SMYD1 and G6PD modulation are critical events for miR-206-mediated differentiation of rhabdomyosarcoma

DM Coda, MF Lingua, D Morena, V Foglizzo… - Cell Cycle, 2015 - Taylor & Francis
DM Coda, MF Lingua, D Morena, V Foglizzo, F Bersani, U Ala, C Ponzetto, R Taulli
Cell Cycle, 2015Taylor & Francis
Rhadomyosarcoma (RMS) is the most common soft tissue sarcoma of childhood. RMS cells
resemble fetal myoblasts but are unable to complete myogenic differentiation. In previous
work we showed that miR-206, which is low in RMS, when induced in RMS cells promotes
the resumption of differentiation by modulating more than 700 genes. To better define the
pathways involved in the conversion of RMS cells into their differentiated counterpart, we
focused on 2 miR-206 effectors emerged from the microarray analysis, SMYD1 and G6PD …
Rhadomyosarcoma (RMS) is the most common soft tissue sarcoma of childhood. RMS cells resemble fetal myoblasts but are unable to complete myogenic differentiation. In previous work we showed that miR-206, which is low in RMS, when induced in RMS cells promotes the resumption of differentiation by modulating more than 700 genes. To better define the pathways involved in the conversion of RMS cells into their differentiated counterpart, we focused on 2 miR-206 effectors emerged from the microarray analysis, SMYD1 and G6PD. SMYD1, one of the most highly upregulated genes, is a H3K4 histone methyltransferase. Here we show that SMYD1 silencing does not interfere with the proliferative block or with the loss anchorage independence imposed by miR-206, but severely impairs differentiation of ERMS, ARMS, and myogenic cells. Thus SMYD1 is essential for the activation of muscle genes. Conversely, among the downregulated genes, we found G6PD, the enzyme catalyzing the rate-limiting step of the pentose phosphate shunt. In this work, we confirmed that G6PD is a direct target of miR-206. Moreover, we showed that G6PD silencing in ERMS cells impairs proliferation and soft agar growth. However, G6PD overexpression does not interfere with the pro-differentiating effect of miR-206, suggesting that G6PD downmodulation contributes to - but is not an absolute requirement for - the tumor suppressive potential of miR-206. Targeting cancer metabolism may enhance differentiation. However, therapeutic inhibition of G6PD is encumbered by side effects. As an alternative, we used DCA in combination with miR-206 to increase the flux of pyruvate into the mitochondrion by reactivating PDH. DCA enhanced the inhibition of RMS cell growth induced by miR-206, and sustained it upon miR-206 de-induction. Altogether these results link miR-206 to epigenetic and metabolic reprogramming, and suggest that it may be worth combining differentiation-inducing with metabolism-directed approaches.
Taylor & Francis Online