Abstract

BackgroundMigration and invasion enhancer 1 (MIEN1) is a novel gene found to be abundantly expressed in breast tumor tissues and functions as a critical regulator of tumor cell migration and invasion to promote systemic metastases. Previous studies have identified post-translational modifications by isoprenylation at the C-terminal tail of MIEN1 to favor its translocation to the inner leaflet of plasma membrane and its function as a membrane-bound adapter molecule. However, the exact molecular events at the membrane interface activating the MIEN1-driven tumor cell motility are vaguely understood.MethodsMIEN1 was first studied using in-silico analysis on available RNA sequencing data of human breast tissues and its expression was ascertained in breast cells. We performed several assays including co-immunoprecipitation, wound healing, western blotting and immunofluorescence to decipher the molecular events involved in MIEN1-mediated tumor cell migration.ResultsClinically, MIEN1 is predominantly overexpressed in Her-2 and luminal B subtypes of breast tumors, and its increased expression correlates with poor disease free survival. Molecular studies identified a phosphorylation-dependent activation signal in the immunoreceptor tyrosine based activation motif (ITAM) of MIEN1 and the phosphorylation-deficient MIEN1-mutants (Y39F/50 F) to regulate filopodia generation, migration and invasion. We found that ITAM-phosphorylation of MIEN1 is significantly impaired in isoprenylation-deficient MIEN1 mutants indicating that prenylation of MIEN1 and membrane association is required for cross-phosphorylation of tyrosine residues. Furthermore, we identified MIEN1 as a novel interactor of Annexin A2 (AnxA2), a Ca2+ -dependent phospholipid binding protein, which serves as an extracellular proteolytic center regulating plasmin generation. Fluorescence resonance energy transfer (FRET) confirmed that MIEN1 physically interacts with AnxA2 and functional studies revealed that they mutually cooperate to accentuate tumor cell motility. Interestingly, our study identified that ectopic overexpression of MIEN1 significantly enhances Tyr23-phosphorylation on AnxA2, thereby stimulating cell surface translocation of AnxA2 and catalyzing the activation of its proteolytic activity.ConclusionOur data show that the presence and interaction of both MIEN1 and AnxA2 in breast tumors are crucial drivers of cell motility. Our study has now deciphered a novel regulatory network governing the vicious process of breast tumor cell invasion-metastasis, and findings suggest MIEN1-AnxA2 as prospective targets to counter the deadly disease.Electronic supplementary materialThe online version of this article (doi:10.1186/s12943-015-0428-8) contains supplementary material, which is available to authorized users.

Highlights

  • Migration and invasion enhancer 1(MIEN1) is located in the chromosomal region 17q12-21, in the ERBB2 amplicon [1,2,3,4]

  • We show that MIEN1 has a functional immunoreceptor tyrosine based activation motif (ITAM) cross-phosphorylated at two tyrosine-residues (Y39 and Y50), which is important for triggering downstream signal transduction

  • MIEN1 is expressed in all subtypes of breast cancer Enhanced expression of MIEN1 is reported in breast cancer compared to normal breast tissues [2]

Read more

Summary

Introduction

Migration and invasion enhancer 1(MIEN1) ( known as C35, C17orf, RDX12, and MGC14832) is located in the chromosomal region 17q12-21, in the ERBB2 amplicon [1,2,3,4]. MIEN1 is frequently amplified along the neighboring genes, ErBB2 and GRB7 in variety of tumors including breast cancer. Our previous studies identified MIEN1 as the prime regulator of cancer cell migration and invasion [5]. The exact molecular events at the membrane interface in MIEN1-driven breast tumor cell motility are poorly understood. Migration and invasion enhancer 1 (MIEN1) is a novel gene found to be abundantly expressed in breast tumor tissues and functions as a critical regulator of tumor cell migration and invasion to promote systemic metastases. Previous studies have identified post-translational modifications by isoprenylation at the C-terminal tail of MIEN1 to favor its translocation to the inner leaflet of plasma membrane and its function as a membrane-bound adapter molecule. The exact molecular events at the membrane interface activating the MIEN1-driven tumor cell motility are vaguely understood

Methods
Results
Discussion
Conclusion
Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.