Abstract

The Wnt and bone morphogenetic protein (BMP) signaling pathways are known to be crucial in the development of neural crest lineages, including the sympathetic nervous system. Surprisingly, their role in paediatric neuroblastoma, the prototypic tumor arising from this lineage, remains relatively uncharacterised. We previously demonstrated that Wnt/β-catenin signaling can have cell-type-specific effects on neuroblastoma phenotypes, including growth inhibition and differentiation, and that BMP4 mRNA and protein were induced by Wnt3a/Rspo2. In this study, we characterised the phenotypic effects of BMP4 on neuroblastoma cells, demonstrating convergent induction of MSX homeobox transcription factors by Wnt and BMP4 signaling and BMP4-induced growth suppression and differentiation. An immunohistochemical analysis of BMP4 expression in primary neuroblastomas confirms a striking absence of BMP4 in poorly differentiated tumors, in contrast to a high expression in ganglion cells. These results are consistent with a tumor suppressive role for BMP4 in neuroblastoma. RNA sequencing following BMP4 treatment revealed induction of Notch signaling, verified by increases of Notch3 and Hes1 proteins. Together, our data demonstrate, for the first time, Wnt-BMP-Notch signaling crosstalk associated with growth suppression of neuroblastoma.

Highlights

  • The canonical Wnt signaling pathway is a critical regulator of differentiation, proliferation, stemness, and determination of cell fate

  • Neuroblastoma is derived from the sympathoadrenal lineage of the neural crest [4,5], and neural crest cell (NCC) fates are contingent on tightly regulated orchestration of many signaling pathways prompting neural crest induction and specification, including Wnt/β-catenin [6,7] and bone morphogenetic protein (BMP) signaling [8]

  • We found that Wnt3a/Rspo2 treatment of neuroblastoma cell-lines did not lead to the induction of MYCN, MYCN and MYC protein levels decreased with Wnt3a/Rspo2 treatment, in contrast to previous reports suggesting induction of MYC in non-MYCN amplified neuroblastomas due to Wnt/β-catenin signaling [15]

Read more

Summary

Introduction

The canonical Wnt signaling pathway is a critical regulator of differentiation, proliferation, stemness, and determination of cell fate. Deregulation of Wnt signaling contributes to many cancers, resulting from both activating mutations of the proto-oncogene CTNNB1 (encoding β-catenin), and loss of function mutations in negative regulators of the pathway, such as APC These changes result in elevated nuclear β-catenin, which acts a co-activator for TCF/LEF transcription factors and a pro-growth/survival gene expression programme, exemplified by transcriptional activation of oncogenes such as MYC and CCND1 [1,2]. Neuroblastoma is derived from the sympathoadrenal lineage of the neural crest [4,5], and neural crest cell (NCC) fates are contingent on tightly regulated orchestration of many signaling pathways prompting neural crest induction and specification, including Wnt/β-catenin [6,7] and BMP signaling [8]

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