Next Article in Journal
Extracellular Vesicles and Epidermal Growth Factor Receptor Activation: Interplay of Drivers in Cancer Progression
Previous Article in Journal
Standardization of Body Composition Status in Patients with Advanced Urothelial Tumors: The Role of a CT-Based AI-Powered Software for the Assessment of Sarcopenia and Patient Outcome Correlation
Previous Article in Special Issue
Genomic and Transcriptomic Analyses of Malignant Pleural Mesothelioma (MPM) Samples Reveal Crucial Insights for Preclinical Testing
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exploring the Expression of the «Dark Matter» of the Genome in Mesothelioma for Potentially Predictive Biomarkers for Prognosis and Immunotherapy

by
Emanuela Felley-Bosco
Laboratory of Molecular Oncology, Department of Thoracic Surgery, Zürich University Hospital, 8091 Zurich, Switzerland
Cancers 2023, 15(11), 2969; https://doi.org/10.3390/cancers15112969
Submission received: 15 April 2023 / Revised: 21 May 2023 / Accepted: 26 May 2023 / Published: 29 May 2023
(This article belongs to the Special Issue Predictive and Prognostic Factors in Mesothelioma)

Abstract

:

Simple Summary

Endogenous retroviruses (ERVs) are integrated retroviral elements that cover 8% of the human genome, representing thereby four-fold the fraction of the genome encoding for protein-coding genes. Overall, their expression represents up to 0.5% of all non-rRNA transcripts in mesothelioma, and some are specifically re-activated. This part of the genome has been considered until recently “junk DNA” or “DNA dark matter”. However, analysis of ERV expression has recently gained attention in several cancers, especially in the context of immunotherapy. In this review, our aim is to raise the curiosity of non-specialists by illustrating the potential of exploring the expression of “DNA dark matter” for prognosis and immunotherapy, potentially as predictive biomarkers in mesothelioma.

Abstract

Recent high-throughput RNA sequencing technologies have confirmed that a large part of the non-coding genome is transcribed. The priority for further investigations is nevertheless generally given in cancer to coding sequences, due to the obvious interest of finding therapeutic targets. In addition, several RNA-sequencing pipelines eliminate repetitive sequences, which are difficult to analyze. In this review, we shall focus on endogenous retroviruses. These sequences are remnants of ancestral germline infections by exogenous retroviruses. These sequences represent 8% of human genome, meaning four-fold the fraction of the genome encoding for proteins. These sequences are generally mostly repressed in normal adult tissues, but pathological conditions lead to their de-repression. Specific mesothelioma-associated endogenous retrovirus expression and their association to clinical outcome is discussed.

1. Introduction: Why It Is Important to Explore Expressed Genome in Mesothelioma

Malignant mesothelioma (reviewed in [1,2]) is a rare cancer with dismal prognosis. It arises in the mesothelium, a tissue with mesodermal origins covering the lungs (pleural mesothelioma), peritoneal cavities (peritoneal mesothelioma), the sacs surrounding the heart (pericardial mesothelioma) and the testes (tunica vaginalis mesothelioma). Exposure to asbestos has been since long-identified as a cause of mesothelioma in the seminal experiments of Wagner [3]. Although the use of asbestos has been banned in several countries, there are several developing nations that continue to use asbestos [1], and the incidence of mesothelioma is still on the rise.
The genetic alteration of tumor suppressors such as CDKN2A and CDKN2B, as observed in many other cancers, and alterations in BRCA-associated protein 1 (BAP1) are frequently observed in mesothelioma tumors (reviewed in [1] and [2]). Another frequently mutated pathway is the NF2/Hippo signaling pathway. Additional less frequent alterations are observed in TP53, in TERT promoter and in genes involved in RNA metabolism. Of note, recent studies have suggested that alterations in epigenetic and splicing regulators [4,5] may represent a novel hallmark of cancer.
Interestingly, for the past 11 years, we have known that 75% of the human genome is transcribed into RNAs, but contrary to what is observed in bacteria, only 2% of these transcripts are translated into proteins [6]. The discovery that a large part of what had been previously called “junk DNA” is actively transcribed and carries out crucial functions inspired the concept of “RNA as epicenter of genetic information” [7], where RNA are the most-influencing molecules in cellular function in eukaryotes. This is supported by the observation that the transcriptome seems to be the best prognostic factor in several cancers [8].
In this review, we focus on the expression of transposable elements (TE), in particular endogenous retroviruses in mesothelioma. Indeed, although they are frequently not investigated in transcriptomics studies because of their repetitive nature, we observed that their expression increases upon mesothelioma development [9] and is associated with better overall survival [10].

2. Endogenous Retroviruses Are the TE with Highest Variation in Mesothelioma

TEs are DNA sequences that move from one location on the genome to another, and they are also known as “jumping genes”. TEs were discovered more than 70 years ago in corn by Barbara McClintock [11]. Then, it was suggested that repeated DNA sequences may affect gene expression through regulatory factors binding sites in their sequences [12]. The total length of these sequences exceeds by a factor greater than 20 that of protein-coding exons [13]. Based on the way they re-transpose in the genome, TEs can be grouped into two major classes: Class I retrotransposons and Class II DNA transposons.
Class I retrotransposons relocate via RNA intermediates, which are reverse-transcribed by a reverse-transcriptase enzyme encoded by the retrotransposon. The DNA is then integrated into the genome [14]. Based on the presence or absence of long terminal repeat (LTR) flanking sequences, Class I retrotransposons are separated into LTR-retrotransposons (also known as endogenous retroviruses or ERV) and non-LTR retrotransposons [15]. The latter cover around 32% of the human genome and include long interspersed nuclear elements (LINEs) and short interspersed nuclear elements (SINEs), per [13] (Figure 1a,b).
LINEs are also autonomous elements, while SINEs are non-autonomous retrotransposons that use the retro-transcription machinery of other TEs (LINEs) [16].
Class II DNA transposons can directly relocate autonomously via a cut and paste mechanism. They are active in many lower organisms, including bacteria [17], but they are inactivated and can no longer transpose in humans, although they cover around 3% of the human genome (Figure 1a). However, DNA transposon-derived sequences affect human biology after having been repurposed over evolutionary time. This is the case in recombination activating genes (RAG1 and RAG2), which derive from Transib DNA TE. Their transposition mechanism mediates V(D)J recombination in B and T cell precursors [18,19].
ERVs are repetitive elements derived from infection by ancient exogenous retroviruses integrated within the genome of germline cells. They have then transmitted through their offspring [20]. ERVs, however, unlike original retroviruses, are deficient in most transposon functions, and represent defective genomic remnants of the retroviral replication cycle. ERVs comprise a significant part of our genomes, covering 8% [13,21] of the human genome (Figure 1a).
The discovery of human ERVs (HERVs) followed the screening of human genomic libraries using either probes from animal retroviruses or by using oligonucleotides with similarity to virus sequences under low-stringency conditions [22]. HERV length varies from 1.5 to 11 kbp [13]. Full-length HERV proviruses (Figure 1b) include gag, which encodes for capside, pol, which encodes the machinery necessary for autonomous replication and env, which encodes for the envelope glycoprotein, flanked by two LTRs. About 90% of ERVs are present in the human genome as single LTRs, termed “solo LTRs”, which originated from recombination events, lacking all open reading frames (ORFs) [23].
A lot of work has been performed for classifying and naming ERVs [24], particularly down to the level of individual proviral loci [25]; however, ERV classification still remains a considerable problem [26]. ERV sequences can be placed into three classes according to their internal homology to exogenous retroviruses, based on phylogenetic analyses of the conserved regions of the pol gene. ERVs clustering with gamma- and epsilonretroviruses are termed Class I; those clustering with lentiviruses, alpha-, beta-, and deltaretroviruses are termed Class II; and those that cluster with spumaviruses are termed Class III, which is the most abundant and also may be the most ancient endogenous retrovirus [24] (Table 1).
MaLR (mammalian apparent LTR retrotransposon) elements, which lack a detectable pol-related sequence, are also considered “Class III” due to the slight homology of some members to Class III gag sequences.
The relative proportions and abundances of Class I, II and III ERVs, as well as MaLR, differ among species, although they are present in all mammalian species tested [27]. For example, Class II ERVs are significantly expanded in mice.
We observed that the expression of ERVs increases during mesothelioma development in mice exposed to asbestos [9]; therefore, our next step was to investigate human samples by comparing available tumor RNA-sequencing (RNA-seq) data from two mesothelioma cohorts: TCGA [28] and Bueno’s [29], or the mesothelioma primary culture cell lines of the FunGeST series [30,31], with human-embryonic-stem-cell-derived mesothelium [32] used as the non-tumor control. Indeed, for the time being, the only RNA-seq data available for normal mesothelial cells were provided in a single-cell mRNA-seq study in the context of a human cell atlas [33]. The polyA RNA-seq protocol that was used has been shown to fail to detect several classes of repeat RNA [34]. We observed that ERVs are the most abundant TEs upregulated or downregulated in mesothelioma patient datasets when compared to mesothelium. ERVs represent on average 74% of the more-than-two-fold upregulated and 48% of the more-than-two-fold downregulated TEs.
In human mesothelioma, we observed that the ERV1 family, which belongs to ERV Class I, has the highest number of upregulated or downregulated loci in ERVs when compared to mesothelial cells [10].
Few ERV-encoded genes, including Class I, are well annotated in databases and can be explored in classical analysis. One of these, env encoding ERVMER34-1 (also called HEMO), is preferentially translated in mesothelioma cells compared to mesothelial cells [35], indicating that proteins from ERVs are produced. Of note, in a pan-cancer analysis, the expression of ERVMER34-1 was associated with immune system repression [36].
Within the ERV1 family, there are three ERV sequences, LTR48B, LTR7Y and LTR6, that are upregulated in all mesothelioma datasets compared to mesothelium. Of note, LTR7Y is an ERV specifically upregulated in early embryo development [37]; therefore, its upregulation in mesothelioma points to developmental signaling re-activation.
A lower number of changes was also observed in the Class III oldest HERV family HERV-L, which has been estimated to be more than 60 to 70 million years old, and the youngest HERV-K (HML-2) family, which is approximately five million years old [38] and belongs to Class II.
The HERV-K family is closely related to the exogenous mouse mammary tumor virus (MMTV) causing breast cancer in mice and has the greatest coding capability [39]. Many HERV-K ERV are still transcriptionally active [40]. There are more than 1000 HERV-K loci in the human genome; however, most of them are “solo-LTRs” [41]. HERV-K family members are less enriched in mesothelioma when compared to mesothelial precursors [10]. ERVmap-k48, also called HERV-K15 [42] had the same expression levels in mesothelioma and mesothelial cells [10]. Gag encoding the ERVmap-k48 locus is near the housekeeping gene SSBP1, and its stable levels in normal vs. tumor tissue might be linked to their coregulation [42].

