"Medicine, Faculty of"@en . "Non UBC"@en . "Medical Genetics, Department of"@en . "DSpace"@en . "Mobile DNA. 2016 Dec 01;7(1):24"@en . "The Author(s)."@en . "Babaian, Artem"@en . "Mager, Dixie L"@en . "2018-05-29T16:39:18Z"@* . "2016-12-01"@en . "Cancer arises from a series of genetic and epigenetic changes, which result in abnormal expression or mutational activation of oncogenes, as well as suppression/inactivation of tumor suppressor genes. Aberrant expression of coding genes or long non-coding RNAs (lncRNAs) with oncogenic properties can be caused by translocations, gene amplifications, point mutations or other less characterized mechanisms. One such mechanism is the inappropriate usage of normally dormant, tissue-restricted or cryptic enhancers or promoters that serve to drive oncogenic gene expression. Dispersed across the human genome, endogenous retroviruses (ERVs) provide an enormous reservoir of autonomous gene regulatory modules, some of which have been co-opted by the host during evolution to play important roles in normal regulation of genes and gene networks. This review focuses on the \u00E2\u0080\u009Cdark side\u00E2\u0080\u009D of such ERV regulatory capacity. Specifically, we discuss a growing number of examples of normally dormant or epigenetically repressed ERVs that have been harnessed to drive oncogenes in human cancer, a process we term onco-exaptation, and we propose potential mechanisms that may underlie this phenomenon."@en . "https://circle.library.ubc.ca/rest/handle/2429/66165?expand=metadata"@en . "REVIEW Open AccessEndogenous retroviral promoter exaptationin human cancerArtem Babaian1,2 and Dixie L. Mager1,2*AbstractCancer arises from a series of genetic and epigenetic changes, which result in abnormal expression or mutationalactivation of oncogenes, as well as suppression/inactivation of tumor suppressor genes. Aberrant expression ofcoding genes or long non-coding RNAs (lncRNAs) with oncogenic properties can be caused by translocations, geneamplifications, point mutations or other less characterized mechanisms. One such mechanism is the inappropriateusage of normally dormant, tissue-restricted or cryptic enhancers or promoters that serve to drive oncogenic geneexpression. Dispersed across the human genome, endogenous retroviruses (ERVs) provide an enormous reservoir ofautonomous gene regulatory modules, some of which have been co-opted by the host during evolution to playimportant roles in normal regulation of genes and gene networks. This review focuses on the \u00E2\u0080\u009Cdark side\u00E2\u0080\u009D of suchERV regulatory capacity. Specifically, we discuss a growing number of examples of normally dormant or epigeneticallyrepressed ERVs that have been harnessed to drive oncogenes in human cancer, a process we term onco-exaptation,and we propose potential mechanisms that may underlie this phenomenon.Keywords: Gene regulation, Endogenous retrovirus, Long terminal repeat, Retrotransposon, Epigenetics, Cancer,Alternative promoter, Exaptation, TranscriptionBackgroundSequences derived from transposable elements (TEs) oc-cupy at least half the human genome [1, 2]. TEs are gen-erally classified into two categories; DNA transposons,which comprise 3.2% of the human genome; and the ret-roelements, short interspersed repeats (SINEs, 12.8% ofthe genome), long interspersed repeats (LINEs, 20.7%)and long terminal repeat (LTR) elements, derived fromendogenous retroviruses (ERVs, 8.6%). Over evolutionarytime, TE sequences in the genome can become func-tional units that confer a fitness advantage, a processcalled \u00E2\u0080\u009Cexaptation\u00E2\u0080\u009D [3, 4]. Exaptation includes proteincoding, non-coding and regulatory effects of TEs. This isin contrast to the designation of \u00E2\u0080\u009Cnonaptations\u00E2\u0080\u009D for gen-etic units that perform some function (such as initiatetranscription) but don\u00E2\u0080\u0099t impact host fitness [4]. Besidestheir roles in shaping genomes during evolution, TEscontinue to have impact in humans through insertionalmutagenesis, inducing rearrangements and affecting generegulation, as discussed in recent reviews [5\u00E2\u0080\u009312].Efforts to explore the role of TEs in human cancerhave focused primarily on LINEs and ERVs. While nearlyall L1s, the major human LINE family, are defective, afew hundred retain the ability to retrotranspose [13] andthese active elements occasionally cause germ line muta-tions [9, 14, 15]. Several recent studies have also docu-mented somatic, cancer-specific L1 insertions [16\u00E2\u0080\u009323],and a few such insertions were shown to contribute tomalignancy [9]. For example, two L1 insertions weredocumented to disrupt the tumor suppressor gene APCin colon cancer [16, 23]. However, it is probable thatmost insertions are non-consequential \u00E2\u0080\u009Cpassenger muta-tions\u00E2\u0080\u009D, as recently discussed by Hancks and Kazazian [9].Thus, the overall biological effect size of LINE retrotran-sposition on the process of oncogenesis may be limited.No evidence for retrotranspositionally active ERVs inhumans has been reported [24\u00E2\u0080\u009326], so it is unlikely thathuman ERVs activate oncogenes or inactivate tumorsuppressor genes by somatic retrotransposition. This isin contrast to the frequent oncogene activation by inser-tions of exogenous and endogenous retroviruses in* Correspondence: dmager@bccrc.ca1Terry Fox Laboratory, British Columbia Cancer Agency, 675 West 10thAvenue, Vancouver, BC V5Z1L3, Canada2Department of Medical Genetics, University of British Columbia, Vancouver,BC, Canada\u00C2\u00A9 The Author(s). 2016 Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, andreproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link tothe Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver(http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.Babaian and Mager Mobile DNA (2016) 7:24 DOI 10.1186/s13100-016-0080-xchickens or mice, where retrotranspositional activity ofERVs is very high [27\u00E2\u0080\u009329]. Therefore, to date, most stud-ies into potential roles for ERVs in human cancer havefocused on their protein products. Indeed, there isstrong evidence that the accessary proteins Np9 andRec, encoded by members of the relatively young HERV-K (HML-2) group, have oncogenic properties, particu-larly in germ cell tumors [30\u00E2\u0080\u009333].Regardless of their retrotranspositional or coding cap-acity, ERVs may play a broader role in oncogenesis involv-ing their intrinsic regulatory capacity. De-repression/activation of cryptic (or normally dormant) promoters todrive ectopic expression is one mechanism that can leadto oncogenic effects [34\u00E2\u0080\u009340]. Because TEs, and especiallyERV LTRs, are an abundant reservoir of natural pro-moters in the human genome [6, 41, 42], inappropriatetranscriptional activation of typically repressed LTRs maycontribute to oncogenesis. Here we review examples ofsuch phenomena, which we term \u00E2\u0080\u009Conco-exaptation\u00E2\u0080\u009D, andpropose two explanatory models to understand the role ofLTRs in oncogenesis.Promoter potential of ERVsHundreds of ERV \u00E2\u0080\u009Cfamilies\u00E2\u0080\u009D or groups, which is themore proper designation [43], are remnants of ancientretroviral infections of the germ line and occupy at least8.67% of the human genome [1, 24, 44]. These rangefrom groups that integrated before the divergence of ro-dents and primates, such as older members of the largeMaLR/ERV-L class, to the youngest HERV-K (HML-2)group, a few members of which are insertionally poly-morphic in humans [45, 46]. While it has been postu-lated that rare \u00E2\u0080\u009Cactive\u00E2\u0080\u009D HERV-K elements exist at verylow allele frequencies [45], there is currently no evi-dence for new somatic or germ line insertions of ERVsin humans and nearly all have lost coding potential[24\u00E2\u0080\u009326]. The situation is starkly different in inbredmice, where at least 10% of documented, phenotype-producing germ line mutations and numerous somatic,cancer-associated insertions are due to ongoing retro-transpositions of ERVs [28, 29, 47]. Table 1 lists selectmajor ERV groups found in humans, members of whichare mentioned in this review.Approximately 90% of the \u00E2\u0080\u009CERV-related\u00E2\u0080\u009D human gen-omic DNA is in the form of solitary LTRs, which are cre-ated over evolutionary time via recombination betweenthe 5\u00E2\u0080\u0099 and 3\u00E2\u0080\u0099 LTRs of an integrated provirus [48, 49].LTRs naturally contain transcriptional promoters andenhancers, and often splice donor sites, required for au-tonomous expression of the integrated LTR element.Furthermore, unlike for LINEs (see below), the integra-tion process nearly always retains the primary tran-scriptional regulatory motifs, i.e. the LTR, even afterrecombination between the LTRs of a full-length proviralform. Mutations will degrade LTR promoter/enhancermotifs over time, but many of the >470,000 ERV/LTR lociin the genome [50] likely still retain some degree of theirancestral promoter/enhancer function, and hence a generegulatory capacity.LTR-mediated regulation of single genes and gene net-works has been increasingly documented in the litera-ture. For example, studies have implicated ERV LTRs inspecies-specific regulatory networks in ES cells [51], inthe interferon response [52], in p53-mediated regulation[53], as tissue-specific enhancers [54, 55] and in regulat-ing pluripotency by promoting genes and lncRNAs instem cells [56\u00E2\u0080\u009360]. LTR regulatory capacity arises fromboth their \u00E2\u0080\u009Cready-to-use\u00E2\u0080\u009D ancestral transcriptional factor(TF) binding sites and by mutation/evolution of novelsites, possibly maintained through epistatic capture [61](recently reviewed in [42]). For more in depth discussionof the evolutionary exaptation of enhancers/promotersof LTRs and other TEs in mammals, we refer the readerto a rapidly growing number of reviews on this subjectTable 1 ERV/LTR groups mentioned in this reviewERV Class ERV Group Associated LTRs (Repbase names) ~Copies of internal regionsa ~Copies of solitary LTRsbI (ERV1) HERV-H LTR7, 7B, 7C, 7Y 1060 1270HERV-9 LTR12, 12B-12 F 450 6500HERV-E LTR2, 2B, 2C 250 720HUERS-P2 LTR1, 1A-1 F 120 3000LOR1 LOR1a, 1b 175 1080MER41 MER41A-41G 275 4110II (ERVK) HERV-K (HML-2) LTR5, 5A, 5B, 5Hs 80 1200III (ERVL) HERV16 LTR16A-16E 860 18100III (MaLR) THE THE1A-1D 7900 21260MLT1 MLT1A-1O 3820 146,550aCopy numbers of internal regions estimated from Dfam (dfam.org) [50]bSolitary LTR numbers estimated from Dfam coverage minus 2x internal region numbers, assuming all internal regions are associated with two LTRsBabaian and Mager Mobile DNA (2016) 7:24 Page 2 of 21[6, 10, 42, 62\u00E2\u0080\u009365]. Suffice it to say that, retrotransposi-tionally incompetent ERV LTRs, long considered the\u00E2\u0080\u009Cpoor cousin\u00E2\u0080\u009D of active L1 elements, have emerged fromthe shadowy realm of junk DNA and are now recognizedas a major source of gene regulatory evolution throughexaptation of their promoters and enhancers.Promoter potential of LINEs and other non-LTR TEsBesides via new retrotransposition events, existing L1 el-ements can also impact genes through promoter dona-tion. Full-length L1 elements harbor two internalpromoters at their 5\u00E2\u0080\u0099 end, a sense promoter that drives ex-pression of the element and an antisense promoter thathas been shown to control expression of nearby genesthrough formation of chimeric transcripts [66\u00E2\u0080\u009369]. Re-cently, this antisense promoter was also shown to promoteexpression of a small protein ORF0, which plays a regula-tory role in retrotransposition [70]. While there are ap-proximately 500,000 L1 loci in the human genome [1], thevast majority of them are 5\u00E2\u0080\u0099 truncated due to incompletereverse transcription during the retrotranspositionprocess. Only ~3500-7000 are full length, retaining theirpromoters and hence, the potential ability to lend thesepromoters to nearby genes [71, 72]. Therefore, irrespectiveof differences in promoter strength, epigenetic regulationor mutational degradation, the vast copy number differ-ence (~500,000 LTRs versus ~5000 promoter-containingL1s), is likely a major reason why the great majority ofTE-initiated transcripts involve LTRs rather than L1s. Ingenome-wide screens of TE-initiated transcripts, smallfragments of old L2 elements, which do not span the ca-nonical L2 promoter, can be found as TSSs of lowlyexpressed transcripts [73] (unpublished data). Such in-stances likely represent \u00E2\u0080\u009Cde novo\u00E2\u0080\u009D promoters, those arisingnaturally from genomic DNA which happens to be de-rived from a TE fragment, (possibly because L2 fragmentshave a GC rich base composition), rather than an \u00E2\u0080\u009Cances-tral\u00E2\u0080\u009D or \u00E2\u0080\u009Cready-made\u00E2\u0080\u009D promoter, one which utilizes a TE\u00E2\u0080\u0099soriginal regulatory sequence.Human SINE elements, namely ALUs and the olderMIRs, can also promote transcription of nearby genesbut these instances are relatively rare [68] given their ex-tremely high copy numbers (~1.85 million fragments)[50]. This likely partly reflects the fact that SINEs, beingderived from small functional RNAs, inherently possessPolIII promoters, rather than PolII, and their autono-mous promoter strength is weak [74, 75]. Old MIR ele-ments, as well as other ancient SINEs and DNA TEs,have been more prominent as enhancers, rather thangenic promoters, as shown in several studies [76\u00E2\u0080\u009381].TEs and the cancer transcriptomeWhile some TE components have assumed cellular func-tions over evolutionary time, such as the syncytin genesin mammalian placenta, derived from independent ERVenv genes in multiple mammals [6, 44, 82\u00E2\u0080\u009384], the vastmajority of TE/ERV insertions will be neutral or detri-mental to the host. Given the potential for harm, mul-tiple host mechanisms to repress these sequences haveevolved. In mammals, ERV and L1 transcription is sup-pressed in normal cells by DNA methylation and/orhistone modifications as well as many other host fac-tors [9, 85\u00E2\u0080\u009392]. The epigenetic regulation of TEs isrelevant in cancer because epigenetic changes are com-mon in malignancy and frequently associated with mu-tations in \u00E2\u0080\u009Cepigenome-modifying\u00E2\u0080\u009D genes [93\u00E2\u0080\u009397]. Whilethe ultimate effects of many such mutations are not yetclear, their prominence indicates a central role for