bims-mirbon Biomed News
on MicroRNAs in bone
Issue of 2021‒11‒14
thirteen papers selected by
Japneet Kaur
Mayo Clinic


  1. J Orthop Surg Res. 2021 Nov 12. 16(1): 665
      BACKGROUND: Osteoporosis is a systemic disease characterized by impaired bone formation, increased bone resorption, and brittle bone fractures. The osteogenic differentiation of human bone marrow-derived mesenchymal stem cells (hBMSCs) is considered to be a vital process for bone formation. Numerous studies have reported that long non-coding RNAs (lncRNAs) are involved in the osteogenic differentiation of hBMSCs. The present study aimed to investigate the effect of FGD5 antisense RNA 1 (FGD5-AS1) on osteogenic differentiation.METHODS: RT-qPCR was performed to detect the expression of FGD5-AS1, miR-506-3p, and osteogenesis-related genes OCN, OPN, OSX, and RUNX2. Western blotting was carried out to detect the protein levels of osteogenesis-related markers. In addition, the regulatory effect of FGD5-AS1 on osteogenic differentiation was detected through alkaline phosphatase (ALP) activity, Alizarin Red S (ARS) staining, and Cell Counting Kit-8 (CCK-8). Bioinformatics analysis and luciferase reporter assay were used to predict and validate the interaction between FGD5-AS1 and miR-506-3p as well as miR-506-3p and bone morphogenetic protein 7 (BMP7).
    RESULTS: The RT-qPCR analysis revealed that FGD5-AS1 was upregulated in hBMSCs following induction of osteogenic differentiation. In addition, FGD5-AS1 knockdown attenuated hBMSC viability and osteogenic differentiation. Bioinformatics analysis and luciferase reporter assays verified that FGD5-AS1 could directly interact with microRNA (miR)-506-3p. Furthermore, miR-506-3p could directly target the 3'-untranslated region (3'-UTR) of BMP7. Additionally, functional assays demonstrated that miR-506-3p silencing could restore the suppressive effect of FGD5-AS1 knockdown on osteogenic differentiation and viability of hBMSCs, and miR-506-3p could attenuate osteogenic differentiation via targeting BMP7.
    CONCLUSIONS: Taken together, the results of the present study suggested that FGD5-AS1 could positively regulate the osteogenic differentiation of hBMSCs via targeting the miR-506-3p/BMP7 axis.
    Keywords:  BMP7; BMSC; FGD5-AS1; Osteoporosis; miR-506-3p
    DOI:  https://doi.org/10.1186/s13018-021-02694-x
  2. Front Cell Dev Biol. 2021 ;9 723759
      Long non-coding RNAs (lncRNAs) play pivotal roles in mesenchymal stem cell differentiation. However, the mechanisms by which non-coding RNA (ncRNA) networks regulate osteogenic differentiation remain unclear. Therefore, our aim was to identify RNA-associated gene and transcript expression profiles during osteogenesis in bone marrow mesenchymal stem cells (BMSCs). Using transcriptome sequencing for differentially expressed ncRNAs and mRNAs between days 0 and 21 of osteogenic differentiation of BMSCs, we found that the microRNA (miRNA) miR-503-5p was significantly downregulated. However, the putative miR-503-5p target, sorbin and SH3 domain containing 1 (SORBS1), was significantly upregulated in osteogenesis. Moreover, through lncRNA-miRNA-mRNA interaction analyses and loss- and gain-of-function experiments, we discovered that the lncRNAs LOC100126784 and POM121L9P were abundant in the cytoplasm and enhanced BMSC osteogenesis by promoting SORBS1 expression. In contrast, miR-503-5p reversed this effect. Ago2 RNA-binding protein immunoprecipitation and dual-luciferase reporter assays further validated the direct binding of miR-503-5p to LOC100126784 and POM121L9P. Furthermore, SORBS1 knockdown suppressed early osteogenic differentiation in BMSCs, and co-transfection with SORBS1 small interfering RNAs counteracted the BMSCs' osteogenic capacity promoted by LOC100126784- and POM121L9P-overexpressing lentivirus plasmids. Thus, the present study demonstrated that the lncRNAs LOC100126784 and POM121L9P facilitate the osteogenic differentiation of BMSCs via the miR-503-5p/SORBS1 axis, providing potential therapeutic targets for treating osteoporosis and bone defects.
