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:

Dissecting the telomere–inner nuclear membrane interface formed in meiosis

Abstract

Tethering telomeres to the inner nuclear membrane (INM) allows homologous chromosome pairing during meiosis. The meiosis-specific protein TERB1 binds the telomeric protein TRF1 to establish telomere–INM connectivity and is essential for mouse fertility. Here we solve the structure of the human TRF1–TERB1 interface to reveal the structural basis for telomere–INM linkage. Disruption of this interface abrogates binding and compromises telomere–INM attachment in mice. An embedded CDK-phosphorylation site within the TRF1-binding region of TERB1 provides a mechanism for cap exchange, a late-pachytene phenomenon involving the dissociation of the TRF1–TERB1 complex. Indeed, further strengthening this interaction interferes with cap exchange. Finally, our biochemical analysis implicates distinct complexes for telomere–INM tethering and chromosome-end protection during meiosis. Our studies unravel the structure, stoichiometry, and physiological implications underlying telomere–INM tethering, thereby providing unprecedented insights into the unique function of telomeres in meiosis.

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

Figure 1: Structural and biochemical dissection of the TRF1TRFH–TERB1TBM interface.
Figure 2: Weakening the TRF1–TERB1 interface compromises telomere–INM tethering.
Figure 3: Further strengthening of the TRF1–TERB1 interface disrupts cap exchange.
Figure 4: Defining protein complexes involved in telomere tethering versus chromosome end protection.

Similar content being viewed by others

Accession codes

Primary accessions

Protein Data Bank

References

  1. Blackburn, E.H. Switching and signaling at the telomere. Cell 106, 661–673 (2001).

    Article  CAS  Google Scholar 

  2. Makarov, V.L., Hirose, Y. & Langmore, J.P. Long G tails at both ends of human chromosomes suggest a C strand degradation mechanism for telomere shortening. Cell 88, 657–666 (1997).

    Article  CAS  Google Scholar 

  3. Palm, W. & de Lange, T. How shelterin protects mammalian telomeres. Annu. Rev. Genet. 42, 301–334 (2008).

    Article  CAS  Google Scholar 

  4. Bilaud, T. et al. Telomeric localization of TRF2, a novel human telobox protein. Nat. Genet. 17, 236–239 (1997).

    Article  CAS  Google Scholar 

  5. Broccoli, D., Smogorzewska, A., Chong, L. & de Lange, T. Human telomeres contain two distinct Myb-related proteins, TRF1 and TRF2. Nat. Genet. 17, 231–235 1997).

    Article  CAS  Google Scholar 

  6. Nishikawa, T. et al. Solution structure of a telomeric DNA complex of human TRF1. Structure 9, 1237–1251 (2001).

    Article  CAS  Google Scholar 

  7. Nandakumar, J. & Cech, T.R. Finding the end: recruitment of telomerase to telomeres. Nat. Rev. Mol. Cell Biol. 14, 69–82 (2013).

    Article  CAS  Google Scholar 

  8. Watanabe, Y. Geometry and force behind kinetochore orientation: lessons from meiosis. Nat. Rev. Mol. Cell Biol. 13, 370–382 (2012).

    Article  CAS  Google Scholar 

  9. Marston, A.L. & Amon, A. Meiosis: cell-cycle controls shuffle and deal. Nat. Rev. Mol. Cell Biol. 5, 983–997 (2004).

    Article  CAS  Google Scholar 

  10. Scherthan, H. A bouquet makes ends meet. Nat. Rev. Mol. Cell Biol. 2, 621–627 (2001).

    Article  CAS  Google Scholar 

  11. Scherthan, H. et al. Centromere and telomere movements during early meiotic prophase of mouse and man are associated with the onset of chromosome pairing. J. Cell Biol. 134, 1109–1125 (1996).

    Article  CAS  Google Scholar 

  12. Hiraoka, Y. & Dernburg, A.F. The SUN rises on meiotic chromosome dynamics. Dev. Cell 17, 598–605 (2009).

    Article  CAS  Google Scholar 

  13. Sato, A. et al. Cytoskeletal forces span the nuclear envelope to coordinate meiotic chromosome pairing and synapsis. Cell 139, 907–919 (2009).

    Article  CAS  Google Scholar 

  14. Conrad, M.N. et al. Rapid telomere movement in meiotic prophase is promoted by NDJ1, MPS3, and CSM4 and is modulated by recombination. Cell 133, 1175–1187 (2008).

    Article  CAS  Google Scholar 

  15. Koszul, R., Kim, K.P., Prentiss, M., Kleckner, N. & Kameoka, S. Meiotic chromosomes move by linkage to dynamic actin cables with transduction of force through the nuclear envelope. Cell 133, 1188–1201 (2008).

    Article  CAS  Google Scholar 

  16. Scherthan, H. et al. Chromosome mobility during meiotic prophase in Saccharomyces cerevisiae. Proc. Natl. Acad. Sci. USA 104, 16934–16939 (2007).

    Article  CAS  Google Scholar 

  17. Trelles-Sticken, E., Adelfalk, C., Loidl, J. & Scherthan, H. Meiotic telomere clustering requires actin for its formation and cohesin for its resolution. J. Cell Biol. 170, 213–223 (2005).

    Article  CAS  Google Scholar 

  18. Ding, X. et al. SUN1 is required for telomere attachment to nuclear envelope and gametogenesis in mice. Dev. Cell 12, 863–872 (2007).

    Article  CAS  Google Scholar 

  19. Chikashige, Y. et al. Meiotic proteins bqt1 and bqt2 tether telomeres to form the bouquet arrangement of chromosomes. Cell 125, 59–69 (2006).

    Article  CAS  Google Scholar 

  20. Conrad, M.N., Dominguez, A.M. & Dresser, M.E. Ndj1p, a meiotic telomere protein required for normal chromosome synapsis and segregation in yeast. Science 276, 1252–1255 (1997).

    Article  CAS  Google Scholar 

  21. Shibuya, H., Ishiguro, K. & Watanabe, Y. The TRF1-binding protein TERB1 promotes chromosome movement and telomere rigidity in meiosis. Nat. Cell Biol. 16, 145–156 (2014).

    Article  CAS  Google Scholar 

  22. Shibuya, H. et al. MAJIN links telomeric DNA to the nuclear membrane by exchanging telomere cap. Cell 163, 1252–1266 (2015).

    Article  CAS  Google Scholar 

  23. Shibuya, H. & Watanabe, Y. The meiosis-specific modification of mammalian telomeres. Cell Cycle 13, 2024–2028 (2014).

    Article  CAS  Google Scholar 

  24. Chen, Y. et al. A shared docking motif in TRF1 and TRF2 used for differential recruitment of telomeric proteins. Science 319, 1092–1096 (2008).

    Article  CAS  Google Scholar 

  25. Blazer, L.L., Roman, D.L., Muxlow, M.R. & Neubig, R.R. Use of flow cytometric methods to quantify protein-protein interactions. Current Protoc. Cytom. 51, 13.11 (2010).

    Google Scholar 

  26. Li, B., Oestreich, S. & de Lange, T. Identification of human Rap1: implications for telomere evolution. Cell 101, 471–483 (2000).

    Article  CAS  Google Scholar 

  27. Arat, N.O. & Griffith, J.D. Human Rap1 interacts directly with telomeric DNA and regulates TRF2 localization at the telomere. J. Biol. Chem. 287, 41583–41594 (2012).

    Article  CAS  Google Scholar 

  28. Frescas, D. & de Lange, T. Binding of TPP1 protein to TIN2 protein is required for POT1a,b protein-mediated telomere protection. J. Biol. Chem. 289, 24180–24187 (2014).

    Article  CAS  Google Scholar 

  29. Takai, K.K., Kibe, T., Donigian, J.R., Frescas, D. & de Lange, T. Telomere protection by TPP1/POT1 requires tethering to TIN2. Mol. Cell 44, 647–659 (2011).

    Article  CAS  Google Scholar 

  30. Banani, S.F., Lee, H.O., Hyman, A.A. & Rosen, M.K. Biomolecular condensates: organizers of cellular biochemistry. Nat. Rev. Mol. Cell Biol. 18, 285–298 (2017).

    Article  CAS  Google Scholar 

  31. Takai, K.K., Hooper, S., Blackwood, S., Gandhi, R. & de Lange, T. In vivo stoichiometry of shelterin components. J. Biol. Chem. 285, 1457–1467 2010).

    Article  CAS  Google Scholar 

  32. Janoušková, E. et al. Human Rap1 modulates TRF2 attraction to telomeric DNA. Nucleic Acids Res. 43, 2691–2700 (2015).

    Article  Google Scholar 

  33. Mossessova, E. & Lima, C.D. Ulp1-SUMO crystal structure and genetic analysis reveal conserved interactions and a regulatory element essential for cell growth in yeast. Mol. Cell 5, 865–876 (2000).

    Article  CAS  Google Scholar 

  34. Collaborative Computational Project, Number 4. The CCP4 suite: programs for protein crystallography. Acta Crystallogr. D Biol. Crystallogr. 50, 760–763 (1994).

  35. Emsley, P. & Cowtan, K. Coot: model-building tools for molecular graphics. Acta Crystallogr. D Biol. Crystallogr. 60, 2126–2132 (2004).

    Article  Google Scholar 

  36. Adams, P.D. et al. PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta Crystallogr. D Biol. Crystallogr. 66, 213–221 (2010).

    Article  CAS  Google Scholar 

  37. Cobb, J., Cargile, B. & Handel, M.A. Acquisition of competence to condense metaphase I chromosomes during spermatogenesis. Dev. Biol. 205, 49–64 (1999).

    Article  CAS  Google Scholar 

  38. Shibuya, H., Morimoto, A. & Watanabe, Y. The dissection of meiotic chromosome movement in mice using an in vivo electroporation technique. PLoS Genet. 10, e1004821 (2014).

    Article  Google Scholar 

  39. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank the Life Sciences Collaborative Access Team at Argonne National Laboratory beamline staff for help with X-ray data collection, M. Iyer for help with protein purifications, and S. Grill for excellent feedback on the manuscript. This work was funded by NIH Grants R00CA167644 (to J.N.), R01GM120094 (to J.N.), R01AG050509 (to J.N., co-investigator), American Cancer Society Research Scholar grant 130882-RSG-17-037-01-DMC (to J.N.), NIH Biology of Aging Training Grant (T32AG000114) awarded to the University of Michigan Geriatrics Center from the National Institute on Aging (fellowship to E.M.S.), and MEXT KAKENHI 25000014 (to Y.W.).

Author information

Authors and Affiliations

Authors

Contributions

J.N., D.F.P., V.M.T., and Y.W. designed experiments; D.F.P., V.M.T., E.M.S., and J.N. conducted the crystallographic experiments; D.F.P. conducted cloning, protein purifications, and in vitro binding studies; Y.F. conducted in vivo cellular localization studies with guidance from H.S.; J.N., D.F.P., and Y.W. analyzed the data; J.N. and D.F.P. wrote the manuscript with input from all authors.

Corresponding author

Correspondence to Jayakrishnan Nandakumar.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 The TRF1-TERB1 interface.

(a) Overlay of the two TRF1TRFH-TERB1TRFB complexes observed in the dimeric crystal structure. The two TRFH domains are shown in cartoon depiction (green and cyan), while the two TBM peptides are shown in stick representation (orange and pink). (b) 2Fo – Fc map of the core of the TRF1-TERB1 interface contoured at 1.0 σ.

Supplementary Figure 2 Mutations in the TRF1-TERB1 interface abrogate binding.

(a) GST pull down of His-Smt3-TERB1TBM WT and mutant proteins using GST-TRF1TRFH as bait. Quantitation at the bottom was performed by dividing the appropriate band intensities by the molecular weight of the species followed by normalization against the molecular weight-normalized GST-TRF1TRFH signal in that line. (b) GST pull down of TRF1TRFH (WT or F142A) using GST-TERB1TBM as bait.

Supplementary Figure 3 Competition data for TERB1 TBM mutants with TRF1TRFH.

(a) And (b) are independent experimental replicates of data shown in Fig. 1j and k, respectively. Mean of technical duplicate is plotted. Mean and s.e.m. of biological duplicates (of technical duplicates) are shown in Table 2. (c) Size-exclusion (Superdex 200) profile of the TERB1TRFB and TERB21-107 complex following Ni-affinity purification and removal of the His-Smt3 tag. (d) Coomassie-stained SDS-PAGE for WT and indicated TERB1 mutant TERB1TRFB - TERB21-107 complexes eluted by size-exclusion chromatography. Yellow boxes highlight fractions indicative of a 1:1 TERB1:TERB2 complex. (e) Molecular weights of WT and TERB1 mutant TERB1TRFB - TERB21-107 complexes were determined by size-exclusion chromatography (Superdex 75). The theoretical molecular weight of a homodimer of the TERB1TRFB - TERB21-107 heterodimer (in kDa) along with the experimentally determined molecular weights (in kDa) is shown in a tabular format. (f) Equal amounts (5 μg) of indicated, purified TERB1TRFB - TERB21-107 complexes were analyzed using coomassie-stained SDS-PAGE.

Supplementary Figure 4 Cap exchange is not prevented in WT spermatocytes expressing TERB1 mutants that disrupt the TRF1-TERB1 interface.

(a) Schematic depiction of TRF1 and TERB1 signal width and localization in early and late pachytene. While the TERB1 signal does not undergo a significant change in width throughout pachytene, the TRF1 signal becomes more diffuse resulting in an increase in its outer diameter. (b) WT late pachytene spermatocytes expressing GFP-TERB1 L647E, GFP-TERB1 T648D, GFP-TERB1 P649E, or GFP-TERB1 R651E-R652E (green) stained with TRF1 (red) and SYCP3 (blue) antibodies.

Supplementary Figure 5 Quantitation of anomalous TRF1 E93K and TERB1 I645F cap exchange.

(a) WT mouse spermatocytes expressing GFP-TRF1 E93K (green) stained with TRF1 (red) antibody in late pachytene. (b and c) Quantitation shows that GFP-TERB1 I645F distributes to the wider area compared to WT TERB1 protein. “n” and P-values indicated on graph in panel b.

Supplementary Figure 6 Composition and stoichiometry of the TRF1-TIN2 and TRF1-TERB1 complexes.

(a) Fluorescence-based competition experiments using Alexa Fluor 488-labeled GST-TIN2TBM pre-bound to biotin-tagged TRF1TRFH on streptavidin beads titrated with varying concentrations of unlabeled full-length TIN2 and TERB1TRFB – TERB21-107 proteins. Mean of technical duplicate is plotted. Error bars indicate s.e.m. (b) Size-exclusion (Superdex 200) profiles confirming intramolecular dimerization of TRFH-TRFH fusions (based on molecular weight determination). (c) Coomassie-stained SDS-PAGE for TRFH-TRFH intramolecular fusion proteins eluted by size-exclusion chromatography. (d) Replicate of experiment shown in Fig. 4c with quantitation shown at the bottom. (e) Mean and s.e.m. of duplicate experiments from panel d and Fig. 4c.

Supplementary Figure 7 Binding of TERB1 to TRF2 in vitro.

(a) Direct binding curves of biotin-tagged TRF1TRFH and TRF2TRFH with Alexa Fluor 488-labeled GST-TERB1TBM. Mean of technical duplicate is plotted. Mean and s.e.m. of duplicate measurements are indicated. (b) Competition experiments demonstrating similar affinity of TRF1 and TRF2 for TERB1TBM peptide. Mean of technical duplicate is plotted. Error bars indicate s.e.m. (c-d) Fluorescence-based competition experiments using Alexa Fluor 488-labeled GST-TERB1TBM pre-bound to biotin-tagged TRF2TRFH on streptavidin beads titrated with varying concentrations of indicated unlabeled TERB1TBM peptides (c) or TERB1TRFB – TERB21-107 complexes (d). For c and d, mean of technical duplicate is plotted and two biological replicates were performed. (e) Ternary pull down on amylose resin showing that TRF2 associates with MBP-tagged TERB1TRFB – TERB21-107 in the absence of Rap1 but not in its presence. (f) Ternary pull down on amylose resin showing that TRF2 associates with MBP-tagged TERB1TRFB – TERB21-107 in the presence or absence of TIN2.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7, Supplementary Table 1 (PDF 1434 kb)

Life Sciences Reporting Summary (PDF 163 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pendlebury, D., Fujiwara, Y., Tesmer, V. et al. Dissecting the telomere–inner nuclear membrane interface formed in meiosis. Nat Struct Mol Biol 24, 1064–1072 (2017). https://doi.org/10.1038/nsmb.3493

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nsmb.3493

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