epi-genomic dysregulation in oncogenesis [94, 98]. Themost well established epigenetic changes are promoterhypermethylation and associated silencing of tumorsuppressor genes [95, 99, 100] as well as genome-wideDNA hypomethylation [101\u00E2\u0080\u0093103]. Hypomethylation ofERVs and L1s in many tumors has been documented[104\u00E2\u0080\u0093106] and general transcriptional up-regulation ofERVs and L1s is often observed in cancers [33, 107\u00E2\u0080\u0093109].However, other studies have shown no significant changesin ERV expression in selected human cancers comparedto corresponding normal tissues [110, 111].General conclusions about overall TE transcriptionalderegulation in malignancy, or in any other biologicalstate, are not always well founded and can depend onthe type and sensitivity of the assay. For example, ex-pression studies that use consensus probes for internalL1 or ERV regions to assay expression by custom micro-arrays or RT-PCR don\u00E2\u0080\u0099t resolve individual loci, so highexpression signals could reflect dispersed transcriptionalactivation of many elements or the high expression ofonly one or a few loci. Such assays typically also cannotdistinguish between expression due to TE promoter de-repression or due to increased transcription of tran-scripts harboring TEs. RNA-Seq has the potential to giveinformation on expression of individual TE loci, but in-terpretations of expression levels can be confounded bymapping difficulties, length of read and sequencingdepth [112]. In any event, in most cases where transcrip-tional up-regulation of TE groups or individual TEs hasbeen detected in cancer, the biological relevance of suchaberrant expression is poorly understood.Onco-exaptation of ERV/TE promotersWe propose that transcriptional up-regulation of LTR(and to a lesser extent L1) promoters is widespread inepigenetically perturbed cells such as cancer cells. Herewe present specific published examples of onco-exaptation of TE-derived promoters affecting protein-coding genes (Table 2, Fig. 1). Although many otherTE-initiated transcripts have been identified in cancerBabaian and Mager Mobile DNA (2016) 7:24 Page 3 of 21cells (see below), in this section we restrict the discus-sion to those cases where some role of the TE-drivengene in cancer or cell growth has been demonstrated.Ectopic and overexpression of protein-coding genesThe most straightforward interaction between a TE pro-moter and a gene is when a TE promoter is activated,initiates transcription, and transcribes a downstreamgene without altering the open reading frame (ORF),thus serving as an alternative promoter. Since the TEpromoter may be regulated differently than the nativepromoter, this can result in ectopic and/or overexpres-sion of the gene, with oncogenic consequences.The first case of such a phenomenon was discoveredin the investigation of a potent oncogene colony stimu-lating factor one receptor (CSF1R) in Hodgkin Lymph-oma (HL). Normally, CSF1R expression is restricted tomacrophages in the myeloid lineage. To understand howthis gene is expressed in HL, a B-cell derived cancer,Lamprecht et al. [113] performed 5\u00E2\u0080\u0099 RACE which re-vealed that the native, myeloid-restricted promoter is si-lent in HL cell lines, with CSF1R expression insteadbeing driven by a solitary THE1B LTR, of the MaLR-ERVL class (Fig. 1a). THE1B LTRs are ancient, found inboth Old and New World primates, and are highlyabundant in the human genome, with a copy number of~17,000 [50, 114] (Table 1). The THE1B-CSF1R tran-script produces a full-length protein in HL, which is re-quired for growth/survival of HL cell lines [113] and isclinically prognostic for poorer patient survival [115].Ectopic CSF1R expression in HL appears to be com-pletely dependent on the THE1B LTR, and CSF1R pro-tein or mRNA is detected in 39\u00E2\u0080\u009348% of HL patientsamples [115, 116].To detect additional cases of onco-exaptation, wescreened whole transcriptomes (RNA-Seq libraries) froma set of HL cell lines as well as from normal human Bcells for TE-initiated transcripts, specifically transcriptsthat were recurrent in HL and not present in normal Bcells [117]. We identified the Interferon RegulatoryFactor 5 gene (IRF5) as a recurrently up-regulated genebeing promoted by a LOR1a LTR located upstream ofthe native/canonical TSS (Fig. 1b). LOR1a LTRs aremuch less abundant compared to THE1 LTRs (Table 1)but are of similar age, with the IRF5 copy havinginserted prior to New World-Old World primate diver-gence. IRF5 has multiple promoters/TSSs and complextranscription [118] and, contrary to the CSF1R case, thenative promoters are not completely silent in HL. How-ever, LTR activity correlates with strong overexpressionof the IRF5 protein and transcript, above normal physio-logical levels [117]. While our study was ongoing, Kreheret al. reported that IRF5 is upregulated in HL and is acentral regulator of the HL transcriptome [119]. More-over, they found that IRF5 is crucial for HL cell survival.Intriguingly, we noted that insertion of the LOR1a LTRcreated an interferon regulatory factor-binding element(IRFE) that overlaps the 5\u00E2\u0080\u0099 end of the LTR. This IRFEwas previously identified to be critical for promoter ac-tivity as a positive feedback loop through binding ofvarious IRFs, including IRF5 itself [120]. Hence, the in-herent promoter motifs of the LTR, coupled with thecreation of the IRFE upon insertion, combined to pro-vide an avenue for ectopic expression of IRF5 in HL.Expression of truncated proteinsIn these cases, a TE-initiated transcript results in the ex-pression of a truncated open reading frame of the af-fected gene, typically because the TE is located in anintron, downstream of the canonical translational startsite. The TE initiates transcription, but the final tran-script structure depends on the position of downstreamsplice sites, and protein expression requires usage of adownstream ATG. Protein truncations can result inoncogenic effects due to loss of regulatory domains orthrough other mechanisms, with a classic example beingTable 2 Activation of oncogenes by Onco-exaptation of TE-derived promotersGenea Gene function Primary result of TE-drivenexpressionTE type TE promoter coordinates(hg38)Cancer type referencesCSF1R Tyrosine kinase receptor Ectopic expression ofnormal protein(ERVL-MaLR) THE1B LTR chr5:150092453\u00E2\u0080\u0093150092809 HL [113]IRF5 Transcription factor Ectopic expression ofnormal protein(ERV1) LOR1a LTR chr7:128936859\u00E2\u0080\u0093128937097 HL [117, 119]MET Tyrosine kinase receptor Protein truncation (L1) L1PA2 chr7:116718498\u00E2\u0080\u0093116724489 CML, others? [124, 125]ALK Tyrosine kinase receptor Protein truncation (ERVL) LTR16B2 chr2:29223783\u00E2\u0080\u009329224196 melanoma [38]ERRB4 Tyrosine kinase receptor Protein truncation (ERVL-MaLR) MLT1C LTR(ERVL-MaLR) MLT1H2 LTRchr2:211693702\u00E2\u0080\u0093211694209chr2:211465146\u00E2\u0080\u0093211465419ALCL [129]SLCO1B3 Anion transporter Chimeric protein (ERV1) LTR7b chr12:20822187\u00E2\u0080\u009320822617 colon, others [133, 136]FABP7 Fatty acid binding Chimeric protein (ERV1) LTR2 chr6:122748805\u00E2\u0080\u0093122749262 DLBCL [138]aOnly those cases with supporting evidence of a role in the cancer are listedbThe fact that the promoter for these isoforms is an LTR was not noted in the cited papersBabaian and Mager Mobile DNA (2016) 7:24 Page 4 of 21Fig. 1 (See legend on next page.)Babaian and Mager Mobile DNA (2016) 7:24 Page 5 of 21v-myb, a truncated form of myb carried by acutely trans-forming animal retroviruses [121, 122].The first such reported case involving a TE was identi-fied in a screen of human ESTs to detect transcriptsdriven by the antisense promoter within L1 elements.M\u00C3\u00A4tlik et al. identified an L1PA2 within the second in-tron of the proto-oncogene MET (MET proto-oncogene,receptor tyrosine kinase) that initiates a transcript bysplicing into downstream MET exons (Fig. 1c) [67]. Notsurprisingly, transcriptional activity of the CpG rich pro-moter of this L1 in bladder and colon cancer cell lines isinversely correlated to its degree of methylation [123, 124].A slightly truncated MET protein is produced by the TE-initiated transcript and one study reported that L1-driventranscription of MET reduces overall MET protein levelsand signaling, although by what mechanism is not clear[124]. Analyses of normal colon tissues and matched pri-mary colon cancers and liver metastasis samples showedthis L1 is progressively demethylated in the metastasissamples, which strongly correlates with increased L1-METtranscripts and protein levels [125]. Since MET levels are anegative prognostic indicator for colon cancer [126], thesefindings suggest an oncogenic role for L1-MET.More recently, Wiesner et al. identified a novel iso-form of the receptor tyrosine kinase (RTK), anaplasticlymphoma kinase (ALK), initiating from an alternativepromoter in its 19th intron [38]. This alternative tran-scription initiation (ATI) isoform or ALKATI was re-ported to be specific to cancer samples and found in~11% of skin cutaneous melanomas. ALKATI transcriptsproduce three protein isoforms encoded by exons 20 to29. These smaller isoforms exclude the extracellular do-main of the protein but contain the catalytic intracellulartyrosine kinase domain. This same region of ALK iscommonly found fused with a range of other genes viachromosomal translocations in lymphomas and a varietyof solid tumors [127]. In the Wiesner et al. study it wasfound that ALKATI stimulates several oncogenic signal-ing pathways, drives cell proliferation in vitro, and pro-motes tumor formation in mice [38].The ALKATI promoter is a sense-oriented solitary LTR(termed LTR16B2) derived from the ancient ERVL fam-ily (Fig. 1d). LTR16B2 elements are found in severalhundred copies in both primates and rodents [50, 114]and this particular element is present in the orthologousposition in mouse. Therefore, the promoter potential ofthis LTR has been retained for at least 70 million years.Although not the first such case, the authors state thattheir findings \u00E2\u0080\u009Csuggest a novel mechanism of oncogeneactivation in cancer through de novo alternative tran-script initiation\u00E2\u0080\u009D. Evidence that this LTR is at least occa-sionally active in normal human cells comes fromCapped Analysis of Gene Expression (CAGE) analysisthrough the FANTOM5 project [128]. A peak of CAGEtags from monocyte-derived macrophages and endothe-lial progenitor cells occurs within this LTR, 60 bp down-stream of the TSS region identified by Wiesner et al.[38] (Fig. 2a), although a biological function, if any, ofthis isoform in normal cells is unknown.To gain a molecular understanding of ALK-negativeanaplastic large-cell lymphoma (ALCL) cases, Scarfo etal. conducted gene expression outlier analysis and identi-fied high ectopic co-expression of ERBB4 and COL29A1in 24% of such cases [129]. Erb-b2 receptor tyrosine kin-ase 4 (ERBB4), also termed HER4, is a member of theERBB family of RTKs, which includes EGFR and HER2,and mutations in this gene have been implicated insome cancers [130]. Analysis of the ERRB4 transcriptsexpressed in these ALCL samples revealed two iso-forms initiated from alternative promoters, one withinintron 12 (I12-ERBB4) and one within intron 20 (I20-ERBB4), with little or no expression from the native/canonical promoter. Both isoforms produce truncatedproteins that show oncogenic potential, either alone(See figure on previous page.)Fig. 1 Examples of Onco-exaptation. Gene models of known TE-derived promoters expressing downstream oncogenes and listed in Table 2.Legend is shown at the top. a 6 kb upstream of CSF1R, a THE1B LTR initiates transcription and contains a splice donor site which joins to an exonwithin a LINE L1MB5 element and then into the first exon of CSF1R. The TE-initiated transcript has a different, longer 5\u00E2\u0080\u0099 UTR than the canonicaltranscript but the same full-length protein coding sequence. b An LOR1a LTR initiates transcription and splices into the canonical second exon ofIRF5 that contains the standard translational initiation site (TIS) to produce a full-length protein. There also is a novel second exon which is non-TE derived which is incorporated into a minor isoform of LOR1a-IRF5. c Within the canonical intron 2 of the proto-oncogene MET, a full lengthLINE L1PA2 element initiates transcription (anti-sense to itself), splicing through a short exon in a SINE MIR element and into the third exon ofMET. The first TIS of the canonical MET transcript is 14 bp into exon 2, although an alternative TIS exists in exon 3, which is believed to also beused by the L1-promoterd isoform. d An LTR16B2 element in intron 19 of the ALK gene initiates transcription and transcribes into the canonicalexon 20 of ALK. An in-frame TIS within the 20th exon results in translation of a shortened oncogenic protein containing only the intra-cellulartyrosine kinase domain, but lacking the transmembrane and extracellular receptor domains of ALK. e There are two TE-promoted isoforms ofERBB4, the minor variant initiates in an MLT1C LTR in the 12th intron and the major variant initiates in a MLT1H LTR in the 20th intron. Bothisoforms produce a truncated protein, although the exact translation start sites are not defined. f In the third exon of SLCO1B3, two adjacent partlyfull-length HERV elements conspire to create a novel first exon. Transcription initiates in the anti-sense orientation from an LTR7 andtranscribes to a sense-oriented splice donor in an adjacent MER4C LTR, which then splices into the fourth exon of SLCO1B3, creating a smallerprotein. g An LTR2 element initiates anti-sense transcription (relative to its own orientation) and splices into the native second exon of FABP7. TheLTR-derived isoform has a non-TE TIS and splice donor which creates a different N-terminal protein sequence of FABP7Babaian and Mager Mobile DNA (2016) 7:24 Page 6 of 21(I12 isoform) or in combination. Remarkably, bothpromoters are LTR elements of the ancient MaLR-ERVL class (Fig. 1e). Of note, Scarfo et al. reportedthat two thirds of ERBB4 positive cases showed a\u00E2\u0080\u009CHodgkin-like\u00E2\u0080\u009D morphology, which is normally foundin only 3% of ALCLs [129]. We therefore examinedour previously published RNA-Seq data from 12 HLcell lines [117] and found evidence for transcriptionfrom the intron 20 MLTH2 LTR in two of these lines(unpublished observations), suggesting that truncatedERBB4 may play a role in some HLs.TE-promoted expression of chimeric proteinsPerhaps the most fascinating examples of onco-exaptationinvolve generation of a novel \u00E2\u0080\u009Cchimeric\u00E2\u0080\u009D ORF via usage ofa TE promoter that fuses otherwise non-coding DNA todownstream gene exons. These cases involve both proteinand transcriptional innovation and the resulting productcan acquire de novo oncogenic potential.The solute carrier organic anion transporter familymember 1B3, encodes organic anion transporting poly-peptide 1B3 (OATP1B3, or SLCO1B3), is a 12-transmembrane transporter with normal expressionand function restricted to the liver [131]. Severalstudies have shown that this gene is ectopicallyexpressed in solid tumors of non-hepatic origin, par-ticularly colon cancer [131\u00E2\u0080\u0093134]. Investigations intothe cause of this ectopic expression revealed that thenormal liver-restricted promoter is silent in these can-cers, with expression of \u00E2\u0080\u009Ccancer-type\u00E2\u0080\u009D (Ct)-OATP1B3being driven from an alternative promoter in the sec-ond canonical intron [133, 134]. While not previouslyreported as being within a TE, we noted that this alter-native promoter maps within the 5\u00E2\u0080\u0099 LTR (LTR7) of apartly full-length antisense HERV-H element that ismissing the 3\u00E2\u0080\u0099 LTR. Expression of HERV-H itself andLTR7-driven chimeric long non-coding RNAs is anoted feature of embryonic stem cells and normal earlyembryogenesis, where several studies indicate an intri-guing role for this ERV group in pluripotency (for re-cent reviews see [8, 10, 60]). A few studies have alsonoted higher general levels of HERV-H transcription incolon cancer [109, 135]. The LTR7-driven isoform ofSLCO1B3 makes a truncated protein lacking the first28 amino acids but also includes protein sequencefrom the LTR7 and an adjacent MER4C LTR (Fig. 1f ).The novel protein is believed to be intracellular and itsrole in cancer remains unclear. However, one studyFig. 