TRIM72, a novel negative feedback regulator of myogenesis, is transcriptionally activated by the synergism of MyoD (or myogenin) and MEF2

https://doi.org/10.1016/j.bbrc.2010.04.072Get rights and content

Abstract

TRIM72 is known to be involved in the negative feedback regulation of myogenesis by targeting insulin receptor substrate-1. Here, we found that TRIM72 was more highly expressed in oxidative muscle with the higher activity of MEF2, compared to glycolytic muscle. Indeed, TRIM72 promoter contained an evolutionarily conserved MEF2 site juxtaposed to E-box. TRIM72 promoter activity was decreased by the site-directed mutagenesis of either E-boxes or a MEF2 site and synergistically enhanced by MyoD (or myogenin) and MEF2, which were associated with proximal E-box, and MEF2 site of the TRIM72 promoter, respectively. Taken together all these data, we concluded that the synergism of MyoD (or myogenin) and MEF2 is necessary for TRIM72 expression during C2C12 differentiation.

Introduction

Skeletal muscle fibers are classified as slow oxidative (type I), fast oxidative (type IIa and type IIx), and fast glycolytic fibers (type IIb) on the basis of their contraction speed, fatigue resistance, and myosin heavy chain (MHC) isoform expression. Gastrocnemius muscle, which is largely composed of type IIb fibers, is used during intense and rapid activities and has a high speed of contraction. Soleus muscle with a high proportion of types I and IIa fibers is used for postural tension and has a high fatigue resistance [1]. Exercise training or motor neuron activity remodels muscle fiber types in adult skeletal muscle [1], [2], [3], [4], [5].

Myocyte enhancer factor 2 (MEF2) transcriptionally activates muscle-specific genes with an A/T-rich consensus sequence (C/TTA(A/T)4TAG/A) called a MEF2 site [6]. MEF2 responds to calcium signaling pathways involved in muscle type remodeling. The number of oxidative fibers is reduced in the soleus of MEF2C- or MEF2D-disrupted mice [7], and exercise training increases β-galactosidase activity in the oxidative fibers of transgenic mice carrying the lacZ gene with MEF2 sites [8], [9], indicating that MEF2 regulates muscle type remodeling. The transcriptional activity of MEF2 is enhanced by Ca2+-induced calcineurin activation but inhibited by class II histone deacetylases (HDACs) [8], [10]. Calcium/calmodulin-dependent kinase (CaMK) and protein kinase D1 induce the phosphorylation and cytoplasmic translocation of HDACs, dissociating MEF2 from HDACs in the nucleus [11], [12], [13], [14].

TRIM72, with a RING finger domain, a B-box, two coiled-coil domains, and a SPRY domain is specifically expressed in the plasma membrane of skeletal and cardiac muscle cells [15], [16]. In addition, the expression level of TRIM72 gradually increases during C2C12 myogenesis. TRIM72 has been known to regulate skeletal muscle differentiation since its adenoviral overexpression prevents, but its knock-down enhances the myogenesis of C2C12 cells and human primary skeletal muscle cells [16]. Since MyoD-dependent TRIM72 promoter activity is regulated by the PI(3)K-Akt pathway, TRIM72 is highly expressed in the plasma membrane of C2C12 myotubes during IGF-mediated myogenesis. The accumulated TRIM72 in the plasma membrane interacts with insulin receptor substrate-1 (IRS-1) and inhibits the IGF-induced activation of IRS-1, blocking excess myogenesis [16]. Thus, TRIM72 might be a negative feedback regulator of myogenesis for regulating muscle fiber size.

In order to understand molecular mechanism of TRIM72 expression during skeletal muscle differentiation, we analyzed the transcriptional regulation of TRIM72, of which promoter contains a MEF site as well as five E-boxes. Here, we demonstrated that TRIM72 transcription was synergistically activated by MyoD (or myogenin), and MEF2 during C2C12 skeletal muscle differentiation, thereby explaining why soleus oxidative muscle with higher activity of MEF2 has a higher expression level of TRIM72 than gastrocnemius glycolytic muscle does.

Section snippets

Antibodies and siRNAs

Rabbit anti-TRIM72 polyclonal antibody was generated from AbFrontier. Anti-MyoD (sc-760X), myogenin (sc-576X), and MEF2 (sc-313X) antibodies used for ChIP were purchased from Santa Cruz biotechnology, and anti-MyoD (554130) and myogenin (556358) antibodies used for EMSA from BD Transduction laboratories. Small interference RNA oligonucleotides for control (si-Con) and myogenin (si-Mgn) were purchased from Santa Cruz Biotechnology.

