Abstract

BackgroundTyrosine kinase inhibitors (TKIs) are effective therapies with demonstrated antineoplastic activity. Nilotinib is a second-generation FDA-approved TKI designed to overcome Imatinib resistance and intolerance in patients with chronic myelogenous leukemia (CML). Interestingly, TKIs have also been shown to be an efficient treatment for several non-malignant disorders such fibrotic diseases, including those affecting skeletal muscles.MethodsWe investigated the role of Nilotinib on skeletal myogenesis using the well-established C2C12 myoblast cell line. We evaluated the impact of Nilotinib during the time course of skeletal myogenesis. We compared the effect of Nilotinib with the well-known p38 MAPK inhibitor SB203580. MEK1/2 UO126 and PI3K/AKT LY294002 inhibitors were used to identify the signaling pathways involved in Nilotinib-related effects on myoblast. Adult primary myoblasts were also used to corroborate the inhibition of myoblasts fusion and myotube-nuclei positioning by Nilotinib.ResultsWe found that Nilotinib inhibited myogenic differentiation, reducing the number of myogenin-positive myoblasts and decreasing myogenin and MyoD expression. Furthermore, Nilotinib-mediated anti-myogenic effects impair myotube formation, myosin heavy chain expression, and compromise myotube-nuclei positioning. In addition, we found that p38 MAPK is a new off-target protein of Nilotinib, which causes inhibition of p38 phosphorylation in a similar manner as the well-characterized p38 inhibitor SB203580. Nilotinib induces the activation of ERK1/2 and AKT on myoblasts but not in myotubes. We also found that Nilotinib stimulates myoblast proliferation, a process dependent on ERK1/2 and AKT activation.ConclusionsOur findings suggest that Nilotinib may have important negative effects on muscle homeostasis, inhibiting myogenic differentiation but stimulating myoblasts proliferation. Additionally, we found that Nilotinib stimulates the activation of ERK1/2 and AKT. On the other hand, we suggest that p38 MAPK is a new off-target of Nilotinib. Thus, there is a necessity for future studies to investigate the long-term effects of TKIs on skeletal muscle homeostasis, along with potential detrimental effects in cell differentiation and proliferation in patients receiving TKI therapies.

Highlights

  • Tyrosine kinase inhibitors (TKIs) are effective therapies with demonstrated antineoplastic activity

  • We found that Nilotinib induces myoblast proliferation, causing impairments in myoblast cell-cycle withdrawal through both ERK1/2 and AKT pathways

  • Nilotinib inhibits skeletal muscle differentiation and muscle regulatory factors (MRFs) expression It has been suggested that Nilotinib does not affect in vitro proliferation of satellite cells [13], but the function of this tyrosine kinase inhibitor has not been tested during skeletal muscle differentiation yet

Read more

Summary

Introduction

Tyrosine kinase inhibitors (TKIs) are effective therapies with demonstrated antineoplastic activity. Nilotinib is a second-generation FDA-approved TKI designed to overcome Imatinib resistance and intolerance in patients with chronic myelogenous leukemia (CML). During skeletal muscle development and regeneration, myogenic precursors undergo several cellular and molecular changes to differentiate into multinucleated myofibers [1]. Muscle specific genes (i.e., myosin heavy chain) began to be expressed along with the appearance of differentiated myotubes due to myoblasts fusion [2]. Muscle stem cells (MuSCs) are a reservoir of quiescent myogenic precursors with an essential role in muscle regeneration [3]. The ablation of Pax7-satelite cells causes a complete loss of muscle regenerative capacity and fibrofatty degeneration after acute damage [4, 5]. Fibro-fatty degeneration is a hallmark of chronic degenerative

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call