2 a UCSC Genome Browser view (hg19) of a portion of the human ALK gene. ALK exon 20 (large blue box) and a part of the upstreamintron are shown, with direction of transcription from right to left. The LTR16B2 alternative promoter shown in the Repeatmasker track as anorange box and the 25 bp region of clustered TSSs in melanoma cells, identified using 5\u00E2\u0080\u0099 RACE by Weiser et al. [38], is shown as a green box TheCAGE track above is from the Fantom5 project [128], with transcriptional direction indicated with a blue arrow. Most CAGE tags are frommonocyte-derived macrophages and endothelial progenitor cells. b UCSC Genome Browser view (hg19) of the region encompassing the SAMMSONlncRNA, which plays an oncogenic role in melanoma [161]. The LTR1A2 promoter is indicated in the Repeatmasker track as an orange box.The ChIP-Seq track for SOX10 was created from a dataset (NCBI Gene Expression Omnibus: GSE61967) generated by Laurette et al. [225] in the501Mel melanoma cell lineBabaian and Mager Mobile DNA (2016) 7:24 Page 7 of 21showed that high expression of this isoform is corre-lated with reduced progression-free survival in coloncancer [136].In another study designed specifically to look for TE-initiated chimeric transcripts, we screened RNA-seq li-braries from 101 patients with diffuse large B-celllymphoma (DLBCL) of different subtypes [137] andcompared to transcriptomes from normal B-cells. Thisscreen resulted in the detection of 98 such transcriptsthat were found in at least two DLBCL cases and nonormals [138]. One of these involved the gene forfatty acid binding protein 7 (FABP7). FABP7, nor-mally expressed in brain, is a member of the FABPfamily of lipid chaperones involved in fatty acid up-take and trafficking [139]. Overexpression of FABP7has been reported in several solid tumor types and isassociated with poorer prognosis in aggressive breastcancer [139, 140]. In 5% of the DLBCL casesscreened, we found that FABP7 is expressed from anantisense LTR2 (the 5\u00E2\u0080\u0099LTR of a HERV-E element)(Fig. 1g). Since the canonical ATG is in the first exonof FABP7, the LTR driven transcript encodes achimeric protein with a different N-terminus (see ac-cession NM_001319042.1) [138]. Functional analysisin DLBCL cell lines revealed that the LTR-FABP7protein isoform is required for optimal cell growthand also has subcellular localization properties dis-tinct from the native form [138].Overall, among all TE types giving rise to chimerictranscripts detected in DLBCL, LTRs were over repre-sented compared to their genomic abundance and,among LTR groups, we found that LTR2 elements andTHE1 LTRs were over represented [138]. As discussedabove, this predominance of LTRs over other TE typesis expected.TE-initiated non-coding RNAs in cancerSince TEs, particularly ERV LTRs, provide a major classof promoters for long non-coding RNAs [56, 141, 142],it is not surprising that multiple LTR-driven lncRNAshave been shown to be involved in cancer. These casescan be broadly divided into those with direct, measur-able oncogenic properties (Table 3) and those with ex-pression correlated with a cancer. It should be notedthat we have likely missed some examples if the natureof the promoter was not highlighted or mentioned inthe original publications. Unlike the coding genes dis-cussed above which have non-TE or native promoters innormal tissues, the lncRNAs described here typicallyhave LTRs as their only promoter in normal or malig-nant cells.TE-initiated LncRNAs with oncogenic propertiesIn an extensive study, Prensner et al. reported that thelncRNA SchLAP1 (SWI/SNF complex antagonist associ-ated with prostate cancer 1) is overexpressed in ~25% ofprostate cancers, is an independent predictor of poorclinical outcomes and is critical for invasiveness and me-tastasis [143]. Intriguingly, they found that SchLAP1 in-hibits the function of the SWI/SNF complex, which isknown to have a tumor suppressor roles [144]. Whilenot mentioned in the main text, the authors report insupplementary data that the promoter for this lncRNAis an LTR (Fig. 3a). Indeed, this LTR is a sense-orientedsolitary LTR12C (of the ERV9 group).Linc-ROR is a non-coding RNA (long intergenic non-protein coding RNA, regulator of reprogramming) pro-moted by the 5\u00E2\u0080\u0099 LTR (LTR7) of a full length HERV-Helement [56] (Fig. 3b) and has been shown to play a rolein human pluripotency [145]. Evidence suggests it actsas a microRNA sponge of miR-145, which is a repressorof the core pluripotency transcription factors Oct4,Nanog and Sox2 [146]. Several recent studies have re-ported an oncogenic role for Linc-ROR in different can-cers by sponging miR-145 [147\u00E2\u0080\u0093149] or through othermechanisms [150, 151].Using Serial Analysis of Gene Expression (SAGE),Rangel et al. identified five Human Ovarian cancer Spe-cific Transcripts (HOSTs) that were expressed in ovariancancer but not in other normal cells or cancer typesTable 3 LTR-driven LncRNAs with oncogenic rolelncRNAa TE type TE promoter coordinates (hg38) Cancer type referencesSchLAP1 (ERV1) LTR12C chr2:180691205\u00E2\u0080\u0093180692425 prostate [143]ROR (ERV1) LTR7 chr18:57072052\u00E2\u0080\u009357072502 breast, others [147, 150]HOST2 (ERV1) LTR2B chr10:84171987\u00E2\u0080\u009384172465 ovarian [154]AFAP1-AS1 (ERVL-MaLR) THE1A LTRb chr4:7753884\u00E2\u0080\u00937754236 several [156, 158]SAMMSON (ERV1) LTR1A2b chr3:69999501\u00E2\u0080\u009370000359 melanoma [161]HULC (ERVL-MaLR) MLT1A LTR chr6:8652095\u00E2\u0080\u00938652454 liver [163]UCA1 (ERV1) LTR7C chr19:15828738\u00E2\u0080\u009315829200 several [165, 167]BANCR (ERV1) MER41B LTR chr9:69306939\u00E2\u0080\u009369307567 melanoma, others [169]aOnly those cases with supporting evidence of a role in the cancer are listedbThe fact that the promoter is an LTR was not previously notedBabaian and Mager Mobile DNA (2016) 7:24 Page 8 of 21examined [152]. One of these, HOST2, is annotated as aspliced lncRNA entirely contained within a full lengthHERV-E and promoted by an LTR2B element (Fig. 3c).Perusal of RNA-Seq from the 9 core ENCODE cell linesshows robust expression of HOST2 in GM12878, a B-lymphoblastoid cell line, which extends beyond theHERV-E. As with Linc-ROR, HOST2 appears to play anoncogenic role by functioning as a miRNA sponge ofmiRNA let-7b, an established tumor suppressor [153], inepithelial ovarian cancer [154].The Ref-Seq annotated lncRNA AFAP1 antisense RNA1 (AFAP1-AS1) runs antisense to the actin filament asso-ciated protein 1 (AFAP1) gene and several publicationsreport its up-regulation and association with poor sur-vival in a number of solid tumor types [155\u00E2\u0080\u0093158]. Whilethe oncogenic mechanism of AFAP1-AS1 has not beenextensively studied, one report presented evidence that itpromotes cell proliferation by upregulating RhoA/Rac2signaling [159] and its expression inversely correlateswith AFAP1. Although clearly annotated as initiatingwithin a solitary THE1A LTR (Fig. 3d), this fact has notbeen mentioned in previous publications. In screens forTE-initiated transcripts using RNA-seq data from HLcell lines, we noted recurrent and cancer-specific up-regulation of AFAP1-AS1 (unpublished observations),suggesting that it is not restricted to solid tumors. Theinverse correlation of expression between AFAP1 andAFAP1-AS1 suggests an interesting potential mechanismby which TE-initiated transcription may suppress a gene;where an anti-sense TE-initiated transcript disrupts thetranscription, translation or stability of a tumor suppres-sor gene transcript through RNA interference [160].Fig. 3 Gene models of select lncRNAs initiating within LTRs that are involved in oncogenesis. a A solitary LTR12C element initiates SChLAP1, along inter-genic non-coding RNA. b The 5\u00E2\u0080\u0099 LTR7 of a full-length HERVH element initiates the lncRNA ROR, with an exon partially incorporatinginternal ERV sequence. c The HOST2 lncRNA is completely derived from components of a Harlequin (or HERV-E) endogenous retrovirus and itsflanking LTR2B. d Anti-sense to the AFAP1 gene, a THE1A LTR initiates transcription of the lncRNA AFAP1-AS1. The second exon of AFAP1-AS1overlaps exons 14\u00E2\u0080\u009316 of AFAP1, possibly leading to RNA interference of the geneBabaian and Mager Mobile DNA (2016) 7:24 Page 9 of 21The SAMMSON lncRNA (survival associated mito-chondrial melanoma specific oncogenic non-codingRNA), which is promoted by a solitary LTR1A2 element,was recently reported as playing an oncogenic role inmelanoma [161]. This lncRNA is located near themelanoma-specific oncogene MITF and is always in-cluded in genomic amplifications involving MITF. Evenin melanomas with no genomic amplification of thislocus, SAMMSON is expressed in most cases, increasesgrowth and invasiveness and is a target for SOX10 [161],a key TF in melanocyte development which is deregu-lated in melanoma [162]. Interestingly, the two SOX10binding sites near the SAMMSON TSS lie just upstreamand downstream of the LTR (Fig. 2b), suggesting thatboth the core promoter motifs provided by the LTR andadjacent enhancer sites combine to regulate SAMMSON.Other examples of LTR-promoted oncogenic lncRNAsinclude HULC for Highly Upregulated in Liver Cancer[163, 164], UCA1 (urothelial cancer associated 1)[165\u00E2\u0080\u0093168] and BANCR (BRAF-regulated lncRNA 1)[169\u00E2\u0080\u0093171]. Although not mentioned in the originalpaper, three of the four exons of BANCR were shownto be derived from a partly full length MER41 ERV,with the promoter within the 5\u00E2\u0080\u0099LTR of this elementannotated MER41B [141]. Intriguingly, MER41 LTRswere recently shown to harbor enhancers responsiveto interferon, indicating a role for this ERV group inshaping the innate immune response in primates [52].It would be interesting to investigate roles for BANCRwith this in mind.TE-initiated lncRNAs as cancer-specific markersThere are many examples of TE-initiated RNAs with po-tential roles in cancer or which are preferentiallyexpressed in malignant cells but for which a direct onco-genic function has not yet been demonstrated. Still, suchtranscripts may underlie a predisposition for transcrip-tion of specific groups of LTRs/TEs in particular malig-nancies and therefore function as a marker for a canceror cancer subtype. Since these events potentially do notconfer a fitness advantage for the cancer cell, they arenot \u00E2\u0080\u009Cexaptations\u00E2\u0080\u009D but \u00E2\u0080\u009Cnonaptations\u00E2\u0080\u009D [4].One of these is a very long RNA initiated by the anti-sense promoter of an L1PA2 element as reported byTufarelli\u00E2\u0080\u0099s group and termed LCT13 [172, 173]. EST evi-dence indicates splicing from the L1 promoter to theGNTG1 gene, located over 300 kb away. The tumor sup-pressor gene, tissue factor pathway inhibitor 2, (TFPI-2),which is often epigenetically silenced in cancers [174], isantisense to LCT13 and it was shown that LCT13 tran-script levels are correlated with down regulation ofTFPI-2 and associated with repressive chromatin marksat the TFPI-2 promoter [172].Gibb et al. analyzed RNA-Seq from colon cancers andmatched normal colon to find cancer-associatedlncRNAs and identified an RNA promoted by a solitaryMER48 LTR, which they termed EVADR, for Endogen-ous retroviral-associated ADenocarcinoma RNA [175].Screening of data from The Cancer Genome Atlas(TCGA) [176] showed that EVADR is highly expressedin several types of adenocarcinomas, it is not associatedwith global activation of MER48 LTRs across the gen-ome and its expression correlated with poorer survival[175]. In another study, Gosenca et al. used a custommicroarray to measure overall expression of severalHERV groups in urothelial carcinoma compared to nor-mal urothelial tissue and generally found no difference[111]. However, they found one full-length HERV-Eelement, located in the antisense direction in an intronof the PLA2G4A gene that is transcribed in urothelialcarcinoma and appears to modulate PLA2G4A expres-sion, thereby possibly contributing to carcinogenesis, al-though the mechanism is not clear.By mining long nuclear RNA datasets from ENCODEcell lines, normal blood and Ewing sarcomas, one groupidentified over 2000 very long (~50\u00E2\u0080\u0093700 kb) non codingtranscripts termed vlincRNAs [142]. They found thepromoters for these vlincRNAs to be enriched in LTRs,particularly for cell type-specific vlincRNAs, and themost common transcribed LTR types varied in differentcell types. Moreover, among the datasets examined, theyreported that the number of LTR-promoted vlincRNAscorrelated with degree of malignant transformation,prompting the