    Keywords:  LOC100126784; POM121L9P; bone marrow mesenchymal stem cell; miR-503-5p; osteogenesis; sorbin and SH3 domain containing-1 gene
    DOI:  https://doi.org/10.3389/fcell.2021.723759
  3. J Bone Miner Metab. 2021 Nov 09.
      INTRODUCTION: Diabetic osteoporosis (DOP) is a chronic diabetic complication, which is attributed to high glucose (HG)-induced dysfunction of bone marrow mesenchymal stem cells (BMSCs). Studies have revealed that microRNAs (miRNAs) play critical roles in osteogenic differentiation of BMSCs in DOP. Here, the role of miR-9-5p in DOP progression was explored.MATERIALS AND METHODS: The rat model of DOP was established by intraperitoneal injection of streptozotocin (STZ). BMSCs were treated with high glucose (HG) to establish in vitro models. Gene expression in BMSCs and bone tissues of rats was tested by RT-qPCR. The degree of osteogenic differentiation of BMSCs was examined by Alizarin Red staining and ALP activity analysis. The protein levels of collagen-I (COL1), osteocalcin (OCN), osteopontin (OPN), runt-related transcription factor-2 (RUNX2), and DEAD-Box Helicase 17 (DDX17) in BMSCs were evaluated by western blotting. The interaction between miR-9-5p and DDX17 was identified by luciferase reporter assay. H&E staining was used to test morphological structure of femurs of rats with STZ treatment.
    RESULTS: MiR-9-5p was overexpressed in HG-treated BMSCs, while DDX17 was downregulated. Functionally, miR-9-5p knockdown promoted BMSCs osteogenic differentiation under HG condition. Mechanically, miR-9-5p targeted DDX17. DDX17 knockdown reversed the effect of miR-9-5p silencing on osteogenic differentiation of HG-treated BMSCs. In in vivo studies, miR-9-5p downregulation ameliorated the DOP condition of rats and miR-9-5p expression was negatively correlated with DDX17 expression in bone tissues of rats with STZ treatment.
    CONCLUSION: MiR-9-5p knockdown promotes HG-induced osteogenic differentiation BMSCs in vitro and mitigates the DOP condition of rats in vivo by targeting DDX17.
    Keywords:  BMSCs; DDX17; Diabetic osteoporosis; miR-9-5p
    DOI:  https://doi.org/10.1007/s00774-021-01280-9
  4. Regen Ther. 2021 Dec;18 430-440
      Background: Circular RNAs (circRNAs) are non-coding RNAs that play a pivotal role in bone diseases. RUNX3 was an essential transcriptional regulator during osteogenesis. However, it is unknown whether RUNX3 regulates hsa_circ_0005752 during osteogenic differentiation.Methods: The levels of hsa_circ_0005752 and RUNX3 were measured by qRT-PCR after osteogenic differentiation of ADSCs. The osteogenic differentiation was analyzed by Alkaline phosphatase (ALP) staining and Alizarin red staining (ARS). qRT-PCR and western blot were used to assess the expressions of osteogenic differentiation-related molecules. RNA pull-down, RIP, and luciferase reporter assays determine the interactions between miR-496 and hsa_circ_0005752 or MDM2 mRNA. CHIP-PCR analyzed the interaction between RUNX3 and LPAR1. Finally, the potential roles of RUNX3 were investigated during osteogenic differentiation with or without hsa_circ_0005752 knockdown.
    Results: Hsa_circ_0005752 and RUNX3 were significantly increased, and miR-496 was remarkably decreased in ADSCs after osteogenic differentiation. Hsa_circ_0005752 could promote osteogenic differentiation, as shown by enhancing ALP and ARS staining intensity. Hsa_circ_0005752 enhanced the expressions of Runx2, ALP, Osx, and OCN. Furthermore, hsa_circ_0005752 directly targeted miR-496, which can directly bind to MDM2. RUNX3 bound to the LPAR1 promoter and enhanced hsa_circ_0005752 expressions. Moreover, the enhanced expression of hsa_circ_0005752 by RUNX3 could promote osteogenic differentiation, whereas knockdown of hsa_circ_0005752 partially antagonized the effects of RUNX3.
    Conclusion: Our study demonstrated that RUNX3 promoted osteogenic differentiation via regulating the hsa_circ_0005752/miR-496/MDM2 axis and thus provided a new therapeutic strategy for osteoporosis.
    Keywords:  3′ UTR, 3′ untranslated region; ADSCs, adipose-derived stem cells; ALP, alkaline phosphatase; ARS, Alizarin Red Staining; Adipose-derived stem cells; BCA, bicinchoninic acid; BM-MSCs, Bone Marrow-Mesenchymal Stem Cells; BMP2, Bone morphogenetic protein 2; ChIP, chromatin immunoprecipitation; Circular RNAs; ECL, enhanced chemiluminescence; H&E staining, Hematoxylin and Eosin staining; LPAR1, lysophosphatidic acid receptor 1; MDM2; MDM2, murine double minute 2; OCN, osteocalcin; OM, osteogenic (differentiation) medium; Osteogenic differentiation; Osx, osterix; PMSF, phenylmethylsulfonyl fluoride; RIP, RNA immunoprecipitation; RUNX3; Runx2, Runt-related transcription factor 2; Runx3, RUNX Family Transcription Factor 3; SDS-PAGE, polyacrylamide gel electrophoresis; UC-MSCs, Umbilical Cord-Mesenchymal Stem Cells; circRNAs, Circular RNAs; miRNAs, microRNA; microRNA; qRT-PCR, quantitative real-time polymerase chain reaction
    DOI:  https://doi.org/10.1016/j.reth.2021.09.006
  5. Mediators Inflamm. 2021 ;2021 9972805
      Background: In the past decade, mesenchymal stem cells (MSCs) have been widely used for the treatment of osteoarthritis (OA), and noncoding RNAs in exosomes may play a major role.Aim: The present study is aimed at exploring the effect and mechanism of miR-326 in exosomes secreted by bone marrow mesenchymal stem cells (BMSCs) on pyroptosis of cartilage and OA improvement.
    Methods: Exosomes from BMSCs (BMSC-Exos) were isolated and identified to incubate with OA chondrocytes. Proliferation, migration, specific gene and miR-326 expression, and pyroptosis of chondrocytes were detected. BMSCs or chondrocytes were transfected with miR-326 mimics or inhibitors to investigate the effect of miR-326 in BMSC-Exos on pyroptosis of chondrocytes and the potential mechanism. Finally, a rat OA model was established to verify the effect and mechanism of miR-326 in BMSC-Exos on cartilage of pyroptosis.
