[HTML][HTML] Identification of Rtl1, a Retrotransposon-Derived Imprinted Gene, as a Novel Driver of Hepatocarcinogenesis

JD Riordan, VW Keng, BR Tschida, TE Scheetz… - PLoS …, 2013 - journals.plos.org
JD Riordan, VW Keng, BR Tschida, TE Scheetz, JB Bell, KM Podetz-Pedersen, CD Moser…
PLoS genetics, 2013journals.plos.org
We previously utilized a Sleeping Beauty (SB) transposon mutagenesis screen to discover
novel drivers of HCC. This approach identified recurrent mutations within the Dlk1-Dio3
imprinted domain, indicating that alteration of one or more elements within the domain
provides a selective advantage to cells during the process of hepatocarcinogenesis. For the
current study, we performed transcriptome and small RNA sequencing to profile gene
expression in SB–induced HCCs in an attempt to clarify the genetic element (s) contributing …
We previously utilized a Sleeping Beauty (SB) transposon mutagenesis screen to discover novel drivers of HCC. This approach identified recurrent mutations within the Dlk1-Dio3 imprinted domain, indicating that alteration of one or more elements within the domain provides a selective advantage to cells during the process of hepatocarcinogenesis. For the current study, we performed transcriptome and small RNA sequencing to profile gene expression in SB–induced HCCs in an attempt to clarify the genetic element(s) contributing to tumorigenesis. We identified strong induction of Retrotransposon-like 1 (Rtl1) expression as the only consistent alteration detected in all SB–induced tumors with Dlk1-Dio3 integrations, suggesting that Rtl1 activation serves as a driver of HCC. While previous studies have identified correlations between disrupted expression of multiple Dlk1-Dio3 domain members and HCC, we show here that direct modulation of a single domain member, Rtl1, can promote hepatocarcinogenesis in vivo. Overexpression of Rtl1 in the livers of adult mice using a hydrodynamic gene delivery technique resulted in highly penetrant (86%) tumor formation. Additionally, we detected overexpression of RTL1 in 30% of analyzed human HCC samples, indicating the potential relevance of this locus as a therapeutic target for patients. The Rtl1 locus is evolutionarily derived from the domestication of a retrotransposon. In addition to identifying Rtl1 as a novel driver of HCC, our study represents one of the first direct in vivo demonstrations of a role for such a co-opted genetic element in promoting carcinogenesis.
PLOS