conclusion that LTR-controlled vlincR-NAs are a \u00E2\u0080\u009Challmark\u00E2\u0080\u009D of cancer [142].In a genome-wide CAGE analysis of 50 hepatocellularcarcinoma (HCC) primary samples and matched non-tumor tissue, Hashimoto et al. found that many LTR-promoted transcripts are upregulated in HCC, most ofthese apparently associated with non-coding RNAs asthe CAGE peaks in the LTRs are far from annotatedprotein coding genes [177]. Similar results were found inmouse HCC. Among the hundreds of human LTRgroups, they found the LTR-associated CAGE peaks tobe significantly enriched in LTR12C (HERV9) LTRs andmapped the common TSS site within these elements,which agrees with older studies on TSS mapping of thisERV group [178]. Moreover, this group reported thatHCCs with highest LTR activity mostly had a viral(Hepatitis B) etiology, were less differentiated and hadhigher risk of recurrence [177]. This study suggestswidespread tissue-inappropriate transcriptional activityof LTRs in HCC.LTR12s as flexible promoters in cancer and normal tissuesMost recent human ERV LTR research has been fo-cused on HERV-H (LTR7/7Y/7B/7C) due to roles forBabaian and Mager Mobile DNA (2016) 7:24 Page 10 of 21HERV-H/LTR7-driven RNAs in pluripotency [56\u00E2\u0080\u009358,60, 179, 180] or on the youngest HERV group,HERV-K (LTR5/5Hs), due to its expression in earlyembryogenesis [181\u00E2\u0080\u0093183], coding capacity of somemembers [30, 184] and potential roles for its proteinsin cancer and other diseases [30\u00E2\u0080\u009333, 185]. LTR12s(including LTR12B,C,D,E and F subtypes), which arethe LTRs associated with the HERV-9 group [186],are generally of similar age to HERV-H [187] but aremuch more numerous than HERV-H or HERV-K,with solitary LTRs numbering over 6000 (Table 1).There are several examples of LTR12s providing pro-moters for coding genes or lncRNAs in various normaltissues [63, 188\u00E2\u0080\u0093191]. LTR12s, particularly LTR12C, arelonger and more CpG rich than most other ERV LTRs,possibly facilitating development of diverse inherenttissue-specificities and flexible combinations of TF bind-ing sites, which may be less probable for other LTR types.For example, the consensus LTR7 (HERV-H) is 450 bpwhereas LTR12C (of similar age) is 1577 bp [114], whichis usually long for retroviral LTRs. As noted above, LTR12elements are among the most enriched LTR types acti-vated as promoters in HCC [177] and appear to be themost active LTR type in K562 cells [142]. It is importantto point out, however, that only a very small fraction ofgenomic LTR12 copies are transcriptionally active in anyof these contexts, so general conclusions about activity of\u00E2\u0080\u0098a family of LTRs\u00E2\u0080\u0099 should be made with caution.A number of other recent investigations on LTR12-driven chimeric transcription have been published. Onestudy specifically screened for and detected numerousLTR12-initiated transcripts in ENCODE cell lines, someof which extend over long genomic regions and emanatefrom bidirectional promoters within these LTRs [192].The group of Dobbelstein discovered that a male germline-specific form of the tumor suppressor TP63 gene isdriven by an LTR12C [190]. Interestingly, they foundthat this LTR is silenced in testicular cancer but reacti-vated upon treatment with histone deacetylase inhibitors(HDACi), which also induces apoptosis [190]. In follow-up studies, this group used 3\u00E2\u0080\u0099 RACE to detect moregenes controlled by LTR12s in primary human testis andin the GH testicular cancer cell line and reported hun-dreds of transcripts, including an isoform of TNFRSF10Bwhich encodes the death receptor DR5 [193]. As withTP63, treating GH or other cancer cell lines with HDACinhibitors such as trichostatin A activated expression ofthe LTR12-driven TNFRSF10B and some other LTR12-chimeric transcripts and induced apoptosis [193, 194].Therefore, in some cases, LTR-driven genes can have aproapoptotic role. In accord with this notion is a studyreporting that LTR12 antisense U3 RNAs wereexpressed at higher levels in non-malignant versus ma-lignant cells [195]. It was proposed that the antisenseU3 RNA may act as a trap for the transcription factorNF-Y, known to bind LTR12s [196], and hence partici-pate in cell cycle arrest [195].Chromosomal translocations involving TEs in cancerActivation or creation of oncogenes via chromosomaltranslocations most commonly involves either the fusionof two coding genes or juxtaposition of new regulatorysequences next to a gene, resulting in oncogenic effectsdue to ectopic expression [197]. One might expect someof the latter cases to involve TE-derived promoters/en-hancers but, to date, there are very few well-documentedexamples of this mechanism in oncogenesis. The ETSfamily member ETV1 (ETS variant 1) is a transcriptionfactor frequently involved in oncogenic translocations,particularly in prostate cancer [198]. Although not acommon translocation, Tomlins et al. identified a pros-tate tumor with the 5\u00E2\u0080\u0099 end of a HERV-K (HML-2) elem-ent on chromosome 22q11.23 fused to ETV1 [199]. Thisparticular HERV-K element is a complex locus with two5\u00E2\u0080\u0099 LTRs and is quite highly expressed in prostate cancer[200]. Indeed, while a possible function is unknown, thisHERV-K locus produces a lncRNA annotated as PCAT-14, for prostate cancer\u00E2\u0080\u0093associated ncRNA transcript-14[201]. In the HERV-K-ETV1 fusion case, the resultanttranscript (Genbank Accession EF632111) initiates inthe upstream 5\u00E2\u0080\u0099LTR, providing evidence that the LTRcontrols expression of ETV1.The fibroblast growth factor receptor 1 (FGFR1) geneon chromosome 8 is involved in translocations with atleast 14 partner genes in stem cell myeloproliferativedisorder and other myeloid and lymphoid cancers [202].One of these involves a HERVK3 element on chromo-some 19 and this event creates a chimeric ORF withHERVK3 gag sequences [203]. While it was reportedthat the LTR promoter may contribute to expression ofthe fusion gene [203], no supporting evidence was pre-sented. Indeed, perusal of public expression data(Expressed sequence tags) from a variety of tissues indi-cates that the HERVK3 element on chromosome 19 ishighly expressed, but from a non-ERV promoter just up-stream (see chr19:58,305,253\u00E2\u0080\u009358,315,303 in human hg38assembly). Therefore, there is little current evidence forLTR/TE promoters playing a role in oncogene activationvia chromosomal translocations or rearrangements.Models for onco-exaptationThe aforementioned cases of onco-exaptation are a dis-tinct mechanism by which proto-oncogenes becomeoncogenic. Classical activating mutations within TEsmay also lead to transcription of downstream oncogenesbut we are unaware of any evidence for DNA mutationsresulting in LTR/TE transcriptional activation, includingcases where local DNA was sequenced [38] (unpublishedBabaian and Mager Mobile DNA (2016) 7:24 Page 11 of 21results). Thus, it is important to consider the etiologythrough which LTRs/TEs become incorporated into newregulatory units in cancer. The mechanism could pos-sibly be therapeutically or diagnostically important andperhaps even model how TEs influence genome regula-tion in evolutionary time.In some of the above examples, there is no or very lit-tle detectable transcription from the LTR/TE in any celltype other than the cancer type in which it was reported,suggesting the activity is specific to a particular TE in aparticular cancer. In other cases, CAGE or EST datashow that the LTR/TE can be expressed in other normalor cancer cell types, perhaps to a lower degree. Hencethe term \u00E2\u0080\u009Ccancer-specific\u00E2\u0080\u009D should be considered a rela-tive one. Indeed, the idea that the same TE-promotedgene transcripts occur recurrently in tumors from inde-pendent individuals is central to understanding howthese transcripts arise. Below we present two modelsthat may explain the phenomenon of onco-exaptation.The De-repression modelLamprecht and co-workers proposed a \u00E2\u0080\u0098De-repressionmodel\u00E2\u0080\u0099 for the LTR driven transcription of CSF1R [204].The distinguishing feature of this model is that onco-exaptations arise deterministically, as a consequence ofmolecular changes that occur during oncogenesis,changes which act to de-repress LTRs or other TEs(Fig. 4). It follows that \u00E2\u0080\u0098activation\u00E2\u0080\u0099 of normally dormantTEs/LTRs could lead to robust oncogene expression. Inthe CSF1R case, the THE1B LTR, which promotesCSF1R in HL, contains binding sites for the transcriptionfactors Sp1, AP-1 and NF-kB, each of which contributesto promoter activity in a luciferase reporter experiment[113]. High NF-kB activity, which is known to be up-regulated in HL, loss of the epigenetic corepressorCBFA2T3 as well as LTR hypomethylation all correlatedwith CSF1R-positive HL driven by the LTR [113]. Underthe de-repression model, the THE1B LTR is repressed bydefault in the cell but under a particular set ofFig. 4 De-repression model for onco-exaptation. In the normal or pre-malignant state TEs (grey triangles) are largely silenced across the genome.There is low transcriptional activity to produce long non-coding RNA (orange box), or express coding genes in the case of evolutionary exaptations(not shown). The example proto-oncogene (green box) is under the regulatory control of its native, restrictive promoter. During the process oftransformation and/or oncogenesis, a change in the molecular state of the cell occurs leading to loss of TE repressors (black circles), i.e. DNAhypomethylation, loss of transcriptional or epigenetic repressive factors. The change could also be accompanied by a change/gain in activating factoractivities (red and purple shapes). Together these de-repression events result in higher TE promoter activity (orange triangles) and more TE-derivedtranscripts based on the factors that become deregulated. Oncogenic activation of proto-oncogenes is a consequence of a particular molecular milieuthat arises in the cancerous cellsBabaian and Mager Mobile DNA (2016) 7:24 Page 12 of 21conditions (gain of NF-kB, loss of CBFA2T3, loss ofDNA methylation) the LTR promoter is remodeled intoan active state [204]. More generally, the model proposesthat a particular LTR activation is a consequence of thepathogenic or disrupted molecular state of the cancercell. In a similar vein, Weber et al. proposed that the L1-driven transcription of MET arose as a consequence ofglobal DNA hypomethylation and loss of repression ofTEs in cancer [124].The LOR1a-IRF5 onco-exaptation in HL [117] can beinterpreted using a de-repression model. An interferonregulatory factor binding element site was created at theintersection of the LOR1a LTR and genomic DNA. Innormal and HL cells negative for LOR1a-IRF5, the LTRis methylated and protected from DNAse digestion, astate that is lost in de-repressed HL cells. This transcrip-tion factor-binding motif is responsive to IRF5 itself andcreates a positive feedback loop between the IRF5 andthe chimeric LOR1a-IRF5 transcript. Thus epigeneticde-repression of this element may reveal an oncogenicexploitation, resulting in high recurrence of LOR1a LTR-driven IRF5 in HL [117].A de-repression model explains several experimentalobservations, such as the necessity for a given set of fac-tors to be present (or absent) for a certain promoter tobe active, especially when those factors differ betweencell states. Indeed, experiments probing the mechanismof TE/LTR activation have used this line of reasoning,often focusing on DNA methylation [113, 117, 125, 129].The limitation of these studies is that they fail to deter-mine if a given condition is sufficient for onco-exaptation to arise. For instance, the human genomecontains >37,000 THE1 LTR loci (Table 1), and indeedthis set of LTRs is generally more active in HL cellscompared to B-cells as would be predicted [113] (un-published results). The critical question is why particu-lar THE1 LTR loci, such as THE1B-CSF1R, arerecurrently de-repressed in HL, yet thousands of hom-ologous LTRs are not.The Epigenetic Evolution modelA central premise in the TE field states that TEs can bebeneficial to a host genome since they increase genetic vari-ation in a population and thus increase the rate at whichevolution (by natural selection) occurs [62, 205, 206]. Theepigenetic evolution model for onco-exaptation (Fig. 5)draws a parallel to this premise within the context oftumor evolution.Key to the epigenetic evolution model is that there ishigh epigenetic variance, both between LTR loci and atthe same LTR locus between cells in a population. Thisepigenetic variance fosters regulatory innovation, and in-creases during oncogenesis. In accord with this idea areseveral studies showing that DNA methylation variation,or heterogeneity, increases in tumor cell populations andthis isn\u00E2\u0080\u0099t simply a global hypomethylation relative tonormal cells [207\u00E2\u0080\u0093209] (reviewed in [210]). In contrastto the de-repression model, a particular pathogenic mo-lecular state is not sufficient or necessary for TE-driventranscripts to arise; instead the given state only dictateswhich sets of TEs in the genome are permissive for tran-scription. Likewise, global de-repression events, such asDNA hypomethylation or mutation of epigenetic regula-tors, are not necessary, but would increase the rate atwhich novel transcriptional regulation evolves.Underpinning this model is the idea that LTRs arehighly abundant and self-contained promoters dispersedacross the genome that can stochastically initiate low ornoisy transcription. This transcriptional noise is a kindof epigenetic variation and thus contributes to cell-cellvariation in a population. Indeed, by re-analyzing CAGEdatasets of retrotransposon-derived TSSs published byFaulkner et al. [73], we observed that TE-derived TSSshave lower expression levels and are less reproducible be-tween biological replicates, compared to non-TE pro-moters (unpublished observations). During malignanttransformation, TFs can become deregulated and genome-wide epigenetic perturbations occur [94, 98, 211] whichwould change the set of LTRs that are potentially active aswell as possibly increasing the total level of LTR-driventranscriptional noise. Up-regulation of specific LTR-driventranscripts would initially be weak and stochastic, fromthe set of permissive LTRs. Those cells gaining an LTR-driven transcript which confers a growth advantage wouldthen be selected for, and the resultant oncogene expres-sion would increase