Abstract

BackgroundWe have found that erbB receptor tyrosine kinases drive Ras hyperactivation and growth in NF1-null malignant peripheral nerve sheath tumors (MPNSTs). However, MPNSTs variably express multiple erbB receptors with distinct functional characteristics and it is not clear which of these receptors drive MPNST pathogenesis. Here, we test the hypothesis that altered erbB4 expression promotes MPNST pathogenesis by uniquely activating key cytoplasmic signaling cascades.MethodsErbB4 expression was assessed using immunohistochemistry, immunocytochemistry, immunoblotting and real-time PCR. To define erbB4 functions, we generated mice that develop MPNSTs with floxed Erbb4 alleles (P0-GGFβ3;Trp53+/−;Erbb4flox/flox mice) and ablated Erbb4 in these tumors. MPNST cell proliferation and survival was assessed using 3H-thymidine incorporation, MTT assays, Real-Time Glo and cell count assays. Control and Erbb4-null MPNST cells were orthotopically xenografted in immunodeficient mice and the growth, proliferation (Ki67 labeling), apoptosis (TUNEL labeling) and angiogenesis of these grafts was analyzed. Antibody arrays querying cytoplasmic kinases were used to identify erbB4-responsive kinases. Pharmacologic or genetic inhibition was used to identify erbB4-responsive kinases that drive proliferation.ResultsAberrant erbB4 expression was evident in 25/30 surgically resected human MPNSTs and in MPNSTs from genetically engineered mouse models (P0-GGFβ3 and P0-GGFβ3;Trp53+/− mice); multiple erbB4 splice variants that differ in their ability to activate PI3 kinase and nuclear signaling were present in MPNST-derived cell lines. Erbb4-null MPNST cells demonstrated decreased proliferation and survival and altered morphology relative to non-ablated controls. Orthotopic allografts of Erbb4-null cells were significantly smaller than controls, with reduced proliferation, survival and vascularization. ERBB4 knockdown in human MPNST cells similarly inhibited DNA synthesis and viability. Although we have previously shown that broad-spectrum erbB inhibitors inhibit Ras activation, Erbb4 ablation did not affect Ras activation, suggesting that erbB4 drives neoplasia via non-Ras dependent pathways. An analysis of 43 candidate kinases identified multiple NRG1β-responsive and erbB4-dependent signaling cascades including the PI3K, WNK1, STAT3, STAT5 and phospholipase-Cγ pathways. Although WNK1 inhibition did not alter proliferation, inhibition of STAT3, STAT5 and phospholipase-Cγ markedly reduced proliferation.ConclusionsErbB4 promotes MPNST growth by activating key non-Ras dependent signaling cascades including the STAT3, STAT5 and phospholipase-Cγ pathways. ErbB4 and its effector pathways are thus potentially useful therapeutic targets in MPNSTs.

Highlights

  • We have found that erbB receptor tyrosine kinases drive Ras hyperactivation and growth in neurofibromatosis type 1 (NF1)-null malignant peripheral nerve sheath tumors (MPNSTs)

  • Aberrant expression of erbB4 is evident in MPNSTs Our previous analyses of two MPNST cell lines and six tumor samples indicated that erbB4 was variably expressed in these neoplasms [16]

  • The membranous staining was often punctate, consistent with our previous observations in MPNST cells [17]; this is consistent with the fact that erbB4 functions as a microenvironment sensor at the plasma membrane but can get proteolytically cleaved and enter the nucleus to function as a transcriptional coactivator

Read more

Summary

Introduction

We have found that erbB receptor tyrosine kinases drive Ras hyperactivation and growth in NF1-null malignant peripheral nerve sheath tumors (MPNSTs). Malignant peripheral nerve sheath tumors (MPNSTs) are highly aggressive spindle cell neoplasms derived from the Schwann cell lineage [1]. These neoplasms are the most common malignancy occurring in patients with neurofibromatosis type 1 (NF1) [2]; they occur sporadically in the general population and at sites of previous radiation therapy. It is generally agreed that in all of these clinical settings, MPNSTs have a poor prognosis, with several academic centers reporting 5-year disease-free survival rates of 34–60% [4, 6,7,8,9,10,11,12] In large part, these poor outcomes reflect the fact that surgical resection is currently the only effective means of treating MPNSTs and achieving a complete surgical resection is often impossible. Developing effective new chemotherapeutic regimens is essential if we are to improve the survival of patients with these aggressive neoplasms

Methods
Results
Discussion
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