Abstract

BackgroundThe ongoing mobilization of mammalian transposable elements (TEs) contributes to natural genetic variation. To survey the epigenetic control and expression of reporter genes inserted by L1 retrotransposition in diverse cellular and genomic contexts, we engineered highly sensitive, real-time L1 retrotransposon reporter constructs.ResultsHere we describe different patterns of expression and epigenetic controls of newly inserted sequences retrotransposed by L1 in various somatic cells and tissues including cultured human cancer cells, mouse embryonic stem cells, and tissues of pseudofounder transgenic mice and their progeny. In cancer cell lines, the newly inserted sequences typically underwent rapid transcriptional gene silencing, but they lacked cytosine methylation even after many cell divisions. L1 reporter expression was reversible and oscillated frequently. Silenced or variegated reporter expression was strongly and uniformly reactivated by treatment with inhibitors of histone deacetylation, revealing the mechanism for their silencing. By contrast, de novo integrants retrotransposed by L1 in pluripotent mouse embryonic stem (ES) cells underwent rapid silencing by dense cytosine methylation. Similarly, de novo cytosine methylation also was identified at new integrants when studied in several distinct somatic tissues of adult founder mice. Pre-existing L1 elements in cultured human cancer cells were stably silenced by dense cytosine methylation, whereas their transcription modestly increased when cytosine methylation was experimentally reduced in cells lacking DNA methyltransferases DNMT1 and DNMT3b. As a control, reporter genes mobilized by piggyBac (PB), a DNA transposon, revealed relatively stable and robust expression without apparent silencing in both cultured cancer cells and ES cells.ConclusionsWe hypothesize that the de novo methylation marks at newly inserted sequences retrotransposed by L1 in early pre-implantation development are maintained or re-established in adult somatic tissues. By contrast, histone deacetylation reversibly silences L1 reporter insertions that had mobilized at later timepoints in somatic development and differentiation, e.g., in cancer cell lines. We conclude that the cellular contexts of L1 retrotransposition can determine expression or silencing of newly integrated sequences. We propose a model whereby reporter expression from somatic TE insertions reflects the timing, molecular mechanism, epigenetic controls and the genomic, cellular and developmental contexts of their integration.

Highlights

  • The ongoing mobilization of mammalian transposable elements (TEs) contributes to natural genetic variation

  • In each of these retrotransposition assays, the L1 donor was marked in its 3′ untranslated region (UTR) by a reporter gene disrupted by an artificial intron (AI)

  • Mimicking the design of other retrotransposition reporter constructs, we introduced the AI into donor cassettes to disrupt the TEM1 or green fluorescent protein (GFP) open reading frames (ORFs), respectively

Read more

Summary

Introduction

The ongoing mobilization of mammalian transposable elements (TEs) contributes to natural genetic variation. Half of the human and mouse genomes is comprised of various classes of transposable elements (TEs) These TE insertions have mobilized by distinct mechanisms and accumulated over evolutionary time [1,2,3,4]. Recent studies established that L1 retrotransposons, along with other classes of mobile genetic elements, can move actively in somatic cells, i.e., in mouse, rat and human neural progenitor cells, in the developing brain, and in certain human cancers [6,7,8,9,10,11] This ongoing movement of endogenous TEs including L1 retrotransposons can result in diverse genetic consequences. The epigenetic marks established at newly mobilized TEs have not been well characterized

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