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:

The Y chromosome may contribute to sex-specific ageing in Drosophila

Abstract

Heterochromatin suppresses repetitive DNA, and a loss of heterochromatin has been observed in aged cells of several species, including humans and Drosophila. Males often contain substantially more heterochromatic DNA than females, due to the presence of a large, repeat-rich Y chromosome, and male flies generally have a shorter average lifespan than females. Here we show that repetitive DNA becomes de-repressed more rapidly in old male flies relative to females, and repeats on the Y chromosome are disproportionally mis-expressed during ageing. This is associated with a loss of heterochromatin at repetitive elements during ageing in male flies, and a general loss of repressive chromatin in aged males away from pericentromeric regions and the Y. By generating flies with different sex chromosome karyotypes (XXY females and X0 and XYY males), we show that repeat de-repression and average lifespan is correlated with the number of Y chromosomes. This suggests that sex-specific chromatin differences may contribute to sex-specific ageing in flies.

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: Ageing and the sex-specific chromatin landscape in Drosophila.
Fig. 2: Sex-specific silencing and expression of repeats during ageing.
Fig. 3: Survivorship of XXY females and X0 and XYY males.
Fig. 4: Expression of repetitive elements in XXY females and X0 and XYY males during ageing.

Similar content being viewed by others

Data availability

All RNA-seq and ChIP–seq reads are deposited at NCBI under BioProject ID PRJNA594556.

References

  1. O’Sullivan, R. J. & Karlseder, J. The great unravelling: chromatin as a modulator of the aging process. Trends Biochem. Sci. 37, 466–476 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Wood, J. G. et al. Chromatin remodeling in the ageing genome of Drosophila. Aging Cell 9, 971–978 (2010).

    Article  CAS  PubMed  Google Scholar 

  3. Tsurumi, A. & Li, W. X. Global heterochromatin loss: a unifying theory of aging? Epigenetics 7, 680–688 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Zhang, W. et al. Aging stem cells. A Werner syndrome stem cell model unveils heterochromatin alterations as a driver of human aging. Science 348, 1160–1163 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Larson, K. et al. Heterochromatin formation promotes longevity and represses ribosomal RNA synthesis. PLoS Genet. 8, e1002473 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Haithcock, E. et al. Age-related changes of nuclear architecture in Caenorhabditis elegans. Proc. Natl Acad. Sci. USA 102, 16690–16695 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Li, W. et al. Activation of transposable elements during aging and neuronal decline in Drosophila. Nat. Neurosci. 16, 529–531 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. De Cecco, M. et al. Genomes of replicatively senescent cells undergo global epigenetic changes leading to gene silencing and activation of transposable elements. Aging Cell 12, 247–256 (2013).

    Article  PubMed  CAS  Google Scholar 

  9. De Cecco, M. et al. Transposable elements become active and mobile in the genomes of ageing mammalian somatic tissues. Aging 5, 867–883 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Wood, J. G. & Helfand, S. L. Chromatin structure and transposable elements in organismal aging. Front. Genet. 4, 274 (2013).

    PubMed  PubMed Central  Google Scholar 

  11. Wood, J. G. et al. Chromatin-modifying genetic interventions suppress age-associated transposable element activation and extend life span in Drosophila. Proc. Natl Acad. Sci. USA 113, 11277–11282 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Van Meter, M. et al. SIRT6 represses LINE1 retrotransposons by ribosylating KAP1 but this repression fails with stress and age. Nat. Commun. 5, 5011 (2014).

    Article  PubMed  CAS  Google Scholar 

  13. Elsner, D., Meusemann, K. & Korb, J. Longevity and transposon defense, the case of termite reproductives. Proc. Natl Acad. Sci. USA 115, 5504–5509 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Hoskins, R. A. et al. Heterochromatic sequences in a Drosophila whole-genome shotgun assembly. Genome Biol. 3, research0085.1 (2002).

    Article  Google Scholar 

  15. Chang, C. H. & Larracuente, A. M. Heterochromatin-enriched assemblies reveal the sequence and organization of the Drosophila melanogaster Y chromosome. Genetics 211, 333–348 (2019).

    Article  CAS  PubMed  Google Scholar 

  16. Yoon, J. S., Gagen, K. P. & Zhu, D. L. Longevity of 68 species of Drosophila. Ohio J. Sci. 90, 16–32 (1990).

    Google Scholar 

  17. Tower, J. & Arbeitman, M. The genetics of gender and life span. J. Biol. 8, 38 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Lehtovaara, A., Schielzeth, H., Flis, I. & Friberg, U. Heritability of life span is largely sex limited in Drosophila. Am. Nat. 182, 653–665 (2013).

    Article  PubMed  Google Scholar 

  19. Pipoly, I. et al. The genetic sex-determination system predicts adult sex ratios in tetrapods. Nature 527, 91–94 (2015).

