MALAT1 in BMSCs remains unclear. Isolated Rat BMSCs were cultured and randomly divided into control group, MALAT1 group and MALAT1 siRNA group, in which lncRNA MALAT1 plasmid and lncRNA MALAT1 siRNA were transfected into BMSCs followed by analysis of lncRNA MALAT1 expression by real time PCR, cell proliferation by MTT assay, Caspase 3 activity, ALP activity was analyzed, calcified nodules by alizarin red staining, expression of Smad1 and Smad7 by Western blot as well as Runx2 expression by real time PCR. In MALAT1 group, MALAT1 expression was significantly increased along with significantly inhibited cell proliferation, increased Caspase 3 activity, decreased ALP activity and calcified nodules, reduced expression of Smad1, Smad7 and Runx2 compared with control group (P < 0 05). MALAT1 expression in MALAT1 siRNA group was decreased with significantly promoted cell proliferation, decreased Caspase 3 activity, increased ALP activity and calcified nodules, as well as significantly elevated expression of Smad1, Smad7 and Runx2 compared with control group (P < 0 05). Up-regulation of lncRNA MALAT1 expression inhibits the Smad/Runx2 signaling pathway, thereby inhibiting BMSCs proliferation and osteogenesis. Down-regulation of lncRNA MALAT1 expression promotes Smad/Runx2 signaling pathway activation, inhibits BMSCs apoptosis, promotes proliferation and osteogenic differentiation.


2019 ◽  
Vol 9 (11) ◽  
pp. 1520-1527
Author(s):  
Xiaoliang Li ◽  
Guofeng Xia ◽  
Hongmei Xin ◽  
Chunsheng Tao ◽  
Weiwei Lai ◽  
...  

ncRNA involves in osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). WNT5A participates in the growth and development of osteogenic differentiation. This study aims to investigate whether lncRNA MALAT1 regulates BMSCs osteogenesis through WNT5A. qRT-PCR was done to detect the lncRNA MALAT1 level and osteogenic markers in osteoporosis patients and control groups. The above markers and WNT5A protein levels were detected by Western blot. The degree of osteogenic differentiation was detected by ALP activity assay and ALP staining. The differentiation ability of BMSCs after lncRNA MALAT1 overexpression was detected by ARS staining. The binding site of lncRNA MALAT1 to WNT5A was determined by dual luciferase reporter assay. lncRNA MALAT1 expression was higher in patients with osteoporosis, and decreased significantly with increased osteogenic induction. Overexpression of lncRNA MALAT1 in BMSCS reduced WNT5A level, while interference with lncRNA MALAT1 increased WNT5A levels. In cells with overexpression of lncRNA MALAT1, transfection of si-WNT5A can significantly downregulate the RUNX2, OSX, ALP, OCN, OPN, and COL1A1, thereby inhibiting osteogenic differentiation, interfering with the regulation of WNT signaling pathway and regulating BMSCs osteogenic differentiation. lncRNA MALAT1 and WNT5A can regulate BMSCs osteogenesis, thus accelerating the progression of osteoporosis.


Sign in / Sign up

Export Citation Format

Share Document