in the tumor population as that epial-lele increases in frequency, in a similar fashion asproposed for the epigenetic silencing of tumor suppressorgenes [95, 99, 100]. Notably, this scenario also means thatwithin a tumor, LTR-driven transcription would be subjectto epigenetic bottleneck effects as well, and that transcrip-tional LTR noise can become \u00E2\u0080\u009Cpassenger\u00E2\u0080\u009D expression sig-nals as the cancer cells undergo somatic, clonal evolution.It may be counter-intuitive to think of evolution andselection as occurring outside the context of geneticvariation, but the fact that both genetic mutations andnon-genetic/epigenetic variants can contribute to somaticevolution of a cancer is becoming clear [209, 212\u00E2\u0080\u0093215].Epigenetic information or variation by definition is trans-mitted from mother to daughter cells. Thus, in the specificcontext of a somatic/asexual cell population such as atumor, this information, which is both variable betweencells in the population and heritable, will be subject toevolutionary changes in frequency. DNA methylation inparticular has a well-established mechanism by which in-formation (mainly gene repression) is transmitted epige-netically from mother to daughter cells [216] and DNAhypomethylation at LTRs often correlates with theirBabaian and Mager Mobile DNA (2016) 7:24 Page 13 of 21Fig. 5 Epigenetic evolution model for onco-exaptation. In the starting cell population there is a dispersed and low/noisy promoter activity at TEs(colored triangles) from a set of transcriptionally permissive TEs (grey triangles). TE-derived transcript expression is low and variable between cells.Some transcripts are more reliably measurable (orange box). Clonal tumor evolutionary forces change the frequency and expression of TE-derivedtranscripts by homogenizing epialleles and use of TE promoters (highlighted haplotype). A higher frequency of \u00E2\u0080\u0098active\u00E2\u0080\u0099 TE epialleles at a locus results inincreased measurable transcripts initiating from that position. TE epialleles that promote oncogenesis, namely onco-exaptations, can be selected forand arise multiple times independently as driver epialleles, in contrast to the more dispersed passenger epialleles, or \u00E2\u0080\u009Cnonaptations\u00E2\u0080\u009DBabaian and Mager Mobile DNA (2016) 7:24 Page 14 of 21expression [113, 117, 217]. Thus, this model suggests thatone important type of \u00E2\u0080\u009Cepigenetic variant\u00E2\u0080\u009D or epiallele isthe transcriptional status of the LTR itself, since thephenotypic impact of LTR transcription may be high inonco-exaptation. Especially in light of the fact that largenumbers of these highly homologous sequences arespread across the genome, epigenetic variation, and pos-sibly selection, at LTRs creates a fascinating system bywhich epigenetic evolution in cancer may occur.ConclusionsHere we have reviewed the growing number of exam-ples of LTR/TE onco-exaptation. Although such TEshave the potential to be deleterious by contributing tooncogenesis if transcriptionally activated, their fixationin the genome and ancient origin suggests that theirpresence is not subject to significant negative selection.This could be due to the low frequency of onco-exaptation at a particular TE locus and/or to the factthat cancer is generally a disease that occurs after thereproductive years. However, it is generally assumedthat negative selection is the reason why TEs are under-represented near or within genes encoding develop-mental regulators [218\u00E2\u0080\u0093220]. Similarly we hypothesizethat LTR/TE insertions predisposed to causing potentonco-exaptations at a high frequency would also be de-pleted by selective forces.In this review we have also presented two models thatmay explain such onco-exaptation events. These twomodels are not mutually exclusive but they do providealternative hypotheses by which TE-driven transcriptionmay be interpreted. This dichotomy is possibly best ex-emplified by the ERBB4 case (Fig. 1e) [129]. There aretwo LTR-derived promoters which result in aberrantERBB4 expression in ALCL. From the de-repressionmodel viewpoint, both LTR elements are grouped MLT1(MLT1C and MLT1H) and thus this group can be inter-preted as de-repressed. From the epigenetic evolutionmodel viewpoint, this is convergent evolution/selectionfor onco-exaptations involving ERBB4.Through application of the de-repression model, TE-derived transcripts could be used as a diagnostic markerin cancer. If the set of TE/LTR derived transcripts are adeterministic consequence of a given molecular state, byunderstanding which set of TEs correspond to whichmolecular state, it might be possible to assay cancersamples for functional molecular phenotypes. In HL forexample, CSF1R status is prognostically important [115]and this is dependent on the transcriptional state of asingle THE1B. HL also has a specific increase in THE1LTR transcription genome-wide (unpublished observa-tions). Thus, it\u00E2\u0080\u0099s reasonable to hypothesize that the prog-nostic power can be increased if the transcriptionalstatus of all THE1 LTRs is considered. A set of LTRs canthen be interpreted as an in situ \u00E2\u0080\u0098molecular sensor\u00E2\u0080\u0099 foraberrant NF-kB function in HL/B-cells for instance.The epigenetic evolution model proposes that LTR-driven transcripts can be interpreted as a set of epimuta-tions in cancer, similar to how oncogenic mutations areanalyzed. Genes that are recurrently (and independently)onco-exapted in multiple different tumors of the samecancer type may be a mark of selective pressure for ac-quiring that transcript. This is distinct from the more di-verse/noisy \u00E2\u0080\u009Cpassenger LTR\u00E2\u0080\u009D transcription occurringacross the genome. These active but \u00E2\u0080\u009Cpassenger LTRs\u00E2\u0080\u009Dmay be expressed to a high level within a single tumorpopulation due to epigenetic drift and population bot-tlenecks but would be more variable across different tu-mors. Thus analysis of recurrent and cancer-specificTE-derived transcripts may enrich for genes of signifi-cance to tumor biology.While we focused in this review on TE-initiated tran-scription in cancer, many of the concepts presented herecan be applied to other regulatory functions of TEs suchas enhancers, insulators, or repressors of transcription.Although less straightforward to measure, it is probablethat perturbations to such TE regulatory functions con-tribute to some malignancies. Furthermore, severalstudies have shown that TEs play substantial roles incryptic splicing in humans [221\u00E2\u0080\u0093223] and thus may bea further substrate of transcriptional innovation in can-cer, particularly since DNA methylation state can affectsplicing [224].Regardless of the underlying mechanism, onco-exaptation offers a tantalizing opportunity to modelevolutionary exaptation. Specifically, questions such as\u00E2\u0080\u009CHow do TEs influence the rate of transcriptional/regu-latory change?\u00E2\u0080\u009D can be tested in cell culture experi-ments. As more studies that focus on regulatoryaberrations in cancer are performed in the comingyears, we predict that this phenomenon will become in-creasingly recognized as a significant force shapingtranscriptional innovation in cancer. Moreover, wepropose that studying such events will provide insightinto how TEs have contributed to reshaping transcrip-tional patterns during species evolution.AbbreviationsAFAP1-AS1: AFAP1 antisense RNA 1; ALCL: Anaplastic large-cell lymphoma;ALK: Anaplastic lymphoma kinase; BANCR: BRAF-regulated lncRNA 1;CAGE: Capped analysis of gene expression; CSFIR: Colony stimulating factorone receptor; DLBCL: Diffuse large B-cell lymphoma; ERBB4: Erb-b2 receptortyrosine kinase 4; ERV: Endogenous retrovirus; EST: Expressed sequence tag;ETV1: ETS variant 1; EVADR: Endogenous retroviral-associatedAdenocarcinoma RNA; FABP7: Fatty acid binding protein 7;HCC: Hepatocellular carcinoma; HL: Hodgkin lymphoma; HOST2: Humanovarian cancer specific transcript-2; HULC: Highly upregulated in liver cancer;IRF5: Interferon regulatory Factor 5; IRFE: Interferon regulatory factor-bindingelement; Linc-ROR: Long intergenic non-protein coding RNA, regulator ofreprogramming; LINE-1: L1: Long interspersed repeat-1; LncRNA: Long non-coding RNA; LTR: Long terminal repeat; MET: MET proto-oncogene, receptortyrosine kinase; OAT1B3: Organic anion transporting polypeptide 1B3;Babaian and Mager Mobile DNA (2016) 7:24 Page 15 of 21SAMMSON: Survival associated mitochondrial melanoma specific oncogenicnon-coding RNA; SchLAP1: SWI/SNF complex antagonist associated withprostate cancer 1; SINE: Short interspersed element; SLCO1B3: Solute carrierorganic anion transporter family member 1B3; TCGA: The cancer genomeatlas; TE: Transposable element; TF: Transcription factor; TFPI-2: Tissue factorpathway inhibitor 2; TIS: Translation initiation site; TSS: Transcriptional startsite; UCA1: Urothelial cancer associated 1AcknowledgmentsWe thank Matt Lorincz and the anonymous reviewers for comments andhelpful suggestions on this manuscript. We apologize to colleagues andother researchers if we failed to cite relevant work on this subject.FundingWork on this topic in our laboratory has been funded by grants from theCanadian Institutes of Health Research (CIHR), the Natural Sciences andEngineering Research Council of Canada (NSERC), the Canadian CancerSociety and the Leukemia and Lymphoma Society of Canada, with coresupport provided by the BC Cancer Agency. AB is supported by astudentship award from NSERC.Availability of data and materialsData sharing not applicable as no datasets were generated or analyzedduring the current study.Authors\u00E2\u0080\u0099 contributionsAB and DLM wrote the manuscript and both authors approved the final version.Competing interestsThe authors declare that they have no competing interests.Consent for publicationNot applicable.Received: 1 September 2016 Accepted: 11 November 2016References1. International Human Genome Sequencing Consortium. Initial sequencingand analysis of the human genome. Nature. 2001;409(6822):860\u00E2\u0080\u0093921.2. Jurka J, Kapitonov VV, Kohany O, Jurka MV. Repetitive Sequences inComplex Genomes: Structure and Evolution. Annu Rev Genomics HumGenet. 2007;8(1):241\u00E2\u0080\u009359.3. Brosius J, Gould SJ. On \"genomenclature\": a comprehensive (and respectful)taxonomy for pseudogenes and other \"junk DNA\". Proc Natl Acad Sci. 1992;89(22):10706\u00E2\u0080\u009310.4. Gould SJ, Vrba ES. Exaptation-A Missing Term in the Science of Form.Paleobiology. 1982;8(1):4\u00E2\u0080\u009315.5. Feschotte C, Gilbert C. Endogenous viruses: insights into viral evolution andimpact on host biology. Nat Rev Genet. 2012;13(4):283\u00E2\u0080\u009396.6. Rebollo R, Romanish MT, Mager DL. Transposable Elements: An Abundantand Natural Source of Regulatory Sequences for Host Genes. Annu RevGenet. 2012;46:21\u00E2\u0080\u009342.7. Richardson SR, Doucet AJ, Kopera HC, Moldovan JB, Garcia-P\u00C3\u00A9rez JL, Moran JV:The Influence of LINE-1 and SINE Retrotransposons on Mammalian Genomes.Microbiol Spectr 2015, 3(2):10.1128/microbiolspec.MDNA1123-0061-20148. Robbez-Masson L, Rowe H. Retrotransposons shape species-specificembryonic stem cell gene expression. Retrovirology. 2015;12(1):45.9. Hancks DC, Kazazian HH. Roles for retrotransposon insertions in humandisease. Mob DNA. 2016;7(1):1\u00E2\u0080\u009328.10. Gerdes P, Richardson SR, Mager DL, Faulkner GJ. Transposable elements inthe mammalian embryo: pioneers surviving through stealth and service.Genome Biol. 2016;17(1):1\u00E2\u0080\u009317.11. Weiss RA. Human endogenous retroviruses: friend or foe? APMIS. 2016;124(1\u00E2\u0080\u00932):4\u00E2\u0080\u009310.12. Elbarbary RA, Lucas BA, Maquat LE. Retrotransposons as regulators of geneexpression. Science. 2016;351(6274):aac7247.13. Brouha B, Schustak J, Badge RM, Lutz-Prigge S, Farley AH, Moran JV,Kazazian Jr HH. Hot L1s account for the bulk of retrotransposition in thehuman population. PNAS. 2003;100(9):5280\u00E2\u0080\u00935.14. Chen J, Stenson P, Cooper D, Ferec C. A systematic analysis of LINE-1endonuclease-dependent retrotranspositional events causing humangenetic disease. Hum Genet. 2005;117(5):411\u00E2\u0080\u009327.15. Kaer K, Speek M. Retroelements in human disease. Gene. 2013;518(2):231\u00E2\u0080\u009341.16. Miki Y, Nishisho I, Horii A, Miyoshi Y, Utsunomiya J, Kinzler KW, Vogelstein B,Nakamura Y. Disruption of the APC gene by a retrotransposal insertion of L1sequence in a colon cancer. Cancer Res. 1992;52(3):643\u00E2\u0080\u00935.17. Lee E, Iskow R, Yang L, Gokcumen O, Haseley P, Luquette LJ, Lohr JG, HarrisCC, Ding L, Wilson RK, et al. Landscape of Somatic Retrotransposition inHuman Cancers. Science. 2012;337(6097):967\u00E2\u0080\u009371.18. Solyom S, Ewing AD, Rahrmann EP, Doucet TT, Nelson HH, Burns MB, HarrisRS, Sigmon DF, Casella A, Erlanger B, et al. Extensive somatic L1retrotransposition in colorectal tumors. Genome Res. 2012;22(12):2328\u00E2\u0080\u009338.19. Shukla R, Upton KR, Munoz-Lopez M, Gerhardt DJ, Fisher ME, Nguyen T,Brennan PM, Baillie JK, Collino A, Ghisletti S, et al. Endogenousretrotransposition activates oncogenic pathways in hepatocellularcarcinoma. Cell. 2013;153(1):101\u00E2\u0080\u009311.20. Tubio JMC, Li Y, Ju YS, Martincorena I, Cooke SL, Tojo M, Gundem G,Pipinikas CP, Zamora J, Raine K, et al. Extensive transduction ofnonrepetitive DNA mediated by L1 retrotransposition in cancer genomes.Science. 2014;345(6196):1251343.21. Rodic N, Steranka JP, Makohon-Moore A, Moyer A, Shen P, Sharma R,Kohutek ZA, Huang CR, Ahn D, Mita P, et al. Retrotransposon insertions inthe clonal evolution of pancreatic ductal adenocarcinoma. Nat Med. 2015;21(9):1060\u00E2\u0080\u00934.22. Ewing AD, Gacita A, Wood LD, Ma F, Xing D, Kim M-S, Manda SS, Abril G,Pereira G, Makohon-Moore A, et al. Widespread somatic L1retrotransposition occurs early during gastrointestinal cancer evolution.Genome Res. 2015;25(10):1536\u00E2\u0080\u009345.23. Scott EC, Gardner EJ, Masood A, Chuang NT, Vertino PM, Devine SE. A hotL1 retrotransposon evades somatic repression and initiates humancolorectal cancer. Genome Res. 2016;26(6):745\u00E2\u0080\u009355.24. Bannert N, Kurth R. The Evolutionary Dynamics of Human EndogenousRetroviral Families. Annu Rev Genomics Hum Genet. 2006;7(1):149\u00E2\u0080\u009373.25. Jern P, Coffin JM. Effects of Retroviruses on Host Genome Function. AnnuRev Genet. 