    Results: Incubation with BMSC-Exos could significantly improve the survival rate, migration ability, and chondrocyte-specific genes (COL2A1, SOX9, Agg, and Prg4) and miR-326 expression of OA chondrocytes and significantly inhibit pyroptosis of chondrocytes by downregulation of the levels of inflammatory cytokines, Caspase-1 activity, and pyroptosis-related proteins such as GSDMD, NLRP3, ASC, IL-1β, and IL-18 (P < 0.01). PKH26 labeling confirmed the uptake of BMSC-Exos by chondrocytes. Incubation with exosomes extracted from BMSCs overexpressing miR-326 can significantly repress the pyroptosis of chondrocytes, while knockdown of miR-326 had the opposite effect (P < 0.01). The same result was also demonstrated by direct interference with the expression level of miR-326 in chondrocytes (P < 0.01). In addition, we found that the overexpression of miR-326 significantly inhibited the expression of HDAC3 and NF-κB p65 and significantly promoted the expression of STAT1, acetylated STAT1, and acetylated NF-κB p65 in chondrocytes (P < 0.01). The targeted relationship between miR-326 and HDAC3 was verified by dual-luciferase reporter assay. Animal experiments confirmed the mechanism by which miR-326 delivered by BMSC-Exos inhibits pyroptosis of cartilage by targeting HDAC3 and STAT1/NF-κB p65 signaling pathway.
    Conclusion: BMSC-Exos can deliver miR-326 to chondrocytes and cartilage and improve OA by targeting HDAC3 and STAT1//NF-κB p65 to inhibit pyroptosis of chondrocytes and cartilage. Our findings provide a new mechanism for BMSC-Exos to treat OA.
    DOI:  https://doi.org/10.1155/2021/9972805
  6. J BUON. 2021 Sep-Oct;26(5):26(5): 1832-1841
      PURPOSE: Recent studies have manifested that bone marrow mesenchymal stem cells (BMSCs) derived exosomes affect the progression of tumors through carrying endogenous molecules. To explore the role of BMSCs-derived exosomes carrying abundant micro ribonucleic acid (miR)-126-3p in non-small-cell lung cancer (NSCLC).METHODS: Firstly, A549 cells were transfected with miR-126-3p to detect the role of miR-126-3p in A549 cells. Next, miR-126-3p was transfected into BMSCs, and BMSCs-derived exosomes with over-expressed miR-126-3p (Exo-miR-126-3p) were isolated through ultracentrifugation. After that, A549 cells were co-incubated with Exo-miR-126-3p to determine the effects of Exo-miR-126-3p on cell proliferation, migration, invasion, and apoptosis. Besides, the targeted relationship between miR-126-3p and protein tyrosine phosphatase non-receptor type 9 (PTPN9) was confirmed via bioinformatics analysis and dual-luciferase reporter gene analysis. Western blotting (WB) was employed to measure the expressions of PTPN9 and related proteins in A549 cells. Additionally, the effects of over-expressed miR-126-3p derived from BMSCs exosomes on tumor growth and apoptosis of NSCLC cells were analyzed in connection with lentiviral packaged miR-126-3p in vivo.
    RESULTS: Overexpressed miR-126-3p suppressed the viability, migration, and invasion of A549 cells in vitro. Based on the results of exosome content analysis, miR-126-3p could mediate the inhibitory effect of exosomes on A549 cells by negative regulation of PTPN9. Notably, over-expressed miR-126-3p derived from BMSCs inhibited the tumor growth and apoptosis in vivo.
    CONCLUSIONS: Taken together, the key finding of the study indicated that over-expressed BMSCs-derived exosomal miR-126-3p can suppress the progression of NSCLC through negatively regulating PTPN9.
