Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Retrotransposons hijack alt-EJ for DNA replication and eccDNA biogenesis

Abstract

Retrotransposons are highly enriched in the animal genome1,2,3. The activation of retrotransposons can rewrite host DNA information and fundamentally impact host biology1,2,3. Although developmental activation of retrotransposons can offer benefits for the host, such as against virus infection, uncontrolled activation promotes disease or potentially drives ageing1,2,3,4,5. After activation, retrotransposons use their mRNA as templates to synthesize double-stranded DNA for making new insertions in the host genome1,2,3,6. Although the reverse transcriptase that they encode can synthesize the first-strand DNA1,2,3,6, how the second-strand DNA is generated remains largely unclear. Here we report that retrotransposons hijack the alternative end-joining (alt-EJ) DNA repair process of the host for a circularization step to synthesize their second-strand DNA. We used Nanopore sequencing to examine the fates of replicated retrotransposon DNA, and found that 10% of them achieve new insertions, whereas 90% exist as extrachromosomal circular DNA (eccDNA). Using eccDNA production as a readout, further genetic screens identified factors from alt-EJ as essential for retrotransposon replication. alt-EJ drives the second-strand synthesis of the long terminal repeat retrotransposon DNA through a circularization process and is therefore necessary for eccDNA production and new insertions. Together, our study reveals that alt-EJ is essential in driving the propagation of parasitic genomic retroelements. Our study uncovers a conserved function of this understudied DNA repair process, and provides a new perspective to understand—and potentially control—the retrotransposon life cycle.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: HMS-Beagle predominantly produces eccDNA after activation.
Fig. 2: Factors from the alt-EJ process drive 1-LTR full-length eccDNA formation.
Fig. 3: Blocking the alt-EJ process abrogates DNA synthesis and all eccDNA production from HMS-Beagle.
Fig. 4: Blocking alt-EJ process abrogates HMS-Beagle mobilization.
Fig. 5: mdg4 and mammalian IAP form eccDNA through the alt-EJ factors.

Data availability

The sequencing data were deposited to the National Center for Biotechnology Information (NCBI) under accession number PRJNA794176. The sequence of the eGFP-tagged HMS-Beagle is available at GitHub (https://github.com/ZhaoZhangZZlab/eccDNA_formation_2021/tree/main/Reference). Source data are provided with this paper.

Code availability

All related code is available at GitHub (https://github.com/ZhaoZhangZZlab/eccDNA_formation_2021).

References

  1. Wells, J. N. & Feschotte, C. A field guide to eukaryotic transposable elements. Ann. Rev. Genet. 54, 539–561 (2020).

    Article  CAS  PubMed  Google Scholar 

  2. Kazazian, H. H. Jr. & Moran, J. V. Mobile DNA in health and disease. New Engl. J. Med. 377, 361–370 (2017).

    Article  CAS  PubMed  Google Scholar 

  3. 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).

    Article  CAS  PubMed  Google Scholar 

  4. Frank, J. A. et al. Evolution and antiviral activity of a human protein of retroviral origin. Science 378, 422–428 (2022).

    Article  ADS  CAS  PubMed  Google Scholar 

  5. Wang, L. et al. Retrotransposon activation during Drosophila metamorphosis conditions adult antiviral responses. Nat. Genet. 54, 1933–1945 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Telesnitsky, A. & Goff, S. P. in Retroviruses (eds Coffin, J. M. et al.) 121–160 (Cold Spring Harbor Laboratory Press, 1997).

  7. Wang, L., Dou, K., Moon, S., Tan, F. J. & Zhang, Z. Z. Hijacking oogenesis enables massive propagation of LINE and retroviral transposons. Cell 174, 1082–1094 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Xie, T. & Spradling, A. C. A niche maintaining germ line stem cells in the Drosophila ovary. Science 290, 328–330 (2000).

    Article  ADS  CAS  PubMed  Google Scholar 

  9. Kaminker, J. S. et al. The transposable elements of the Drosophila melanogaster euchromatin: a genomics perspective. Genome Biol. 3, research0084.1 (2002).

    Article  Google Scholar 

  10. Wicker, T. et al. A unified classification system for eukaryotic transposable elements. Nat. Rev. Genet. 8, 973–982 (2007).

