Abstract

Simple SummaryThe miR-29 family is subjected to complex regulation by tumor suppressors and oncogenes and has tumor suppressive potential in several cancers. We demonstrate that, in melanoma, oncogenic BRAF paradoxically induces miR-29 in concert with p53, thereby forming a barrier to melanoma progression. This barrier is overcome by reduced expression of miR-29, likely via diminished p53 activity. We further identify the transcription factors MAFG and MYBL2 as targets of miR-29 and show that their repression is detrimental for melanoma cells. Targeting MAFG- and MYBL2-regulated processes may therefore represent a promising therapeutic strategy to treat miR-29-low melanoma.The miR-29 family of microRNAs is encoded by two clusters, miR-29b1~a and miR-29b2~c, and is regulated by several oncogenic and tumor suppressive stimuli. While in vitro evidence suggests a tumor suppressor role for miR-29 in melanoma, the mechanisms underlying its deregulation and contribution to melanomagenesis have remained elusive. Using various in vitro systems, we show that oncogenic MAPK signaling paradoxically stimulates transcription of pri-miR-29b1~a and pri-miR-29b2~c, the latter in a p53-dependent manner. Expression analyses in melanocytes, melanoma cells, nevi, and primary melanoma revealed that pri-miR-29b2~c levels decrease during melanoma progression. Inactivation of miR-29 in vivo with a miRNA sponge in a rapid melanoma mouse model resulted in accelerated tumor development and decreased overall survival, verifying tumor suppressive potential of miR-29 in melanoma. Through integrated RNA sequencing, target prediction, and functional assays, we identified the transcription factors MAFG and MYBL2 as bona fide miR-29 targets in melanoma. Our findings suggest that attenuation of miR-29b2~c expression promotes melanoma development, at least in part, by derepressing MAFG and MYBL2.

Highlights

  • While genetic and genomic alterations are established drivers of melanoma formation, aberrant control of gene expression is emerging as a major contributor to melanoma progression. microRNAs bind to the 3 UTRs of target mRNAs to negatively regulate gene expression [1]

  • Given the regulation of miR-29 by several cancer-associated pathways, we first examined if BRAFV600E modulates miR-29 levels in mouse embryonic fibroblasts (MEFs) carrying a Cre-inducible endogenous BrafV600E allele (LSL-BrafV600E) [18]

  • The previously reported increased expression of mature miR-29a in response to p53 activity is likely explained by the inability of the mature miRNA qRT-PCR assay to distinguish between miR-29a and miR-29c, which only differ in one nucleotide, as has been suggested previously [16]

Read more

Summary

Introduction

While genetic and genomic alterations are established drivers of melanoma formation, aberrant control of gene expression is emerging as a major contributor to melanoma progression. microRNAs (miRNAs) bind to the 3 UTRs of target mRNAs to negatively regulate gene expression [1]. Various miRNAs have been reported to regulate the biology of melanoma [2,3] including miR-29a, for which melanoma suppressive potential has recently been proposed based on in vitro analyses [4]. MiR-29 is considered a tumor suppressor miRNA given its ability to repress genes involved in proliferation and cell survival, such as AKT3 [7,8], DNMT3A/B [9], MCL1 [10], and CDK6 [11], and its frequent downregulation in cancer [5], including leukemia [12], ovarian [13], or breast. MiR-29 has emerged as a major regulatory hub that integrates signaling from potent oncogenes and tumor suppressors. The regulation of miR-29 and its function in melanoma biology are poorly understood

Methods
Results
Discussion
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