Abstract
Transposable elements (TEs) are mobile repetitive nucleic acid sequences that have been incorporated into the genome through spontaneous integration, accounting for almost 50% of human DNA. Even though most TEs are no longer mobile today, studies have demonstrated that they have important roles in different biological processes, such as ageing, embryonic development, and cancer. TEs influence these processes through various mechanisms, including active transposition of TEs contributing to ongoing evolution, transposon transcription generating RNA or protein, and by influencing gene regulation as enhancers. However, how TEs interact with the immune system remains a largely unexplored field. In this Perspective, we describe how TEs might influence different aspects of the immune system, such as innate immune responses, T cell activation and differentiation, and tissue adaptation. Furthermore, TEs can serve as a source of neoantigens for T cells in antitumour immunity. We suggest that TE biology is an important emerging field of immunology and discuss the potential to harness the TE network therapeutically, for example, to improve immunotherapies for cancer and autoimmune and inflammatory diseases.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$259.00 per year
only $21.58 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout


Similar content being viewed by others
References
Feschotte, C. Transposable elements: McClintock’s legacy revisited. Nat. Rev. Genet. 24, 797–800 (2023).
Bruno, M., Mahgoub, M. & Macfarlan, T. S. The arms race between KRAB-zinc finger proteins and endogenous retroelements and its impact on mammals. Annu. Rev. Genet. 53, 393–416 (2019).
Wells, J. N. & Feschotte, C. A field guide to eukaryotic transposable elements. Annu. Rev. Genet. 54, 539–561 (2020).
Kojima, S. et al. Mobile element variation contributes to population-specific genome diversification, gene regulation and disease risk. Nat. Genet. 55, 939–951 (2023).
Ostertag, E. M. & Kazazian, H. H. Jr Biology of mammalian L1 retrotransposons. Annu. Rev. Genet. 35, 501–538 (2001).
Szak, S. T. et al. Molecular archeology of L1 insertions in the human genome. Genome Biol. 3, research0052 (2002).
Loreto, E. L. S. & Pereira, C. M. Somatizing the transposons action. Mob. Genet. Elem. 7, 1–9 (2017).
Haig, D. Transposable elements: self-seekers of the germline, team-players of the soma. Bioessays 38, 1158–1166 (2016).
Gusa, A. et al. Genome-wide analysis of heat stress-stimulated transposon mobility in the human fungal pathogen Cryptococcus deneoformans. Proc. Natl Acad. Sci. USA 120, e2209831120 (2023).
Hirochika, H. Activation of tobacco retrotransposons during tissue culture. EMBO J. 12, 2521–2528 (1993).
Hirochika, H., Sugimoto, K., Otsuki, Y., Tsugawa, H. & Kanda, M. Retrotransposons of rice involved in mutations induced by tissue culture. Proc. Natl Acad. Sci. USA 93, 7783–7788 (1996).
Paquin, C. E. & Williamson, V. M. Temperature effects on the rate of ty transposition. Science 226, 53–55 (1984).
Gusa, A. et al. Transposon mobilization in the human fungal pathogen Cryptococcus is mutagenic during infection and promotes drug resistance in vitro. Proc. Natl Acad. Sci. USA 117, 9973–9980 (2020).
Sehgal, A., Lee, C. Y. & Espenshade, P. J. SREBP controls oxygen-dependent mobilization of retrotransposons in fission yeast. PLoS Genet. 3, e131 (2007).
Gusa, A. & Jinks-Robertson, S. Mitotic recombination and adaptive genomic changes in human pathogenic fungi. Genes 10, 901 (2019).
Brouha, B. et al. Hot L1s account for the bulk of retrotransposition in the human population. Proc. Natl Acad. Sci. USA 100, 5280–5285 (2003).
Zhang, T. et al. Heterologous survey of 130 DNA transposons in human cells highlights their functional divergence and expands the genome engineering toolbox. Cell 187, 3741–3760.e3730 (2024).
Thawani, A., Ariza, A. J. F., Nogales, E. & Collins, K. Template and target-site recognition by human LINE-1 in retrotransposition. Nature 626, 186–193 (2024).
Baldwin, E. T. et al. Structures, functions and adaptations of the human LINE-1 ORF2 protein. Nature 626, 194–206 (2024).
Ewing, A. D. & Kazazian, H. H. Jr. High-throughput sequencing reveals extensive variation in human-specific L1 content in individual human genomes. Genome Res. 20, 1262–1270 (2010).
Sekine, K., Onoguchi, M. & Hamada, M. Transposons contribute to the acquisition of cell type-specific cis-elements in the brain. Commun. Biol. 6, 631 (2023).
Muotri, A. R. et al. Somatic mosaicism in neuronal precursor cells mediated by L1 retrotransposition. Nature 435, 903–910 (2005).
Coufal, N. G. et al. L1 retrotransposition in human neural progenitor cells. Nature 460, 1127–1131 (2009).
Muotri, A. R., Zhao, C., Marchetto, M. C. & Gage, F. H. Environmental influence on L1 retrotransposons in the adult hippocampus. Hippocampus 19, 1002–1007 (2009).
Muotri, A. R. & Gage, F. H. Generation of neuronal variability and complexity. Nature 441, 1087–1093 (2006).
Maggioni, E. et al. Investigating the impact of L1 retrotransposons on behavior: a pilot study on young twins. Schizophr. Res. 271, 353–354 (2024).
van Dongen, J. et al. Identical twins carry a persistent epigenetic signature of early genome programming. Nat. Commun. 12, 5618 (2021).
Muotri, A. R. et al. L1 retrotransposition in neurons is modulated by MeCP2. Nature 468, 443–446 (2010).
Takahashi, T. et al. LINE-1 activation in the cerebellum drives ataxia. Neuron 110, 3278–3287.e3278 (2022).
Shpyleva, S., Melnyk, S., Pavliv, O., Pogribny, I. & Jill James, S. Overexpression of LINE-1 retrotransposons in autism brain. Mol. Neurobiol. 55, 1740–1749 (2018).
Guo, C. et al. Tau activates transposable elements in Alzheimer’s disease. Cell Rep. 23, 2874–2880 (2018).
Bundo, M. et al. Increased l1 retrotransposition in the neuronal genome in schizophrenia. Neuron 81, 306–313 (2014).
Cajuso, T. et al. Retrotransposon insertions can initiate colorectal cancer and are associated with poor survival. Nat. Commun. 10, 4022 (2019).
Helman, E. et al. Somatic retrotransposition in human cancer revealed by whole-genome and exome sequencing. Genome Res. 24, 1053–1063 (2014).
Tubio, J. M. C. et al. Mobile DNA in cancer. Extensive transduction of nonrepetitive DNA mediated by L1 retrotransposition in cancer genomes. Science 345, 1251343 (2014).
Rodic, N. et al. Retrotransposon insertions in the clonal evolution of pancreatic ductal adenocarcinoma. Nat. Med. 21, 1060–1064 (2015).
Doucet-O’Hare, T. T. et al. Somatically acquired LINE-1 insertions in normal esophagus undergo clonal expansion in esophageal squamous cell carcinoma. Hum. Mutat. 37, 942–954 (2016).
Lee, E. et al. Landscape of somatic retrotransposition in human cancers. Science 337, 967–971 (2012).
Gozuacik, D. & Kimchi, A. DAPk protein family and cancer. Autophagy 2, 74–79 (2006).
Michie, A. M., McCaig, A. M., Nakagawa, R. & Vukovic, M. Death-associated protein kinase (DAPK) and signal transduction: regulation in cancer. FEBS J. 277, 74–80 (2010).
Lu, Y., Wajapeyee, N., Turker, M. S. & Glazer, P. M. Silencing of the DNA mismatch repair gene MLH1 induced by hypoxic stress in a pathway dependent on the histone demethylase LSD1. Cell Rep. 8, 501–513 (2014).
Machado, H. E. et al. Diverse mutational landscapes in human lymphocytes. Nature 608, 724–732 (2022).
Jaiswal, S. & Ebert, B. L. Clonal hematopoiesis in human aging and disease. Science 366, eaan4673 (2019).
Gorbunova, V. et al. The role of retrotransposable elements in ageing and age-associated diseases. Nature 596, 43–53 (2021).
De Cecco, M. et al. L1 drives IFN in senescent cells and promotes age-associated inflammation. Nature 566, 73–78 (2019).
Zhu, X., Fang, H., Gladysz, K., Barbour, J. A. & Wong, J. W. H. Overexpression of transposable elements is associated with immune evasion and poor outcome in colorectal cancer. Eur. J. Cancer 157, 94–107 (2021).
Szpakowski, S. et al. Loss of epigenetic silencing in tumors preferentially affects primate-specific retroelements. Gene 448, 151–167 (2009).
Kong, Y. et al. Transposable element expression in tumors is associated with immune infiltration and increased antigenicity. Nat. Commun. 10, 5228 (2019).
Li, W. et al. Human endogenous retrovirus-K contributes to motor neuron disease. Sci. Transl. Med. 7, 307ra153 (2015).
Hornung, V. et al. 5′-Triphosphate RNA is the ligand for RIG-I. Science 314, 994–997 (2006).
Li, X. et al. Structural basis of double-stranded RNA recognition by the RIG-I like receptor MDA5. Arch. Biochem. Biophys. 488, 23–33 (2009).
Chen, Q., Sun, L. & Chen, Z. J. Regulation and function of the cGAS–STING pathway of cytosolic DNA sensing. Nat. Immunol. 17, 1142–1149 (2016).
Simon, M. et al. LINE1 derepression in aged wild-type and SIRT6-deficient mice drives inflammation. Cell Metab. 29, 871–885 e875 (2019).
Bonte, P. E. et al. Selective control of transposable element expression during T cell exhaustion and anti-PD-1 treatment. Sci. Immunol. 8, eadf8838 (2023).
Larouche, J. D. et al. Transposable elements regulate thymus development and function. eLife 12, e91037 (2024).
Marasca, F. et al. LINE1 are spliced in non-canonical transcript variants to regulate T cell quiescence and exhaustion. Nat. Genet. 54, 180–193 (2022).
Young, G. R., Mavrommatis, B. & Kassiotis, G. Microarray analysis reveals global modulation of endogenous retroelement transcription by microbes. Retrovirology 11, 59 (2014).
Macchietto, M. G., Langlois, R. A. & Shen, S. S. Virus-induced transposable element expression up-regulation in human and mouse host cells. Life Sci. Alliance 3, e201900536 (2020).
Sorek, M., Meshorer, E. & Schlesinger, S. Impaired activation of transposable elements in SARS-CoV-2 infection. EMBO Rep. 23, e55101 (2022).
Lima-Junior, D. S. et al. Endogenous retroviruses promote homeostatic and inflammatory responses to the microbiota. Cell 184, 3794–3811.e3719 (2021).
Jang, H. J. et al. Epigenetic therapy potentiates transposable element transcription to create tumor-enriched antigens in glioblastoma cells. Nat. Genet. 56, 1903–1913 (2024).
Bonaventura, P. et al. Identification of shared tumor epitopes from endogenous retroviruses inducing high-avidity cytotoxic T cells for cancer immunotherapy. Sci. Adv. 8, eabj3671 (2022).
Attig, J. et al. LTR retroelement expansion of the human cancer transcriptome and immunopeptidome revealed by de novo transcript assembly. Genome Res. 29, 1578–1590 (2019).
Cosby, R. L. et al. Recurrent evolution of vertebrate transcription factors by transposase capture. Science 371, eabc6405 (2021).
Larouche, J. D. et al. Widespread and tissue-specific expression of endogenous retroelements in human somatic tissues. Genome Med. 12, 40 (2020).
Shah, N. M. et al. Pan-cancer analysis identifies tumor-specific antigens derived from transposable elements. Nat. Genet. 55, 631–639 (2023).
Kobayashi, S. et al. Proteogenomic identification of an immunogenic antigen derived from human endogenous retrovirus in renal cell carcinoma. JCI Insight 8, e167712 (2023).
Saini, S. K. et al. Human endogenous retroviruses form a reservoir of T cell targets in hematological cancers. Nat. Commun. 11, 5660 (2020).
Laumont, C. M. et al. Noncoding regions are the main source of targetable tumor-specific antigens. Sci. Transl. Med. 10, eaau5516 (2018).
Merlotti, A. et al. Noncanonical splicing junctions between exons and transposable elements represent a source of immunogenic recurrent neo-antigens in patients with lung cancer. Sci. Immunol. 8, eabm6359 (2023).
Ng, K. W. et al. Antibodies against endogenous retroviruses promote lung cancer immunotherapy. Nature 616, 563–573 (2023).
Arribas, Y. A. et al. Transposable element exonization generates a reservoir of evolving and functional protein isoforms. Cell 187, 7603–7620.e7622 (2024).
Pasquesi, G. I. M. et al. Regulation of human interferon signaling by transposon exonization. Cell 187, 7621–7636.e7619 (2024).
Ueda, M. T. et al. Functional and dynamic profiling of transcript isoforms reveals essential roles of alternative splicing in interferon response. Cell Genom. 4, 100654 (2024).
Goyal, A. et al. DNMT and HDAC inhibition induces immunogenic neoantigens from human endogenous retroviral element-derived transcripts. Nat. Commun. 14, 6731 (2023).
Chiappinelli, K. B. et al. Inhibiting DNA methylation causes an interferon response in cancer via dsRNA including endogenous retroviruses. Cell 162, 974–986 (2015).
Roulois, D. et al. DNA-demethylating agents target colorectal cancer cells by inducing viral mimicry by endogenous transcripts. Cell 162, 961–973 (2015).
Brocks, D. et al. DNMT and HDAC inhibitors induce cryptic transcription start sites encoded in long terminal repeats. Nat. Genet. 49, 1052–1060 (2017).
Daskalakis, M. et al. Reactivation of endogenous retroviral elements via treatment with DNMT- and HDAC-inhibitors. Cell Cycle 17, 811–822 (2018).
Cordaux, R., Udit, S., Batzer, M. A. & Feschotte, C. Birth of a chimeric primate gene by capture of the transposase gene from a mobile element. Proc. Natl Acad. Sci. USA 103, 8101–8106 (2006).
Newman, J. C., Bailey, A. D., Fan, H. Y., Pavelitz, T. & Weiner, A. M. An abundant evolutionarily conserved CSB–PiggyBac fusion protein expressed in Cockayne syndrome. PLoS Genet. 4, e1000031 (2008).
Lin, R. et al. Transposase-derived transcription factors regulate light signaling in Arabidopsis. Science 318, 1302–1305 (2007).
Henaff, E. et al. Extensive amplification of the E2F transcription factor binding sites by transposons during evolution of Brassica species. Plant. J. 77, 852–862 (2014).
Sundaram, V. et al. Functional cis-regulatory modules encoded by mouse-specific endogenous retrovirus. Nat. Commun. 8, 14550 (2017).
Mukherjee, K. & Moroz, L. L. Transposon-derived transcription factors across metazoans. Front. Cell Dev. Biol. 11, 1113046 (2023).
Majumdar, S., Singh, A. & Rio, D. C. The human THAP9 gene encodes an active P-element DNA transposase. Science 339, 446–448 (2013).
Henssen, A. G. et al. Genomic DNA transposition induced by human PGBD5. eLife 4, e10565 (2015).
Agrawal, A., Eastman, Q. M. & Schatz, D. G. Transposition mediated by RAG1 and RAG2 and its implications for the evolution of the immune system. Nature 394, 744–751 (1998).
Fugmann, S. D. The origins of the Rag genes-from transposition to V(D)J recombination. Semin. Immunol. 22, 10–16 (2010).
Hiom, K., Melek, M. & Gellert, M. DNA transposition by the RAG1 and RAG2 proteins: a possible source of oncogenic translocations. Cell 94, 463–470 (1998).
Fueyo, R., Judd, J., Feschotte, C. & Wysocka, J. Roles of transposable elements in the regulation of mammalian transcription. Nat. Rev. Mol. Cell Biol. 23, 481–497 (2022).
Feschotte, C. Transposable elements and the evolution of regulatory networks. Nat. Rev. Genet. 9, 397–405 (2008).
Jacques, P. E., Jeyakani, J. & Bourque, G. The majority of primate-specific regulatory sequences are derived from transposable elements. PLoS Genet. 9, e1003504 (2013).
Brini, A. T., Lee, G. M. & Kinet, J. P. Involvement of Alu sequences in the cell-specific regulation of transcription of the gamma chain of Fc and T cell receptors. J. Biol. Chem. 268, 1355–1361 (1993).
Hambor, J. E., Mennone, J., Coon, M. E., Hanke, J. H. & Kavathas, P. Identification and characterization of an Alu-containing, T-cell-specific enhancer located in the last intron of the human CD8 alpha gene. Mol. Cell. Biol. 13, 7056–7070 (1993).
Chuong, E. B., Elde, N. C. & Feschotte, C. Regulatory evolution of innate immunity through co-option of endogenous retroviruses. Science 351, 1083–1087 (2016).
Donnard, E. et al. Comparative analysis of immune cells reveals a conserved regulatory lexicon. Cell Syst. 6, 381–394.e387 (2018).
Adoue, V. et al. The histone methyltransferase SETDB1 controls T helper cell lineage integrity by repressing endogenous retroviruses. Immunity 50, 629–644.e628 (2019).
Simon, M. et al. Single-cell chromatin accessibility and transposable element landscapes reveal shared features of tissue-residing immune cells. Immunity 57, 1975–1993.e1910 (2024).
Ye, M. et al. Specific subfamilies of transposable elements contribute to different domains of T lymphocyte enhancers. Proc. Natl Acad. Sci. USA 117, 7905–7916 (2020).
O’Shea, J. J. & Paul, W. E. Mechanisms underlying lineage commitment and plasticity of helper CD4+ T cells. Science 327, 1098–1102 (2010).
Wilson, C. B., Rowell, E. & Sekimata, M. Epigenetic control of T-helper-cell differentiation. Nat. Rev. Immunol. 9, 91–105 (2009).
Delacher, M. et al. Precursors for nonlymphoid-tissue treg cells reside in secondary lymphoid organs and are programmed by the transcription factor BATF. Immunity 52, 295–312.e211 (2020).
Delacher, M. et al. Genome-wide DNA-methylation landscape defines specialization of regulatory T cells in tissues. Nat. Immunol. 18, 1160–1172 (2017).
Delacher, M. et al. Single-cell chromatin accessibility landscape identifies tissue repair program in human regulatory T cells. Immunity 54, 702–720.e717 (2021).
DiSpirito, J. R. et al. Molecular diversification of regulatory T cells in nonlymphoid tissues. Sci. Immunol. 3, eaat5861 (2018).
Miragaia, R. J. et al. Single-cell transcriptomics of regulatory T cells reveals trajectories of tissue adaptation. Immunity 50, 493–504.e497 (2019).
Burton, O. T. et al. The tissue-resident regulatory T cell pool is shaped by transient multi-tissue migration and a conserved residency program. Immunity 57, 1586–1602.e1510 (2024).
Hayatsu, N. et al. Analyses of a mutant Foxp3 allele reveal BATF as a critical transcription factor in the differentiation and accumulation of tissue regulatory T cells. Immunity 47, 268–283.e269 (2017).
Li, C. et al. TCR transgenic mice reveal stepwise, multi-site acquisition of the distinctive fat-Treg phenotype. Cell 174, 285–299e212 (2018).
Karttunen, K. et al. Transposable elements as tissue-specific enhancers in cancers of endodermal lineage. Nat. Commun. 14, 5313 (2023).
Sundaram, V. et al. Widespread contribution of transposable elements to the innovation of gene regulatory networks. Genome Res. 24, 1963–1976 (2014).
Li, X. et al. LINE-1 transcription activates long-range gene expression. Nat. Genet. 56, 1494–1502 (2024).
Lu, J. Y. et al. Homotypic clustering of L1 and B1/Alu repeats compartmentalizes the 3D genome. Cell Res. 31, 613–630 (2021).
Attig, J. et al. Heteromeric RNP assembly at LINEs controls lineage-specific RNA processing. Cell 174, 1067–1081 e1017 (2018).
Liu, N. et al. Selective silencing of euchromatic L1s revealed by genome-wide screens for L1 regulators. Nature 553, 228–232 (2018).
Sakashita, A. et al. Transcription of MERVL retrotransposons is required for preimplantation embryo development. Nat. Genet. 55, 484–495 (2023).
Wallin, J. et al. Pax1 is expressed during development of the thymus epithelium and is required for normal T-cell maturation. Development 122, 23–30 (1996).
Yamazaki, Y. et al. PAX1 is essential for development and function of the human thymus. Sci. Immunol. 5, eaax1036 (2020).
Akhtar, M. Z., Sutherland, A. I., Huang, H., Ploeg, R. J. & Pugh, C. W. The role of hypoxia-inducible factors in organ donation and transplantation: the current perspective and future opportunities. Am. J. Transpl. 14, 1481–1487 (2014).
Smith, Z. D. et al. DNA methylation dynamics of the human preimplantation embryo. Nature 511, 611–615 (2014).
Deniz, O., Frost, J. M. & Branco, M. R. Regulation of transposable elements by DNA modifications. Nat. Rev. Genet. 20, 417–431 (2019).
Moore, L. D., Le, T. & Fan, G. DNA methylation and its basic function. Neuropsychopharmacology 38, 23–38 (2013).
Walsh, C. P., Chaillet, J. R. & Bestor, T. H. Transcription of IAP endogenous retroviruses is constrained by cytosine methylation. Nat. Genet. 20, 116–117 (1998).
Bertozzi, T. M., Elmer, J. L., Macfarlan, T. S. & Ferguson-Smith, A. C. KRAB zinc finger protein diversification drives mammalian interindividual methylation variability. Proc. Natl Acad. Sci. USA 117, 31290–31300 (2020).
Silver, M. J. et al. Independent genomewide screens identify the tumor suppressor VTRNA2-1 as a human epiallele responsive to periconceptional environment. Genome Biol. 16, 118 (2015).
Yoder, J. A., Walsh, C. P. & Bestor, T. H. Cytosine methylation and the ecology of intragenomic parasites. Trends Genet. 13, 335–340 (1997).
Barau, J. et al. The DNA methyltransferase DNMT3C protects male germ cells from transposon activity. Science 354, 909–912 (2016).
Grunstein, M. Histone acetylation in chromatin structure and transcription. Nature 389, 349–352 (1997).
Sengupta, N. & Seto, E. Regulation of histone deacetylase activities. J. Cell. Biochem. 93, 57–67 (2004).
Greger, J. G., Katz, R. A., Ishov, A. M., Maul, G. G. & Skalka, A. M. The cellular protein daxx interacts with avian sarcoma virus integrase and viral DNA to repress viral transcription. J. Virol. 79, 4610–4618 (2005).
Katz, R. A. et al. High-frequency epigenetic repression and silencing of retroviruses can be antagonized by histone deacetylase inhibitors and transcriptional activators, but uniform reactivation in cell clones is restricted by additional mechanisms. J. Virol. 81, 2592–2604 (2007).
Peek, G. W. & Tollefsbol, T. O. The transposon-driven evolutionary origin and basis of histone deacetylase functions and limitations in disease prevention. Clin. Epigenetics 2, 97–112 (2011).
Daujat, S. et al. H3K64 trimethylation marks heterochromatin and is dynamically remodeled during developmental reprogramming. Nat. Struct. Mol. Biol. 16, 777–781 (2009).
Martens, J. H. et al. The profile of repeat-associated histone lysine methylation states in the mouse epigenome. EMBO J. 24, 800–812 (2005).
Mikkelsen, T. S. et al. Genome-wide maps of chromatin state in pluripotent and lineage-committed cells. Nature 448, 553–560 (2007).
Matsui, T. et al. Proviral silencing in embryonic stem cells requires the histone methyltransferase ESET. Nature 464, 927–931 (2010).
Leung, D. C. 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. USA 108, 5718–5723 (2011).
Macfarlan, T. S. et al. Endogenous retroviruses and neighboring genes are coordinately repressed by LSD1/KDM1A. Genes Dev. 25, 594–607 (2011).
Zarnack, K. et al. Direct competition between hnRNP C and U2AF65 protects the transcriptome from the exonization of Alu elements. Cell 152, 453–466 (2013).
Ilik, I. A. et al. Autonomous transposons tune their sequences to ensure somatic suppression. Nature 626, 1116–1124 (2024).
Farmiloe, G., Lodewijk, G. A., Robben, S. F., van Bree, E. J. & Jacobs, F. M. J. Widespread correlation of KRAB zinc finger protein binding with brain-developmental gene expression patterns. Philos. Trans. R. Soc. Lond. B 375, 20190333 (2020).
Truby, N. L. et al. A zinc finger transcription factor enables social behaviors while controlling transposable elements and immune response in prefrontal cortex. Transl. Psychiatry 14, 59 (2024).
Huntley, S. et al. A comprehensive catalog of human KRAB-associated zinc finger genes: insights into the evolutionary history of a large family of transcriptional repressors. Genome Res. 16, 669–677 (2006).
Yang, P., Wang, Y. & Macfarlan, T. S. The role of KRAB-ZFPs in transposable element repression and mammalian evolution. Trends Genet. 33, 871–881 (2017).
Wolf, G. et al. KRAB-zinc finger protein gene expansion in response to active retrotransposons in the murine lineage. eLife 9, e56337 (2020).
Fabry, M. H. et al. piRNA-guided co-transcriptional silencing coopts nuclear export factors. eLife 8, e47999 (2019).
Le Thomas, A. et al. Piwi induces piRNA-guided transcriptional silencing and establishment of a repressive chromatin state. Genes Dev. 27, 390–399 (2013).
Brennecke, J. et al. Discrete small RNA-generating loci as master regulators of transposon activity in Drosophila. Cell 128, 1089–1103 (2007).
Thomas, C. A. et al. Modeling of TREX1-dependent autoimmune disease using human stem cells highlights L1 accumulation as a source of neuroinflammation. Cell Stem Cell 21, 319–331.e318 (2017).
Lynch, V. J., Leclerc, R. D., May, G. & Wagner, G. P. Transposon-mediated rewiring of gene regulatory networks contributed to the evolution of pregnancy in mammals. Nat. Genet. 43, 1154–1159 (2011).
Senft, A. D. & Macfarlan, T. S. Transposable elements shape the evolution of mammalian development. Nat. Rev. Genet. 22, 691–711 (2021).
Eckersley-Maslin, M. A., Alda-Catalinas, C. & Reik, W. Dynamics of the epigenetic landscape during the maternal-to-zygotic transition. Nat. Rev. Mol. Cell Biol. 19, 436–450 (2018).
Modzelewski, A. J. et al. A mouse-specific retrotransposon drives a conserved Cdk2ap1 isoform essential for development. Cell 184, 5541–5558.e5522 (2021).
Oomen, M. E. et al. An atlas of transcription initiation reveals regulatory principles of gene and transposable element expression in early mammalian development. Cell 188, 1156–1174.e1120 (2025).
Lavialle, C. et al. Paleovirology of ‘syncytins’, retroviral env genes exapted for a role in placentation. Philos. Trans. R. Soc. Lond. B 368, 20120507 (2013).
Blaise, S., de Parseval, N., Benit, L. & Heidmann, T. Genomewide screening for fusogenic human endogenous retrovirus envelopes identifies syncytin 2, a gene conserved on primate evolution. Proc. Natl Acad. Sci. USA 100, 13013–13018 (2003).
Blaise, S., de Parseval, N. & Heidmann, T. Functional characterization of two newly identified Human Endogenous Retrovirus coding envelope genes. Retrovirology 2, 19 (2005).
Lokossou, A. G. et al. Endogenous retrovirus-encoded Syncytin-2 contributes to exosome-mediated immunosuppression of T cells†. Biol. Reprod. 102, 185–198 (2020).
Tolosa, J. M. et al. The endogenous retroviral envelope protein syncytin-1 inhibits LPS/PHA-stimulated cytokine responses in human blood and is sorted into placental exosomes. Placenta 33, 933–941 (2012).
Hu, G. et al. A genome-wide RNAi screen identifies a new transcriptional module required for self-renewal. Genes Dev. 23, 837–848 (2009).
Cheng, B., Ren, X. & Kerppola, T. K. KAP1 represses differentiation-inducible genes in embryonic stem cells through cooperative binding with PRC1 and derepresses pluripotency-associated genes. Mol. Cell Biol. 34, 2075–2091 (2014).
Rowe, H. M. et al. KAP1 controls endogenous retroviruses in embryonic stem cells. Nature 463, 237–240 (2010).
Lu, X. et al. The retrovirus HERVH is a long noncoding RNA required for human embryonic stem cell identity. Nat. Struct. Mol. Biol. 21, 423–425 (2014).
Wang, J. et al. Primate-specific endogenous retrovirus-driven transcription defines naive-like stem cells. Nature 516, 405–409 (2014).
Anzalone, A. V., Koblan, L. W. & Liu, D. R. Genome editing with CRISPR–Cas nucleases, base editors, transposases and prime editors. Nat. Biotechnol. 38, 824–844 (2020).
Acknowledgements
This work was supported by the Deutsche Forschungsgemeinschaft (German Research Foundation) Projektnummer 324392634-TRR 221 to M.F.
Author information
Authors and Affiliations
Contributions
L.S., P.S. and M.F. conceptualized the review. L.S., P.S. and M.F. wrote the main body. All authors contributed to editing of the manuscript and providing references. All authors approved the submitted version of the article.
Corresponding author
Ethics declarations
Competing interests
The authors declare no competing interests.
Peer review
Peer review information
Nature Reviews Immunology thanks J. Mathai, D. Schatz and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Glossary
- CRISPR activation
-
(CRISPRa). A modified CRISPR–Cas9 system using a deactivated Cas9 (dCas9) fused to a transcriptional activator to enhance the expression of genes without affecting the DNA sequence.
- CRISPR–Cas9
-
A genetic engineering system that induces double-stranded breaks in a sequence-specific manner by duplexing a Cas9 nuclease with a guide RNA. Genetic alterations are induced through non-homologous end joining and homology-directed repair.
- CRISPR–Cas-derived base editors
-
A catalytically impaired CRISPR–Cas system that induces targeted point mutations without double-stranded DNA breaks.
- CRISPR interference
-
(CRISPRi). A modified CRISPR–Cas9 system using a deactivated Cas9 (dCas9) fused to a repressor construct that can be used to silence the transcription of genes without altering the DNA sequence.
- Domestication
-
An evolutionary process during which foreign material is integrated into the genome to fulfil a function that is beneficial to the host organism, for instance, the integration of transposons into the genome to form the RAG genes.
- Endogenous retroviruses
-
(ERVs). Retrotransposons of the long-terminal repeat (LTR) subclass derived from retroviruses.
- Host–transposase fusion (HTF) genes or proteins
-
Genes or proteins that are comprised partially of host and partially of transposon material. Many transcription factors are HTF genes, combining transposable element-derived DNA-binding domains with host regulatory domains.
- JETs
-
Non-canonical splice junctions involving exons and transposable elements (junctions between exons and transposable elements).
- Lancelets
-
A group of primitive chordates; a frequently used animal model system used to study evolution.
- LINE-1
-
Long interspersed nuclear element-1 also known as L1, both a retrotransposon and a group of retrotransposons of the non-long-terminal repeat (LTR) family and LINE subfamily.
- Metastable epialleles
-
Loci that show systemic variable DNA methylation among individuals without underlying genetic differences.
- MHC class I-associated peptides
-
(MAPs). Peptides that can be presented by MHC class I molecules and recognized by CD8 T cells.
- Micro-evolution
-
Small-scale evolutionary changes that occur over a relatively short period of time. An example of micro-evolution is the development of antibiotic resistance in bacteria.
- TE exonization
-
The process of incorporating transposable elements (TEs) from non-coding regions, predominantly introns, into new exons, using splicing signal sequences.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Schmidleithner, L., Stüve, P. & Feuerer, M. Transposable elements as instructors of the immune system. Nat Rev Immunol 25, 696–706 (2025). https://doi.org/10.1038/s41577-025-01172-3
Accepted:
Published:
Issue date:
DOI: https://doi.org/10.1038/s41577-025-01172-3