    Article  CAS  PubMed  Google Scholar 

  11. Lammel, U. & Klambt, C. Specific expression of the Drosophila midline-jumper retro-transposon in embryonic CNS midline cells. Mech. Dev. 100, 339–342 (2001).

    Article  CAS  PubMed  Google Scholar 

  12. Siomi, M. C., Sato, K., Pezic, D. & Aravin, A. A. PIWI-interacting small RNAs: the vanguard of genome defence. Nat. Rev. Mol. Cell Biol. 12, 246–258 (2011).

    Article  CAS  PubMed  Google Scholar 

  13. Li, C. et al. Collapse of germline piRNAs in the absence of Argonaute3 reveals somatic piRNAs in flies. Cell 137, 509–521 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Vagin, V. V. et al. A distinct small RNA pathway silences selfish genetic elements in the germline. Science 313, 320–324 (2006).

    Article  ADS  CAS  PubMed  Google Scholar 

  15. Wang, Y. et al. eccDNAs are apoptotic products with high innate immunostimulatory activity. Nature 599, 308–314 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  16. Henriksen, R. A. et al. Circular DNA in the human germline and its association with recombination. Mol. Cell 82, 209–217 (2022).

    Article  CAS  PubMed  Google Scholar 

  17. Boeke, J. D., Garfinkel, D. J., Styles, C. A. & Fink, G. R. Ty elements transpose through an RNA intermediate. Cell 40, 491–500 (1985).

    Article  CAS  PubMed  Google Scholar 

  18. Shoshani, O. et al. Chromothripsis drives the evolution of gene amplification in cancer. Nature 591, 137–141 (2021).

    Article  ADS  CAS  PubMed  Google Scholar 

  19. Libuda, D. E. & Winston, F. Amplification of histone genes by circular chromosome formation in Saccharomyces cerevisiae. Nature 443, 1003–1007 (2006).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  20. Moller, H. D. et al. Formation of extrachromosomal circular DNA from long terminal repeats of retrotransposons in Saccharomyces cerevisiae. G3 6, 453–462 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Brown, P. O. in Retroviruses (eds Coffin, J. M. et al.) 161–204 (Cold Spring Harbor Laboratory Press, 1997).

  22. Brambati, A., Barry, R. M. & Sfeir, A. DNA polymerase theta (Polθ)—an error-prone polymerase necessary for genome stability. Curr. Opin. Genet. Dev. 60, 119–126 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Mateos-Gomez, P. A. et al. Mammalian polymerase θ promotes alternative NHEJ and suppresses recombination. Nature 518, 254–257 (2015).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  24. Ramsden, D. A., Carvajal-Garcia, J. & Gupta, G. P. Mechanism, cellular functions and cancer roles of polymerase-theta-mediated DNA end joining. Nat. Rev. Mol. Cell Biol. 23, 125–140 (2022).

    Article  CAS  PubMed  Google Scholar 

  25. Lauermann, V. & Boeke, J. D. Plus-strand strong-stop DNA transfer in yeast Ty retrotransposons. EMBO J. 16, 6603–6612 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Heyman, T., Agoutin, B., Friant, S., Wilhelm, F. X. & Wilhelm, M. L. Plus-strand DNA synthesis of the yeast retrotransposon Ty1 is initiated at two sites, PPT1 next to the 3′ LTR and PPT2 within the pol gene. PPT1 is sufficient for Ty1 transposition. J. Mol. Biol. 253, 291–303 (1995).

    Article  CAS  PubMed  Google Scholar 

  27. Tanese, N., Telesnitsky, A. & Goff, S. P. Abortive reverse transcription by mutants of Moloney murine leukemia virus deficient in the reverse transcriptase-associated RNase H function. J. Virol. 65, 4387–4397 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Finston, W. I. & Champoux, J. J. RNA-primed initiation of Moloney murine leukemia virus plus strands by reverse transcriptase in vitro. J. Virol. 51, 26–33 (1984).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Rhim, H., Park, J. & Morrow, C. D. Deletions in the tRNA(Lys) primer-binding site of human immunodeficiency virus type 1 identify essential regions for reverse transcription. J. Virol. 65, 4555–4564 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Le Grice, S. F. “In the beginning”: initiation of minus strand DNA synthesis in retroviruses and LTR-containing retrotransposons. Biochemistry 42, 14349–14355 (2003).

