Articles

< Previous         Next >  
Smad1/5/8 are myogenic regulators of murine and human mesoangioblasts
Domiziana Costamagna1,2, Mattia Quattrocelli1, Florence van Tienen3, Lieve Umans4,5, Irineus F. M. de Coo6, An Zwijsen4, Danny Huylebroeck5,7, and Maurilio Sampaolesi1,8,*
1Translational Cardiomyology Laboratory, Stem Cell Biology and Embryology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
2Laboratory of Experimental Medicine and Clinical Pathology, Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
3Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
4Laboratory for Developmental Signalling, VIB Center for the Biology of Disease, Department of Human Genetics, KU Leuven, Leuven, Belgium
5Laboratory of Molecular Biology (Celgen), Department of Development and Regeneration, KU Leuven, Leuven, Belgium
6Department of Neurology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
7Department of Cell Biology, Erasmus MC, Rotterdam, The Netherlands
8Division of Human Anatomy, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy *Correspondence to:Maurilio Sampaolesi, E-mail: maurilio.sampaolesi{at}med.kuleuven.be
J Mol Cell Biol, Volume 8, Issue 1, February 2016, 73-87,  https://doi.org/10.1093/jmcb/mjv059
Keyword: BMP signalling, mesoangioblast, pericytes, skeletal muscle, regeneration, SMAD

Mesoangioblasts (MABs) are vessel-associated stem cells that express pericyte marker genes and participate in skeletal muscle regeneration. Molecular circuits that regulate the myogenic commitment of MABs are still poorly characterized. The critical role of bone morphogenetic protein (BMP) signalling during proliferation and differentiation of adult myogenic precursors, such as satellite cells, has recently been established. We evaluated whether BMP signalling impacts on the myogenic potential of embryonic and adult MABs both in vitro and in vivo. Addition of BMP inhibited MAB myogenic differentiation, whereas interference with the interactions between BMPs and receptor complexes induced differentiation. Similarly, siRNA-mediated knockdown of Smad8 in Smad1/5-null MABs or inhibition of SMAD1/5/8 phosphorylation with Dorsomorphin (DM) also improved myogenic differentiation, demonstrating a novel role of SMAD8. Moreover, using a transgenic mouse model of Smad8 deletion, we demonstrated that the absence of SMAD8 protein improved MAB myogenic differentiation. Furthermore, once injected into α-Sarcoglycan (Sgca)-null muscles, DM-treated MABs were more efficacious to restore α-sarcoglycan (αSG) protein levels and re-establish functional muscle properties. Similarly, in acute muscle damage, DM-treated MABs displayed a better myogenic potential compared with BMP-treated and untreated cells. Finally, SMADs also control the myogenic commitment of human MABs (hMABs). BMP signalling antagonists are therefore novel candidates to improve the therapeutic effects of hMABs.