2008;42(1):709\u00E2\u0080\u009332.26. Magiorkinis G, Blanco-Melo D, Belshaw R. The decline of humanendogenous retroviruses: extinction and survival. Retrovirology. 2015;12(1):1\u00E2\u0080\u009312.27. Rosenberg N, Jolicoeur P. Retroviral pathogenesis. In: Coffin JM, Hughes SH,Varmus H, editors. Retroviruses. Cold Spring Harbor: Cold Spring HarborLaboratory Press; 1997. p. 475\u00E2\u0080\u0093586.28. Howard G, Eiges R, Gaudet F, Jaenisch R, Eden A. Activation andtransposition of endogenous retroviral elements in hypomethylationinduced tumors in mice. Oncogene. 2008;27(3):404\u00E2\u0080\u00938.29. Fan H, Johnson C. Insertional Oncogenesis by Non-Acute Retroviruses:Implications for Gene Therapy. Viruses. 2011;3(4):398\u00E2\u0080\u0093422.30. Hohn O, Hanke K, Bannert N. HERV-K(HML-2), the best preserved family ofHERVs: endogenisation, expression and implications in health and disease.Front Oncol. 2013;3:246.31. Chen T, Meng Z, Gan Y, Wang X, Xu F, Gu Y, Xu X, Tang J, Zhou H, Zhang X,et al. The viral oncogene Np9 acts as a critical molecular switch for co-activating beta-catenin, ERK, Akt and Notch1 and promoting the growth ofhuman leukemia stem/progenitor cells. Leukemia. 2013;27(7):1469\u00E2\u0080\u009378.32. Downey RF, Sullivan FJ, Wang-Johanning F, Ambs S, Giles FJ, Glynn SA.Human endogenous retrovirus K and cancer: Innocent bystander ortumorigenic accomplice? Int J Cancer. 2015;137(6):1249\u00E2\u0080\u009357.33. Kassiotis G. Endogenous Retroviruses and the Development of Cancer.J Immunol. 2014;192(4):1343\u00E2\u0080\u00939.34. Gomez-del Arco P, Kashiwagi M, Jackson AF, Naito T, Zhang J, Liu F, Kee B,Vooijs M, Radtke F, Redondo JM, et al. Alternative promoter usage at theNotch1 locus supports ligand-independent signaling in T cell developmentand leukemogenesis. Immunity. 2010;33(5):685\u00E2\u0080\u009398.35. Thorsen K, Schepeler T, \u00C3\u0098ster B, Rasmussen MH, Vang S, Wang K, HansenKQ, Lamy P, Pedersen JS, Eller A, et al. Tumor-specific usage of alternativetranscription start sites in colorectal cancer identified by genome-wide exonarray analysis. BMC Genomics. 2011;12(1):1\u00E2\u0080\u009314.36. Muratani M, Deng N, Ooi WF, Lin SJ, Xing M, Xu C, Qamra A, Tay ST, Malik S,Wu J, et al. Nanoscale chromatin profiling of gastric adenocarcinomareveals cancer-associated cryptic promoters and somatically acquiredregulatory elements. Nat Commun. 2014;5:4361.Babaian and Mager Mobile DNA (2016) 7:24 Page 16 of 2137. Nagarajan RP, Zhang B, Bell RJA, Johnson BE, Olshen AB, Sundaram V,Li D, Graham AE, Diaz A, Fouse SD, et al. Recurrent epimutationsactivate gene body promoters in primary glioblastoma. Genome Res.2014;24(5):761\u00E2\u0080\u009374.38. Wiesner T, Lee W, Obenauf AC, Ran L, Murali R, Zhang QF, Wong EWP, HuW, Scott SN, Shah RH, et al. Alternative transcription initiation leads toexpression of a novel ALK isoform in cancer. Nature. 2015;526(7573):453\u00E2\u0080\u00937.39. O\u00E2\u0080\u0099Connell MR, Sarkar S, Luthra GK, Okugawa Y, Toiyama Y, Gajjar AH, Qiu S,Goel A, Singh P. Epigenetic changes and alternate promoter usage byhuman colon cancers for expressing DCLK1-isoforms: Clinical Implications.Scie Rep. 2015;5:14983.40. Grassilli E, Pisano F, Cialdella A, Bonomo S, Missaglia C, Cerrito MG, MasieroL, Ianzano L, Giordano F, Cicirelli V, et al. A novel oncogenic BTK isoform isoverexpressed in colon cancers and required for RAS-mediatedtransformation. Oncogene. 2016;35:4368\u00E2\u0080\u009378.41. Maeso I, Tena JJ. Favorable genomic environments for cis-regulatoryevolution: A novel theoretical framework. Semin Cell Dev Biol. 2016;57:2\u00E2\u0080\u009310.42. Thompson Peter J, Macfarlan Todd S, Lorincz Matthew C. Long TerminalRepeats: From Parasitic Elements to Building Blocks of the TranscriptionalRegulatory Repertoire. Mol Cell. 2016;62(5):766\u00E2\u0080\u009376.43. Blomberg J, Benachenhou F, Blikstad V, Sperber G, Mayer J. Classificationand nomenclature of endogenous retroviral sequences (ERVs): Problemsand recommendations. Gene. 2009;448(2):115\u00E2\u0080\u009323.44. Mager DL, Stoye JP: Mammalian Endogenous Retroviruses. Microbiol Spectr2015, 3(1). doi: 10.1128/microbiolspec.MDNA3-0009-2014.45. Belshaw R, Dawson ALA, Woolven-Allen J, Redding J, Burt A, Tristem M.Genomewide Screening Reveals High Levels of Insertional Polymorphism inthe Human Endogenous Retrovirus Family HERV-K(HML2): Implications forPresent-Day Activity. J Virol. 2005;79(19):12507\u00E2\u0080\u009314.46. Wildschutte JH, Williams ZH, Montesion M, Subramanian RP, Kidd JM, CoffinJM. Discovery of unfixed endogenous retrovirus insertions in diverse humanpopulations. Proc Natl Acad Sci. 2016;113(16):E2326\u00E2\u0080\u009334.47. Maksakova IA, Romanish MT, Gagnier L, Dunn CA, van de Lagemaat LN,Mager DL. Retroviral Elements and Their Hosts: Insertional Mutagenesis inthe Mouse Germ Line. PLoS Genet. 2006;2(1):e2.48. Belshaw R, Watson J, Katzourakis A, Howe A, Woolven-Allen J, Burt A,Tristem M. Rate of Recombinational Deletion among Human EndogenousRetroviruses. J Virol. 2007;81(17):9437\u00E2\u0080\u009342.49. Gemmell P, Hein J, Katzourakis A. Phylogenetic Analysis Reveals That ERVs\"Die Young\" but HERV-H Is Unusually Conserved. PLoS Comput Biol. 2016;12(6):e1004964.50. Hubley R, Finn RD, Clements J, Eddy SR, Jones TA, Bao W, Smit AFA,Wheeler TJ. The Dfam database of repetitive DNA families. Nucleic AcidsRes. 2016;44(D1):D81\u00E2\u0080\u00939.51. Kunarso G, Chia NY, Jeyakani J, Hwang C, Lu X, Chan YS, Ng HH, Bourque G.Transposable elements have rewired the core regulatory network of humanembryonic stem cells. Nat Genet. 2010;42(7):631\u00E2\u0080\u00934.52. Chuong EB, Elde NC, Feschotte C. Regulatory evolution of innate immunitythrough co-option of endogenous retroviruses. Science. 2016;351(6277):1083\u00E2\u0080\u00937.53. Wang T, Zeng J, Lowe CB, Sellers RG, Salama SR, Yang M, Burgess SM,Brachmann RK, Haussler D. Species-specific endogenous retroviruses shapethe transcriptional network of the human tumor suppressor protein p53.Proc Natl Acad Sci. 2007;104(47):18613\u00E2\u0080\u00938.54. Jacques P-E, Jeyakani J, Bourque G. The Majority of Primate-SpecificRegulatory Sequences Are Derived from Transposable Elements. PLoSGenet. 2013;9(5):e1003504.55. Xie M, Hong C, Zhang B, Lowdon RF, Xing X, Li D, Zhou X, Lee HJ, Maire CL,Ligon KL, et al. DNA hypomethylation within specific transposable elementfamilies associates with tissue-specific enhancer landscape. Nat Genet. 2013;45(7):836\u00E2\u0080\u009341.56. Kelley D, Rinn J. Transposable elements reveal a stem cell specific class oflong noncoding RNAs. Genome Biol. 2012;13(11):R107.57. Lu X, Sachs F, Ramsay L, Jacques PE, Goke J, Bourque G, Ng HH. Theretrovirus HERVH is a long noncoding RNA required for human embryonicstem cell identity. Nat Struct Mol Biol. 2014;21(4):423\u00E2\u0080\u00935.58. Wang J, Xie G, Singh M, Ghanbarian AT, Rasko T, Szvetnik A, Cai H, Besser D,Prigione A, Fuchs NV, et al. Primate-specific endogenous retrovirus-driventranscription defines naive-like stem cells. Nature. 2014;516(7531):405\u00E2\u0080\u00939.59. Durruthy-Durruthy J, Sebastiano V, Wossidlo M, Cepeda D, Cui J, Grow EJ,Davila J, Mall M, Wong WH, Wysocka J, et al. The primate-specificnoncoding RNA HPAT5 regulates pluripotency during humanpreimplantation development and nuclear reprogramming. Nat Genet.2016;48:44\u00E2\u0080\u009352.60. Izsv\u00C3\u00A1k Z, Wang J, Singh M, Mager DL, Hurst LD. Pluripotency and theendogenous retrovirus HERVH: Conflict or serendipity? BioEssays. 2016;38(1):109\u00E2\u0080\u009317.61. Emera D, Wagner GP. Transposable element recruitments in the mammalianplacenta: impacts and mechanisms. Brief Funct Genomics. 2012;11(4):267\u00E2\u0080\u009376.62. Feschotte C. Transposable elements and the evolution of regulatorynetworks. Nat Rev Genet. 2008;9(5):397\u00E2\u0080\u0093405.63. Cohen CJ, Lock WM, Mager DL. Endogenous retroviral LTRs as promoters forhuman genes: a critical assessment. Gene. 2009;448(2):105\u00E2\u0080\u009314.64. Rayan NA, del Rosario RCH, Prabhakar S. Massive contribution oftransposable elements to mammalian regulatory sequences. Semin Cell DevBiol. 2016;57:51\u00E2\u0080\u00936.65. Friedli M, Trono D. The Developmental Control of Transposable Elements andthe Evolution of Higher Species. Annu Rev Cell Dev Biol. 2015;31(1):429\u00E2\u0080\u009351.66. Nigumann P, Redik K, Matlik K, Speek M. Many human genes are transcribedfrom the antisense promoter of L1 retrotransposon. Genomics. 2002;79(5):628\u00E2\u0080\u009334.67. M\u00C3\u00A4tlik K, Redik K, Speek M. L1 Antisense Promoter Drives Tissue-SpecificTranscription of Human Genes. J Biomed Biotechnol. 2006;2006:71753.68. Rebollo R, Farivar S, Mager DL. C-GATE - catalogue of genes affected bytransposable elements. Mob DNA. 2012;3(1):9.69. Criscione SW, Theodosakis N, Micevic G, Cornish TC, Burns KH, Neretti N,Rodi\u00C4\u0087 N. Genome-wide characterization of human L1 antisense promoter-driven transcripts. BMC Genomics. 2016;17(1):1\u00E2\u0080\u009315.70. Denli Ahmet M, Narvaiza I, Kerman Bilal E, Pena M, Benner C, MarchettoMaria CN, Diedrich Jolene K, Aslanian A, Ma J, Moresco James J, et al.Primate-Specific ORF0 Contributes to Retrotransposon-Mediated Diversity.Cell. 2015;163(3):583\u00E2\u0080\u009393.71. Szak ST, Pickeral OK, Makalowski W, Boguski MS, Landsman D, Boeke JD.Molecular archeology of L1 insertions in the human genome. Genome Biol.2002;3(10):research0052. 0051\u00E2\u0080\u0093research0052.0018.72. Khan H, Smit A, Boissinot S. Molecular evolution and tempo of amplificationof human LINE-1 retrotransposons since the origin of primates. GenomeRes. 2006;16(1):78\u00E2\u0080\u009387.73. Faulkner GJ, Kimura Y, Daub CO, Wani S, Plessy C, Irvine KM, Schroder K,Cloonan N, Steptoe AL, Lassmann T, et al. The regulated retrotransposontranscriptome of mammalian cells. Nat Genet. 2009;41(5):563\u00E2\u0080\u009371.74. Roy AM, West NC, Rao A, Adhikari P, Alem\u00C3\u00A1n C, Barnes AP, Deininger PL.Upstream flanking sequences and transcription of SINEs1. J Mol Biol. 2000;302(1):17\u00E2\u0080\u009325.75. Deininger P. Alu elements: know the SINEs. Genome Biol. 2011;12(12):1\u00E2\u0080\u009312.76. Bejerano G, Lowe CB, Ahituv N, King B, Siepel A, Salama SR, Rubin EM,James Kent W, Haussler D. A distal enhancer and an ultraconserved exonare derived from a novel retroposon. Nature. 2006;441(7089):87\u00E2\u0080\u009390.77. Lowe CB, Bejerano G, Haussler D. Thousands of human mobile elementfragments undergo strong purifying selection near developmental genes.Proc Natl Acad Sci. 2007;104(19):8005\u00E2\u0080\u009310.78. Sasaki T, Nishihara H, Hirakawa M, Fujimura K, Tanaka M, Kokubo N, Kimura-Yoshida C, Matsuo I, Sumiyama K, Saitou N, et al. Possible involvement ofSINEs in mammalian-specific brain formation. Proc Natl Acad Sci. 2008;105(11):4220\u00E2\u0080\u00935.79. Jjingo D, Conley AB, Wang J, Mari\u00C3\u00B1o-Ram\u00C3\u00ADrez L, Lunyak VV, Jordan IK:Mammalian-wide interspersed repeat (MIR)-derived enhancers and theregulation of human gene expression. Mobile DNA 2014, 5:14\u00E2\u0080\u00931480. Lynch VJ, Leclerc RD, May G, Wagner GP. Transposon-mediated rewiring ofgene regulatory networks contributed to the evolution of pregnancy inmammals. Nat Genet. 2011;43(11):1154\u00E2\u0080\u00939.81. Lynch Vincent J, Nnamani Mauris C, Kapusta A, Brayer K, Plaza Silvia L, MazurErik C, Emera D, Sheikh Shehzad Z, Gr\u00C3\u00BCtzner F, Bauersachs S, et al. AncientTransposable Elements Transformed the Uterine Regulatory Landscape andTranscriptome during the Evolution of Mammalian Pregnancy. Cell Rep.2015;10(4):551\u00E2\u0080\u009361.82. Sinzelle L, Izsvak Z, Ivics Z. Molecular domestication of transposableelements: From detrimental parasites to useful host genes. Cell Mol Life Sci.2009;66(6):1073\u00E2\u0080\u009393.83. Bowen NJ, Jordan IK. Transposable elements and the evolution ofeukaryotic complexity. Curr Issues Mol Biol. 2002;4(3):65\u00E2\u0080\u009376.84. Dupressoir A, Lavialle C, Heidmann T. From ancestral infectious retrovirusesto bona fide cellular genes: Role of the captured syncytins in placentation.Placenta. 2012;33(9):663\u00E2\u0080\u009371.Babaian and Mager Mobile DNA (2016) 7:24 Page 17 of 2185. Wolf D, Goff SP. Host Restriction Factors Blocking Retroviral Replication.Annu Rev Genet. 2008;42(1):143\u00E2\u0080\u009363.86. Friedli M, Turelli P, Kapopoulou A, Rauwel B, Castro-Diaz N, Rowe HM, EccoG, Unzu C, Planet E, Lombardo A, et al. Loss of transcriptional control overendogenous retroelements during reprogramming to pluripotency.Genome Res. 2014;24(8):1251\u00E2\u0080\u00939.87. Jacobs FMJ, Greenberg D, Nguyen N, Haeussler M, Ewing AD, Katzman S,Paten B, Salama SR, Haussler D. An evolutionary arms race between KRABzinc-finger genes ZNF91/93 and SVA/L1 retrotransposons. Nature. 2014;516(7530):242\u00E2\u0080\u00935.88. Wolf G, Greenberg D, Macfarlan T. Spotting the enemy within: Targetedsilencing of foreign DNA in mammalian genomes by the Kruppel-associatedbox zinc finger protein family. Mob DNA. 2015;6(1):17.89. Rowe HM, Trono D. Dynamic control of endogenous retroviruses duringdevelopment. Virology. 2011;411(2):273\u00E2\u0080\u009387.90. Leung DC, Lorincz MC. Silencing of endogenous retroviruses: when andwhy do histone marks predominate? Trends Biochem Sci. 2012;37(4):127\u00E2\u0080\u009333.91. Liu S, Brind\u00E2\u0080\u0099Amour J, Karimi MM, Shirane K, Bogutz A, Lefebvre L, Sasaki H,Shinkai Y, Lorincz MC. Setdb1 is required for germline development andsilencing of H3K9me3-marked endogenous retroviruses in primordial germcells. Genes Dev. 2014;28(18):2041\u00E2\u0080\u009355.92. Yang F, Wang PJ: Multiple LINEs of retrotransposon silencing mechanisms inthe mammalian germline. Semin Cell Dev Biol 2016, in press.93. Baylin SB, Jones PA. A decade of exploring the cancer epigenome - biologicaland translational implications. Nat Rev Cancer. 2011;11(10):726\u00E2\u0080\u009334.94. Timp W, Feinberg AP. Cancer as a dysregulated epigenome allowing cellulargrowth advantage at the expense of the host. Nat Rev Cancer. 2013;13(7):497\u00E2\u0080\u0093510.95. Berdasco M, Esteller M. Aberrant Epigenetic Landscape in Cancer: HowCellular Identity Goes Awry. Dev Cell. 2010;19(5):698\u00E2\u0080\u0093711.96. Skulte KA, Phan L, Clark SJ, Taberlay PC. Chromatin remodeler mutations inhuman cancers: epigenetic implications. Epigenomics. 