3. ERV and Epigenetic Regulation in Mesothelioma

Various mechanisms, including accumulation of mutations, RNA silencing and epigenetic regulation control ERV expression and activity [43]. HERV expression is repressed in the pre-implantation embryo and is maintained in most adult tissues [44]. Most of the mechanisms of HERV repression have been inferred from observations in mouse ERVs. In mice, ERV transcription is suppressed by DNA methylation and/or repressive histone modifications depending on tissue type [45]. Some ERV are normally expressed in various developmental stages of human embryogenesis, and their activity is regulated by alterations in epigenetic regulation in different human tissues [43,46,47].
DNA methylation, which is used by cells to silence transcription, is one of the primary epigenetic mechanisms by which ERV expression is regulated. Indeed, DNA methylation plays an important role in different stages of embryogenesis and in somatic cells in regulating ERV transcription [43,47,48,49,50]. In humans, some HERV transcripts are detectable in many cell types [51], but most ERVs are heavily methylated in somatic cells [52,53,54,55,56]. We observed that ERVs with higher expression in mesothelioma are the consequence of the demethylation of nearby CpG islands [9,10]. In addition, it is worth noting that some of the ERVs that are specifically overexpressed in mesothelioma are near genes such as, e.g., the MSLN locus on chromosome 16, which is especially enriched in ERV expression. Mesothelin expression is driven by both YAP activation [57] and promoter de-methylation [58]. The mouse Msln promoter, which has been used to generate transgenic mice expressing large T-antigen upon asbestos exposure [59], contains Mer54b TE; therefore, one could make the hypothesis that the asbestos-induced demethylation-driven expression of Mer54b contributes to large-T antigen expression in MexTag mice.
Besides DNA methylation, histone modifications also play major roles, particularly in undifferentiated and/or stem cells [26]. SETDB1 (SET domain bifurcated histone lysine methyltransferase 1) is a protein lysine methyltransferase that adds methyl moieties to histone H3 at lysine 9 (H3K9). SETDB1-mediated H3K9 tri-methylation (H3K9me3) is responsible for the silencing of ERVs [60,61,62]. H3K9me3 is deposited on ERVs by the SETDB1/KAP1 complex (KAP1 is also known as TRIM28) [63] (Figure 2a).
Of note in the mesothelioma TCGA patients, the quartile with the lowest KAP1 expression has improved overall survival compared with the patients in the higher quartile [64]. It would be interesting to learn whether this is associated with any change in ERV expression.
KAP1 interacts with Krüppel-associated box zinc finger proteins (KRAB-ZFPs), and it has been proposed that the SETDB1/KAP1 complex is recruited to distinct ERVs by different KRAB-ZFPs [65,66,67] (Figure 2a). Therefore, the level of variation in HERV-associated KZFP can potentially explain the differential expressions of HERV in mesothelioma. For example, the expression of ZNF534, which is associated with pluripotency [68], is higher in sarcomatoid compared to epithelioid mesothelioma [29]. In addition, since increased expression of ERVs is observed upon loss of function of SETDB1 [69], it is likely that the subset of mesothelioma patients with mutated SETDB1 display different levels of ERV expression [28,29,70,71]; however, this remains to be demonstrated. An additional regulator of SETDB1 is the human silencing hub (HUSH) complex, which, although mostly involved in LINE silencing, contributes to the regulation of ERV expression (reviewed in [38]).
Global H3K9me3 levels are also decreased and are accompanied by increased ERV expression downstream from the loss of function of lysine acetyltransferase Tat-interactive protein 60. Indeed, the acetylated lysines resulting from the activity of this enzyme recruit bromodomain-containing protein 4, resulting in the increased expression of H3K9 methyltransferases SUV39H1 and SETDB1 and higher levels of H3K9me3 [72].
Some ERVs have promoter or enhancer activity and are associated with active epigenetic marks [73] such as H3K4me1 (an enhancer mark) and exhibit enhancer activity in reporter assays [74]. The depletion of Kap1 in mouse embryonic stem cells leads to the loss of the repressive mark H3K9me3 at selected ERVs, but also the gain of enhancer marks H3K27ac and H3K4me1, which correlates with the induction of a subset of nearby genes [75] (Figure 2b).