    Article  CAS  PubMed  Google Scholar 

  20. Hoskins, R. A. et al. The Release 6 reference sequence of the Drosophila melanogaster genome. Genome Res. 25, 445–458 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Li, X. Y., Harrison, M. M., Villalta, J. E., Kaplan, T. & Eisen, M. B. Establishment of regions of genomic activity during the Drosophila maternal to zygotic transition. eLife 3, e03737 (2014).

    Article  PubMed Central  CAS  Google Scholar 

  22. Brown, E. J., Nguyen, A. H. & Bachtrog, D. The Drosophila Y chromosome affects heterochromatin integrity genome-wide. Mol. Biol. Evol. https://doi.org/10.1093/molbev/msaa082 (2020).

  23. Bonhoure, N. et al. Quantifying ChIP-seq data: a spiking method providing an internal reference for sample-to-sample normalization. Genome Res. 24, 1157–1168 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Lu, B. Y., Emtage, P. C., Duyf, B. J., Hilliker, A. J. & Eissenberg, J. C. Heterochromatin protein 1 is required for the normal expression of two heterochromatin genes in Drosophila. Genetics 155, 699–708 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Carlson, K. A. et al. Genome-wide gene expression in relation to age in large laboratory cohorts of Drosophila melanogaster. Genet. Res. Int. 2015, 835624 (2015).

    PubMed  PubMed Central  Google Scholar 

  26. Garschall, K. & Flatt, T. The interplay between immunity and aging in Drosophila. F1000Res. 7, 160 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Pletcher, S. D. et al. Genome-wide transcript profiles in aging and calorically restricted Drosophila melanogaster. Curr. Biol. 12, 712–723 (2002).

    Article  CAS  PubMed  Google Scholar 

  28. Chen, H., Zheng, X. & Zheng, Y. Age-associated loss of lamin-B leads to systemic inflammation and gut hyperplasia. Cell 159, 829–843 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Chen, H., Zheng, X., Xiao, D. & Zheng, Y. Age-associated de-repression of retrotransposons in the Drosophila fat body, its potential cause and consequence. Aging Cell 15, 542–552 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Tran, J. R., Chen, H., Zheng, X. & Zheng, Y. Lamin in inflammation and aging. Curr. Opin. Cell Biol. 40, 124–130 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Pindyurin, A. V. et al. The large fraction of heterochromatin in Drosophila neurons is bound by both B-type lamin and HP1a. Epigenetics Chromatin 11, 65 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Salz, H. K. & Erickson, J. W. Sex determination in Drosophila: the view from the top. Fly 4, 60–70 (2010).

    Article  CAS  PubMed  Google Scholar 

  33. Carvalho, A. B., Koerich, L. B. & Clark, A. G. Origin and evolution of Y chromosomes: Drosophila tales. Trends Genet. 25, 270–277 (2009).

    Article  PubMed Central  CAS  Google Scholar 

  34. Ganley, A. R. & Kobayashi, T. Ribosomal DNA and cellular senescence: new evidence supporting the connection between rDNA and aging. FEMS Yeast Res. 14, 49–59 (2014).

    Article  CAS  PubMed  Google Scholar 

  35. Sinclair, D. A., Mills, K. & Guarente, L. Accelerated aging and nucleolar fragmentation in yeast sgs1 mutants. Science 277, 1313–1316 (1997).

    Article  CAS  PubMed  Google Scholar 

  36. Lu, K. L., Nelson, J. O., Watase, G. J., Warsinger-Pepe, N. & Yamashita, Y. M. Transgenerational dynamics of rDNA copy number in Drosophila male germline stem cells. eLife 7, e32421 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Peng, J. C. & Karpen, G. H. H3K9 methylation and RNA interference regulate nucleolar organization and repeated DNA stability. Nat. Cell Biol. 9, 25–35 (2007).

    Article  CAS  PubMed  Google Scholar 

  38. Buchwalter, A. & Hetzer, M. W. Nucleolar expansion and elevated protein translation in premature aging. Nat. Commun. 8, 328 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Tiku, V. et al. Small nucleoli are a cellular hallmark of longevity. Nat. Commun. 8, 16083 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Helmrich, A., Ballarino, M., Nudler, E. & Tora, L. Transcription-replication encounters, consequences and genomic instability. Nat. Struct. Mol. Biol. 20, 412–418 (2013).

    Article  CAS  PubMed  Google Scholar 

  41. Greil, F. & Ahmad, K. Nucleolar dominance of the Y chromosome in Drosophila melanogaster. Genetics 191, 1119–1128 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Tower, J. Sex-specific gene expression and life span regulation. Trends Endocrinol. Metab. 28, 735–747 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Lyckegaard, E. M. & Clark, A. G. Ribosomal DNA and stellate gene copy number variation on the Y chromosome of Drosophila melanogaster. Proc. Natl Acad. Sci. USA 86, 1944–1948 (1989).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Repping, S. et al. Polymorphism for a 1.6-Mb deletion of the human Y chromosome persists through balance between recurrent mutation and haploid selection. Nat. Genet. 35, 247–251 (2003).