2014;6(4):397\u00E2\u0080\u0093414.97. Schwartzentruber J, Korshunov A, Liu XY, Jones DT, Pfaff E, Jacob K, SturmD, Fontebasso AM, Quang DA, Tonjes M, et al. Driver mutations in histoneH3.3 and chromatin remodelling genes in paediatric glioblastoma. Nature.2012;482(7384):226\u00E2\u0080\u009331.98. Shen H, Laird PW. Interplay between the Cancer Genome and Epigenome.Cell. 2013;153(1):38\u00E2\u0080\u009355.99. Nephew KP, Huang TH-M. Epigenetic gene silencing in cancer initiation andprogression. Cancer Lett. 2003;190(2):125\u00E2\u0080\u009333.100. Kazanets A, Shorstova T, Hilmi K, Marques M, Witcher M. Epigenetic silencing oftumor suppressor genes: Paradigms, puzzles, and potential. Biochimica etBiophysica Acta (BBA) - Reviews on Cancer. 2016;1865(2):275\u00E2\u0080\u009388.101. Ehrlich M. DNA methylation in cancer: too much, but also too little.Oncogene. 2002;21(35):5400\u00E2\u0080\u009313.102. Hoffmann MJ, Schulz WA. Causes and consequences of DNAhypomethylation in human cancer. Biochem Cell Biol. 2005;83(3):296\u00E2\u0080\u0093321.103. De Smet C, Loriot A. DNA hypomethylation in cancer: Epigenetic scars of aneoplastic journey. Epigenetics. 2010;5(3):206\u00E2\u0080\u009313.104. Ross JP, Rand KN, Molloy PL. Hypomethylation of repeated DNA sequencesin cancer. Epigenomics. 2010;2(2):245\u00E2\u0080\u009369.105. Szpakowski S, Sun X, Lage JM, Dyer A, Rubinstein J, Kowalski D, Sasaki C,Costa J, Lizardi PM. Loss of epigenetic silencing in tumors preferentiallyaffects primate-specific retroelements. Gene. 2009;448(2):151\u00E2\u0080\u009367.106. Barchitta M, Quattrocchi A, Maugeri A, Vinciguerra M, Agodi A. LINE-1Hypomethylation in Blood and Tissue Samples as an Epigenetic Marker forCancer Risk: A Systematic Review and Meta-Analysis. PLoS One. 2014;9(10):e109478.107. Romanish MT, Cohen CJ, Mager DL. Potential mechanisms of endogenousretroviral-mediated genomic instability in human cancer. Semin Cancer Biol.2010;20(4):246\u00E2\u0080\u009353.108. Piskareva O, Lackington W, Lemass D, Hendrick C, Doolan P, Barron N. Thehuman L1 element: a potential biomarker in cancer prognosis, currentstatus and future directions. Curr Mol Med. 2011;11(4):286\u00E2\u0080\u0093303.109. P\u00C3\u00A9rot P, Mullins CS, Naville M, Bressan C, H\u00C3\u00BChns M, Gock M, K\u00C3\u00BChn F, Volff J-N, Trillet-Lenoir V, Linnebacher M, et al. Expression of young HERV-H loci inthe course of colorectal carcinoma and correlation with molecular subtypes.Oncotarget. 2015;6(37):40095\u00E2\u0080\u0093111.110. Haupt S, Tisdale M, Vincendeau M, Clements MA, Gauthier DT, Lance R,Semmes OJ, Turqueti-Neves A, Noessner E, Leib-M\u00C3\u00B6sch C, et al. Humanendogenous retrovirus transcription profiles of the kidney and kidney-derived cell lines. J Gen Virol. 2011;92(10):2356\u00E2\u0080\u009366.111. Gosenca D, Gabriel U, Steidler A, Mayer J, Diem O, Erben P, Fabarius A,Leib-M\u00CB\u0086sch C, Hofmann W-K, Seifarth W. HERV-E-Mediated Modulation ofPLA2G4A Transcription in Urothelial Carcinoma. PLoS One. 2012;7(11):e49341.112. Haase K, Mosch A, Frishman D. Differential expression analysis of humanendogenous retroviruses based on ENCODE RNA-seq data. BMC Med Genet.2015;8(1):71.113. Lamprecht B, Walter K, Kreher S, Kumar R, Hummel M, Lenze D, Kochert K,Bouhlel MA, Richter J, Soler E, et al. Derepression of an endogenous longterminal repeat activates the CSF1R proto-oncogene in human lymphoma.Nat Med. 2010;16(5):571\u00E2\u0080\u00939.114. Bao W, Kojima KK, Kohany O. Repbase Update, a database of repetitiveelements in eukaryotic genomes. Mob DNA. 2015;6(1):1\u00E2\u0080\u00936.115. Steidl C, Diepstra A, Lee T, Chan FC, Farinha P, Tan K, Telenius A, Barclay L,Shah SP, Connors JM, et al. Gene expression profiling of microdissectedHodgkin Reed-Sternberg cells correlates with treatment outcome in classicalHodgkin lymphoma. Blood. 2012;120(17):3530\u00E2\u0080\u009340.116. Mart\u00C3\u00ADn-Moreno AM, Roncador G, Maestre L, Mata E, Jim\u00C3\u00A9nez S, Mart\u00C3\u00ADnez-Torrecuadrada JL, Reyes-Garc\u00C3\u00ADa AI, Rubio C, Tom\u00C3\u00A1s JF, Est\u00C3\u00A9vez M, et al.CSF1R Protein Expression in Reactive Lymphoid Tissues and Lymphoma: ItsRelevance in Classical Hodgkin Lymphoma. PLoS One. 2015;10(6):e0125203.117. Babaian A, Romanish MT, Gagnier L, Kuo LY, Karimi MM, Steidl C, Mager DL.Onco-exaptation of an endogenous retroviral LTR drives IRF5 expression inHodgkin lymphoma. Oncogene. 2016;35(19):2542\u00E2\u0080\u00936.118. Clark DN, Read RD, Mayhew V, Petersen SC, Argueta LB, Stutz LA, Till RE,Bergsten SM, Robinson BS, Baumann DG, et al. Four Promoters of IRF5Respond Distinctly to Stimuli and are Affected by Autoimmune-RiskPolymorphisms. Front Immunol. 2013;4:360.119. Kreher S, Bouhlel MA, Cauchy P, Lamprecht B, Li S, Grau M, Hummel F,K\u00C3\u00B6chert K, Anagnostopoulos I, J\u00C3\u00B6hrens K, et al. Mapping of transcriptionfactor motifs in active chromatin identifies IRF5 as key regulator in classicalHodgkin lymphoma. Proc Natl Acad Sci. 2014;111(42):E4513\u00E2\u0080\u009322.120. Mancl ME, Hu G, Sangster-Guity N, Olshalsky SL, Hoops K, Fitzgerald-BocarslyP, Pitha PM, Pinder K, Barnes BJ. Two Discrete Promoters Regulate theAlternatively Spliced Human Interferon Regulatory Factor-5 Isoforms:Multiple isoforms with distinct cell type-specific expression, localization,regulation, and function. J Biol Chem. 2005;280(22):21078\u00E2\u0080\u009390.121. Introna M, Luchetti M, Castellano M, Arsura M, Golay J. The myb oncogenefamily of transcription factors: potent regulators of hematopoietic cellproliferation and differentiation. Semin Cancer Biol. 1994;5(2):113\u00E2\u0080\u009324.122. Ramsay RG, Gonda TJ. MYB function in normal and cancer cells. Nat RevCancer. 2008;8(7):523\u00E2\u0080\u009334.123. Wolff EM, Byun H-M, Han HF, Sharma S, Nichols PW, Siegmund KD, Yang AS,Jones PA, Liang G. Hypomethylation of a LINE-1 Promoter Activates anAlternate Transcript of the MET Oncogene in Bladders with Cancer. PLoSGenet. 2010;6(4):e1000917.124. Weber B, Kimhi S, Howard G, Eden A, Lyko F. Demethylation of a LINE-1antisense promoter in the cMet locus impairs Met signalling throughinduction of illegitimate transcription. Oncogene. 2010;29(43):5775\u00E2\u0080\u009384.125. Hur K, Cejas P, Feliu J, Moreno-Rubio J, Burgos E, Boland CR, Goel A.Hypomethylation of long interspersed nuclear element-1 (LINE-1) leads toactivation of proto-oncogenes in human colorectal cancer metastasis. Gut.2014;63(4):635\u00E2\u0080\u009346.126. Gao H, Guan M, Sun Z, Bai C. High c-Met expression is a negativeprognostic marker for colorectal cancer: a meta-analysis. Tumor Biol. 2015;36(2):515\u00E2\u0080\u009320.127. Mari\u00C3\u00B1o-Enr\u00C3\u00ADquez A, Dal Cin P. ALK as a paradigm of oncogenic promiscuity:different mechanisms of activation and different fusion partners drivetumors of different lineages. Cancer Genetics. 2013;206(11):357\u00E2\u0080\u009373.128. Fantom-Consortium. A promoter-level mammalian expression atlas. Nature.2014;507(7493):462\u00E2\u0080\u009370.129. Scarf\u00C3\u00B2 I, Pellegrino E, Mereu E, Kwee I, Agnelli L, Bergaggio E, Garaffo G,Vitale N, Caputo M, Machiorlatti R, et al. Identification of a new subclass ofALK-negative ALCL expressing aberrant levels of ERBB4 transcripts. Blood.2016;127(2):221\u00E2\u0080\u009332.130. Arteaga Carlos L, Engelman Jeffrey A. ERBB Receptors: From OncogeneDiscovery to Basic Science to Mechanism-Based Cancer Therapeutics.Cancer Cell. 2014;25(3):282\u00E2\u0080\u0093303.131. Obaidat A, Roth M, Hagenbuch B. The Expression and Function of OrganicAnion Transporting Polypeptides in Normal Tissues and in Cancer. AnnuRev Pharmacol Toxicol. 2012;52(1):135\u00E2\u0080\u009351.Babaian and Mager Mobile DNA (2016) 7:24 Page 18 of 21132. Lee W, Belkhiri A, Lockhart AC, Merchant N, Glaeser H, Harris EI, WashingtonMK, Brunt EM, Zaika A, Kim RB, et al. Overexpression of OATP1B3 ConfersApoptotic Resistance in Colon Cancer. Cancer Res. 2008;68(24):10315\u00E2\u0080\u009323.133. Nagai M, Furihata T, Matsumoto S, Ishii S, Motohashi S, Yoshino I, Ugajin M,Miyajima A, Matsumoto S, Chiba K. Identification of a new organic aniontransporting polypeptide 1B3 mRNA isoform primarily expressed in humancancerous tissues and cells. Biochem Biophys Res Commun. 2012;418(4):818\u00E2\u0080\u009323.134. Imai S, Kikuchi R, Tsuruya Y, Naoi S, Nishida S, Kusuhara H, Sugiyama Y.Epigenetic Regulation of Organic Anion Transporting Polypeptide 1B3 inCancer Cell Lines. Pharm Res. 2013;30(11):2880\u00E2\u0080\u009390.135. Liang Q, Xu Z, Xu R, Wu L, Zheng S. Expression Patterns of Non-CodingSpliced Transcripts from Human Endogenous Retrovirus HERV-H Elements inColon Cancer. PLoS One. 2012;7(1):e29950.136. Teft WA, Welch S, Lenehan J, Parfitt J, Choi YH, Winquist E, Kim RB. OATP1B1and tumour OATP1B3 modulate exposure, toxicity, and survival afteririnotecan-based chemotherapy. Br J Cancer. 2015;112(5):857\u00E2\u0080\u009365.137. Morin RD, Mendez-Lago M, Mungall AJ, Goya R, Mungall KL, Corbett RD,Johnson NA, Severson TM, Chiu R, Field M, et al. Frequent mutation ofhistone-modifying genes in non-Hodgkin lymphoma. Nature. 2011;476(7360):298\u00E2\u0080\u0093303.138. Lock FE, Rebollo R, Miceli-Royer K, Gagnier L, Kuah S, Babaian A, Sistiaga-Poveda M, Lai CB, Nemirovsky O, Serrano I, et al. Distinct isoform of FABP7revealed by screening for retroelement-activated genes in diffuse large B-cell lymphoma. Proc Natl Acad Sci. 2014;111(34):E3534\u00E2\u0080\u009343.139. Thumser AE, Moore JB, Plant NJ. Fatty acid binding proteins: tissue-specificfunctions in health and disease. Curr Opin Clin Nutr Metab Care. 2014;17(2):124\u00E2\u0080\u00939.140. Liu R-Z, Graham K, Glubrecht DD, Lai R, Mackey JR, Godbout R. A fatty acid-binding protein 7/RXR\u00CE\u00B2 pathway enhances survival and proliferation intriple-negative breast cancer. J Pathol. 2012;228(3):310\u00E2\u0080\u009321.141. Kapusta A, Kronenberg Z, Lynch VJ, Zhuo X, Ramsay L, Bourque G, YandellM, Feschotte C. Transposable Elements Are Major Contributors to theOrigin, Diversification, and Regulation of Vertebrate Long Noncoding RNAs.PLoS Genet. 2013;9(4):e1003470.142. St Laurent G, Shtokalo D, Dong B, Tackett M, Fan X, Lazorthes S, Nicolas E,Sang N, Triche T, McCaffrey T, et al. VlincRNAs controlled by retroviralelements are a hallmark of pluripotency and cancer. Genome Biol. 2013;14(7):R73.143. Prensner JR, Iyer MK, Sahu A, Asangani IA, Cao Q, Patel L, Vergara IA,Davicioni E, Erho N, Ghadessi M, et al. The long noncoding RNA SChLAP1promotes aggressive prostate cancer and antagonizes the SWI/SNFcomplex. Nat Genet. 2013;45(11):1392\u00E2\u0080\u00938.144. Masliah-Planchon J, Bi\u00C3\u00A8che I, Guinebreti\u00C3\u00A8re J-M, Bourdeaut F, Delattre O.SWI/SNF Chromatin Remodeling and Human Malignancies. Ann Rev PatholMech Dis. 2015;10(1):145\u00E2\u0080\u009371.145. Loewer S, Cabili MN, Guttman M, Loh Y-H, Thomas K, Park IH, Garber M,Curran M, Onder T, Agarwal S, et al. Large intergenic non-coding RNA-RoRmodulates reprogramming of human induced pluripotent stem cells. NatGenet. 2010;42(12):1113\u00E2\u0080\u00937.146. Wang Y, Xu Z, Jiang J, Xu C, Kang J, Xiao L, Wu M, Xiong J, Guo X, Liu H.Endogenous miRNA sponge lincRNA-RoR regulates Oct4, Nanog, and Sox2in human embryonic stem cell self-renewal. Dev Cell. 2013;25(1):69\u00E2\u0080\u009380.147. Eades G, Wolfson B, Zhang Y, Li Q, Yao Y, Zhou Q. lincRNA-RoR and miR-145Regulate Invasion in Triple-Negative Breast Cancer via Targeting ARF6. MolCancer Res. 2015;13(2):330\u00E2\u0080\u00938.148. Gao S, Wang P, Hua Y, Xi H, Meng Z, Liu T, Chen Z, Liu L. ROR functions asa ceRNA to regulate Nanog expression by sponging miR-145 and predictspoor prognosis in pancreatic cancer. Oncotarget. 2016;7(2):1608\u00E2\u0080\u009318.149. Zhou P, Sun L, Liu D, Liu C, Sun L. Long Non-Coding RNA lincRNA-RORPromotes the Progression of Colon Cancer and Holds Prognostic Value byAssociating with miR-145. Pathol Oncol Res. 2016;22(4):733\u00E2\u0080\u009340.150. Fan J, Xing Y, Wen X, Jia R, Ni H, He J, Ding X, Pan H, Qian G, Ge S, et al.Long non-coding RNA ROR decoys gene-specific histone methylation topromote tumorigenesis. Genome Biol. 2015;16(1):139.151. Huang J, Zhang A, Ho T-T, Zhang Z, Zhou N, Ding X, Zhang X, Xu M, Mo Y-Y.Linc-RoR promotes c-Myc expression through hnRNP I and AUF1. NucleicAcids Res. 2016;44(7):3059\u00E2\u0080\u009369.152. Rangel LBA, Sherman-Baust CA, Wernyj RP, Schwartz DR, Cho KR, Morin PJ.Characterization of novel human ovarian cancer-specific transcripts (HOSTs)identified by serial analysis of gene expression. Oncogene. 2003;22(46):7225\u00E2\u0080\u009332.153. Adams Brian D, Kasinski Andrea L, Slack Frank J. Aberrant Regulation andFunction of MicroRNAs in Cancer. Curr Biol. 2014;24(16):R762\u00E2\u0080\u009376.154. Gao Y, Meng H, Liu S, Hu J, Zhang Y, Jiao T, Liu Y, Ou J, Wang D, Yao L, etal. LncRNA-HOST2 regulates cell biological behaviors in epithelial ovariancancer through a mechanism involving microRNA let-7b. Hum Mol Genet.2015;24(3):841\u00E2\u0080\u009352.155. Yang F, Lyu S, Dong S, Liu Y, Zhang X, Wang O. Expression profile analysis oflong noncoding RNA in HER-2-enriched subtype breast cancer by next-generation sequencing and bioinformatics. OncoTargets Ther. 2016;9:761\u00E2\u0080\u009372.156. Zeng Z, Bo H, Gong Z, Lian Y, Li X, Li X, Zhang W, Deng H, Zhou M, Peng S,et al. AFAP1-AS1, a long noncoding RNA upregulated in lung cancer andpromotes invasion and metastasis. Tumor Biol. 2016;37(1):729\u00E2\u0080\u009337.157. Deng J, Liang Y, Liu C, He S, Wang S. The up-regulation of long non-codingRNA AFAP1-AS1 is associated with the poor prognosis of NSCLC patients.Biomed Pharmacother. 2015;75:8\u00E2\u0080\u009311.158. Wu W, Bhagat TD, Yang X, Song JH, Cheng Y, Agarwal R, Abraham JM,Ibrahim S, Bartenstein M, Hussain Z, et al. Hypomethylation of NoncodingDNA Regions and Overexpression of the Long Noncoding RNA, AFAP1-AS1,in Barrett's Esophagus and Esophageal Adenocarcinoma. Gastroenterology.2013;144(5):956\u00E2\u0080\u009366. e954.159. Zhang J-Y, Weng M-Z, Song F-B, Xu Y-G, Liu Q, Wu J-Y, Qin J, Jin T, Xu J-M.Long noncoding RNA AFAP1-AS1 indicates a poor prognosis ofhepatocellular carcinoma and promotes cell proliferation and invasion viaupregulation of the RhoA/Rac2 signaling. Int J Oncol. 2016;48:1590.160. Guil S, Esteller M. Cis-acting noncoding RNAs: friends and foes. Nat StructMol Biol. 2012;19(11):1068\u00E2\u0080\u009375.161. Leucci E, Vendramin R, Spinazzi M, Laurette P, Fiers M, Wouters J, Radaelli E,Eyckerman S, Leonelli C, Vanderheyden K, et al. Melanoma addiction to thelong non-coding RNA SAMMSON. Nature. 2016;531(7595):518\u00E2\u0080\u009322.162. Harris ML, Baxter LL, Loftus SK, Pavan WJ. Sox proteins in melanocytedevelopment and melanoma. Pigment Cell Melanoma Res. 2010;23(4):496\u00E2\u0080\u0093513.163. Panzitt K, Tschernatsch MMO, Guelly C, Moustafa T, Stradner M, StrohmaierHM, Buck CR, Denk H, Schroeder R, Trauner M, et al. Characterization ofHULC, a Novel Gene With Striking Up-Regulation in HepatocellularCarcinoma, as Noncoding RNA. Gastroenterology. 