    Article  PubMed  Google Scholar 

  31. Hu, Z., Leppla, S. H., Li, B. & Elkins, C. A. Antibodies specific for nucleic acids and applications in genomic detection and clinical diagnostics. Expert Rev. Mol. Diagn. 14, 895–916 (2014).

    Article  CAS  PubMed  Google Scholar 

  32. Gagnier, L., Belancio, V. P. & Mager, D. L. Mouse germ line mutations due to retrotransposon insertions. Mobile DNA 10, 15 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Dewannieux, M., Dupressoir, A., Harper, F., Pierron, G. & Heidmann, T. Identification of autonomous IAP LTR retrotransposons mobile in mammalian cells. Nat. Genet. 36, 534–539 (2004).

    Article  CAS  PubMed  Google Scholar 

  34. Schorn, A. J., Gutbrod, M. J., LeBlanc, C. & Martienssen, R. LTR-retrotransposon control by tRNA-derived small RNAs. Cell 170, 61–71 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Shank, P. R. et al. Mapping unintegrated avian sarcoma virus DNA: termini of linear DNA bear 300 nucleotides present once or twice in two species of circular DNA. Cell 15, 1383–1395 (1978).

    Article  CAS  PubMed  Google Scholar 

  36. Grow, E. J. et al. Intrinsic retroviral reactivation in human preimplantation embryos and pluripotent cells. Nature 522, 221–225 (2015).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  37. Wang, J. et al. Primate-specific endogenous retrovirus-driven transcription defines naive-like stem cells. Nature 516, 405–409 (2014).

    Article  ADS  CAS  PubMed  Google Scholar 

  38. Macfarlan, T. S. et al. Embryonic stem cell potency fluctuates with endogenous retrovirus activity. Nature 487, 57–63 (2012).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  39. Pang, M., McConnell, M. & Fisher, P. A. The Drosophila mus308 gene product, implicated in tolerance of DNA interstrand crosslinks, is a nuclear protein found in both ovaries and embryos. DNA Repair 4, 971–982 (2005).

    Article  CAS  PubMed  Google Scholar 

  40. Vaidya, A. et al. Knock-in reporter mice demonstrate that DNA repair by non-homologous end joining declines with age. PLoS Genet. 10, e1004511 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Danecek, P. et al. Twelve years of SAMtools and BCFtools. Gigascience 10, giab008 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Gu, Z., Gu, L., Eils, R., Schlesner, M. & Brors, B. circlize implements and enhances circular visualization in R. Bioinformatics 30, 2811–2812 (2014).

    Article  CAS  PubMed  Google Scholar 

  43. Li, H. Minimap2: pairwise alignment for nucleotide sequences. Bioinformatics 34, 3094–3100 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparinggenomic features. Bioinformatics 26, 841–842 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Robinson, J. T. et al. Integrative genomics viewer. Nat. Biotechnol. 29, 24–26 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank M. Dewannieux and K. Wood for providing plasmids; J. Brennecke, X. Chen, J. Sekelsky and the members of the BDSC for providing fly stocks; the members of the Z.Z.Z. laboratory and D. MacAlpine for suggestions; D. Fox, X.-F. Wang, B. Cullen, L. Lin and D. MacAlpine for reading the manuscript; and D. Erie and P. Marszalek for suggestions on AFM sample preparation. This work was supported by grants to Z.Z.Z. from the Pew Biomedical Scholars Program and the National Institutes of Health (DP5 OD021355 and R01 GM141018); and to D.A.R. from the National Cancer Institute (P01CA247773).

Author information

Authors and Affiliations

Authors

Contributions

Z.Z.Z., F.Y. and W.S. conceived the project. All of the authors designed the experiments. O.W.C. performed experiments for Fig. 5d and Extended Data Fig. 9c,d. L.T. performed experiments for Extended Data Fig. 9e. L.W. generated data for Fig. 1. F.Y. performed the rest of the experiments. W.S. performed all of the bioinformatics. Z.Z.Z., F.Y. and W.S. wrote the manuscript. All of the authors read and approved the manuscript.