Animals and Western blotting

C57BL/6 mice were housed in plastic cages on a 12:12 h light–dark

TRIM72 promoter contains an evolutionarily conserved MEF2 site

Oxidative fibers have smaller diameters than glycolytic fibers do and therefore the diffusion distance for oxygen to travel to the mitochondria is reduced. Since TRIM72 is proposed to be a negative size regulator in skeletal muscle [16], it is tempting to speculate that TRIM72 might be highly expressed in oxidative fibers. In order to address the issue, TRIM72 expression levels were determined in mouse soleus, tibialis anterior (TA), and gastrocnemius muscles by Western blotting. Among the

Acknowledgments

This work was supported by grants to Y.-G. Ko from Korea Research Foundation (KRF-2007-313-C00582).

References (23)

  • M.J. Potthoff et al.

    Histone deacetylase degradation and MEF2 activation promote the formation of slow-twitch myofibers

    J. Clin. Invest.

    (2007)
  • Cited by (34)

    • Proteomics discovery of protein biomarkers linked to meat quality traits in post-mortem muscles: Current trends and future prospects: A review

      2020, Trends in Food Science and Technology
      Citation Excerpt :

      TRIM72 eliminates the harmful agents during the apoptotic process and its levels are reduced in the apoptotic phase, which would explain its negative correlation with meat tenderness (Grabež et al., 2015). The negative relationship between TRIM72 and L* value affirms the anti-oxidative properties of this protein (Jung & Ko, 2010). However, further research is needed to confirm the roles of FHL1 and TRIM72 proteins in the development of meat quality traits.

    • Reverse Phase Protein array for the quantification and validation of protein biomarkers of beef qualities: The case of meat color from Charolais breed

      2018, Meat Science
      Citation Excerpt :

      In the same manner, Tripartite motif-containing 72 (TRIM72), which is exclusively expressed in skeletal muscle, might act as a sensor of oxidation on membrane damage (Cai et al., 2009), thereby influencing meat color aspects. The negative relation between TRIM72 and L* confirms the anti-oxidative properties of this protein (Jung & Ko, 2010), which may contribute to the clearance of harmful agents that accumulate during apoptosis. Further investigations are needed to clarify the roles of these two proteins in relation to meat quality traits and color development/stability.

    • MG53-IRS-1 (mitsugumin 53-insulin receptor substrate-1) interaction disruptor sensitizes insulin signaling in skeletal muscle

      2016, Journal of Biological Chemistry
      Citation Excerpt :

      Insulin-like growth factor-1 (IGF-1) initiates MyoD activation via an IGF-1 receptor-PI3K-Akt pathway during skeletal myogenesis (5, 6). MyoD and myocyte enhancer factor 2 (MEF2) binds to two proximal E-boxes and an MEF2 site in MG53 promoter, activating MG53 gene transcription (7). The MG53 protein interacts with IRS-1 and focal adhesion kinase (FAK), inducing IRS-1 and FAK ubiquitination and degradation in skeletal muscle with the help of E2 ligase UBE2H (8–10).

    • Mitsugumin 53 (MG53) Ligase ubiquitinates Focal Adhesion Kinase during Skeletal Myogenesis

      2014, Journal of Biological Chemistry
      Citation Excerpt :

      It has been proposed that mitsugumin 53 (MG53), also known as tripartite motif-containing 72 (TRIM72), is a novel E3 ligase that induces ubiquitination and proteasomal degradation of insulin receptor substrate 1 (IRS-1) in skeletal muscle (12–14). MG53 is highly up-regulated during skeletal myogenesis because its promoter contains E-box- and myocyte enhancer factor-binding sites, which are binding sites for MyoD and myocyte enhancer factor, respectively (15). With the catalytic RING finger domain in its N terminus, MG53 induces IRS-1 ubiquitination with the aid of the E2 enzyme UBE2H, negatively regulating myogenesis and insulin signaling in C2C12 cells, mouse embryonic fibroblasts (MEFs), and mouse skeletal muscles (13).

    • Mitochondrial complex I deficiency enhances skeletal myogenesis but impairs insulin signaling through SIRT1 inactivation

      2014, Journal of Biological Chemistry
      Citation Excerpt :

      As shown in Fig. 3A, the expression levels of these CI subunits were dramatically reduced after NDUFV1 knockdown, suggesting that NDUFV1 knockdown induces CI deficiency in mitochondria. We differentiated the NDUFV1-knockdown C2C12 myoblasts to myotubes and monitored skeletal myogenesis using MyHC immunofluorescence, the myogenic index and immunoblotting for myogenic marker proteins such as MyHC, myogenin (Mgn), caveolin-3 (Cav-3), and MG53 (40, 45, 46). As shown in Fig. 1, A and B, the nuclei number of MyHC-positive cells was increased ∼4-fold in 3-day-differentiated myotubes after NDUFV1 knockdown.

    View all citing articles on Scopus
    View full text