  7. Biomaterials. 2021 Nov 05. pii: S0142-9612(21)00599-8. [Epub ahead of print]279 121242
      Osteolysis at the tendon-bone interface can impair pullout strength during tendon-bone healing and lead to surgery failure, but the effects of clinical treatments are not satisfactory. Mesenchymal stem cell (MSC)-derived exosomes have been used as potent and feasible natural nanocarriers for drug delivery and have been proven to enhance tendon-bone healing strength, indicating that MSC-derived exosomes could be a promising therapeutic strategy. In this study, we explored Scleraxis (Scx) dynamically expressed in PDGFRα(+) bone marrow-derived mesenchymal stem cells (BMMSCs) during natural tendon-bone healing. Then, we investigated the role of PDGFRα(+) BMMSCs in tendon-bone healing after Scx overexpression as well as the underlying mechanisms. Our data demonstrated that Scx-overexpressing PDGFRα(+) BMMSCs (BMMSCScx) could efficiently inhibit peritunnel osteolysis and enhance tendon-bone healing strength by preventing osteoclastogenesis in an exosomes-dependent manner. Exosomal RNA-seq revealed that the abundance of a novel miRNA, miR-6924-5p, was highest among miRNAs. miR-6924-5p could directly inhibit osteoclast formation by binding to the 3'-untranslated regions (3'UTRs) of OCSTAMP and CXCL12. Inhibition of miR-6924-5p expression reversed the prevention of osteoclastogenic differentiation by BMMSCScx derived exosomes (BMMSCScx-exos). Local injection of BMMSCScx-exos or miR-6924-5p dramatically reduced osteoclast formation and improved tendon-bone healing strength. Furthermore, delivery of miR-6924-5p efficiently inhibited the osteoclastogenesis of human monocytes. In brief, our study demonstrates that BMMSCScx-exos or miR-6924-5p could serve as a potential therapy for the treatment of osteolysis during tendon-bone healing and improve the outcome.
    Keywords:  Exosomes; Osteoclastogenesis; PDGFRα(+) BMMSCs; Scx; Tendon-bone healing; miR-6924–5p
    DOI:  https://doi.org/10.1016/j.biomaterials.2021.121242
  8. Exp Ther Med. 2021 Dec;22(6): 1478
      The present study aimed to investigate the expression and clinical significance of miR-519d-3p in patients with post-traumatic osteoarthritis (PTOA). The levels of miR-519d-3p in the synovium and synovial fluid (SF) of all subjects were detected by reverse transcription-quantitative polymerase chain reaction. The results of the present study demonstrated that the levels of miR-519d-3p in the synovium and SF of patients with PTOA were significantly lower, but that the VEGF content was significantly higher, compared with that of control group. Dual-luciferase reporter and Western blot assays demonstrated that VEGF was a target gene of miR-519d-3p. Furthermore, miR-519d-3p inhibitor-induced cell apoptosis, and cell cycle arrest could be partially reversed by silencing VEGF. Additionally, the level of miR-519d-3p in the synovium and SF of patients with PTOA was negatively correlated with the level of VEGF. ROC analysis demonstrated that miR-519d-3p levels in the synovium and SF could effectively differentiate patients with PTOA from healthy controls, with areas under the ROC curve of 0.928 and 0.896, respectively. In conclusion, reduction of miR-519d-3p in the synovium and SF resulted in the upregulation of VEGF in patients with PTOA, and miR-519d-3p may be a potential therapeutic target of PTOA.
    Keywords:  miR-519d-3p; post-traumatic osteoarthritis; synovial fluid; synovium; vascular endothelial growth factor
    DOI:  https://doi.org/10.3892/etm.2021.10913
  9. Biochimie. 2021 Nov 03. pii: S0300-9084(21)00255-8. [Epub ahead of print]
      Osteoblast differentiation is an important process in skeletal development and bone remodelling. Serious bone diseases occur from any delay, defect, or imbalance in osteoblastic differentiation. Non-coding RNAs (ncRNAs) play a regulatory role in controlling the expression of proteins under physiological and pathological conditions via inhibiting mRNA translation or degrading mRNA. Circular RNAs (circRNAs) and microRNAs (miRNAs) are the long and small ncRNAs, respectively, which have been reported to regulate the expression of osteoblast marker genes directly or indirectly. Also, recent studies identified the regulatory mechanisms involving the crosstalk among circRNAs, miRNAs, and mRNAs during osteoblast differentiation. Understanding these regulatory mechanisms behind osteoblastic differentiation would help to diagnose or treat bone and bone-related disorders. Hence, the current review comprehensively discussed the regulatory relationship of circRNAs, miRNAs and mRNAs, and their functional role as circRNA-miRNA-mRNA axis in osteoblast differentiation.
