Published in

National Academy of Sciences, Proceedings of the National Academy of Sciences, 47(109), p. 19280-19285, 2012

DOI: 10.1073/pnas.1214394109

Links

Tools

Export citation

Search in Google Scholar

Splicing program of human MENA produces a previously undescribed isoform associated with invasive, mesenchymal-like breast tumors

This paper is made freely available by the publisher.
This paper is made freely available by the publisher.

Full text: Download

Red circle
Preprint: archiving forbidden
Green circle
Postprint: archiving allowed
Red circle
Published version: archiving forbidden
Data provided by SHERPA/RoMEO

Abstract

Human mena (hMENA), a member of the actin cytoskeleton regulators Ena/VASP, is overexpressed in high-risk preneoplastic lesions and in primary breast tumors and has been identified as playing a role in invasiveness and poor prognosis in breast cancers that express HER2. Here we identify a unique isoform, hMENAΔv6, derived from the hMENA alternative splicing program. In an isogenic model of human breast cancer progression, we show that hMENA 11a is expressed in premalignant cells, whereas hMENAΔv6 expression is restricted to invasive cancer cells. “Reversion” of the malignant phenotype leads to concurrent down-regulation of all hMENA isoforms. In breast cancer cell lines, isoform-specific hMENA overexpression or knockdown revealed that in the absence of hMENA 11a , overexpression of hMENAΔv6 increased cell invasion, whereas overexpression of hMENA 11a reduced the migratory and invasive ability of these cells. hMENA 11a splicing was shown to be dependent on the epithelial regulator of splicing 1 (ESRP1), and forced expression of ESRP1 in invasive mesenchymal breast cancer cells caused a phenotypic switch reminiscent of a mesenchymal-to-epithelial transition (MET) characterized by changes in the cytoskeletal architecture, reexpression of hMENA 11a , and a reduction in cell invasion. hMENA-positive primary breast tumors, which are hMENA 11a -negative, are more frequently E-cadherin low in comparison with tumors expressing hMENA 11a . These data suggest that polarized and growth-arrested cellular architecture correlates with absence of alternative hMENA isoform expression, and that the hMENA splicing program is relevant to malignant progression in invasive disease.