[HTML][HTML] Autophagy-regulating TP53INP2 mediates muscle wasting and is repressed in diabetes

D Sala, S Ivanova, N Plana, V Ribas… - The Journal of …, 2014 - Am Soc Clin Investig
D Sala, S Ivanova, N Plana, V Ribas, J Duran, D Bach, S Turkseven, M Laville, H Vidal…
The Journal of clinical investigation, 2014Am Soc Clin Investig
A precise balance between protein degradation and synthesis is essential to preserve
skeletal muscle mass. Here, we found that TP53INP2, a homolog of the Drosophila
melanogaster DOR protein that regulates autophagy in cellular models, has a direct impact
on skeletal muscle mass in vivo. Using different transgenic mouse models, we demonstrated
that muscle-specific overexpression of Tp53inp2 reduced muscle mass, while deletion of
Tp53inp2 resulted in muscle hypertrophy. TP53INP2 activated basal autophagy in skeletal …
A precise balance between protein degradation and synthesis is essential to preserve skeletal muscle mass. Here, we found that TP53INP2, a homolog of the Drosophila melanogaster DOR protein that regulates autophagy in cellular models, has a direct impact on skeletal muscle mass in vivo. Using different transgenic mouse models, we demonstrated that muscle-specific overexpression of Tp53inp2 reduced muscle mass, while deletion of Tp53inp2 resulted in muscle hypertrophy. TP53INP2 activated basal autophagy in skeletal muscle and sustained p62-independent autophagic degradation of ubiquitinated proteins. Animals with muscle-specific overexpression of Tp53inp2 exhibited enhanced muscle wasting in streptozotocin-induced diabetes that was dependent on autophagy; however, TP53INP2 ablation mitigated experimental diabetes-associated muscle loss. The overexpression or absence of TP53INP2 did not affect muscle wasting in response to denervation, a condition in which autophagy is blocked, further indicating that TP53INP2 alters muscle mass by activating autophagy. Moreover, TP53INP2 expression was markedly repressed in muscle from patients with type 2 diabetes and in murine models of diabetes. Our results indicate that TP53INP2 negatively regulates skeletal muscle mass through activation of autophagy. Furthermore, we propose that TP53INP2 repression is part of an adaptive mechanism aimed at preserving muscle mass under conditions in which insulin action is deficient.
The Journal of Clinical Investigation