Abstract

We have previously shown that the Wnt canonical pathway (WCP) is constitutively active in most cases of mantle cell lymphoma (MCL). Here, we aimed to elucidate the mechanisms underlying this biochemical deregulation. We hypothesized that gene methylation/silencing of WIF1 (Wnt inhibitory factor-1), a physiologic inhibitor of WCP, contributes to the deregulation of WCP and promotes cell growth in MCL. In support of this hypothesis, we found that the expression of WIF1 was detectable in none of the 4 MCL cell lines, and in only 2 of 5 tumors (40%) examined. Using methylation-specific PCR, we found evidence of gene methylation of WIF1 in 4 of 5 cell lines (80%) and in 24 of 29 (82%) tumors. The addition of the demethylation agent 5-aza-2′-deoxycytidine to Mino and JeKo-1, two WIF1-negative cell lines, restored the expression of WIF1 mRNA in these cells. Gene transfection of WIF1 into JeKo-1 and Mino cells significantly reduced cell growth, and this finding correlated with substantial downregulations of various proteins in WCP, such as β-catenin and pGSK-3β. In conclusion, our results support the concept that gene methylation/silencing of WIF1 is a frequent event in MCL, and this abnormality contributes to the aberrant activation of WCP. These results have provided further evidence that aberrant Wnt signaling is pathogenetically important in MCL and it may represent a potential therapeutic target.

Highlights

  • Mantle cell lymphoma (MCL) is a type of aggressive B-cell non-Hodgkin lymphoma that carries a guarded prognosis despite a high rate of initial remission induced by conventional chemotherapy regimens [1,2]

  • Wnt inhibitory factor-1 (WIF1) Is Hypermethylated in Most MCL Cell Lines and Tumors

  • We speculated that WIF1 is not expressed in most MCL cell lines and patient samples due to gene methylation and silencing

Read more

Summary

Introduction

Mantle cell lymphoma (MCL) is a type of aggressive B-cell non-Hodgkin lymphoma that carries a guarded prognosis despite a high rate of initial remission induced by conventional chemotherapy regimens [1,2]. Most of these tumors harbor the characteristic chromosomal translocation, t(11;14) (q13;q32), which brings the cyclin D1 gene under the influence of the enhancer of IgH, resulting in aberrant cyclin D1 expression in these neoplastic B-cells [3]. In view of its normal function, cyclin D1 is believed to drive tumorigenesis by promoting cell-cycle progression. Oncogenic events additional to the aberrant over-expression of cyclin D1 are likely to be important for the development of MCL

Objectives
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