    Keywords:  Osteoblast; Runx2; ncRNAs
    DOI:  https://doi.org/10.1016/j.biochi.2021.11.001
  10. Bioengineered. 2021 Nov 09.
      MicroRNAs (miRNAs) are powerful modulators of fracture healing. The research explored the level of serum miR-223-3p in fracture patients and its potential mechanism in fracture healing. In the study, miR-223-3p levels in 42 patients with intra-articular fracture and 40 patients with hand fracture were detected by real-time fluorescence quantitative PCR reaction (qRT-PCR). Subsequently, osteoblasts MC3T3-E1 was transfected with miR-223-3p mimic or inhibitor, and cell function was detected by Cell counting kit (CCK-8) assay and flow cytometry. Dual-luciferase reporter assay verified the regulation mechanism of miR-223-3p and its target genes. We found that miR-223-3p was significantly elevated over time in patients with intra-articular fracture and hand fracture patients compared with healthy individuals. Moreover, increased miR-223-3p significantly reduced cell viability and promoted cell apoptosis. The fibroblast growth factor receptor 2 (FGFR2) was the target of miR-223-3p. Serum FGFR2 was significantly decreased in patients, which was contrary to the expression of miR-223-3p. Moreover, FGFR2 levels in cells were negatively regulated by miR-223-3p. Finally, si-FGFR2 significantly reversed the promotion of miR-223-3p inhibitor on cell viability and the inhibition of cell apoptosis. Our research suggested that miR-223-3p is highly expressed in fracture patients, and regulates osteoblast cell viability and apoptosis by targeting FGFR2. This may be a valuable target for fracture healing therapy and provide a new perspective for its treatment.
    Keywords:  FGFR2; fracture healing; miR-223-3p
    DOI:  https://doi.org/10.1080/21655979.2021.2002498
  11. Immun Inflamm Dis. 2021 Nov 11.
      BACKGROUND: Dysregulation of BZRAP1-AS1 was associated with immune statuses of cancer or Alzheimer's disease patients, yet little is known about its role in rheumatoid arthritis.METHODS: RT-qPCR and western blot were applied to assess the expression of indicated expression. CCK-8 and BrdU proliferation assays were used to measure the proliferation of RA-HFLS. Apoptosis in RA-HFLS was evidenced by the alteration of caspase-3 activity and apoptosis-related factors. ELISA was performed to detect IL-6, IL-1β, and TNF-α level. Luciferase reporter, RIP, and pull-down assays were used to confirm the BZRAP1-AS1/miR-1286/COL5A2 cascade predicted by bioinformatics analysis.
    RESULTS: BZRAP1-AS1 and COL5A2 were downregulated in RA tissues and RA-HFLS while miR-1286 was amplified. Overexpression of BZRAP1-AS1 reduced the RA-HFLS proliferation, IL-6, IL-1β, and TNF-α level and induced cell apoptosis while BZRAP1-AS1 silence produced an opposite effect. Overexpression of BZRAP1-AS1 reduced the miR-1286 expression which in turn increased the COL5A2 expression, thereby relieving the excessive proliferation and limited apoptosis in RA-HFLS.
    CONCLUSION: Our findings suggested that BZRAP1-AS1 sequestered miR-1286 and reshaped the COL5A2 expression, thereby suppressed RA-HFLS proliferation and inflammation, and triggered cell apoptosis, resulting in the attenuation of RA progression.
    Keywords:  apoptosis; inflammation; lncRNA; miRNA; proliferation; rheumatoid arthritis
    DOI:  https://doi.org/10.1002/iid3.558
  12. J Orthop Surg Res. 2021 Nov 06. 16(1): 658
      BACKGROUND: Chondrogenic differentiation of human adipose-derived stem cells (hADSCs) is important for cartilage generation and degradation. LncRNAs play an essential role in stem cell differentiation. However, the role and mechanism of lncRNA in hADSCs remain unclear. Our previous study showed that miR-490-5p was downregulated during chondrogenic differentiation of hADSCs. In this study, we investigated the effect and mechanism of lncRNA NONHSAT030515 interacting with miR-490-5p on chondrogenic differentiation of hADSCs.METHODS: Alcian blue staining was used to assess the deposition of chondromatrix proteins following chondrogenic differentiation of human adipose stem cells. Immunohistochemistry was used to evaluate the expression of collagenII. TargetScan, miRTarBase and miRDB database analyses were applied to find the miRNA and target genes of lncRNA NONHSAT030515. A dual luciferase experiment was conducted to identify the direct target of NONHSAT030515. pcDNA3.1- NONHSAT030515 transfection and sh- NONHSAT030515 treatment were conducted to verify the role of lncRNA NONHSAT030515 in chondrogenic differentiation. The levels of Aggrecan, SOX9 and COL2A1 were analyzed by qRT-PCR and Western blot assay.
    RESULTS: Alcian blue staining, immunocytochemical, qRT-PCR, and Western blot have determined that lncRNA NONHSAT030515 can promote the chondrogenic differentiation of hADSCs. MiR-490- 5p was the direct target of NONHSAT030515, while BMPR2 was the target gene. This result was confirmed by luciferase reporter assay. Up-regulation of NONHSAT030515 promoted BMPR2 protein expression and promoted chondrogenic differentiation, whereas down-regulation of NONHSAT030515 caused completely opposite results.
    CONCLUSION: LncRNA NONHSAT030515 promotes the chondrogenic differentiation of hADSCs through increasing BMPR2 expression by regulating miR-490- 5p.
    Keywords:  Chondrogenic differentiation; Human adipose-derived stem cells; lncRNA NONHSAT030515
    DOI:  https://doi.org/10.1186/s13018-021-02757-z
  13. Pathobiology. 2021 Nov 09. 1-11
      BACKGROUND: Osteoarthritis (OA) is a common type of degenerative joint diseases that is regulated by a combination of complex intercellular signals and modulators, including non-coding RNAs. Mounting evidence suggests that miR-31-5p is physiologically involved in the regulation of chondrocytes, but the mechanism remains unclear.METHODS: Expression levels of miR-31-5p and SOX4 in OA cartilage tissues and in IL-1β-stimulated chondrocytes were examined by quantification polymerase chain reaction (q-PCR) or immunohistochemistry assays. Cell proliferation and apoptosis were detected by Cell Counting Kit-8 (CCK-8) and flow cytometry assays, respectively. Expression of LC3 was detected using immunofluorescence staining. Expressions of autophagy-related proteins and extracellular regulated protein kinase (ERK)/mechanical target of rapamycin kinase (mTORC1) signal-related proteins were measured by Western blot analysis. Molecular interaction was validated by dual luciferase reporter assay.
    RESULTS: Downregulation of miR-31-5p and upregulation of SOX4 were observed in both OA patients and OA chondrocytes. Mechanistic experiments revealed that miR-31-5p negatively modulated SOX4 expression by directly targeting its 3'- untranslated region. Moreover, overexpression of miR-31-5p suppressed the activation of mTORC1 in an ERK-dependent manner by inhibiting SOX4. Further functional experiments demonstrated that overexpressing miR-31-5p in OA chondrocytes markedly promoted its proliferation and autophagy while inhibiting apoptosis. However, these effects were abolished by overexpression of SOX4 or treatment with 3BDO, an mTOR activator.
    CONCLUSION: These results demonstrated that miR-31-5p enhanced survival and autophagy of OA chondrocytes through inactivation of mTORC1 via directly targeting SOX4, suggesting that miR-31-5p may play a protective role in OA progression.
    Keywords:  Apoptosis; Autophagy; Chondrocytes; SOX4; miR-31-5p
    DOI:  https://doi.org/10.1159/000519006