2007;132(1):330\u00E2\u0080\u009342.164. Li C, Chen J, Zhang K, Feng B, Wang R, Chen L. Progress and Prospects ofLong Noncoding RNAs (lncRNAs) in Hepatocellular Carcinoma. Cell PhysiolBiochem. 2015;36(2):423\u00E2\u0080\u009334.165. Wang X-S, Zhang Z, Wang H-C, Cai J-L, Xu Q-W, Li M-Q, Chen Y-C, Qian X-P,Lu T-J, Yu L-Z, et al. Rapid Identification of UCA1 as a Very Sensitive andSpecific Unique Marker for Human Bladder Carcinoma. Clin Cancer Res.2006;12(16):4851\u00E2\u0080\u00938.166. Wang F, Li X, Xie X, Zhao L, Chen W. UCA1, a non-protein-coding RNA up-regulated in bladder carcinoma and embryo, influencing cell growth andpromoting invasion. FEBS Lett. 2008;582(13):1919\u00E2\u0080\u009327.167. Xue M, Chen W, Li X. Urothelial cancer associated 1: a long noncoding RNAwith a crucial role in cancer. J Cancer Res Clin Oncol. 2016;142(7):1407\u00E2\u0080\u009319.168. Hu J-J, Song W, Zhang S-D, Shen X-H, Qiu X-M, Wu H-Z, Gong P-H, Lu S,Zhao Z-J, He M-L, et al. HBx-upregulated lncRNA UCA1 promotes cellgrowth and tumorigenesis by recruiting EZH2 and repressing p27Kip1/CDK2signaling. Sci Rep. 2016;6:23521.169. Flockhart RJ, Webster DE, Qu K, Mascarenhas N, Kovalski J, Kretz M, KhavariPA. BRAFV600E remodels the melanocyte transcriptome and induces BANCRto regulate melanoma cell migration. Genome Res. 2012;22(6):1006\u00E2\u0080\u009314.170. Guo Q, Zhao YAN, Chen J, Hu JUN, Wang S, Zhang D, Sun Y. BRAF-activatedlong non-coding RNA contributes to colorectal cancer migration byinducing epithelial-mesenchymal transition. Oncol Lett. 2014;8(2):869\u00E2\u0080\u009375.171. Wang Y, Guo Q, Zhao YAN, Chen J, Wang S, Hu JUN, Sun Y. BRAF-activatedlong non-coding RNA contributes to cell proliferation and activatesautophagy in papillary thyroid carcinoma. Oncol Lett. 2014;8(5):1947\u00E2\u0080\u009352.172. Cruickshanks HA, Vafadar-Isfahani N, Dunican DS, Lee A, Sproul D, Lund JN,Meehan RR, Tufarelli C. Expression of a large LINE-1-driven antisense RNA islinked to epigenetic silencing of the metastasis suppressor gene TFPI-2 incancer. Nucleic Acids Res. 2013;41(14):6857\u00E2\u0080\u009369.173. Cruickshanks HA, Tufarelli C. Isolation of cancer-specific chimeric transcriptsinduced by hypomethylation of the LINE-1 antisense promoter. Genomics.2009;94(6):397\u00E2\u0080\u0093406.174. Nigro CL, Wang H, McHugh A, Lattanzio L, Matin R, Harwood C, Syed N,Hatzimichael E, Briasoulis E, Merlano M, et al. Methylated Tissue FactorPathway Inhibitor 2 (TFPI2) DNA in Serum Is a Biomarker of MetastaticMelanoma. J Investig Dermatol. 2013;133(5):1278\u00E2\u0080\u009385.Babaian and Mager Mobile DNA (2016) 7:24 Page 19 of 21175. Gibb E, Warren R, Wilson G, Brown S, Robertson G, Morin G, Holt R.Activation of an endogenous retrovirus-associated long non-coding RNA inhuman adenocarcinoma. Genome Med. 2015;7(1):22.176. Tomczak K, Czerwi\u00C5\u0084ska P, Wiznerowicz M. The Cancer Genome Atlas (TCGA):an immeasurable source of knowledge. Contemp Oncol. 2015;19(1A):A68\u00E2\u0080\u009377.177. Hashimoto K, Suzuki AM, Dos Santos A, Desterke C, Collino A, Ghisletti S,Braun E, Bonetti A, Fort A, Qin X-Y, et al. CAGE profiling of ncRNAs inhepatocellular carcinoma reveals widespread activation of retroviral LTRpromoters in virus-induced tumors. Genome Res. 2015;25(12):1812\u00E2\u0080\u009324.178. Lania L, Di Cristofano A, Strazzullo M, Pengue G, Majello B, La Mantia G.Structural and functional organization of the human endogenous retroviralERV9 sequences. Virology. 1992;191(1):464\u00E2\u0080\u00938.179. Santoni F, Guerra J, Luban J. HERV-H RNA is abundant in human embryonicstem cells and a precise marker for pluripotency. Retrovirology. 2012;9(1):111.180. Ohnuki M, Tanabe K, Sutou K, Teramoto I, Sawamura Y, Narita M, NakamuraM, Tokunaga Y, Nakamura M, Watanabe A, et al. Dynamic regulation ofhuman endogenous retroviruses mediates factor-induced reprogrammingand differentiation potential. Proc Natl Acad Sci. 2014;111(34):12426\u00E2\u0080\u009331.181. Fuchs N, Loewer S, Daley G, Izsvak Z, Lower J, Lower R. Human endogenousretrovirus K (HML-2) RNA and protein expression is a marker for humanembryonic and induced pluripotent stem cells. Retrovirology. 2013;10(1):115.182. Grow EJ, Flynn RA, Chavez SL, Bayless NL, Wossidlo M, Wesche DJ, Martin L,Ware CB, Blish CA, Chang HY, et al. Intrinsic retroviral reactivation in humanpreimplantation embryos and pluripotent cells. Nature. 2015;522(7555):221\u00E2\u0080\u00935.183. G\u00C3\u00B6ke J, Lu X, Chan Y-S, Ng H-H, Ly L-H, Sachs F, Szczerbinska I. DynamicTranscription of Distinct Classes of Endogenous Retroviral Elements MarksSpecific Populations of Early Human Embryonic Cells. Cell Stem Cell. 2015;16(2):135\u00E2\u0080\u009341.184. Bannert N, Kurth R. Retroelements and the human genome: new perspectiveson an old relation. Proc Natl Acad Sci U S A. 2004;101 Suppl 2:14572\u00E2\u0080\u00939.185. Li W, Lee M-H, Henderson L, Tyagi R, Bachani M, Steiner J, Campanac E,Hoffman DA, von Geldern G, Johnson K, et al. Human endogenousretrovirus-K contributes to motor neuron disease. Sci Transl Med. 2015;7(307):307ra153. 307ra153.186. Costas J, Naveira H. Evolutionary History of the Human EndogenousRetrovirus Family ERV9. Mol Biol Evol. 2000;17(2):320\u00E2\u0080\u009330.187. Mager DL, Medstrand P. Retroviral Repeat Sequences. In: eLS. edn. Hoboken:Wiley; 2005.188. Di Cristofano A, Strazullo M, Longo L, La Mantia G. Characterization andgenomic mapping of the ZNF80 locus: Expression of this zinc-finger gene isdriven by a solitary LTR of ERV9 endogenous retroviral family. Nucleic AcidsRes. 1995;23:2823.189. Chen H-J, Carr K, Jerome RE, Edenberg HJ. A Retroviral Repetitive ElementConfers Tissue-Specificity to the Human Alcohol Dehydrogenase 1C(ADH1C) Gene. DNA Cell Biol. 2002;21(11):793\u00E2\u0080\u0093801.190. Beyer U, Moll-Rocek J, Moll UM, Dobbelstein M. Endogenous retrovirusdrives hitherto unknown proapoptotic p63 isoforms in the male germ lineof humans and great apes. Proc Natl Acad Sci U S A. 2011;108(9):3624\u00E2\u0080\u00939.191. Pi W, Zhu X, Wu M, Wang Y, Fulzele S, Eroglu A, Ling J, Tuan D. Long-rangefunction of an intergenic retrotransposon. Proc Natl Acad Sci U S A. 2010;107(29):12992\u00E2\u0080\u00937.192. Sokol M, Jessen KM, Pedersen FS. Human endogenous retroviruses sustaincomplex and cooperative regulation of gene-containing loci andunannotated megabase-sized regions. Retrovirology. 2015;12:32.193. Beyer U, Kronung SK, Leha A, Walter L, Dobbelstein M. Comprehensiveidentification of genes driven by ERV9-LTRs reveals TNFRSF10B as a re-activatable mediator of testicular cancer cell death. Cell Death Differ. 2016;23:64\u00E2\u0080\u009375.194. Kr\u00C3\u00B6nung SK, Beyer U, Chiaramonte ML, Dolfini D, Mantovani R, DobbelsteinM. LTR12 promoter activation in a broad range of human tumor cells byHDAC inhibition. Oncotarget. 2016;7(23):33484\u00E2\u0080\u0093497.195. Xu L, Elkahloun AG, Candotti F, Grajkowski A, Beaucage SL, Petricoin EF,Calvert V, Juhl H, Mills F, Mason K, et al. A Novel Function of RNAs ArisingFrom the Long Terminal Repeat of Human Endogenous Retrovirus 9 in CellCycle Arrest. J Virol. 2013;87(1):25\u00E2\u0080\u009336.196. Yu X, Zhu X, Pi W, Ling J, Ko L, Takeda Y, Tuan D. The Long Terminal Repeat(LTR) of ERV-9 Human Endogenous Retrovirus Binds to NF-Y in theAssembly of an Active LTR Enhancer Complex NF-Y/MZF1/GATA-2. J BiolChem. 2005;280(42):35184\u00E2\u0080\u009394.197. Gasparini P, Sozzi G, Pierotti MA. The role of chromosomal alterations inhuman cancer development. J Cell Biochem. 2007;102(2):320\u00E2\u0080\u009331.198. Oh S, Shin S, Janknecht R. ETV1, 4 and 5: An oncogenic subfamily of ETStranscription factors. Biochim Biophys Acta. 2012;1826(1):1\u00E2\u0080\u009312.199. Tomlins SA, Laxman B, Dhanasekaran SM, Helgeson BE, Cao X, Morris DS,Menon A, Jing X, Cao Q, Han B, et al. Distinct classes of chromosomalrearrangements create oncogenic ETS gene fusions in prostate cancer.Nature. 2007;448(7153):595\u00E2\u0080\u00939.200. Goering W, Schmitt K, Dostert M, Schaal H, Deenen R, Mayer J, Schulz WA.Human endogenous retrovirus HERV-K(HML-2) activity in prostate cancer isdominated by a few loci. Prostate. 2015;75(16):1958\u00E2\u0080\u009371.201. Prensner JR, Iyer MK, Balbin OA, Dhanasekaran SM, Cao Q, Brenner JC, LaxmanB, Asangani IA, Grasso CS, Kominsky HD, et al. Transcriptome sequencingacross a prostate cancer cohort identifies PCAT-1, an unannotated lincRNAimplicated in disease progression. Nat Biotech. 2011;29(8):742\u00E2\u0080\u00939.202. Kumar KR, Chen W, Koduru PR, Luu HS. Myeloid and Lymphoid NeoplasmWith Abnormalities of FGFR1 Presenting With Trilineage Blasts and RUNX1Rearrangement. Am J Clin Pathol. 2015;143(5):738\u00E2\u0080\u009348.203. Guasch G, Popovici C, Mugneret F, Chaffanet M, Pontarotti P, Birnbaum D,P\u00C3\u00A9busque M-J. Endogenous retroviral sequence is fused to FGFR1 kinase inthe 8p12 stem-cell myeloproliferative disorder with t(8;19)(p12;q13.3). Blood.2002;101(1):286\u00E2\u0080\u00938.204. Lamprecht B, Bonifer C, Mathas S. Repeat element-driven activation ofproto-oncogenes in human malignancies. Cell Cycle. 2010;9(21):4276\u00E2\u0080\u009381.205. B\u00C3\u00B6hne A, Brunet F, Galiana-Arnoux D, Schultheis C, Volff J-N. Transposableelements as drivers of genomic and biological diversity in vertebrates.Chromosom Res. 2008;16(1):203\u00E2\u0080\u009315.206. Rebollo R, Horard B, Hubert B, Vieira C. Jumping genes and epigenetics:Towards new species. Gene. 2010;454(1\u00E2\u0080\u00932):1\u00E2\u0080\u00937.207. Hansen KD, Timp W, Bravo HC, Sabunciyan S, Langmead B, McDonald OG,Wen B, Wu H, Liu Y, Diep D, et al. Increased methylation variation inepigenetic domains across cancer types. Nat Genet. 2011;43(8):768\u00E2\u0080\u009375.208. Landau Dan A, Clement K, Ziller Michael J, Boyle P, Fan J, Gu H, StevensonK, Sougnez C, Wang L, Li S, et al. Locally Disordered Methylation Forms theBasis of Intratumor Methylome Variation in Chronic Lymphocytic Leukemia.Cancer Cell. 2014;26(6):813\u00E2\u0080\u009325.209. Li S, Garrett-Bakelman FE, Chung SS, Sanders MA, Hricik T, Rapaport F, PatelJ, Dillon R, Vijay P, Brown AL, et al. Distinct evolution and dynamics ofepigenetic and genetic heterogeneity in acute myeloid leukemia. Nat Med.2016;22(7):792\u00E2\u0080\u00939.210. Mazor T, Pankov A, Song Jun S, Costello Joseph F. IntratumoralHeterogeneity of the Epigenome. Cancer Cell. 2016;29(4):440\u00E2\u0080\u009351.211. Hanahan D, Weinberg Robert A. Hallmarks of Cancer: The Next Generation.Cell. 2011;144(5):646\u00E2\u0080\u009374.212. Brock A, Chang H, Huang S. Non-genetic heterogeneity - a mutation-independent driving force for the somatic evolution of tumours. Nat RevGenet. 2009;10(5):336\u00E2\u0080\u009342.213. Werfel J, Krause S, Bischof AG, Mannix RJ, Tobin H, Bar-Yam Y, Bellin RM,Ingber DE. How Changes in Extracellular Matrix Mechanics and GeneExpression Variability Might Combine to Drive Cancer Progression. PLoSOne. 2013;8(10):e76122.214. Pisco AO, Brock A, Zhou J, Moor A, Mojtahedi M, Jackson D, Huang S. Non-Darwinian dynamics in therapy-induced cancer drug resistance. NatCommun. 2013;4:2467.215. Marusyk A, Almendro V, Polyak K. Intra-tumour heterogeneity: a lookingglass for cancer? Nat Rev Cancer. 2012;12(5):323\u00E2\u0080\u009334.216. Bashtrykov P, Jankevicius G, Smarandache A, Jurkowska Renata Z, Ragozin S,Jeltsch A. Specificity of Dnmt1 for Methylation of Hemimethylated CpGSites Resides in Its Catalytic Domain. Chem Biol. 2012;19(5):572\u00E2\u0080\u00938.217. Lavie L, Kitova M, Maldener E, Meese E, Mayer J. CpG methylation directlyregulates transcriptional activity of the human endogenous retrovirus familyHERV-K(HML-2). J Virol. 2005;79(2):876\u00E2\u0080\u009383.218. van de Lagemaat LN, Landry J-R, Mager DL, Medstrand P. Transposableelements in mammals promote regulatory variation and diversification ofgenes with specialized functions. Trends Genet. 2003;19(10):530\u00E2\u0080\u00936.219. Simons C, Pheasant M, Makunin IV, Mattick JS. Transposon-free regions inmammalian genomes. Genome Res. 2006;16(2):164\u00E2\u0080\u009372.220. Mortada H, Vieira C, Lerat E. Genes Devoid of Full-Length TransposableElement Insertions are Involved in Development and in the Regulation ofTranscription in Human and Closely Related Species. J Mol Evol. 2010;71(3):180\u00E2\u0080\u009391.221. Sorek R, Ast G, Graur D. Alu-Containing Exons are Alternatively Spliced.Genome Res. 2002;12(7):1060\u00E2\u0080\u00937.Babaian and Mager Mobile DNA (2016) 7:24 Page 20 of 21222. Vorechovsky I. Transposable elements in disease-associated cryptic exons.Hum Genet. 2010;127(2):135\u00E2\u0080\u009354.223. Darby MM, Leek JT, Langmead B, Yolken RH, Sabunciyan S: Widespreadsplicing of repetitive element loci into coding regions of gene transcripts.Hum Mol Gen 2016, in press.224. Lev Maor G, Yearim A, Ast G. The alternative role of DNA methylation insplicing regulation. Trends Genet. 2015;31(5):274\u00E2\u0080\u009380.225. Laurette P, Strub T, Koludrovic D, Keime C, Le Gras S, Seberg H, VanOtterloo E, Imrichova H, Siddaway R, Aerts S, et al. Transcription factor MITFand remodeller BRG1 define chromatin organisation at regulatory elementsin melanoma cells. eLife. 2015;4:e06857.\u00E2\u0080\u00A2 We accept pre-submission inquiries \u00E2\u0080\u00A2 Our selector tool helps you to find the most relevant journal\u00E2\u0080\u00A2 We provide round the clock customer support \u00E2\u0080\u00A2 Convenient online submission\u00E2\u0080\u00A2 Thorough peer review\u00E2\u0080\u00A2 Inclusion in PubMed and all major indexing services \u00E2\u0080\u00A2 Maximum visibility for your researchSubmit your manuscript atwww.biomedcentral.com/submitSubmit your next manuscript to BioMed Central and we will help you at every step:Babaian and Mager Mobile DNA (2016) 7:24 Page 21 of 21"@en . "Article"@en . "10.14288/1.0367866"@en . "eng"@en . "Reviewed"@en . "Vancouver : University of British Columbia Library"@en . "BioMed Central"@en . "10.1186/s13100-016-0080-x"@en . "Attribution 4.0 International (CC BY 4.0)"@en . "http://creativecommons.org/licenses/by/4.0/"@en . "Faculty"@en . "Gene regulation"@en . "Endogenous retrovirus"@en . "Long terminal repeat"@en . "Retrotransposon"@en . "Epigenetics"@en . "Cancer"@en . "Alternative promoter"@en . "Exaptation"@en . "Transcription"@en . "Endogenous retroviral promoter exaptation in human cancer"@en . "Text"@en . "http://hdl.handle.net/2429/66165"@en .