Abstract

FYN is a non-receptor tyrosine kinase belonging to the SRC family of kinases, which are frequently over-expressed in human cancers, and play key roles in cancer biology. SRC has long been recognized as an important oncogene, but little attention has been given to its other family members. In this report, we have studied the role of FYN in FLT3 signaling in respect to acute myeloid leukemia (AML). We observed that FYN displays a strong association with wild-type FLT3 as well as oncogenic FLT3-ITD and is dependent on the kinase activity of FLT3 and the SH2 domain of FYN. We identified multiple FYN binding sites in FLT3, which partially overlapped with SRC binding sites. To understand the role of FYN in FLT3 signaling, we generated FYN overexpressing cells. We observed that expression of FYN resulted in slightly enhanced phosphorylation of AKT, ERK1/2 and p38 in response to ligand stimulation. Furthermore, FYN expression led to a slight increase in FLT3-ITD-dependent cell proliferation, but potent enhancement of STAT5 phosphorylation as well as colony formation. We also observed that FYN expression is deregulated in AML patient samples and that higher expression of FYN, in combination with FLT3-ITD mutation, resulted in enrichment of the STAT5 signaling pathway and correlated with poor prognosis in AML. Taken together our data suggest that FYN cooperates with oncogenic FLT3-ITD in cellular transformation by selective activation of the STAT5 pathway. Therefore, inhibition of FYN, in combination with FLT3 inhibition, will most likely be beneficial for this group of AML patients.

Highlights

  • Expression of receptor tyrosine kinases has been shown to be deregulated in many cancers

  • We suggest that FYN may play a role in acute myeloid leukemia (AML) patients carrying FLT3-internal tandem duplication (ITD)

  • We aimed to understand the role of FYN in FLT3-ITD - dependent AML

Read more

Summary

Introduction

Expression of receptor tyrosine kinases has been shown to be deregulated in many cancers. Activation of FLT3 is mediated through ligand binding and dimerization followed by phosphorylation of several tyrosine residues in the intracellular domain. Phosphorylation of tyrosine residues creates docking sites for the SH2 domain-containing proteins that further propagate or terminate signaling. For example association of non-receptor tyrosine kinases SRC and SYK, adaptor proteins GRB10 and SLAP resulted in enhancement of FLT3 signaling [3,4,5]. The association of other SH2 domain containing proteins such as CBL ubiquitin ligases, SOCS2 and SOCS6 adaptor proteins, LNK and the non-receptor tyrosine kinase CSK negatively regulates FLT3 signaling [6,7,8,9]. Understanding the role of FLT3 binding proteins is of importance to understand FLT3 mediated pathogenesis in AML

Objectives
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