    Article  CAS  PubMed  Google Scholar 

  45. Griffin, R. M., Le Gall, D., Schielzeth, H. & Friberg, U. Within-population Y-linked genetic variation for lifespan in Drosophila melanogaster. J. Evol. Biol. 28, 1940–1947 (2015).

    Article  CAS  PubMed  Google Scholar 

  46. Lemos, B., Branco, A. T. & Hartl, D. L. Epigenetic effects of polymorphic Y chromosomes modulate chromatin components, immune response, and sexual conflict. Proc. Natl Acad. Sci. USA 107, 15826–15831 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Lemos, B., Araripe, L. O. & Hartl, D. L. Polymorphic Y chromosomes harbor cryptic variation with manifold functional consequences. Science 319, 91–93 (2008).

    Article  CAS  PubMed  Google Scholar 

  48. Sackton, T. B., Montenegro, H., Hartl, D. L. & Lemos, B. Interspecific Y chromosome introgressions disrupt testis-specific gene expression and male reproductive phenotypes in Drosophila. Proc. Natl Acad. Sci. USA 108, 17046–17051 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Bosco, G., Campbell, P., Leiva-Neto, J. T. & Markow, T. A. Analysis of Drosophila species genome size and satellite DNA content reveals significant differences among strains as well as between species. Genetics 177, 1277–1290 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Ellis, L. et al. Intrapopulation genome size variation in D. melanogaster reflects life history. PLoS Genet. 10, e1004522 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Linford, N. J., Bilgir, C., Ro, J. & Pletcher, S. D. Measurement of lifespan in Drosophila melanogaster. J. Vis. Exp. 71, e50068 (2013).

    Google Scholar 

  53. Alekseyenko, A., Larschan, E., Lai, W., Park, P. & Kuroda, M. High-resolution ChIP-chip analysis reveals that the Drosophila MSL complex selectively identifies active genes on the male X chromosome. Genes Dev. 20, 848–857 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Ellison, C. E. & Bachtrog, D. Dosage compensation via transposable element mediated rewiring of a regulatory network. Science 342, 846–850 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  57. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    Article  CAS  PubMed  Google Scholar 

  58. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Eden, E., Navon, R., Steinfeld, I., Lipson, D. & Yakhini, Z. GOrilla: a tool for discovery and visualization of enriched GO terms in ranked gene lists. BMC Bioinformatics 10, 48 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Supek, F., Bošnjak, M., Škunca, N. & Šmuc, T. REVIGO summarizes and visualizes long lists of gene ontology terms. PLoS ONE 6, e21800 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Koch, P., Platzer, M. & Downie, B. R. RepARK—de novo creation of repeat libraries from whole-genome NGS reads. Nucleic Acids Res. 42, e80 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Quinlan, A. R. BEDTools: the Swiss-army tool for genome feature analysis. Curr. Protoc. Bioinformatics 47, 11.12.1–34 (2014).

    Article  Google Scholar 

  63. Kaplan, E. L. & Meier, P. Nonparametric estimation from incomplete observations. J. Am. Stat. Assoc. 53, 457–481 (1958).

    Article  Google Scholar 

Download references

Acknowledgements

D.B. was funded by NIH grants (nos. R01GM076007, GM101255 and R01AG057029).

Author information

Authors and Affiliations

Authors

Contributions

D.B. and E.J.B. conceived the study and wrote the paper. E.J.B. and A.H.N. collected and analysed the data.

Corresponding author

Correspondence to Doris Bachtrog.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Extended data

Extended Data Fig. 1 Survivorship curves of additional D. melanogaster strains.

Shown are Kaplan-Meier survivorship curves for line 2549 males and females ((C(1;Y),y1cv1v1B/0 & C(1)RM,y1v1/0) and Oregon-R wild-type males and females.

Extended Data Fig. 2 Genome-wide enrichment of H3K9me2 for replicate young and old D. melanogaster males and females along the different chromosome arms.

Pearson correlation coefficients for replicate H3K9me2 datasets for old males and females, and boxplots of normalized enrichment values for the replicates. Genome-wide plots were generated using biological replicate data as in Fig. 1b,d.

Extended Data Fig. 3 Loss and gain of heterochromatin during ageing.

Shown are chromosomal locations of 50 kb windows that gain (red) or lose (blue) at least 1.5-fold H3K9me2 signal during ageing for males and females. Pericentromeric regions are indicated by the red portion of the line beneath each chromosome.

Supplementary information

Supplementary Information

Supplementary Figs. 1–19 and Tables 1 and 2.

Reporting Summary

Supplementary Table 3 and 4

Gene expression changes during ageing in (A) XX females, (B) XY males, (C) X0 males, (D) XXY females and (E) XYY males. Enriched GO categories of genes that significantly changed expression during ageing in (A) XX females, (B) XY males, (C) X0 males, (D) XXY females and (E) XYY males.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Brown, E.J., Nguyen, A.H. & Bachtrog, D. The Y chromosome may contribute to sex-specific ageing in Drosophila. Nat Ecol Evol 4, 853–862 (2020). https://doi.org/10.1038/s41559-020-1179-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41559-020-1179-5

This article is cited by

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