Downregulated LncRNA-ANCR promotes osteoblast differentiation by targeting EZH2 and regulating Runx2 expression

https://doi.org/10.1016/j.bbrc.2013.02.036Get rights and content

Abstract

Long noncoding RNAs (lncRNAs) are key regulators of diverse biological processes such as transcriptional regulation, cell growth and differentiation. Previous studies have demonstrated that the lncRNA-ANCR (anti-differentiation ncRNA) is required to maintain the undifferentiated cell state within the epidermis. However, little is known about whether ANCR regulates osteoblast differentiation. In this study, we found that the ANCR expression level is significantly decreased during hFOB1.19 cell differentiation. ANCR-siRNA blocks the expression of endogenous ANCR, resulting in osteoblast differentiation, whereas ANCR overexpression is sufficient to inhibit osteoblast differentiation. We further demonstrated that ANCR is associated with enhancer of zeste homolog 2 (EZH2) and that this association results in the inhibition of both Runx2 expression and subsequent osteoblast differentiation. These data suggest that ANCR is an essential mediator of osteoblast differentiation, thus offering a new target for the development of therapeutic agents to treat bone diseases.

Highlights

► ANCR level is significantly decreased during osteoblastic differentiation. ► ANCR downregulation promotes osteoblastic differentiation. ► ANCR is associated with enhancer of zeste homolog 2 (EZH2). ► ANCR regulates Runx2 expression and osteoblast differentiation by EZH2.

Introduction

Mesenchymal stem cells (MSCs) have the potential to differentiate into multiple connective tissue cell types, including osteoblasts, myoblasts, adipocytes, and chondrocytes [1], [2]. The differentiation of MSCs into specific lineage depends on the extracellular cues received by these cells. Bone morphogenetic protein (BMP)-2, parathyroid hormone (PTH), and Runt-related transcription factor 2 (Runx2) plays an important roles in the regulation of osteoblast differentiation [2], [3], [4]. The alteration or malfunction of this regulation results in pathological consequences [5].

Long noncoding RNAs (lncRNAs) have been proposed to be a key regulators of diverse biological processes, such as transcriptional regulation, cell growth, and differentiation [6], [7]. Ng et al., demonstrated that lncRNAs promote neuronal differentiation by associating with chromatin modifiers and transcription factors [6]. Some hESC-specific lncRNAs involved in pluripotency maintenance were shown to physically interact with SOX2 and SUZ12 [6]. Sunwoo et al., found that MEN ε/β lncRNAs are upregulated during muscle differentiation and are essential components of paraspeckles [8]. The knockdown of MEN ε/β results in the disruption of nuclear paraspeckles. In the epidermis, lncRNA-ANCR (anti-differentiation ncRNA) loss abolishes the normal blockade of differentiation in the progenitor-containing compartment. Thus, ANCR is required to maintain the undifferentiated cell state within the epidermis. However, little is known about the role of ANCR in the osteoblast differentiation of hMSCs.

EZH2 is highly expressed in embryonic stem cells (ESCs) and is involved in the differentiation of these cells into different cell lineages, including osteoblasts, hepatocytes and neurons [9], [10]. EZH2 catalyzes histone H3 trimethylation at lysine 27 (H3K27me3) in target gene promoters to silence gene expression [11]. Wei et al., demonstrated that the CDK1-dependent phosphorylation of EZH2 disrupts methyltransferase activity of EZH2 and suppresses the methylation of H3K27, promoting the differentiation of MSCs into osteoblasts. Yu et al., found that EZH2 regulates the neuronal differentiation of MSCs through PIP5K1C-dependent calcium signaling [9]. EZH2 also regulates muscle gene expression and skeletal muscle differentiation [12]. Recent studies have demonstrated that some lncRNAs physically interact with EZH2 to regulate gene expression. lncRNA-HOTAIR targets Polycomb Repressive Complex 2 (which contains the H3K27 methylase EZH2) to silence the HOXD gene [13]. Therefore, we speculated that ANCR may regulate the osteoblast differentiation of ESCs by associating with EZH2.

Based on these findings, we investigated the molecular mechanism by which ANCR regulates osteoblast differentiation. We found that ANCR is an important regulator of osteoblast differentiation. ANCR associates with EZH2, and this association results in the inhibition of both Runx2 expression and subsequent osteoblast differentiation.

Section snippets

Cell culture

The human fetal osteoblastic cell line hFOB1.19 was obtained from the American Type Culture Collection (ATCC, Manassas, VA) and maintained at 33.5 °C in complete medium consisting of 1:1 Dulbecco’s modified Eagle’s medium (DMEM)/Ham’s F-12 medium supplemented with 10% FBS (Atlas Biologicals, Fort Collins, CO) and 0.3 mg/mL G418/geneticin (Calbiochem, Gibbstown, NJ). For osteoblastic differentiation, confluent cultures of hFOB1.19 cells were maintained in complete medium with the addition of the

The downregulation of ANCR promotes osteoblast differentiation

Previous studies have indicated that ANCR is required to maintain the undifferentiated cell state within the epidermis [16]. To investigate whether ANCR regulates osteoblast differentiation, we first assayed the ANCR levels during hFOB1.19 cell differentiation. We found that the ANCR levels were significantly decreased in hFOB1.19 cells cultured in osteoblastic differentiation medium (Fig. 1(A)). To investigate the biological role of ANCR in the regulation of hFOB1.19 cell differentiation,

Discussion

The human transcriptome has been shown to be more complex than simply a collection of protein-coding genes and their splice variants [18], [19]. Using whole-transcriptome and whole-genome sequencing technologies, it has been demonstrated that approximately 90% of the genome is transcribed [19]. Although the transcription of non-coding RNAs was initially argued to be spurious transcriptional noise, new evidence suggests that this transcriptional activity of the genome may play a major biological

Conclusion

Our data demonstrates that ANCR downregulation promotes osteoblast differentiation by regulating EZH2/Runx2. Thus, ANCR should be considered as an important candidate for a molecular target in osteoblastic differentiation and could be used in the development of therapeutic agents to treat osteogenic disorders.

References (26)

  • J. Huang et al.

    MicroRNA-204 regulates Runx2 protein expression and mesenchymal progenitor cell differentiation

    Stem Cells

    (2010)
  • S.Y. Ng et al.

    Human long non-coding RNAs promote pluripotency and neuronal differentiation by association with chromatin modifiers and transcription factors

    EMBO J.

    (2012)
  • H. Sunwoo et al.

    MEN epsilon/beta nuclear-retained non-coding RNAs are up-regulated upon muscle differentiation and are essential components of paraspeckles

    Genome Res.

    (2009)
  • Cited by (220)

    • Exosomes in bone remodeling and breast cancer bone metastasis

      2022, Progress in Biophysics and Molecular Biology
    • Epigenetic modifications of histones during osteoblast differentiation

      2022, Biochimica et Biophysica Acta - Gene Regulatory Mechanisms
    • Contribution of miRNAs and lncRNAs in osteogenesis and related disorders

      2021, Biomedicine and Pharmacotherapy
      Citation Excerpt :

      Notably, the canonical WNT signaling pathway has been induced by ANCR silencing in the course of proliferation and osteogenic induction [187]. Down-regulation of ANCR can also affect expressions of EZH2 and Runx2, thus promoting osteoblast differentiation [188]. In addition, MIR31HG/NF-κB axis has been found to improve the osteogenic capacity of human adipose-derived stem cells under an inflammatory microenvironment [189].

    View all citing articles on Scopus
    View full text