Regulation of tumor cell migration and invasion by the H19/let-7 axis is antagonized by metformin-induced DNA methylation

L Yan, J Zhou, Y Gao, S Ghazal, L Lu, S Bellone… - Oncogene, 2015 - nature.com
L Yan, J Zhou, Y Gao, S Ghazal, L Lu, S Bellone, Y Yang, N Liu, X Zhao, AD Santin, H Taylor
Oncogene, 2015nature.com
The imprinted, developmentally regulated H19 long noncoding RNA has been implicated in
the pathogenesis of diverse human cancers, but the underlying mechanisms have remained
poorly understood. Here, we report that H19 promotes tumor cell migration and invasion by
inhibiting let-7, a potent tumor suppressor microRNA that functions to posttranscriptionally
suppress the expression of oncogenes that regulate cell growth and motility. We show that
H19 depletion impairs, whereas its overexpression enhances the motility and invasiveness …
Abstract
The imprinted, developmentally regulated H19 long noncoding RNA has been implicated in the pathogenesis of diverse human cancers, but the underlying mechanisms have remained poorly understood. Here, we report that H19 promotes tumor cell migration and invasion by inhibiting let-7, a potent tumor suppressor microRNA that functions to posttranscriptionally suppress the expression of oncogenes that regulate cell growth and motility. We show that H19 depletion impairs, whereas its overexpression enhances the motility and invasiveness of tumor cells. These phenomena occur, at least in part through affecting let-7-mediated regulation of metastasis-promoting genes, including Hmga2, c-Myc and Igf2bp3. This H19/let-7-dependent regulation is recapitulated in vivo where co-expressions of oncogenes and H19 exist in both primary human ovarian and endometrial cancers. Furthermore, we provide evidence that the anti-diabetic drug metformin inhibits tumor cell migration and invasion, partly by downregulating H19 via DNA methylation. Our results reveal a novel mechanism underpinning H19-mediated regulation in metastasis and may explain why in some cases increased let-7 expression unexpectedly correlates with poor prognosis, given the widely accepted role for let-7 as a tumor suppressor. Targeting this newly identified pathway might offer therapeutic opportunities.
nature.com