Corresponding author

Correspondence to ZZ Zhao Zhang.

Ethics declarations

Competing interests

Z.Z.Z., F.Y. and W.S. are listed co-inventors on a US provisional patent application (no. 63/309,136) filed by Duke University related to this work.

Peer review

Peer review information

Nature thanks Todd Macfarlan and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data figures and tables

Extended Data Fig. 1 The engineered HMS-Beagle reporter dominantly forms eccDNA.

a, Schematic design of the HMS-Beagle reporter. An eGFP reporter is inserted into the 3′ UTR of HMS-Beagle sequence in an antisense direction. b, Fly cross scheme to collect samples for measuring the potential integration and eccDNA events from transposon-silenced and transposon-activated flies. c, Integrative Genomics Viewer (IGV) alignments showing reads mapped to HMS-Beagle reporter locus in the genome from embryos laid by transposon-silenced females. Individual purple horizonal bar represents a unique Nanopore read containing eGFP sequence. All of the reads contained at least one of the LTRs and extended to the adjacent region, indicating they were aligned to the original genomic locus of the reporter. d, The distribution of new integrations from engineered HMS-Beagle reporter on Drosophila genome. Each triangle represents a new integration event.

Extended Data Fig. 2 PCR based assay to measure HMS-Beagle eccDNA.

a, schematic of the design of divergent primers to identify retrotransposon eccDNA. b, AFM imaging to visualize the shapes of DNA. Exonuclease digestion significantly enrich eccDNA for detection in panel c. Scale bar, 500 nm. c, the representative gel image showing retrotransposons predominantly form 1-LTR circles. Performing PCR using total DNA as template produced non-specific bands, likely resulting from the nested transposon fragments resided within the linear genome. Using exonuclease to enrich eccDNA generated two PCR products corresponding to 1-LTR and 2-LTR eccDNA respectively. d and e, Sanger sequencing to validate the formation of HMS-Beagle eccDNA. The PCR products for the very right lane of panel c were cloned into plasmid vector and 11 corresponding colonies were sequenced. Ten of the 11 colonies are from 1-LTR eccDNA (d). One colony is from 2-LTR eccDNA (e). Notably, this 2-LTR eccDNA has 34 bp deletion at the end-end junction site, indicating it is formed by the error-prone NHEJ pathway. This conclusion is further supported by Extended Data Fig. 9.

Extended Data Fig. 3 eccDNA-seq to provide direct evidence of circle formation.

a, Schematic of the eccDNA-seq workflow. After extracting total DNA, linear DNA was removed by Plasmid-Safe DNase digestion. eccDNA was amplified by Phi29 DNA polymerase through rolling circle amplification. And the sequencing libraries were prepared and sequenced on a Nanopore instrument. b, The proportion of eccDNA-seq and genome-seq reads mapped to mitochondria (black regions), transposons (pink regions), and the rest of the genome (grey regions). All samples were from fly ovaries. The genome-seq libraries were made by the tagmentation method and sequenced by the Nanopore platform to capture circular DNA, such as the mitochondrial genome. c, Bar graph showing qPCR results of mitochondrial DNA copies detected by two sets of primers respectively. The relative abundances are normalized to the spike-in plasmid. The mitochondrial DNA copies are essentially unchanged upon transposon activation in Drosophila ovary. The bars report mean ± standard deviation from three biological replicates (n = 3). p values were calculated with a two-tailed, two-sample unequal variance t test. d, Circos plots showing the number of the eccDNA-seq reads for the four classes of HMS-Beagle circles. From the outer layer to inward: 1-LTR full-length circles, 2-LTR full-length circles, 1-LTR-rearranged circle, and non-LTR rearranged circles.

Source data

Extended Data Fig. 4 eccDNA production from HMS-Beagle requires its mRNA intermediates.

a, RNA-FISH to detect HMS-Beagle mRNA. All flies carrying sh-aub to activate transposons in germline cells. Further introducing sh-white (serving as a control) into the animals does not change transposon activity: HMS-Beagle remains activated. Upon introducing shHMS-Beagle construct to silence it, its RNA was undetectable by RNA-FISH. Scale bar, 20 µm, b, Top: primer design to detect HMS-Beagle eccDNA (Extended Data Fig. 2). Bottom: The representative gel image of PCR products showing that HMS-Beagle eccDNA production was abolished when its mRNA production was suppressed by RNAi. Each genotype has three biological replicates.