4. HERV and Type 1 Interferon Activation

HERVs have been considered as “junk” DNA without biological functions for a long time. However, some HERVs have been co-opted into physiological roles in the host [76]. The best-described example is the production of syncytin-1, a retroviral protein coded by the env gene of a provirus belonging to the HERV-W group, expressed in human trophoblasts [77,78,79,80,81]. In addition, HERVs are important determinants of the pluripotency of human embryonic stem cells and of the reprogramming process of induced pluripotent stem cells [37,82].
We documented that ERV transcripts with increased expression in mesothelioma form double stranded RNA (dsRNA) [9,10]. dsRNA is part of the molecular patterns activating the type-I IFN response, and in the tissue of mice exposed to asbestos, we observed 27 interferon-stimulated genes (ISGs) with an expression higher in tumor samples when compared to the samples from asbestos-exposed mice with no tumors [9]. When we investigated the association of the expression of these ISGs with clinical outcome in mesothelioma patients in the TCGA study [28], we found that overexpression of six of them (DDX58, IFIT2, IFIT3, IFITM1, IRF1, RSAD2), is associated with best overall survival, as it has been previously described for the type I interferon signaling pathway [83]. In human mesothelioma cell lines, the increased expression of ISGs is dependent on the activity of dsRNA sensors and the presence of interferon B1 gene [10]. The latter is located on chromosome 9, near the CDKN2A gene, a gene altered in 50–60% of mesothelioma ([84,85]). The IFNB1 gene is co-deleted in 70% of mesothelioma with CDKN2A deletion [86].
The increased expression of ERV transcripts forming dsRNA structures resulting in the activation of dsRNA sensors is consistent with previous observations of MDA5 activation observed due to increased synthesis of ERV upon suppression of DNA methyltransferases [87,88]. Downstream of MDA5 activation, type 1 interferon signaling is stimulated (reviewed in [89]).
Viral mimicry response is widely associated with the expression of TE in several cancers [90] and in a mesothelioma development model [9]. In agreement with these observations, we observed that human mesothelioma tumors expressing high levels of ERVmap_1248, which is the near-full-length ERV with the highest expression in mesothelioma compared to mesothelial cells, show a basal activation of type-I IFN signaling and are associated with longer overall survival (Table 2) in three mesothelioma datasets [10].
The samples with the highest ERVmap_1248 expression also had the highest inflammatory signature score (based on the expression of CD8A, STAT1, LAG3 and CD274), which predicts response to checkpoint inhibitors in the CheckMate743 trial, where previously untreated mesothelioma patients received either the immune checkpoint inhibitors anti-PD1 and anti-CTLA-4 or chemotherapy [91]. Future studies should prospectively assess the value of ERVmap_1248 expression as predictive of sensitivity to immunotherapy including immune checkpoint inhibition. However, for therapies inducing type-I IFN signaling, if the contribution of the production of IFNB1 by tumor cells themselves has a role, it should not be forgotten that some forms of mesothelioma have lost type-I IFN genes, as mentioned above [86].
There are several examples where the expression of ERV has been associated with better outcome and/or response to immunotherapy. The activation of type-I IFN is associated with responses to immune checkpoint inhibitors in clear cell renal cancer, which originates in organs from embryonic-related tissue as mesothelioma [52]. In an urothelial cancer cohort, ERV expression is a better predictor of patient response to anti-PDL1 therapy when compared to type-I IFN signatures [34]. The expression of some well-defined [92] ERVs is associated with both immune activation and immune checkpoint signaling upregulation [93] in clear cell renal cell carcinoma. The expression levels of one of these ERVs, ERV3-2, predicted response to single-agent PD-1/PD-L1 antibody, and it has recently been demonstrated that this particular ERV is mostly expressed in immune cells [94], which shows that selected ERVs point to responsive immune cells. In an additional clear cell renal cell carcinoma study, ERV_4700 (or ERVmap_4700) expression predicted response to immunocheckpoint blockade and was associated with translation, meaning the production of neoantigens [95]. In another clear cell renal cell carcinoma study, RNAseq performed on more than 100 formalin-fixed paraffin-embedded patient samples from the two clinical trials (CheckMate-010 and CheckMate-025) identified a link between ERV_2282 (or ERVmap_2282) expression with better overall survival in the group treated with anti-PD-1 [96]. Differences between the different studies in this cancer type have been suggested to be linked not only to RNA sequencing approaches, as previously mentioned, but also to cohort differences and ERV mapping and annotation (reviewed in [97]).
In melanoma patients, high ERV and other TE expression is associated with low-risk tumors with better outcomes, while the opposite was associated with repression of TE [98]. Complete response to anti-PD1 treatment has been shown to be associated with high ERVmap_2637 expression in melanoma patients [60] and negatively correlated with KDM5B expression, which recruits SETDB1. In non-small-cell lung cancer, high expression of the MER4 ERV is associated with better clinical outcome and efficacy of anti-PD1/anti-PDL1 [99] therapy. In addition, ERV envelope glycoproteins have been recently identified as the dominant anti-tumor antibody target in non-small lung cancer [100].
Therapeutic approaches depending on the type-I IFN signaling pathway have already been implemented in the clinic [101] to treat mesothelioma patients. Preclinical studies have suggested the role of this signaling either to improve sensitivity to chemotherapy or in the sensitivity to oncolytic therapy [102,103].
Because of the correlation between the expression of the selected ERVs and the signature predictive of the response to immunocheckpoint inhibition (Table 2) and type I interferon, ERV expression could be useful to select those mesothelioma patients with epithelioid histotype that have the potential to respond to immune checkpoint inhibition, since this tumor histotype responds less compared to the sarcomatoid histotype [104].

5. Conclusions

In this review, we summarized the current knowledge on ERVs and their contribution to mesothelioma phenotypes. Besides being associated with the activation of type 1 interferon, HERVs contribute to the expression of tumor-specific antigens in several cancers including mesothelioma [105]. Indeed, HERVs can function as alternative promoters for nearby genes in tumor cells, and cryptic transcription start sites within HERV can generate aberrant protein-coding mRNAs. HLA-pull-down experiments have confirmed that these proteins act as neoantigens, and it will be of interest if some of these overlap with tumor-cell-derived vaccines currently used in the clinic [106].
In addition, these novel ORFs may lead to uncontrolled cell proliferation [107]. For example, aberrant high activity of LTR THE1B contributes to a high degree of transcription of colony stimulation factor receptor 1 in Hodgkin’s lymphoma [108]. HERV-induced chromosomal translocation is another oncogenic mechanism as has been shown in prostate cancer, where HERV-K_22q11.3 is fused to the oncogene E26 transformation-specific family gene ETV1 [109]. We already mentioned that the choice of RNA-sequencing strategy (polyA enrichment vs. rRNA depletion) introduces a bias in the analysis of TE expression. Novel long-read technologies [110], which allow the capture of full-length sequences, will improve the detection and the annotation of expressed TE, including co-transcripts and chimeric transcripts.
It has recently been acknowledged that classical immunotherapy sensitivity biomarkers such as tumor mutation burden and PD-L1 expression have limited predictive value in mesothelioma immunotherapy (reviewed in [111]). Ongoing studies investigating tumor microenvironment, including single-cell sequencing and novel analysis pipelines for neoantigen discovery, will help address the remaining questions about the importance of the expression of selected ERVs in mesothelioma tumor cells or in the tumor microenvironment and their predictive value for immunotherapy.
Finally, besides ERVs, other TEs such as selected LINE1 are overexpressed in mesothelioma [10], and their contribution to pathology and evaluation as biomarkers has yet to be evaluated.

Funding

This research was funded by the Swiss National Science Foundation, grant 320030_182690, Walter-Bruckerhoff Stiftung and Stiftung für Angewandte Krebsforchung.

Data Availability Statement

Publicly available datasets were analyzed in this study. These data can be found at zenodo.org: https://doi.org/10.5281/zenodo.7220103.

Acknowledgments

The authors are grateful to the many scientists who have contributed to this field whose work was not cited due to space limitations.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Felley-Bosco, E.; MacFarlane, M. Asbestos: Modern Insights for Toxicology in the Era of Engineered Nanomaterials. Chem. Res. Toxicol. 2018, 31, 994–1008. [Google Scholar] [CrossRef]
  2. Carbone, M.; Adusumilli, P.S.; Alexander, H.R., Jr.; Baas, P.; Bardelli, F.; Bononi, A.; Bueno, R.; Felley-Bosco, E.; Galateau-Salle, F.; Jablons, D.; et al. Mesothelioma: Scientific Clues for Prevention, Diagnosis, and Therapy. CA Cancer J. Clin. 2019, 69, 402–429. [Google Scholar] [CrossRef]
  3. Wagner, J.C. Experimental Production of Mesothelial Tumours of the Pleura by Implantation of Dusts in Laboratory Animals. Nature 1962, 196, 180–181. [Google Scholar] [CrossRef] [PubMed]
  4. Imielinski, M.; Berger, A.H.; Hammerman, P.S.; Hernandez, B.; Pugh, T.J.; Hodis, E.; Cho, J.; Suh, J.; Capelletti, M.; Sivachenko, A.; et al. Mapping the Hallmarks of Lung Adenocarcinoma with Massively Parallel Sequencing. Cell 2012, 150, 1107–1120. [Google Scholar] [CrossRef] [PubMed]
  5. Elsasser, S.J.; Allis, C.D.; Lewis, P.W. Cancer. New Epigenetic Drivers of Cancers. Science 2011, 331, 1145–1146. [Google Scholar] [CrossRef] [PubMed]
  6. Bernstein, B.E.; Birney, E.; Dunham, I.; Green, E.D.; Gunter, C.; Snyder, M. An Integrated Encyclopedia of DNA Elements in the Human Genome. Nature 2012, 489, 57–74. [Google Scholar] [CrossRef]
  7. Amaral, P.; Mattick, J. RNA, the Epicenter of Genetic Information; CRC Press: Boca Raton, FL, USA, 2023; ISBN 978-1-00-310924-2. [Google Scholar]
  8. Smith, J.C.; Sheltzer, J.M. Genome-Wide Identification and Analysis of Prognostic Features in Human Cancers. Cell Rep. 2022, 38, 110569. [Google Scholar] [CrossRef]
  9. Sun, S.; Frontini, F.; Qi, W.; Hariharan, A.; Ronner, M.; Wipplinger, M.; Blanquart, C.; Rehrauer, H.; Fonteneau, J.F.; Felley-Bosco, E. Endogenous Retrovirus Expression Activates Type-I Interferon Signaling in an Experimental Mouse Model of Mesothelioma Development. Cancer Lett. 2021, 507, 26–38. [Google Scholar] [CrossRef]
  10. Sun, S. Viral Mimicry Response Is Associated with Clinical Outcome in Pleural Mesothelioma. J. Thorac. Oncol. Clin. Res. Rep. 2022, 3, 100430. [Google Scholar] [CrossRef]
  11. Mc, C.B. The Origin and Behavior of Mutable Loci in Maize. Proc. Natl. Acad. Sci. USA 1950, 36, 344–355. [Google Scholar] [CrossRef]
  12. Britten, R.J.; Davidson, E.H. Gene Regulation for Higher Cells: A Theory. Science 1969, 165, 349–357. [Google Scholar] [CrossRef] [PubMed]
  13. Lander, E.S.; Linton, L.M.; Birren, B.; Nusbaum, C.; Zody, M.C.; Baldwin, J.; Devon, K.; Dewar, K.; Doyle, M.; FitzHugh, W.; et al. Initial Sequencing and Analysis of the Human Genome. Nature 2001, 409, 860–921. [Google Scholar] [CrossRef] [PubMed]
  14. Boeke, J.D.; Garfinkel, D.J.; Styles, C.A.; Fink, G.R. Ty Elements Transpose through an RNA Intermediate. Cell 1985, 40, 491–500. [Google Scholar] [CrossRef] [PubMed]
  15. Makałowski, W.; Gotea, V.; Pande, A.; Makałowska, I. Transposable Elements: Classification, Identification, and Their Use As a Tool For Comparative Genomics. Methods Mol. Biol. 2019, 1910, 177–207. [Google Scholar] [CrossRef]
  16. Kramerov, D.A.; Vassetzky, N.S. Origin and Evolution of SINEs in Eukaryotic Genomes. Hered. Edinb. 2011, 107, 487–495. [Google Scholar] [CrossRef]
  17. Kazazian, H.H., Jr.; Moran, J.V. Mobile DNA in Health and Disease. N. Engl. J. Med. 2017, 377, 361–370. [Google Scholar] [CrossRef]
  18. Huang, S.; Tao, X.; Yuan, S.; Zhang, Y.; Li, P.; Beilinson, H.A.; Zhang, Y.; Yu, W.; Pontarotti, P.; Escriva, H.; et al. Discovery of an Active RAG Transposon Illuminates the Origins of V(D)J Recombination. Cell 2016, 166, 102–114. [Google Scholar] [CrossRef]
  19. Kapitonov, V.V.; Jurka, J. RAG1 Core and V(D)J Recombination Signal Sequences Were Derived from Transib Transposons. PLoS Biol. 2005, 3, e181. [Google Scholar] [CrossRef]
  20. Stocking, C.; Kozak, C.A. Murine Endogenous Retroviruses. Cell Mol. Life Sci. 2008, 65, 3383–3398. [Google Scholar] [CrossRef]
  21. Hoyt, S.J.; Storer, J.M.; Hartley, G.A.; Grady, P.G.S.; Gershman, A.; de Lima, L.G.; Limouse, C.; Halabian, R.; Wojenski, L.; Rodriguez, M.; et al. From Telomere to Telomere: The Transcriptional and Epigenetic State of Human Repeat Elements. Science 2022, 376, eabk3112. [Google Scholar] [CrossRef]
  22. Belshaw, R.; Pereira, V.; Katzourakis, A.; Talbot, G.; Paces, J.; Burt, A.; Tristem, M. Long-Term Reinfection of the Human Genome by Endogenous Retroviruses. Proc. Natl. Acad. Sci. USA 2004, 101, 4894–4899. [Google Scholar] [CrossRef] [PubMed]
  23. Thomas, J.; Perron, H.; Feschotte, C. Variation in Proviral Content among Human Genomes Mediated by LTR Recombination. Mob. DNA 2018, 9, 36. [Google Scholar] [CrossRef] [PubMed]
  24. Vargiu, L.; Rodriguez-Tome, P.; Sperber, G.O.; Cadeddu, M.; Grandi, N.; Blikstad, V.; Tramontano, E.; Blomberg, J. Classification and Characterization of Human Endogenous Retroviruses; Mosaic Forms Are Common. Retrovirology 2016, 13, 7. [Google Scholar] [CrossRef] [PubMed]
  25. Tokuyama, M.; Kong, Y.; Song, E.; Jayewickreme, T.; Kang, I.; Iwasaki, A. ERVmap Analysis Reveals Genome-Wide Transcription of Human Endogenous Retroviruses. Proc. Natl. Acad. Sci. USA 2018, 115, 12565–12572. [Google Scholar] [CrossRef] [PubMed]
  26. Mager, D.L.; Stoye, J.P. Mammalian Endogenous Retroviruses. Microbiol. Spectr. 2015, 3, Mdna3-0009–2014. [Google Scholar] [CrossRef]
  27. Gifford, R.; Tristem, M. The Evolution, Distribution and Diversity of Endogenous Retroviruses. Virus Genes 2003, 26, 291–315. [Google Scholar] [CrossRef]
  28. Hmeljak, J.; Sanchez-Vega, F.; Hoadley, K.A.; Shih, J.; Stewart, C.; Heiman, D.; Tarpey, P.; Danilova, L.; Drill, E.; Gibb, E.A.; et al. Integrative Molecular Characterization of Malignant Pleural Mesothelioma. Cancer Discov. 2018, 8, 1548–1565. [Google Scholar] [CrossRef]
  29. Bueno, R.; Stawiski, E.W.; Goldstein, L.D.; Durinck, S.; De Rienzo, A.; Modrusan, Z.; Gnad, F.; Nguyen, T.T.; Jaiswal, B.S.; Chirieac, L.R.; et al. Comprehensive Genomic Analysis of Malignant Pleural Mesothelioma Identifies Recurrent Mutations, Gene Fusions and Splicing Alterations. Nat. Genet. 2016, 48, 407–416. [Google Scholar] [CrossRef]
  30. de Reynies, A.; Jaurand, M.C.; Renier, A.; Couchy, G.; Hysi, I.; Elarouci, N.; Galateau-Salle, F.; Copin, M.C.; Hofman, P.; Cazes, A.; et al. Molecular Classification of Malignant Pleural Mesothelioma: Identification of a Poor Prognosis Subgroup Linked to the Epithelial-to-Mesenchymal Transition. Clin. Cancer Res. 2014, 20, 1323–1334. [Google Scholar] [CrossRef]
  31. Quetel, L.; Meiller, C.; Assié, J.B.; Blum, Y.; Imbeaud, S.; Montagne, F.; Tranchant, R.; de Wolf, J.; Caruso, S.; Copin, M.C.; et al. Genetic Alterations of Malignant Pleural Mesothelioma: Association with Tumor Heterogeneity and Overall Survival. Mol. Oncol. 2020, 14, 1207–1223. [Google Scholar] [CrossRef]
  32. Colunga, T.; Hayworth, M.; Kress, S.; Reynolds, D.M.; Chen, L.; Nazor, K.L.; Baur, J.; Singh, A.M.; Loring, J.F.; Metzger, M.; et al. Human Pluripotent Stem Cell-Derived Multipotent Vascular Progenitors of the Mesothelium Lineage Have Utility in Tissue Engineering and Repair. Cell Rep. 2019, 26, 2566–2579.e10. [Google Scholar] [CrossRef] [PubMed]
  33. Han, X.; Zhou, Z.; Fei, L.; Sun, H.; Wang, R.; Chen, Y.; Chen, H.; Wang, J.; Tang, H.; Ge, W.; et al. Construction of a Human Cell Landscape at Single-Cell Level. Nature 2020, 581, 303–309. [Google Scholar] [CrossRef] [PubMed]
  34. Solovyov, A.; Vabret, N.; Arora, K.S.; Snyder, A.; Funt, S.A.; Bajorin, D.F.; Rosenberg, J.E.; Bhardwaj, N.; Ting, D.T.; Greenbaum, B.D. Global Cancer Transcriptome Quantifies Repeat Element Polarization between Immunotherapy Responsive and T Cell Suppressive Classes. Cell Rep. 2018, 23, 512–521. [Google Scholar] [CrossRef] [PubMed]
  35. Grosso, S.; Marini, A.; Gyuraszova, K.; Voorde, J.V.; Sfakianos, A.; Garland, G.D.; Tenor, A.R.; Mordue, R.; Chernova, T.; Morone, N.; et al. The Pathogenesis of Mesothelioma Is Driven by a Dysregulated Translatome. Nat. Commun. 2021, 12, 4920. [Google Scholar] [CrossRef]
  36. Kasperek, A.; Béguin, A.; Bawa, O.; De Azevedo, K.; Job, B.; Massard, C.; Scoazec, J.; Heidmann, T.; Heidmann, O. Therapeutic Potential of the Human Endogenous Retroviral Envelope Protein HEMO: A Pan-cancer Analysis. Mol. Oncol. 2022, 16, 1451–1473. [Google Scholar] [CrossRef]
  37. Goke, J.; Lu, X.; Chan, Y.S.; Ng, H.H.; Ly, L.H.; Sachs, F.; Szczerbinska, I. Dynamic Transcription of Distinct Classes of Endogenous Retroviral Elements Marks Specific Populations of Early Human Embryonic Cells. Cell Stem. Cell 2015, 16, 135–141. [Google Scholar] [CrossRef]
  38. Geis, F.K.; Goff, S.P. Silencing and Transcriptional Regulation of Endogenous Retroviruses: An Overview. Viruses 2020, 12, 884. [Google Scholar] [CrossRef]
  39. Xue, B.; Sechi, L.A.; Kelvin, D.J. Human Endogenous Retrovirus K (HML-2) in Health and Disease. Front. Microbiol. 2020, 11, 1690. [Google Scholar] [CrossRef]
  40. Garcia-Montojo, M.; Doucet-O’Hare, T.; Henderson, L.; Nath, A. Human Endogenous Retrovirus-K (HML-2): A Comprehensive Review. Crit. Rev. Microbiol. 2018, 44, 715–738. [Google Scholar] [CrossRef]
  41. Hughes, J.F.; Coffin, J.M. Human Endogenous Retrovirus K Solo-LTR Formation and Insertional Polymorphisms: Implications for Human and Viral Evolution. Proc. Natl. Acad. Sci. USA 2004, 101, 1668–1672. [Google Scholar] [CrossRef]
  42. Schmitt, K.; Reichrath, J.; Roesch, A.; Meese, E.; Mayer, J. Transcriptional Profiling of Human Endogenous Retrovirus Group HERV-K(HML-2) Loci in Melanoma. Genome Biol. Evol. 2013, 5, 307–328. [Google Scholar] [CrossRef] [PubMed]
  43. Rowe, H.M.; Trono, D. Dynamic Control of Endogenous Retroviruses during Development. Virology 2011, 411, 273–287. [Google Scholar] [CrossRef]
  44. Friedli, M.; Trono, D. The Developmental Control of Transposable Elements and the Evolution of Higher Species. Annu. Rev. Cell Dev. Biol. 2015, 31, 429–451. [Google Scholar] [CrossRef] [PubMed]
  45. Leung, D.C.; Lorincz, M.C. Silencing of Endogenous Retroviruses: When and Why Do Histone Marks Predominate? Trends Biochem. Sci. 2012, 37, 127–133. [Google Scholar] [CrossRef]
  46. Pisano, M.P.; Grandi, N.; Tramontano, E. High-Throughput Sequencing Is a Crucial Tool to Investigate the Contribution of Human Endogenous Retroviruses (HERVs) to Human Biology and Development. Viruses 2020, 12, 633. [Google Scholar] [CrossRef]
  47. Hurst, T.P.; Magiorkinis, G. Epigenetic Control of Human Endogenous Retrovirus Expression: Focus on Regulation of Long-Terminal Repeats (LTRs). Viruses 2017, 9, 130. [Google Scholar] [CrossRef] [PubMed]
  48. Howard, G.; Eiges, R.; Gaudet, F.; Jaenisch, R.; Eden, A. Activation and Transposition of Endogenous Retroviral Elements in Hypomethylation Induced Tumors in Mice. Oncogene 2008, 27, 404–408. [Google Scholar] [CrossRef]
  49. Jaenisch, R.; Schnieke, A.; Harbers, K. Treatment of Mice with 5-Azacytidine Efficiently Activates Silent Retroviral Genomes in Different Tissues. Proc. Natl. Acad. Sci. USA 1985, 82, 1451–1455. [Google Scholar] [CrossRef]
  50. Bourc’his, D.; Bestor, T.H. Meiotic Catastrophe and Retrotransposon Reactivation in Male Germ Cells Lacking Dnmt3L. Nature 2004, 431, 96–99. [Google Scholar] [CrossRef]
  51. Seifarth, W.; Frank, O.; Zeilfelder, U.; Spiess, B.; Greenwood, A.D.; Hehlmann, R.; Leib-Mösch, C. Comprehensive Analysis of Human Endogenous Retrovirus Transcriptional Activity in Human Tissues with a Retrovirus-Specific Microarray. J. Virol. 2005, 79, 341–352. [Google Scholar] [CrossRef]
  52. de Cubas, A.A.; Dunker, W.; Zaninovich, A.; Hongo, R.A.; Bhatia, A.; Panda, A.; Beckermann, K.E.; Bhanot, G.; Ganesan, S.; Karijolich, J.; et al. DNA Hypomethylation Promotes Transposable Element Expression and Activation of Immune Signaling in Renal Cell Cancer. JCI Insight 2020, 5, 137569. [Google Scholar] [CrossRef] [PubMed]
  53. Lavie, L.; Kitova, M.; Maldener, E.; Meese, E.; Mayer, J. CpG Methylation Directly Regulates Transcriptional Activity of the Human Endogenous Retrovirus Family HERV-K(HML-2). J. Virol. 2005, 79, 876–883. [Google Scholar] [CrossRef] [PubMed]
  54. Stengel, S.; Fiebig, U.; Kurth, R.; Denner, J. Regulation of Human Endogenous Retrovirus-K Expression in Melanomas by CpG Methylation. Genes. Chromosomes Cancer 2010, 49, 401–411. [Google Scholar] [CrossRef]
  55. Xie, Q.; Chen, S.; Tian, R.; Huang, X.; Deng, R.; Xue, B.; Qin, Y.; Xu, Y.; Wang, J.; Guo, M.; et al. Long Noncoding RNA ITPRIP-1 Positively Regulates the Innate Immune Response through Promotion of Oligomerization and Activation of MDA5. J. Virol. 2018, 92, e00507-18. [Google Scholar] [CrossRef] [PubMed]
  56. Kong, Y.; Rose, C.M.; Cass, A.A.; Williams, A.G.; Darwish, M.; Lianoglou, S.; Haverty, P.M.; Tong, A.J.; Blanchette, C.; Albert, M.L.; et al. Transposable Element Expression in Tumors Is Associated with Immune Infiltration and Increased Antigenicity. Nat. Commun. 2019, 10, 5228. [Google Scholar] [CrossRef]
  57. Ren, Y.R.; Patel, K.; Paun, B.C.; Kern, S.E. Structural Analysis of the Cancer-Specific Promoter in Mesothelin and in Other Genes Overexpressed in Cancers. J. Biol. Chem. 2011, 286, 11960–11969. [Google Scholar] [CrossRef]
  58. Nelson, H.H.; Almquist, L.M.; LaRocca, J.L.; Plaza, S.L.; Lambert-Messerlian, G.M.; Sugarbaker, D.J.; Bueno, R.; Godleski, J.J.; Marsit, C.J.; Christensen, B.C.; et al. The Relationship between Tumor MSLN Methylation and Serum Mesothelin (SMRP) in Mesothelioma. Epigenetics 2011, 6, 1029–1034. [Google Scholar] [CrossRef]
  59. Robinson, C.; van Bruggen, I.; Segal, A.; Dunham, M.; Sherwood, A.; Koentgen, F.; Robinson, B.W.; Lake, R.A. A Novel SV40 TAg Transgenic Model of Asbestos-Induced Mesothelioma: Malignant Transformation Is Dose Dependent. Cancer Res. 2006, 66, 10786–10794. [Google Scholar] [CrossRef]
  60. Zhang, S.M.; Cai, W.L.; Liu, X.; Thakral, D.; Luo, J.; Chan, L.H.; McGeary, M.K.; Song, E.; Blenman, K.R.M.; Micevic, G.; et al. KDM5B Promotes Immune Evasion by Recruiting SETDB1 to Silence Retroelements. Nature 2021, 598, 682–687. [Google Scholar] [CrossRef]
  61. Karimi, M.M.; Goyal, P.; Maksakova, I.A.; Bilenky, M.; Leung, D.; Tang, J.X.; Shinkai, Y.; Mager, D.L.; Jones, S.; Hirst, M.; et al. DNA Methylation and SETDB1/H3K9me3 Regulate Predominantly Distinct Sets of Genes, Retroelements, and Chimeric Transcripts in MESCs. Cell Stem Cell 2011, 8, 676–687. [Google Scholar] [CrossRef]
  62. Fukuda, K.; Shinkai, Y. SETDB1-Mediated Silencing of Retroelements. Viruses 2020, 12, 596. [Google Scholar] [CrossRef]
  63. Pontis, J.; Planet, E.; Offner, S.; Turelli, P.; Duc, J.; Coudray, A.; Theunissen, T.W.; Jaenisch, R.; Trono, D. Hominoid-Specific Transposable Elements and KZFPs Facilitate Human Embryonic Genome Activation and Control Transcription in Naive Human ESCs. Cell Stem Cell 2019, 24, 724–735.e5. [Google Scholar] [CrossRef]
  64. Lorenzini, E.; Torricelli, F.; Zamponi, R.; Donati, B.; Manicardi, V.; Sauta, E.; Faria do Valle, I.; Reggiani, F.; Gugnoni, M.; Manzotti, G.; et al. KAP1 Is a New Non-Genetic Vulnerability of Malignant Pleural Mesothelioma (MPM). NAR Cancer 2022, 4, zcac024. [Google Scholar] [CrossRef] [PubMed]
  65. Shi, H.; Strogantsev, R.; Takahashi, N.; Kazachenka, A.; Lorincz, M.C.; Hemberger, M.; Ferguson-Smith, A.C. ZFP57 Regulation of Transposable Elements and Gene Expression within and beyond Imprinted Domains. Epigenet. Chromatin 2019, 12, 49. [Google Scholar] [CrossRef] [PubMed]
  66. Turelli, P.; Castro-Diaz, N.; Marzetta, F.; Kapopoulou, A.; Raclot, C.; Duc, J.; Tieng, V.; Quenneville, S.; Trono, D. Interplay of TRIM28 and DNA Methylation in Controlling Human Endogenous Retroelements. Genome Res. 2014, 24, 1260–1270. [Google Scholar] [CrossRef] [PubMed]
  67. Frietze, S.; O’Geen, H.; Blahnik, K.R.; Jin, V.X.; Farnham, P.J. ZNF274 Recruits the Histone Methyltransferase SETDB1 to the 3’ Ends of ZNF Genes. PLoS ONE 2010, 5, e15082. [Google Scholar] [CrossRef]
  68. Carter, T.A.; Singh, M.; Dumbovic, G.; Chobirko, J.D.; Rinn, J.L.; Feschotte, C. Mosaic Cis-Regulatory Evolution Drives Transcriptional Partitioning of HERVH Endogenous Retrovirus in the Human Embryo. eLife 2022, 11, 76257. [Google Scholar] [CrossRef]
  69. Cuellar, T.L.; Herzner, A.M.; Zhang, X.; Goyal, Y.; Watanabe, C.; Friedman, B.A.; Janakiraman, V.; Durinck, S.; Stinson, J.; Arnott, D.; et al. Silencing of Retrotransposons by SETDB1 Inhibits the Interferon Response in Acute Myeloid Leukemia. J. Cell Biol. 2017, 216, 3535–3549. [Google Scholar] [CrossRef]
  70. Pagano, M.; Ceresoli, L.G.; Zucali, P.A.; Pasello, G.; Garassino, M.; Grosso, F.; Tiseo, M.; Soto Parra, H.; Zanelli, F.; Cappuzzo, F.; et al. Mutational Profile of Malignant Pleural Mesothelioma (MPM) in the Phase II RAMES Study. Cancers 2020, 12, 2948. [Google Scholar] [CrossRef]
  71. Kang, H.C.; Kim, H.K.; Lee, S.; Mendez, P.; Kim, J.W.; Woodard, G.; Yoon, J.H.; Jen, K.Y.; Fang, L.T.; Jones, K.; et al. Whole Exome and Targeted Deep Sequencing Identify Genome-Wide Allelic Loss and Frequent SETDB1 Mutations in Malignant Pleural Mesotheliomas. Oncotarget 2016, 7, 8321–8331. [Google Scholar] [CrossRef]
  72. Rajagopalan, D.; Tirado-Magallanes, R.; Bhatia, S.S.; Teo, W.S.; Sian, S.; Hora, S.; Lee, K.K.; Zhang, Y.; Jadhav, S.P.; Wu, Y.; et al. TIP60 Represses Activation of Endogenous Retroviral Elements. Nucleic Acids Res. 2018, 46, 9456–9470. [Google Scholar] [CrossRef] [PubMed]
  73. Huda, A.; Bowen, N.J.; Conley, A.B.; Jordan, I.K. Epigenetic Regulation of Transposable Element Derived Human Gene Promoters. Gene 2011, 475, 39–48. [Google Scholar] [CrossRef] [PubMed]
  74. Xie, M.; Hong, C.; Zhang, B.; Lowdon, R.F.; Xing, X.; Li, D.; Zhou, X.; Lee, H.J.; Maire, C.L.; Ligon, K.L.; et al. DNA Hypomethylation within Specific Transposable Element Families Associates with Tissue-Specific Enhancer Landscape. Nat. Genet. 2013, 45, 836–841. [Google Scholar] [CrossRef] [PubMed]
  75. Rowe, H.M.; Kapopoulou, A.; Corsinotti, A.; Fasching, L.; Macfarlan, T.S.; Tarabay, Y.; Viville, S.; Jakobsson, J.; Pfaff, S.L.; Trono, D. TRIM28 Repression of Retrotransposon-Based Enhancers Is Necessary to Preserve Transcriptional Dynamics in Embryonic Stem Cells. Genome Res. 2013, 23, 452–461. [Google Scholar] [CrossRef]
  76. Grandi, N.; Pisano, M.P.; Tramontano, E. The Emerging Field of Human Endogenous Retroviruses: Understanding Their Physiological Role and Contribution to Diseases. Future Virol. 2019, 14, 441–444. [Google Scholar] [CrossRef]
  77. Blond, J.L.; Lavillette, D.; Cheynet, V.; Bouton, O.; Oriol, G.; Chapel-Fernandes, S.; Mandrand, B.; Mallet, F.; Cosset, F.L. An Envelope Glycoprotein of the Human Endogenous Retrovirus HERV-W Is Expressed in the Human Placenta and Fuses Cells Expressing the Type D Mammalian Retrovirus Receptor. J. Virol. 2000, 74, 3321–3329. [Google Scholar] [CrossRef]
  78. Mi, S.; Lee, X.; Li, X.; Veldman, G.M.; Finnerty, H.; Racie, L.; LaVallie, E.; Tang, X.Y.; Edouard, P.; Howes, S.; et al. Syncytin Is a Captive Retroviral Envelope Protein Involved in Human Placental Morphogenesis. Nature 2000, 403, 785–789. [Google Scholar] [CrossRef]
  79. Lavialle, C.; Cornelis, G.; Dupressoir, A.; Esnault, C.; Heidmann, O.; Vernochet, C.; Heidmann, T. Paleovirology of “Syncytins”, Retroviral Env Genes Exapted for a Role in Placentation. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2013, 368, 20120507. [Google Scholar] [CrossRef]
  80. Dupressoir, A.; Lavialle, C.; Heidmann, T. From Ancestral Infectious Retroviruses to Bona Fide Cellular Genes: Role of the Captured Syncytins in Placentation. Placenta 2012, 33, 663–671. [Google Scholar] [CrossRef]
  81. Griffiths, D.J. Endogenous Retroviruses in the Human Genome Sequence. Genome Biol. 2001, 2, REVIEWS1017. [Google Scholar] [CrossRef]
  82. Grow, E.J.; Flynn, R.A.; Chavez, S.L.; Bayless, N.L.; Wossidlo, M.; Wesche, D.J.; Martin, L.; Ware, C.B.; Blish, C.A.; Chang, H.Y.; et al. Intrinsic Retroviral Reactivation in Human Preimplantation Embryos and Pluripotent Cells. Nature 2015, 522, 221–225. [Google Scholar] [CrossRef] [PubMed]
  83. Broad Institute TCGA Genome Data Analysis Center (2014): Correlation between Aggregated Molecular Cancer Subtypes and Selected Clinical Features. Broad Institute of MIT and Harvard. Available online: http://gdac.broadinstitute.org/runs/analyses__2014_10_17/reports/cancer/MESO-TP/Correlate_Clinical_vs_Molecular_Subtypes/nozzle.html (accessed on 12 May 2015).
  84. Hiltbrunner, S.; Fleischmann, Z.; Sokol, E.S.; Zoche, M.; Felley-Bosco, E.; Curioni Fontecedro, A. Genomic Landscape of Pleural and Peritoneal Mesothelioma Tumors. Br. J. Cancer 2022, 127, 1997–2005. [Google Scholar] [CrossRef] [PubMed]
  85. Mangiante, L.; Alcala, N.; Sexton-Oates, A.; Di Genova, A.; Gonzalez-Perez, A.; Khandekar, A.; Bergstrom, E.N.; Kim, J.; Liu, X.; Blazquez-Encinas, R.; et al. Multiomic Analysis of Malignant Pleural Mesothelioma Identifies Molecular Axes and Specialized Tumor Profiles Driving Intertumor Heterogeneity. Nat. Genet. 2023, 55, 607–618. [Google Scholar] [CrossRef] [PubMed]
  86. Delaunay, T.; Achard, C.; Boisgerault, N.; Grard, M.; Petithomme, T.; Chatelain, C.; Dutoit, S.; Blanquart, C.; Royer, P.J.; Minvielle, S.; et al. Frequent Homozygous Deletions of Type I Interferon Genes in Pleural Mesothelioma Confer Sensitivity to Oncolytic Measles Virus. J. Thorac. Oncol. 2020, 15, 827–842. [Google Scholar] [CrossRef]
  87. Roulois, D.; Loo Yau, H.; Singhania, R.; Wang, Y.; Danesh, A.; Shen, S.Y.; Han, H.; Liang, G.; Jones, P.A.; Pugh, T.J.; et al. DNA-Demethylating Agents Target Colorectal Cancer Cells by Inducing Viral Mimicry by Endogenous Transcripts. Cell 2015, 162, 961–973. [Google Scholar] [CrossRef] [PubMed]
  88. Chiappinelli, K.B.; Strissel, P.L.; Desrichard, A.; Li, H.; Henke, C.; Akman, B.; Hein, A.; Rote, N.S.; Cope, L.M.; Snyder, A.; et al. Inhibiting DNA Methylation Causes an Interferon Response in Cancer via DsRNA Including Endogenous Retroviruses. Cell 2015, 162, 974–986. [Google Scholar] [CrossRef]
  89. Felley-Bosco, E. Non-Coding RNA Regulatory Networks in Mesothelioma: A Narrative Review of Their Implication in Innate Immune Signaling Pathway. Precis Cancer Med. 2021, 4, pcm-21-4. [Google Scholar] [CrossRef]
  90. Chen, R.; Ishak, C.A.; De Carvalho, D.D. Endogenous Retroelements and the Viral Mimicry Response in Cancer Therapy and Cellular Homeostasis. Cancer Discov. 2021, 11, 2707–2725. [Google Scholar] [CrossRef]
  91. Peters, S.; Scherpereel, A.; Cornelissen, R.; Oulkhouir, Y.; Greillier, L.; Kaplan, M.A.; Talbot, T.; Monnet, I.; Hiret, S.; Baas, P.; et al. First-Line Nivolumab plus Ipilimumab versus Chemotherapy in Patients with Unresectable Malignant Pleural Mesothelioma: 3-Year Outcomes from CheckMate 743. Ann. Oncol. 2022, 33, 488–499. [Google Scholar] [CrossRef]
  92. Mayer, J.; Blomberg, J.; Seal, R.L. A Revised Nomenclature for Transcribed Human Endogenous Retroviral Loci. Mob. DNA 2011, 2, 7. [Google Scholar] [CrossRef]
  93. Panda, A.; de Cubas, A.A.; Stein, M.; Riedlinger, G.; Kra, J.; Mayer, T.; Smith, C.C.; Vincent, B.G.; Serody, J.S.; Beckermann, K.E.; et al. Endogenous Retrovirus Expression Is Associated with Response to Immune Checkpoint Blockade in Clear Cell Renal Cell Carcinoma. JCI Insight 2018, 3, 121522. [Google Scholar] [CrossRef] [PubMed]
  94. Au, L.; Hatipoglu, E.; Robert de Massy, M.; Litchfield, K.; Beattie, G.; Rowan, A.; Schnidrig, D.; Thompson, R.; Byrne, F.; Horswell, S.; et al. Determinants of Anti-PD-1 Response and Resistance in Clear Cell Renal Cell Carcinoma. Cancer Cell 2021, 39, 1497–1518.e11. [Google Scholar] [CrossRef] [PubMed]
  95. Smith, C.C.; Beckermann, K.E.; Bortone, D.S.; De Cubas, A.A.; Bixby, L.M.; Lee, S.J.; Panda, A.; Ganesan, S.; Bhanot, G.; Wallen, E.M.; et al. Endogenous Retroviral Signatures Predict Immunotherapy Response in Clear Cell Renal Cell Carcinoma. J. Clin. Investig. 2018, 128, 4804–4820. [Google Scholar] [CrossRef] [PubMed]
  96. Braun, D.A.; Hou, Y.; Bakouny, Z.; Ficial, M.; Sant’ Angelo, M.; Forman, J.; Ross-Macdonald, P.; Berger, A.C.; Jegede, O.A.; Elagina, L.; et al. Interplay of Somatic Alterations and Immune Infiltration Modulates Response to PD-1 Blockade in Advanced Clear Cell Renal Cell Carcinoma. Nat. Med. 2020, 26, 909–918. [Google Scholar] [CrossRef]
  97. Wolf, M.M.; Rathmell, W.K.; De Cubas, A.A. Immunogenicity in Renal Cell Carcinoma: Shifting Focus to Alternative Sources of Tumour-Specific Antigens. Nat. Rev. Nephrol. 2023; Online ahead of print. [Google Scholar] [CrossRef]
  98. Badal, B.; Solovyov, A.; Di Cecilia, S.; Chan, J.M.; Chang, L.W.; Iqbal, R.; Aydin, I.T.; Rajan, G.S.; Chen, C.; Abbate, F.; et al. Transcriptional Dissection of Melanoma Identifies a High-Risk Subtype Underlying TP53 Family Genes and Epigenome Deregulation. JCI Insight 2017, 2, 92102. [Google Scholar] [CrossRef]
  99. Lecuelle, J.; Favier, L.; Fraisse, C.; Lagrange, A.; Kaderbhai, C.; Boidot, R.; Chevrier, S.; Joubert, P.; Routy, B.; Truntzer, C.; et al. MER4 Endogenous Retrovirus Correlated with Better Efficacy of Anti-PD1/PD-L1 Therapy in Non-Small Cell Lung Cancer. J. Immunother. Cancer 2022, 10, e004241. [Google Scholar] [CrossRef]
  100. Ng, K.W.; Boumelha, J.; Enfield, K.S.S.; Almagro, J.; Cha, H.; Pich, O.; Karasaki, T.; Moore, D.A.; Salgado, R.; Sivakumar, M.; et al. Antibodies against Endogenous Retroviruses Promote Lung Cancer Immunotherapy. Nature 2023, 616, 563–573. [Google Scholar] [CrossRef]
  101. Sterman, D.H.; Recio, A.; Haas, A.R.; Vachani, A.; Katz, S.I.; Gillespie, C.T.; Cheng, G.; Sun, J.; Moon, E.; Pereira, L.; et al. A Phase I Trial of Repeated Intrapleural Adenoviral-Mediated Interferon-Beta Gene Transfer for Mesothelioma and Metastatic Pleural Effusions. Mol. Ther. 2010, 18, 852–860. [Google Scholar] [CrossRef]
  102. Vanbervliet-Defrance, B.; Delaunay, T.; Daunizeau, T.; Kepenekian, V.; Glehen, O.; Weber, K.; Estornes, Y.; Ziverec, A.; Djemal, L.; Delphin, M.; et al. Cisplatin Unleashes Toll-like Receptor 3-Mediated Apoptosis through the Downregulation of c-FLIP in Malignant Mesothelioma. Cancer Lett. 2020, 472, 29–39. [Google Scholar] [CrossRef]
  103. Achard, C.; Boisgerault, N.; Delaunay, T.; Roulois, D.; Nedellec, S.; Royer, P.J.; Pain, M.; Combredet, C.; Mesel-Lemoine, M.; Cellerin, L.; et al. Sensitivity of Human Pleural Mesothelioma to Oncolytic Measles Virus Depends on Defects of the Type I Interferon Response. Oncotarget 2015, 6, 44892–44904. [Google Scholar] [CrossRef]
  104. Baas, P.; Scherpereel, A.; Nowak, A.K.; Fujimoto, N.; Peters, S.; Tsao, A.S.; Mansfield, A.S.; Popat, S.; Jahan, T.; Antonia, S.; et al. First-Line Nivolumab plus Ipilimumab in Unresectable Malignant Pleural Mesothelioma (CheckMate 743): A Multicentre, Randomised, Open-Label, Phase 3 Trial. Lancet 2021, 397, 375–386. [Google Scholar] [CrossRef] [PubMed]
  105. Shah, N.M.; Jang, H.J.; Liang, Y.; Maeng, J.H.; Tzeng, S.-C.; Wu, A.; Basri, N.L.; Qu, X.; Fan, C.; Li, A.; et al. Pan-Cancer Analysis Identifies Tumor-Specific Antigens Derived from Transposable Elements. Nat. Genet. 2023, 55, 631–639. [Google Scholar] [CrossRef] [PubMed]
  106. Dumoulin, D.W.; Cornelissen, R.; Bezemer, K.; Baart, S.J.; Aerts, J.G.J.V. Long-Term Follow-Up of Mesothelioma Patients Treated with Dendritic Cell Therapy in Three Phase I/II Trials. Vaccines 2021, 9, 525. [Google Scholar] [CrossRef] [PubMed]
  107. Jansz, N.; Faulkner, G.J. Endogenous Retroviruses in the Origins and Treatment of Cancer. Genome Biol. 2021, 22, 147. [Google Scholar] [CrossRef]
  108. Warming, S.; Liu, P.; Suzuki, T.; Akagi, K.; Lindtner, S.; Pavlakis, G.N.; Jenkins, N.A.; Copeland, N.G. Evi3, a Common Retroviral Integration Site in Murine B-Cell Lymphoma, Encodes an EBFAZ-Related Krüppel-like Zinc Finger Protein. Blood 2003, 101, 1934–1940. [Google Scholar] [CrossRef] [PubMed]
  109. Tomlins, S.A.; Laxman, B.; Dhanasekaran, S.M.; Helgeson, B.E.; Cao, X.; Morris, D.S.; Menon, A.; Jing, X.; Cao, Q.; Han, B.; et al. Distinct Classes of Chromosomal Rearrangements Create Oncogenic ETS Gene Fusions in Prostate Cancer. Nature 2007, 448, 595–599. [Google Scholar] [CrossRef] [PubMed]
  110. Shahid, S.; Slotkin, R.K. The Current Revolution in Transposable Element Biology Enabled by Long Reads. Curr. Opin. Plant Biol. 2020, 54, 49–56. [Google Scholar] [CrossRef]
  111. Perrino, M.; De Vincenzo, F.; Cordua, N.; Borea, F.; Aliprandi, M.; Santoro, A.; Zucali, P.A. Immunotherapy with Immune Checkpoint Inhibitors and Predictive Biomarkers in Malignant Mesothelioma: Work Still in Progress. Front. Immunol. 2023, 14, 1121557. [Google Scholar] [CrossRef]
Figure 1. (a) Percentage of the human genome covered by transposable elements (TEs) compared to protein-coding sequences. (b) The classification of Class I TE. EN: endonuclease; RT: reverse transcriptase.
Figure 1. (a) Percentage of the human genome covered by transposable elements (TEs) compared to protein-coding sequences. (b) The classification of Class I TE. EN: endonuclease; RT: reverse transcriptase.
Cancers 15 02969 g001
Figure 2. (a) A model for SETDB1 recruitment. Shown is a schematic illustrating the current model for KAP1-mediated transcriptional repression. KAP1 is thought to be brought to the genome by interaction with a KRAB-ZNF, which binds specific sites in the DNA. KAP1 in turn has been suggested to recruit the histone methyltransferase SETDB1, which then specifically mediates trimethylation of lysine 9 of histone H3 near the KRAB-ZNF binding sites. (b) Upper scheme: most ERVs are epigenetically silenced by DNA methylation and/or repressive histone modifications. Middle scheme: some ERVs function as promoters and are marked by promoter-associated histone modifications. Lower scheme: some ERVs have enhancer activity and are marked by enhancer-associated histone modifications and transcription factors (TF). LTR: long terminal repeat.
Figure 2. (a) A model for SETDB1 recruitment. Shown is a schematic illustrating the current model for KAP1-mediated transcriptional repression. KAP1 is thought to be brought to the genome by interaction with a KRAB-ZNF, which binds specific sites in the DNA. KAP1 in turn has been suggested to recruit the histone methyltransferase SETDB1, which then specifically mediates trimethylation of lysine 9 of histone H3 near the KRAB-ZNF binding sites. (b) Upper scheme: most ERVs are epigenetically silenced by DNA methylation and/or repressive histone modifications. Middle scheme: some ERVs function as promoters and are marked by promoter-associated histone modifications. Lower scheme: some ERVs have enhancer activity and are marked by enhancer-associated histone modifications and transcription factors (TF). LTR: long terminal repeat.
Cancers 15 02969 g002
Table 1. Representative human ERVs.
Table 1. Representative human ERVs.
ClassRelated Exogenous GroupGroup
IGammaHERV1, HERV-H *, HERV-W *, HERV-E *, ERVMER34-1
IIBetaHERV-K *
IIISpumaHERV-L *, MaLR
*: ERVs are also sometimes defined according to the cellular tRNA molecule that is used as a primer for reverse transcription via annealing to an 18 bp binding site sequence. The letter indicates the specific tRNA, e.g., H: His tRNA.
Table 2. Human ERVs identified as prognostic or immunotherapy biomarkers in various cancer types.
Table 2. Human ERVs identified as prognostic or immunotherapy biomarkers in various cancer types.
ERVCancerBiomarker TypeReference
ERVmap_1248MesotheliomaPrognostic[10]
ERV3-2Clear cell renal cell carcinomaImmunotherapy response [93,94]
ERV_4700Clear cell renal cell carcinomaImmunotherapy response[95]
ERV_2282Clear cell renal cell carcinomaPrognostic[96]
ERVmap_2637MelanomaImmunotherapy response[60]
MER4Non-small cell lung cancerPrognostic and immunotherapy response[99]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Felley-Bosco, E. Exploring the Expression of the «Dark Matter» of the Genome in Mesothelioma for Potentially Predictive Biomarkers for Prognosis and Immunotherapy. Cancers 2023, 15, 2969. https://doi.org/10.3390/cancers15112969

AMA Style

Felley-Bosco E. Exploring the Expression of the «Dark Matter» of the Genome in Mesothelioma for Potentially Predictive Biomarkers for Prognosis and Immunotherapy. Cancers. 2023; 15(11):2969. https://doi.org/10.3390/cancers15112969

Chicago/Turabian Style

Felley-Bosco, Emanuela. 2023. "Exploring the Expression of the «Dark Matter» of the Genome in Mesothelioma for Potentially Predictive Biomarkers for Prognosis and Immunotherapy" Cancers 15, no. 11: 2969. https://doi.org/10.3390/cancers15112969

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop