Abstract
Accounting for approximately 8% of the human genome, human endogenous retroviruses (HERVs) have been implicated in a variety of cancers including gliomas. In normal cells, tight epigenetic regulation of HERVs prevent aberrant expression; however, in cancer cells, HERVs expression remains pervasive, suggesting a role of HERVs in oncogenic transformation. HERVs may contribute to oncogenesis in several ways including insertional mutagenesis, chromosomal rearrangements, proto-oncogene formation, and maintenance of stemness. On the other hand, recent data has suggested that reversing epigenetic silencing of HERVs may induce robust anti-tumor immune responses, suggesting HERVs’ potential therapeutic utility in gliomas. By reversing epigenetic modifications that silence HERVs, DNA methyltransferase, and histone deacetylase inhibitors may stimulate a viral-mimicry cascade via HERV-derived dsRNA formation that induces interferon-mediated apoptosis. Leveraging this anti-tumor autoimmune response may be a unique avenue to target certain subsets of epigenetically-dysregulated gliomas. Nevertheless, the role of HERVs in gliomas as either arbitrators of oncogenesis or forerunners of the innate anti-tumor immune response remains unclear. Here, we review the role of HERVs in gliomas, their potential dichotomous function in propagating oncogenesis and stimulating the anti-tumor immune response, and identify future directions for research.
Keywords: etiology, glioma, human endogenous retrovirus, viral mimicry
First described in 1981, human endogenous retroviruses (HERV) account for approximately 8% of the human genome and serve as relics of ancestral germline retroviral infection.1–3 HERVs can be identified by the presence of canonical retroviral genes: gag (core structural proteins), pol (viral replication enzymes including reverse transcriptase) env (envelope protein), and LTR (long terminal repeats containing promoters, enhancers, and other regulatory proteins; Figure 1). Currently, there are 31 HERV families (classified according to their tRNA primer-binding site) that are divided into three main classes based on their homology to exogenous retroviruses (see Table 1). Due to their accumulation of coding deletions and postinsertional mutations, many HERVs have inherently lost their ability to replicate and form functional viral particles. However, a few HERVs (mainly HERV-K, subtype HML-2) have retained near full-length sequences with some degree of transcriptional activity.4–6 Polymorphisms have also been identified in HERVs however, their clinical significance remains unclear.7 The transcription of these “semi-active” HERVs remains tightly regulated by epigenetic modification of retroviral LTRs and nuclear transcriptional factors. As such, many HERV elements are expressed in early embryogenesis and are critical in organ development but remain silent in normal adults.8 However, HERVs may be re-expressed in pathologic conditions with epigenetic dysregulation such as amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), and cancer. Over the last several years, an association between human cancers (melanoma, breast, prostate, and lymphoma) and HERV transcriptional activation has been reported, but a clear etiologic role of HERVs in cancer has yet to be established.9–12 A thorough examination of HERVs in brain neoplasms, specifically glioma, has yet to be performed. Here, we review the potential role of HERVs in glioma pathophysiology and highlight therapeutic options to leverage HERVs for immunotherapy.
Fig. 1.
Schema of original HERV infection and integration into human genome. HERVs maintain similar retroviral phylogeny with conserved RNA sequences gag (group antigens), pol (polymerases), and env (envelope proteins). Retroviral integration into the genome requires reverse transcriptase and integrase. Once integrated, HERVs are typically silenced by epigenetic machinery, but occasionally, HERV env splice products (HERV-K hml) may be transcribed into functional proteins, rec, and np9.
Table 1.
Human Endogenous Retrovirus Classification
HERV Class | Homology | Type |
---|---|---|
HERV Class I | Mammalian Type C retroviruses (murine leukemia virus) | HERV-H, HERV-F, HERV-RW, HERV-W, HERV-P, HERV-E,HERV-ERI, HERV-R, HERV-T, HERV-I, HERV-FRD |
HERV Class II | Type D retroviruses Mammalian Type B (Mouse mammary tumor virus) |
HERV-K (HML1-10) |
HERV Class III | Spumavirus (foamy virus) | HERV-L |
HERV Expression in Gliomas
Currently, the presence or significance of HERV expression in glioma samples remains uncertain. Most of the extensive bioinformatic HERV analyses do not include gliomas due to the heterogeneity of these cancers, limitations caused by the RNA preparation method, lack of deep sequencing, and inadequate control samples. However, a few bioinformatics studies have suggested that HERV protein expression is increased in gliomas. A large study of over 300 glioma samples from the Chinese Glioma Genome Atlas (CGGA) demonstrated that HERVK-113 was detected in the genome of over 90% of samples, although no association between virus expression and survival was noted.13 Additionally, it has also been suggested that HERV LTRs are differentially upregulated in glioma tissue compared to normal brain samples, albeit these results are preliminary.14 Despite these findings, quantitative mRNA qPCR analysis from established glioma cell lines and patient tissue samples failed to demonstrate a robust expression of either full-length HERV-K envelope protein or its splice products. Since HERV-K envelope splice products have been associated with tumor transformation, their absence suggests that HERV-K splice products may not be associated with human astrocytic tumors. Aside from HERV-K, the association of other families of HERVs and gliomas has not been investigated thoroughly. Although limited in nature, the aforementioned studies support the need for a more thorough investigation of the association of HERVs and gliomas.
HERVs as Oncogenic Drivers in Glioma
Research over the last four decades has implicated endogenous retroviruses in cellular de-differentiation and malignant transformations.15 HERVs may contribute to tumor formation/pathogenesis in two main ways: indirect transcriptional regulation of oncogenes/tumor suppressors and expression of oncogenic HERV proteins. However, these oncogenic properties have not been methodically investigated in gliomas. Therefore, we aim to highlight potential mechanisms by which HERVs may influence glioma oncogenesis (Figure 2).
Fig. 2.
Theoretical oncogenic role of HERVs in cancer.16 HERV LTR can be rearranged to act as cis-promoters of downstream oncogenes. Nonhomologous recombination of HERVs can lead to chromosomal rearrangements, leading to disruption of functional cell regulator genes. Insertional mutagenesis of HERV elements can also inactivate tumor suppressor genes. HERV-K envelop splice products (np9 and rec) have also been associated with oncogenic activity. HERV-FRD and HERV-W have been implicated in cell-cell fusion via syncitin-1, and have been implicated in several gynecological cancers.17
Oncogenic HERV Proteins
Although none of the HERVs possess established oncogenes, protein products from HERV transcripts have been implicated in inducing tumorigenesis by directly altering cellular signaling pathways, promoting tumor immune evasion, and inducing cellular fusion.5 Splice products from the envelope protein of HERV-K (HML-2), rec, and Np9 have been linked to tumorigenesis in a variety of cancers due to their activation of the c-MYC proto-oncogene.18 By binding to the transcriptional repressor promyelocytic leukemia zinc finger (PLZF), rec, and np9 may dysregulate the cell cycle and modulate cellular proliferation in specific cancers. Aside from mediating the expression of the protoconcogene C-MYC, PLZF also regulates tumor suppressor genes p53 and p21.19 Of note, PLZF was noted to be increased more than three-fold compared to normal controls in glioblastoma samples, suggesting that this is a potential target for HERV-K proteins in human high-grade gliomas.19,20 Nevertheless, the role of PLZF in human gliomas as a tumor initiator has not yet been established. The other splice product np9 has also been associated with disinhibiting Notch signaling by binding with the Ligand of Numb Protein X (LNX) and initiating proteasome degradation.21 Since the LNX protein has been shown to be downregulated in glioma cells, an investigation of the role of np9-LNX complex may help understand aberrant Notch signaling pathways in gliomas.22
Aside from cellular proliferation, HERVs have also been implicated in promoting tumor invasion and metastasis for specific cancer types. Most notably, HERV-H has been associated with expansion of a immune-evasive CD271+ cell populations in metastatic colon cancer cells undergoing epithelial-mesenchymal transitions. In contrast, depletion of the HERV-H-mediated CD271 populations resulted in tumor regression.23 CD271 (p75 neurotrophin receptor) is a well-known mediator of cellular migration and invasion in gliomas and medulloblastomas; furthermore, CD271+ cells also form a stem-cell niche of treatment-refractory glioma cells that resist conventional chemoradiation and evade immunosurveillance.24–27 As such, investigations into the role of HERV-H in regulating CD271 expression in gliomas may be warranted.
Indirect Gene Dysregulation
HERVs have also been implicated in indirectly dysregulating proto-oncogenes due to nonhomologous recombination in various cancers. In glioma, HERV-K derived sequences were discovered interspersed within tumor suppressor and DNA repair genes, BRCA2, and XRCC1, respectively.28 These viral-derived interspersed rearrangements share homology to HERV-K and may be implicated in gene inactivation of regulatory genetic machinery. Additionally, chromosomal translocation of HERV LTRs has also been associated with enhanced expression of downstream oncogenes in several cancers.29 In prostate cancer, the enhanced expression of the ETV1 oncogene has been associated with upstream HERV-K 22q11.23 5′-UTR LTR translocation; the ETV1 oncogene has also been noted to be overexpressed in a variety of capicua (CIC) mutated oligodendrogliomas and has been associated with an aggressive phenotype.30–32 Similar, oncogenic chromosomal rearrangements of HERVs have been described in childhood glioma forming syndromes such as constitutional mismatch repair deficiency (CMMRD) due to biallelic inactivation of the mismatch repair gene, PMS2.33,34
The promoter function of HERV LTRs also serves as a well-established mechanism of HERV-induced cellular dysregulation. Through cis activation of a downstream oncogene, an HERV-derived LTR can induce tumorigenesis.35,36 In such a manner, HERV LTRs can promote overexpression of proteins such as CSF1R and ERBB4 in certain lymphomas. These proteins have also been linked to an aggressive phenotype in glioblastoma and medulloblastoma, respectively, but a causal link between HERVs and their upregulation has not been explored.36–40 The transcriptional regulation of cancer-promoting genes by LTRs has been well described in a variety of cancers. Still, cis LTR regulation of oncogenes in gliomas has yet to be definitively established (see Table 2).
Table 2.
Potential Linkages of HERVs as Cancer-Promoting Agents in Glioma
Study | HERV-element | Mechanism | Cancer Cellular Gene | Gene Role in Glioma |
---|---|---|---|---|
18 | Rec/Np9 | Activation of C-MYC oncogene | PLZF | Upregulated three-fold in glioma samples |
21 | Np9 | Amplifying Notch signaling via LNX degradation | LNX | Downregulated in gliomas |
28 | HERV-K | Gene inactivation by insertional mutagenesis | BRCA2 | Germline inactivation mutations noted in some samples |
28 | HERV-K | Gene inactivation by insertional mutagenesis | XRCC1 | Polymorphisms associated with increased risk of gliomas |
32 | HERV-K LTR | Chromosomal rearrangement activating ETV1 proto-oncogene | ETV1 | ETV1 overexpression associated with aggressive phenotype in oligodendrogliomas |
33 | HERV LTR | Chromosomal rearrangement | PMS2 | Association with CMMRD |
37 | HERV LTR | cis activation of promoter | CSF1R | Implicated in Glioma invasion41 |
40 | HERV LTR | cis activation of promoter | ERBB4 | Associated with aggressive glioma phenotype42 |
43 | HERV-K | Correlation to CD133+ stem-cell markers in melanoma | CD133 | Glioma stem-cell marker, associated with aggressive phenotype |
8 | HERV-K (Hml-2) | Maintenance of stemness through interactions with CD98HC/mTOR | CD98HC | CD98 upregulated in astrocytic tumors, facilitates tumor proliferation, amino acid transport44 |
Abbreviations: CSF1R, colony-stimulating factor 1 receptor; ERBB4, receptor tyrosine-protein kinase; ETV1, ETS translocation variant 1; LNX, E3 ubiquitin-protein ligase; PLZF, promyelocytic leukemia zinc finger protein; PMS2, Mismatch repair endonuclease; XRCC1, X-ray repair cross-complementing protein 1.
HERVs May Be Responsible for Stem-Cell Phenotype in Cancer Cells
One of the earliest examples of HERV function was discovered in embryonic stem cells, which activate HERV-K expression to maintain pluripotency and an undifferentiated state. Through epigenetic modifications (histone/DNA hypomethylation), HERV-K expression is induced during embryogenesis/development to protect the host by inducing antiviral defense mechanisms through the envelope splice product, rec.45 Additionally, envelope proteins from HERV-W and FRD, syncitin-1 and 2, both facilitate cell-cell fusion during embryogenesis and trophectoderm development.46 In cancer, associations between HERVs and cancer stem cells have been described. CD133 (a common glioma stem-cell marker) was found to be tightly correlated to HERV expression in melanoma cell lines, and treatment with reverse transcriptase inhibitors restricted both HERV-K expression and CD133+ cancer cell populations.43 The HERK-K envelope protein (HML-2) is found to be overexpressed in human pluripotent stem cells (PSC), and consequently downregulated during neuronal differentiation. The HERV-K (HML-2) interacts with CD98HC and activates the mTOR pathway, while concurrently inducing epigenetic changes through lysophosphatidylcholine acyltransferase (LPCAT1).8 CD98 is widely expressed in astrocytic tumors, where it may impact tumor biology by facilitating amino acid transport and promoting oncogenic transformation.44 Accordingly, epigenetic silencing or downregulation of HML-2 reverses stemness and promotes neuronal differentiation, suggesting a potential role of HERV-K envelope protein in neurogenerative conditions and neuro-oncology.8
HERVs as a Biomarker of Cancer
The association of HERV expression and cancer suggests that HERVs could be utilized as a prognostic biomarker for certain cancers. The most notable prognostic biomarker HERV-K (HML-2) has been studied in breast and ovarian cancer patients; HERV-K (HML-2) expression in serum and tissues was quantitatively correlated to both breast and ovarian cancer histopathology (low-grade lesions with demonstrably lower expression of HERV-K (HML-2) and antibodies compared to higher-grade lesions).12,47,48 Additionally, HERV expression was also noted to increase after induction of isocitrate dehydrogenase (IDH) R132 mutation in glioma cells. In this model, immortalized astrocytes with inducible IDH1 mutant enzyme were noted to have a global increase in HERV expression with concomitant epigenetic changes.49 Similarly, HERV upregulation (HERV-K, T, W, F) was also noted to increase in glioma-initiating cells after exposure to cytomegalovirus. Given the potential etiologic role of CMV in glioma, there is a suggestion that HERVs may have a bystander role in glioma tumorigenesis.50 Analogous findings were noted in non-HIV permissive glioma cell lines (U87) after exposure to HIV-1.51 However, it is unclear if HERV expression is a consequence of oncogenesis or an intrinsic cellular immune defense mechanism. Nevertheless, these findings suggest a potential role for HERVs as a biomarker of disease in gliomas.
Therapeutic Implications of HERVs in Gliomas
In addition to the potential tumorigenic role of HERVs, recent studies suggest that HERV expression may help augment immunotherapy for gliomas.
Adaptive Immune Response Against HERVs
The adaptive immune response to HERVs is inherently suppressed because of two factors: the presence of immune tolerance to intrinsic HERV protein and epigenetic silencing of HERV-specific lymphocytes. However, in cancer, upregulation of HERV expression activates the immune response to a certain extent producing an antibody response against HERVs proteins.52 Anti-HERV-K antibodies were discovered in several cancers, including prostate, breast, germ-cell tumors, and melanoma.36,48,53–55 To amplify the efficacy of this intrinsic antibody response, direct antibody therapy, and vaccination strategies have been suggested for a variety of cancers. Indeed, exogenous monoclonal antibodies against HERV env protein have demonstrated considerable efficacy in a variety of experimental models of cancers and in a human study of multiple sclerosis.52,56 Preclinical vaccination strategies against HERV envelope or gag proteins have also demonstrated considerable safety and efficacy in reducing the metastatic tumor burden in renal carcinoma and other cancers.5,57,58 Of note, vaccination strategies against HERV proteins in gliomas have not yet been investigated.
HERV-Mediated Viral-Mimicry
As mentioned previously, HERV expression in normal cells is tightly mediated by epigenetic repressive changes that silence retroviral transcription. Epigenetic modifications, including histone/DNA hypermethylation and histone deacetylation, prevent HERV transcription, thereby silencing innate immune responses. Repressive histone markers such as H3K9me3 have been associated with HERV silencing.59,60 These repressive histone markers are mainly mediated by histone methyltransferases (SUV39h1, SUV39h2, SETDB1), and knock-down of histone methyltransferase SETDB1 has been associated with increased HERV expression in embryonic stem cells.61–63 As such, a retroviral silencing complex (RSC) comprising SETDB1 (histone methyltransferases), HUSH complex, tripartite motif-containing protein 28 (TRIM28), and others has been established to regulate the expression of transposable elements and HERVs.64 Of interest, H3K9me3 and SETDB1 have both been demonstrated to play a critical role in gliomagenesis, especially in gliomas with a hypermethylator CIMP (CpG island methylator) phenotype, and TRIM28 has also been shown to increase the proliferation of glioma cells.65–67
Given these findings, there has been considerable interest in capitalizing on HERV expression by reversing epigenetic changes with the fundamental premise that transient HERV expression may induce an innate antiviral immune response (i.e. viral mimicry). Since most of the epigenetic silencing hinges on DNA/histone hypermethylation or deacetylation, targeted pharmaceutical blockade of these enzymes has been proposed. For example, recent evidence has demonstrated that DNA methyltransferase inhibitors (DNMTi) could induce pro-apoptotic interferon responses that are mediated by increased HERV expression.68,69 Specifically, DNMTi therapy elicited dsRNA from HERVs that are sensed by TLR3 and MDA5, which precipitate a robust interferon cytokine response that ultimately suppresses proliferation and induces apoptosis (Figure 3). Similarly, it was demonstrated that DNMTi increased transposable element expression and HERV-derived peptides in established glioblastoma cell lines, suggesting a potential synergistic role of DNMTi-induced overexpression of HERVs and immunotherapy.70 Early phase 1 studies using DNMTi (5-Azacitidine) monotherapy in recurrent high-grade IDHm gliomas achieved disease stabilization in approximately 40% of patients but failed to achieve a durable radiographic response (PFS and OS of 4.7 and 25 months, respectively).71 Of note, the study was relatively limited by the small sample size (n = 12), histological heterogeneity and concomitant use of bevacizumab. Similar to DNMTi, activation of a viral-mimicry cascade can also be elicited through histone deacetylase inhibitors (HDACi) which relieve repressive histone marks. Currently, early-phase clinical trials using HDACi (vorinostat) in high-grade gliomas have demonstrated safety/tolerability in treating recurrent glioblastoma, albeit with some dose-limiting toxicity.72–74 However, HDACi and immunotherapy have not been combined in a clinical setting to treat high-grade gliomas, and the clinical efficacy of HDACi in improving overall survival/progression-free survival has yet to be demonstrated. The clinical role of epigenetic therapy in actuating HERVs in gliomas requires further exploration, which optimally would include a “window of opportunity” study to confirm that HDACi therapy alters the epigenetic profile of the tumor and increase HERV expression.
Fig. 3.
Viral mimicry cascade. Epigenetic therapies may de-repress HERV expression, leading to formation and release of nuclear dsRNA. dsRNA binds MDA5 which signals MAVS (mitochondrial antiviral signaling protein) to activate IRF and NF- kB. The end-result of this cascade is the induction of a pro-inflammatory immune response mediated by interferon (IFN). Anti-HERV antibodies are also produced by activation of TLR by dsRNA.
The Dichotomous Role of HERVs in Oncogenesis and Immunotherapy
The divergent roles of HERVs as either oncogenic drivers or arbitrators of the immune system have yet to be defined in gliomas. Most data suggest that specific subsets of HERVs may contribute to oncogenesis through envelope proteins (most commonly HERV-K, hml-2), insertional mutagenesis, and nonhomologous recombination.5,29,35 In gliomas particularly, increases in HERV expression were concomitantly found after induction of IDH mutations, a precursor of gliomagenesis; however, these associations have not established causality.49 Therefore, HERV’s direct contribution to oncogenic transformation in gliomas has yet to be established. On the other hand, the reactivation of HERVs through viral-mimicry pathways in gliomas has been demonstrated in several studies through epigenetic reprogramming.70 Exposure to demethylating agents or histone modifiers induces total HERV expression and secondary endogenous antiviral immune responses in gliomas through secondary dsRNA-mediated activation interferon pathways. However, selectively inducing viral-mimicry responses in gliomas with epigenetic drugs may be complicated since aberrant reactivation of HERVs in normal cells may prompt de novo mutations/transformations and unregulated autoimmune responses. Therefore, there must be a delicate balance when considering activating HERVs for immunomodulation to circumvent systemic toxicity. Ultimately, reactivation of HERVs through a viral-mimicry state in glioma remains an appealing concept for the future of glioma immunotherapy.
Limitations of Classifying HERVs in Gliomas
As mentioned previously, identifying and quantifying HERV expression in human brain tumors remains challenging. Primarily, restricting analysis to the tumor component remains a principal issue given the intrinsic heterogeneity of glioblastoma and a tumor microenvironment containing intervening normal brain tissue. Any computational platform for transposable elements must account for this heterogeneity and adjust for tumor sample purity. Analysis of tumors with poor tumor purity and intervening normal parenchyma/non-tumor proteins must be amended to accurately reflect the parent tumor phenotype. Additionally, epigenetic changes within glioma specimens vary widely; IDH-mutant gliomas carry a distinctive epigenetic phenotype that may significantly alter HERV expression, so separate analyses of each tumor subset may be necessary. Single-cell analysis may be necessary to identify heterogeneity within the tumor and to identify any somatic mutations and novel insertions. As mentioned previously, HERV expression is tightly regulated by epigenetic silencing mechanisms including TRIM28, HMTs, HDAC, and HUSH complexes; hence, analysis of tumor tissues should consider the tumor’s distinct epigenetic phenotype that may regulate HERV expression. Tumors with pervasive histone/DNA modifications could theoretically repress HERV expression and should be accounted for when comparing results among multiple patients. Lastly, typical RNA-seq platforms and datasets are not optimized to accurately detect HERVs and their associated polymorphisms. Large-scale bioinformatic studies on HERV expression in known databases such as TCGA or CCGA have insufficient reading depth (<100 bp, <50 million reads) or inadequate preparation (non-rRNA depleted) to accurately detect HERV sequences; therefore, false-negative rates may be relatively high. To date, no comprehensive database of HERV expression exists for gliomas due to the aforementioned reasons. Only a handful of established total RNA-seq datasets are appropriately prepared to quantify HERV expression, such as the encyclopedia of DNA elements (ENCODE) for certain tumor cell lines; however, the applicability of cell-line specific analyses has yet to be demonstrated for primary tumor tissues.75,76 Additionally, users must carefully select computational tools to adequately estimate the retrotranscriptome of these RNA-seq data sets; several software algorithms have been proposed to adequately quantify retrotransposable elements including TE-transcripts, Telescope, Salmon-TE, etc.77–79 As such, focused bioinformatic analyses of glioma specimens may be required to better understand the role of HERVs in gliomas.
Conclusion
Research data suggest several mechanisms by which HERVs could play a role in glioma pathogenesis. HERVs may facilitate tumor formation through direct protein formation, its splice products or indirectly dysregulating proto-oncogene/tumor suppressor expression. In addition to maintaining stemness of pluripotent cells, HERVs may also act as biomarkers of glioma progression or acquisition of mutations (IDH1 mutation). Although there is decent data to implicate the role of HERVs in glioma pathogenesis, in-depth -omics on the genome, epigenome, and transcriptome may help further elucidate their function. Aside from their role in oncogenic transformation, HERVs can also elicit robust anti-tumor immune responses in gliomas through a viral-mimicry cascade which may synergize with immunotherapy. New strategies to capitalize on HERV-induced anti-tumor immune responses in glioma should be considered therapeutic options.
Funding
This work was supported by National Institute of Neurological Disorders and Stroke (R25NS108937-01).
Conflict of interest statement. The authors have no financial or personal conflicts of interest.
Authorship statement. A.H.S. contributed to design and preparation of manuscript. M.G., A.N., and J.H. critically reviewed paper and provided senior mentorship.
References
- 1.Martin MA, Bryan T, Rasheed S, Khan AS. Identification and cloning of endogenous retroviral sequences present in human DNA. Proc Natl Acad Sci U S A. 1981;78(8):4892–4896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Griffiths DJ. Endogenous retroviruses in the human genome sequence. Genome Biol. 2001;2(6):REVIEWS1017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Weiss RA. The discovery of endogenous retroviruses. Retrovirology. 2006;3:67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Balestrieri E, Pica F, Matteucci C, et al. Transcriptional activity of human endogenous retroviruses in human peripheral blood mononuclear cells. Biomed Res Int. 2015;2015:164529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Bannert N, Hofmann H, Block A, Hohn O. HERVs new role in cancer: from accused perpetrators to cheerful protectors. Front Microbiol. 2018;9:178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Bannert N, Kurth R. Retroelements and the human genome: new perspectives on an old relation. Proc Natl Acad Sci U S A. 2004;101(Supplement 2):14572–14579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Wildschutte JH, Williams ZH, Montesion M, Subramanian RP, Kidd JM, Coffin JM. Discovery of unfixed endogenous retrovirus insertions in diverse human populations. Proc Natl Acad Sci U S A. 2016;113(16):E2326–E2334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Wang T, Medynets M, Johnson KR, et al. Regulation of stem cell function and neuronal differentiation by HERV-K via mTOR pathway. Proc Natl Acad Sci U S A. 2020;117(30):17842–17853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Voisset C, Weiss RA, Griffiths DJ. Human RNA “rumor” viruses: the search for novel human retroviruses in chronic disease. Microbiol Mol Biol Rev. 2008;72(1):157–96, table of contents. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Cegolon L, Salata C, Weiderpass E, Vineis P, Palù G, Mastrangelo G. Human endogenous retroviruses and cancer prevention: evidence and prospects. BMC Cancer. 2013;13:4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Humer J, Waltenberger A, Grassauer A, et al. Identification of a melanoma marker derived from melanoma-associated endogenous retroviruses. Cancer Res. 2006;66(3):1658–1663. [DOI] [PubMed] [Google Scholar]
- 12.Wang-Johanning F, Frost AR, Johanning GL, et al. Expression of human endogenous retrovirus k envelope transcripts in human breast cancer. Clin Cancer Res. 2001;7(6):1553–1560. [PubMed] [Google Scholar]
- 13.Wang Z, Hao Y, Zhang C, et al. The Landscape of Viral Expression Reveals Clinically Relevant Viruses with Potential Capability of Promoting Malignancy in Lower-Grade Glioma. Clin Cancer Res. 2017;23(9):2177–2185. [DOI] [PubMed] [Google Scholar]
- 14.Yuan Z, Zhang N, An Z, Zheng W. Abstract B37: Analysis of the differential expression of human endogenous retrovirus in glioblastoma multiforme. Cancer Research. 2020;80(8 Supplement):B37–B37. [Google Scholar]
- 15.Chumakov IM, Zabarovsky ER, Prassolov VS, Mett VL, Kisselev LL. Human nucleotide sequences related to the transforming gene of a murine sarcoma virus: studies with cloned viral and cellular DNAs. Gene. 1982;17(1):19–26. [DOI] [PubMed] [Google Scholar]
- 16.Zhang M, Liang JQ, Zheng S. Expressional activation and functional roles of human endogenous retroviruses in cancers. Rev Med Virol. 2019;29(2):e2025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Gasent Blesa J, Candel V. Cell-cell fusion as a potential target in cancer therapy. Ecancermedicalscience. 2009;3:145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Kessler AF, Wiesner M, Denner J, et al. Expression-analysis of the human endogenous retrovirus HERV-K in human astrocytic tumors. BMC Res Notes. 2014;7:159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Choi WI, Kim MY, Jeon BN, et al. Role of promyelocytic leukemia zinc finger (PLZF) in cell proliferation and cyclin-dependent kinase inhibitor 1A (p21WAF/CDKN1A) gene repression. J Biol Chem. 2014;289(27):18625–18640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Downey RF, Sullivan FJ, Wang-Johanning F, Ambs S, Giles FJ, Glynn SA. Human endogenous retrovirus K and cancer: Innocent bystander or tumorigenic accomplice? Int J Cancer. 2015;137(6):1249–1257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Armbruester V, Sauter M, Roemer K, et al. Np9 protein of human endogenous retrovirus K interacts with ligand of numb protein X. J Virol. 2004;78(19):10310–10319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Chen J, Xu J, Zhao W, et al. Characterization of human LNX, a novel ligand of Numb protein X that is downregulated in human gliomas. Int J Biochem Cell Biol. 2005;37(11):2273–2283. [DOI] [PubMed] [Google Scholar]
- 23.Kudo-Saito C, Yura M, Yamamoto R, Kawakami Y. Induction of immunoregulatory CD271+ cells by metastatic tumor cells that express human endogenous retrovirus H. Cancer Res. 2014;74(5):1361–1370. [DOI] [PubMed] [Google Scholar]
- 24.Ahn BY, Saldanha-Gama RF, Rahn JJ, et al. Glioma invasion mediated by the p75 neurotrophin receptor (p75(NTR)/CD271) requires regulated interaction with PDLIM1. Oncogene. 2016;35(11):1411–1422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Alshehri MM, Robbins SM, Senger DL. The role of neurotrophin signaling in gliomagenesis: a focus on the p75 neurotrophin receptor (p75NTR/CD271). Vitam Horm. 2017;104:367–404. [DOI] [PubMed] [Google Scholar]
- 26.Liang L, Coudière-Morrison L, Tatari N, et al. CD271+ cells are diagnostic and prognostic and exhibit elevated MAPK activity in SHH medulloblastoma. Cancer Res. 2018;78(16):4745–4759. [DOI] [PubMed] [Google Scholar]
- 27.Johnston AL, Lun X, Rahn JJ, et al. The p75 neurotrophin receptor is a central regulator of glioma invasion. PLoS Biol. 2007;5(8):e212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Misra A, Chosdol K, Sarkar C, Mahapatra AK, Sinha S. Alteration of a sequence with homology to human endogenous retrovirus (HERV-K) in primary human glioma: implications for viral repeat mediated rearrangement. Mutat Res. 2001;484(1-2):53–59. [DOI] [PubMed] [Google Scholar]
- 29.Yu HL, Zhao ZK, Zhu F. The role of human endogenous retroviral long terminal repeat sequences in human cancer (Review). Int J Mol Med. 2013;32(4):755–762. [DOI] [PubMed] [Google Scholar]
- 30.Gleize V, Alentorn A, Connen de Kérillis L, et al. ; POLA network . CIC inactivating mutations identify aggressive subset of 1p19q codeleted gliomas. Ann Neurol. 2015;78(3):355–374. [DOI] [PubMed] [Google Scholar]
- 31.Padul V, Epari S, Moiyadi A, Shetty P, Shirsat NV. ETV/Pea3 family transcription factor-encoding genes are overexpressed in CIC-mutant oligodendrogliomas. Genes Chromosomes Cancer. 2015;54(12):725–733. [DOI] [PubMed] [Google Scholar]
- 32.Tomlins SA, Laxman B, Dhanasekaran SM, et al. Distinct classes of chromosomal rearrangements create oncogenic ETS gene fusions in prostate cancer. Nature. 2007;448(7153):595–599. [DOI] [PubMed] [Google Scholar]
- 33.Vogt J, Wernstedt A, Ripperger T, et al. PMS2 inactivation by a complex rearrangement involving an HERV retroelement and the inverted 100-kb duplicon on 7p22.1. Eur J Hum Genet. 2016;24(11):1598–1604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Ramchander NC, Ryan NA, Crosbie EJ, Evans DG. Homozygous germ-line mutation of the PMS2 mismatch repair gene: a unique case report of constitutional mismatch repair deficiency (CMMRD). BMC Med Genet. 2017;18(1):40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Romanish MT, Cohen CJ, Mager DL. Potential mechanisms of endogenous retroviral-mediated genomic instability in human cancer. Semin Cancer Biol. 2010;20(4):246–253. [DOI] [PubMed] [Google Scholar]
- 36.Kassiotis G, Stoye JP. Making a virtue of necessity: the pleiotropic role of human endogenous retroviruses in cancer. Philos Trans R Soc Lond B Biol Sci. 2017;372(1732):20160277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Lamprecht B, Walter K, Kreher S, et al. Derepression of an endogenous long terminal repeat activates the CSF1R proto-oncogene in human lymphoma. Nat Med. 2010;16(5):571–9, 1p following 579. [DOI] [PubMed] [Google Scholar]
- 38.Forget A, Martignetti L, Puget S, et al. Aberrant ERBB4-SRC signaling as a hallmark of group 4 medulloblastoma revealed by integrative phosphoproteomic profiling. Cancer Cell. 2018;34(3):379–395.e7. [DOI] [PubMed] [Google Scholar]
- 39.Sun L, Liang H, Yu W, Jin X. Increased invasive phenotype of CSF-1R expression in glioma cells via the ERK1/2 signaling pathway. Cancer Gene Ther. 2019;26(5-6):136–144. [DOI] [PubMed] [Google Scholar]
- 40.Scarfò I, Pellegrino E, Mereu E, et al. ; European T-Cell Lymphoma Study Group . Identification of a new subclass of ALK-negative ALCL expressing aberrant levels of ERBB4 transcripts. Blood. 2016;127(2):221–232. [DOI] [PubMed] [Google Scholar]
- 41.Pyonteck SM, Akkari L, Schuhmacher AJ, et al. CSF-1R inhibition alters macrophage polarization and blocks glioma progression. Nat Med. 2013;19(10):1264–1272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Berezowska S, Schlegel J. Targeting ErbB receptors in high-grade glioma. Curr Pharm Des. 2011;17(23):2468–2487. [DOI] [PubMed] [Google Scholar]
- 43.Argaw-Denboba A, Balestrieri E, Serafino A, et al. HERV-K activation is strictly required to sustain CD133+ melanoma cells with stemness features. J Exp Clin Cancer Res. 2017;36(1):20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Nawashiro H, Otani N, Shinomiya N, et al. The role of CD98 in astrocytic neoplasms. Hum Cell. 2002;15(1):25–31. [DOI] [PubMed] [Google Scholar]
- 45.Grow EJ, Flynn RA, Chavez SL, et al. Intrinsic retroviral reactivation in human preimplantation embryos and pluripotent cells. Nature. 2015;522(7555):221–225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Guo L, Gu F, Xu Y, Zhou C. Increased copy number of syncytin-1 in the trophectoderm is associated with implantation of the blastocyst. PeerJ. 2020;8:e10368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Wang-Johanning F, Liu J, Rycaj K, et al. Expression of multiple human endogenous retrovirus surface envelope proteins in ovarian cancer. Int J Cancer. 2007;120(1):81–90. [DOI] [PubMed] [Google Scholar]
- 48.Wang-Johanning F, Radvanyi L, Rycaj K, et al. Human endogenous retrovirus K triggers an antigen-specific immune response in breast cancer patients. Cancer Res. 2008;68(14):5869–5877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Turcan S, Makarov V, Taranda J, et al. Mutant-IDH1-dependent chromatin state reprogramming, reversibility, and persistence. Nat Genet. 2018;50(1):62–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Assinger A, Yaiw KC, Göttesdorfer I, Leib-Mösch C, Söderberg-Nauclér C. Human cytomegalovirus (HCMV) induces human endogenous retrovirus (HERV) transcription. Retrovirology. 2013;10:132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Contreras-Galindo R, López P, Vélez R, Yamamura Y. HIV-1 infection increases the expression of human endogenous retroviruses type K (HERV-K) in vitro. AIDS Res Hum Retroviruses. 2007;23(1):116–122. [DOI] [PubMed] [Google Scholar]
- 52.Grandi N, Tramontano E. HERV envelope proteins: physiological role and pathogenic potential in cancer and autoimmunity. Front Microbiol. 2018;9:462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Sauter M, Schommer S, Kremmer E, et al. Human endogenous retrovirus K10: expression of Gag protein and detection of antibodies in patients with seminomas. J Virol. 1995;69(1):414–421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Sauter M, Roemer K, Best B, et al. Specificity of antibodies directed against Env protein of human endogenous retroviruses in patients with germ cell tumors. Cancer Res. 1996;56(19):4362–4365. [PubMed] [Google Scholar]
- 55.Hahn S, Ugurel S, Hanschmann KM, et al. Serological response to human endogenous retrovirus K in melanoma patients correlates with survival probability. AIDS Res Hum Retroviruses. 2008;24(5):717–723. [DOI] [PubMed] [Google Scholar]
- 56.Wang-Johanning F, Rycaj K, Plummer JB, et al. Immunotherapeutic potential of anti-human endogenous retrovirus-K envelope protein antibodies in targeting breast tumors. J Natl Cancer Inst. 2012;104(3):189–210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Kraus B, Fischer K, Sliva K, Schnierle BS. Vaccination directed against the human endogenous retrovirus-K (HERV-K) gag protein slows HERV-K gag expressing cell growth in a murine model system. Virol J. 2014;11:58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Sacha JB, Kim IJ, Chen L, et al. Vaccination with cancer- and HIV infection-associated endogenous retrotransposable elements is safe and immunogenic. J Immunol. 2012;189(3):1467–1479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Maksakova IA, Mager DL, Reiss D. Keeping active endogenous retroviral-like elements in check: the epigenetic perspective. Cell Mol Life Sci. 2008;65(21):3329–3347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Leung DC, Dong KB, Maksakova IA, et al. Lysine methyltransferase G9a is required for de novo DNA methylation and the establishment, but not the maintenance, of proviral silencing. Proc Natl Acad Sci U S A. 2011;108(14):5718–5723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Bulut-Karslioglu A, De La Rosa-Velázquez IA, Ramirez F, et al. Suv39h-dependent H3K9me3 marks intact retrotransposons and silences LINE elements in mouse embryonic stem cells. Mol Cell. 2014;55(2):277–290. [DOI] [PubMed] [Google Scholar]
- 62.Karimi MM, Goyal P, Maksakova IA, et al. DNA methylation and SETDB1/H3K9me3 regulate predominantly distinct sets of genes, retroelements, and chimeric transcripts in mESCs. Cell Stem Cell. 2011;8(6):676–687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Kato M, Takemoto K, Shinkai Y. A somatic role for the histone methyltransferase Setdb1 in endogenous retrovirus silencing. Nat Commun. 2018;9(1):1683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Geis FK, Goff SP. Silencing and transcriptional regulation of endogenous retroviruses: an overview. Viruses. 2020;12(8):884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Sepsa A, Levidou G, Gargalionis A, et al. Emerging role of linker histone variant H1x as a biomarker with prognostic value in astrocytic gliomas. A multivariate analysis including trimethylation of H3K9 and H4K20. PLoS One. 2015;10(1):e0115101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Spyropoulou A, Gargalionis A, Dalagiorgou G, et al. Role of histone lysine methyltransferases SUV39H1 and SETDB1 in gliomagenesis: modulation of cell proliferation, migration, and colony formation. Neuromolecular Med. 2014;16(1):70–82. [DOI] [PubMed] [Google Scholar]
- 67.Peng Y, Zhang M, Jiang Z, Jiang Y. TRIM28 activates autophagy and promotes cell proliferation in glioblastoma. Onco Targets Ther. 2019;12:397–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Chiappinelli KB, Strissel PL, Desrichard A, et al. Inhibiting DNA methylation causes an interferon response in cancer via dsRNA including endogenous retroviruses. Cell. 2015;162(5):974–986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Roulois D, Loo Yau H, Singhania R, et al. DNA-demethylating agents target colorectal cancer cells by inducing viral mimicry by endogenous transcripts. Cell. 2015;162(5):961–973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Kong Y, Rose CM, Cass AA, et al. Transposable element expression in tumors is associated with immune infiltration and increased antigenicity. Nat Commun. 2019;10(1):5228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Federici L, Capelle L, Annereau M, et al. 5-Azacitidine in patients with IDH1/2-mutant recurrent glioma. Neuro Oncol. 2020;22(8):1226–1228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Lee EQ, Puduvalli VK, Reid JM, et al. Phase I study of vorinostat in combination with temozolomide in patients with high-grade gliomas: North American Brain Tumor Consortium Study 04-03. Clin Cancer Res. 2012;18(21):6032–6039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Friday BB, Anderson SK, Buckner J, et al. Phase II trial of vorinostat in combination with bortezomib in recurrent glioblastoma: a north central cancer treatment group study. Neuro Oncol. 2012;14(2):215–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Peters KB, Lipp ES, Miller E, et al. Phase I/II trial of vorinostat, bevacizumab, and daily temozolomide for recurrent malignant gliomas. J Neurooncol. 2018;137(2):349–356. [DOI] [PubMed] [Google Scholar]
- 75.Davis CA, Hitz BC, Sloan CA, et al. The Encyclopedia of DNA elements (ENCODE): data portal update. Nucleic Acids Res. 2018;46(D1):D794–D801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Consortium EP. An integrated encyclopedia of DNA elements in the human genome. Nature. 2012;489(7414):57–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Bendall ML, de Mulder M, Iñiguez LP, et al. Telescope: Characterization of the retrotranscriptome by accurate estimation of transposable element expression. PLoS Comput Biol. 2019;15(9):e1006453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Jin Y, Tam OH, Paniagua E, Hammell M. TEtranscripts: a package for including transposable elements in differential expression analysis of RNA-seq datasets. Bioinformatics. 2015;31(22):3593–3599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Jeong HH, Yalamanchili HK, Guo C, Shulman JM, Liu Z. An ultra-fast and scalable quantification pipeline for transposable elements from next generation sequencing data. Pac Symp Biocomput. 2018;23:168–179. [PubMed] [Google Scholar]