Extended Data Fig. 5 Confirmation of the RNAi silencing efficiency in oocytes.

RT-qPCR showing the depletion efficiency of indicated genes by germline-specific RNAi. Relative mRNA levels were normalized to rp49 gene. The bars report mean ± standard deviation from four biological replicates (n = 4). p values were calculated with a two-tailed, two-sample unequal variance t test. Silencing Lig3 or Fen1 made flies barely lay eggs/oocytes, impeding a validation of the RNAi silencing efficiency for them.

Source data

Extended Data Fig. 6 HMS-Beagle mRNA remains unchanged upon depletion of the components from alt-EJ process.

Transposon activation was achieved by silencing Aub in germline cells. The Y-axis is normalized reads count.

Extended Data Fig. 7 Immunoprecipitation assay to measure the accumulation of HMS-Beagle single-stranded DNA upon alt-EJ suppression.

The bars report mean ± standard deviation from four biological replicates (n = 4). P values were calculated with a two-tailed, two-sample unequal variance t test. Although Mab3034 antibodies used in this experiment have 10-fold higher affinity for single-stranded DNA than double-stranded DNA31, they still can bind HMS-Beagle genomic double-stranded DNA across all samples. This would mask the difference of single-stranded DNA across samples and lead to underestimation of the amount of accumulated single-stranded DNA upon alt-EJ inhibition.

Source data

Extended Data Fig. 8 IAP needs its reverse transcriptase, but not integrase, activity for eccDNA biogenesis.

a, Sanger sequencing to validate the IAP reverse transcriptase mutant. b, Sanger sequencing to validate the IAP integrase mutant. c, PCR based assay to measure the production of IAP eccDNA. The very left lane was the condition without introducing IAP plasmid. de, Either immunoblotting (d and e) or RT-qPCR (f) to test the silencing efficiency of CRISPRi on depleting the alt-EJ factors. For each gene, two gRNAs were designed. NT (non-targeting) is a random gRNA without a targeting site. For RT-qPCR, relative mRNA levels were normalized to the RR18S gene. The bars report mean ± standard deviation from four biological replicates (n = 4). Statistical significance were calculated with a two-tailed, two-sample unequal variance t test. The p value for gRNA-1 is 0.0011, while for gRNA-2 is 0.0014.

Source data

Extended Data Fig. 9 NHEJ pathway is essential for 2-LTR eccDNA biogenesis.

a, Mutating Lig4 abolishes 2-LTR eccDNA production for mdg4 retrotransposon. b, Silencing Ku80 or Lig4 by RNAi reduces mdg4 2-LTR eccDNA formation. c, Sanger sequencing to validate lig4 mutation of the 293T cells. d, Sanger sequencing to validate XRCC4 mutation of the 293T cells. e, Mutating either Lig4 or XRCC4 abolishes 2-LTR eccDNA production for IAP retrotransposon.

Extended Data Fig. 10 Detailed model of the replication cycle of LTR-retrotransposons supported by our study.

Our data support alt-EJ factors mediate a circularization step for retrotransposon 2-nd strand DNA synthesis. While this step can generate full-length linear double-stranded DNA for integration, it appears to dominantly produce 1-LTR eccDNA.

Supplementary information

Supplementary Figs. 1–6

Supplementary Figs. 1–6.

Reporting Summary

Peer Review File

Supplementary Table 1

Statistical summary of the deep-sequencing results.

Supplementary Table 2

RNAi screen to identify factors required for HMS-Beagle 1-LTR eccDNA biogenesis.

Supplementary Table 3

List of flies used in this study.

Supplementary Table 4

List of oligos used in this study and related information.

Source data

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.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yang, F., Su, W., Chung, O.W. et al. Retrotransposons hijack alt-EJ for DNA replication and eccDNA biogenesis. Nature 620, 218–225 (2023). https://doi.org/10.1038/s41586-023-06327-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-023-06327-7

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing