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:

Structure of Tetrahymena telomerase-bound CST with polymerase α-primase

Abstract

Telomeres are the physical ends of linear chromosomes. They are composed of short repeating sequences (such as TTGGGG in the G-strand for Tetrahymena thermophila) of double-stranded DNA with a single-strand 3′ overhang of the G-strand and, in humans, the six shelterin proteins: TPP1, POT1, TRF1, TRF2, RAP1 and TIN21,2. TPP1 and POT1 associate with the 3′ overhang, with POT1 binding the G-strand3 and TPP1 (in complex with TIN24) recruiting telomerase via interaction with telomerase reverse transcriptase5 (TERT). The telomere DNA ends are replicated and maintained by telomerase6, for the G-strand, and subsequently DNA polymerase α–primase7,8 (PolαPrim), for the C-strand9. PolαPrim activity is stimulated by the heterotrimeric complex CTC1–STN1–TEN110,11,12 (CST), but the structural basis of the recruitment of PolαPrim and CST to telomere ends remains unknown. Here we report cryo-electron microscopy (cryo-EM) structures of Tetrahymena CST in the context of the telomerase holoenzyme, in both the absence and the presence of PolαPrim, and of PolαPrim alone. Tetrahymena Ctc1 binds telomerase subunit p50, a TPP1 orthologue, on a flexible Ctc1 binding motif revealed by cryo-EM and NMR spectroscopy. The PolαPrim polymerase subunit POLA1 binds Ctc1 and Stn1, and its interface with Ctc1 forms an entry port for G-strand DNA to the POLA1 active site. We thus provide a snapshot of four key components that are required for telomeric DNA synthesis in a single active complex—telomerase-core ribonucleoprotein, p50, CST and PolαPrim—that provides insights into the recruitment of CST and PolαPrim and the handoff between G-strand and C-strand synthesis.

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: Cryo-EM structure of TtCST in the telomerase holoenzyme.
Fig. 2: Interface between TtCST and p50.
Fig. 3: Structure of Tetrahymena telomerase holoenzyme in complex with PolαPrim.
Fig. 4: Interface between TtCST and POLA1core.
Fig. 5: Structural comparison of Tetrahymena and human CST.

Similar content being viewed by others

Data availability

Cryo-EM density maps have been deposited in the Electron Microscopy Data Bank under accession numbers EMD-26863 (telomerase with CST), EMD-26864 (telomerase with CST–PolαPrim), EMD-26865 (telomerase), EMD-26866 (CST–PolαPrim), EMD-26867 (PolαPrim platform), EMD-26868 (POLA2–POLA1CTD–PRIM2N) and EMD-26869 (PRIM2N–PRIM1). The atomic models have been deposited in the Protein Data Bank under accession codes 7UY5 (telomerase with CST), 7UY6 (telomerase), 7UY7 (CST–PolαPrim) and 7UY8 (PolαPrim platform). Backbone chemical shifts have been deposited in BMRB under accession codes 51441 (Ctc1 OB-A), 51442 (Ctc1 OB-A with p50 peptide 228–250) and 51443 (p50 peptide 228–250).

References

  1. de Lange, T. Shelterin-mediated telomere protection. Annu. Rev. Genet. 52, 223–247 (2018).

    Article  PubMed  CAS  Google Scholar 

  2. Lim, C. J. & Cech, T. R. Shaping human telomeres: from shelterin and CST complexes to telomeric chromatin organization. Nat. Rev. Mol. Cell Biol. 22, 283–298 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Baumann, P. & Cech, T. R. Pot1, the putative telomere end-binding protein in fission yeast and humans. Science 292, 1171–1175 (2001).

    Article  ADS  CAS  PubMed  Google Scholar 

  4. Abreu, E. et al. TIN2-tethered TPP1 recruits human telomerase to telomeres in vivo. Mol. Cell. Biol. 30, 2971–2982 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Nandakumar, J. et al. The TEL patch of telomere protein TPP1 mediates telomerase recruitment and processivity. Nature 492, 285–289 (2012).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  6. Greider, C. W. & Blackburn, E. H. Identification of a specific telomere terminal transferase activity in Tetrahymena extracts. Cell 43, 405–413 (1985).

    Article  CAS  PubMed  Google Scholar 

  7. Pellegrini, L. The Pol α-primase complex. Subcell. Biochem. 62, 157–169 (2012).

    Article  CAS  PubMed  Google Scholar 

  8. Baranovskiy, A. G. & Tahirov, T. H. Elaborated action of the human primosome. Genes 8, 62 (2017).

    Article  PubMed Central  CAS  Google Scholar 

  9. Qi, H. & Zakian, V. A. The Saccharomyces telomere-binding protein Cdc13p interacts with both the catalytic subunit of DNA polymerase α and the telomerase-associated Est1 protein. Genes Dev. 14, 1777–1788 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Wang, F. et al. Human CST has independent functions during telomere duplex replication and C-strand fill-in. Cell Rep. 2, 1096–1103 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Surovtseva, Y. V. et al. Conserved telomere maintenance component 1 interacts with STN1 and maintains chromosome ends in higher eukaryotes. Mol. Cell 36, 207–218 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Chen, L. Y., Redon, S. & Lingner, J. The human CST complex is a terminator of telomerase activity. Nature 488, 540–544 (2012).

    Article  ADS  CAS  PubMed  Google Scholar 

  13. Zaug, A. J. et al. CST does not evict elongating telomerase but prevents initiation by ssDNA binding. Nucleic Acids Res. 49, 11653–11665 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Stewart, J. A. et al. Human CST promotes telomere duplex replication and general replication restart after fork stalling. EMBO J. 31, 3537–3549 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Mirman, Z. et al. 53BP1–RIF1–shieldin counteracts DSB resection through CST- and Polα-dependent fill-in. Nature 560, 112–116 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  16. Holohan, B., Wright, W. E. & Shay, J. W. Telomeropathies: an emerging spectrum disorder. J. Cell Biol. 205, 289–299 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Chen, L. Y., Majerska, J. & Lingner, J. Molecular basis of telomere syndrome caused by CTC1 mutations. Genes Dev. 27, 2099–2108 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Prakash, A. & Borgstahl, G. E. The structure and function of replication protein A in DNA replication. Subcell. Biochem. 62, 171–196 (2012).

    Article  CAS  PubMed  Google Scholar 

  19. Lim, C. J. et al. The structure of human CST reveals a decameric assembly bound to telomeric DNA. Science 368, 1081–1085 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  20. Ge, Y. et al. Structural insights into telomere protection and homeostasis regulation by yeast CST complex. Nat. Struct. Mol. Biol. 27, 752–762 (2020).

    Article  CAS  PubMed  Google Scholar 

  21. Bryan, C., Rice, C., Harkisheimer, M., Schultz, D. C. & Skordalakes, E. Structure of the human telomeric Stn1–Ten1 capping complex. PLoS ONE 8, e66756 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  22. Jiang, J. et al. Structure of Tetrahymena telomerase reveals previously unknown subunits, functions, and interactions. Science 350, aab4070 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Wan, B. et al. The Tetrahymena telomerase p75–p45–p19 subcomplex is a unique CST complex. Nat. Struct. Mol. Biol. 22, 1023–1026 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Kratz, K. & de Lange, T. Protection of telomeres 1 proteins POT1a and POT1b can repress ATR signaling by RPA exclusion, but binding to CST limits ATR repression by POT1b. J. Biol. Chem. 293, 14384–14392 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Wan, M., Qin, J., Songyang, Z. & Liu, D. OB fold-containing protein 1 (OBFC1), a human homolog of yeast Stn1, associates with TPP1 and is implicated in telomere length regulation. J. Biol. Chem. 284, 26725–26731 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Kelich, J. M., Papaioannou, H. & Skordalakes, E. Pol α-primase dependent nuclear localization of the mammalian CST complex. Commun. Biol. 4, 349 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Ganduri, S. & Lue, N. F. STN1–POLA2 interaction provides a basis for primase-pol α stimulation by human STN1. Nucleic Acids Res. 45, 9455–9466 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Feng, X., Hsu, S. J., Kasbek, C., Chaiken, M. & Price, C. M. CTC1-mediated C-strand fill-in is an essential step in telomere length maintenance. Nucleic Acids Res. 45, 4281–4293 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Feng, X. et al. CTC1–STN1 terminates telomerase while STN1–TEN1 enables C-strand synthesis during telomere replication in colon cancer cells. Nat. Commun. 9, 2827 (2018).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  30. Wang, Y., Susac, L. & Feigon, J. Structural biology of telomerase. Cold Spring Harb. Perspect. Biol. 11, a032383 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Wang, F. et al. The POT1–TPP1 telomere complex is a telomerase processivity factor. Nature 445, 506–510 (2007).

    Article  ADS  CAS  PubMed  Google Scholar 

  32. Sekne, Z., Ghanim, G. E., van Roon, A. M. & Nguyen, T. H. D. Structural basis of human telomerase recruitment by TPP1–POT1. Science 375, 1173–1176 (2022).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  33. Liu, B. et al. Structure of active human telomerase with telomere shelterin protein TPP1. Nature 604, 578–583 (2022).

    Article  ADS  CAS  PubMed  Google Scholar 

  34. Upton, H. E., Chan, H., Feigon, J. & Collins, K. Shared subunits of Tetrahymena telomerase holoenzyme and replication protein A have different functions in different cellular complexes. J. Biol. Chem. 292, 217–228 (2017).

    Article  CAS  PubMed  Google Scholar 

  35. Zeng, Z. et al. Structural basis for Tetrahymena telomerase processivity factor Teb1 binding to single-stranded telomeric-repeat DNA. Proc. Natl Acad. Sci. USA 108, 20357–20361 (2011).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  36. Lei, M., Podell, E. R. & Cech, T. R. Structure of human POT1 bound to telomeric single-stranded DNA provides a model for chromosome end-protection. Nat. Struct. Mol. Biol. 11, 1223–1229 (2004).

    Article  CAS  PubMed  Google Scholar 

  37. Chen, C. et al. Structural insights into POT1–TPP1 interaction and POT1 C-terminal mutations in human cancer. Nat. Commun. 8, 14929 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  38. Rice, C. et al. Structural and functional analysis of the human POT1–TPP1 telomeric complex. Nat. Commun. 8, 14928 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  39. Jiang, J. et al. Structure of telomerase with telomeric DNA. Cell 173, 1179–1190.e1113 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. He, Y. et al. Structures of telomerase at several steps of telomere repeat synthesis. Nature 593, 454–459 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  41. Ghanim, G. E. et al. Structure of human telomerase holoenzyme with bound telomeric DNA. Nature 593, 449–453 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  42. Wang, Y., Gallagher-Jones, M., Sušac, L., Song, H. & Feigon, J. A structurally conserved human and Tetrahymena telomerase catalytic core. Proc. Natl Acad. Sci. USA 117, 31078–31087 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  43. Hong, K. et al. Tetrahymena telomerase holoenzyme assembly, activation, and inhibition by domains of the p50 central hub. Mol. Cell. Biol. 33, 3962–3971 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Lue, N. F., Chan, J., Wright, W. E. & Hurwitz, J. The CDC13–STN1–TEN1 complex stimulates Pol α activity by promoting RNA priming and primase-to-polymerase switch. Nat. Commun. 5, 5762 (2014).

    Article  ADS  CAS  PubMed  Google Scholar 

  45. Baranovskiy, A. G. et al. Mechanism of concerted RNA–DNA primer synthesis by the human primosome. J. Biol. Chem. 291, 10006–10020 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Kilkenny, M. L. et al. Structural basis for the interaction of SARS-CoV-2 virulence factor nsp1 with DNA polymerase α-primase. Protein Sci. 31, 333–344 (2022).

    Article  CAS  PubMed  Google Scholar 

  47. Coloma, J., Johnson, R. E., Prakash, L., Prakash, S. & Aggarwal, A. K. Human DNA polymerase α in binary complex with a DNA:DNA template–primer. Sci Rep. 6, 23784 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  48. Beseiso, D. et al. The first crystal structures of hybrid and parallel four-tetrad intramolecular G-quadruplexes. Nucleic Acids Res. 50, 2959–2972 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Bryan, T. M. G-quadruplexes at telomeres: friend or foe? Molecules 25, 3686 (2020).

    Article  CAS  PubMed Central  Google Scholar 

  50. Williamson, J. R., Raghuraman, M. K. & Cech, T. R. Monovalent cation-induced structure of telomeric DNA: the G-quartet model. Cell 59, 871–880 (1989).

    Article  CAS  PubMed  Google Scholar 

  51. Lee, W., Tonelli, M. & Markley, J. L. NMRFAM-SPARKY: enhanced software for biomolecular NMR spectroscopy. Bioinformatics 31, 1325–1327 (2015).

    Article  PubMed  Google Scholar 

  52. Herrmann, T., Guntert, P. & Wuthrich, K. Protein NMR structure determination with automated NOE-identification in the NOESY spectra using the new software ATNOS. J. Biomol. NMR 24, 171–189 (2002).

    Article  CAS  PubMed  Google Scholar 

  53. Guntert, P. & Buchner, L. Combined automated NOE assignment and structure calculation with CYANA. J. Biomol. NMR 62, 453–471 (2015).

    Article  PubMed  CAS  Google Scholar 

  54. Shen, Y., Delaglio, F., Cornilescu, G. & Bax, A. TALOS+: a hybrid method for predicting protein backbone torsion angles from NMR chemical shifts. J. Biomol. NMR 44, 213–223 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Shen, Y. et al. Consistent blind protein structure generation from NMR chemical shift data. Proc. Natl Acad. Sci. USA 105, 4685–4690 (2008).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  56. Williamson, M. P. Using chemical shift perturbation to characterise ligand binding. Prog. Nucl. Magn. Reson. Spectrosc. 73, 1–16 (2013).

    Article  CAS  PubMed  Google Scholar 

  57. Jiang, J. et al. The architecture of Tetrahymena telomerase holoenzyme. Nature 496, 187–192 (2013).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  58. Mastronarde, D. N. Automated electron microscope tomography using robust prediction of specimen movements. J. Struct. Biol. 152, 36–51 (2005).

    Article  PubMed  Google Scholar 

  59. Zivanov, J. et al. New tools for automated high-resolution cryo-EM structure determination in RELION-3. eLife 7, e42166 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Zheng, S. Q. et al. MotionCor2: anisotropic correction of beam-induced motion for improved cryo-electron microscopy. Nat. Methods 14, 331–332 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Rohou, A. & Grigorieff, N. CTFFIND4: Fast and accurate defocus estimation from electron micrographs. J. Struct. Biol. 192, 216–221 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Bepler, T. et al. Positive-unlabeled convolutional neural networks for particle picking in cryo-electron micrographs. Nat. Methods 16, 1153–1160 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Punjani, A., Rubinstein, J. L., Fleet, D. J. & Brubaker, M. A. cryoSPARC: algorithms for rapid unsupervised cryo-EM structure determination. Nat. Methods 14, 290–296 (2017).

    Article  CAS  PubMed  Google Scholar 

  64. Kucukelbir, A., Sigworth, F. J. & Tagare, H. D. Quantifying the local resolution of cryo-EM density maps. Nat. Methods 11, 63–65 (2014).

    Article  CAS  PubMed  Google Scholar 

  65. Tan, Y. Z. et al. Addressing preferred specimen orientation in single-particle cryo-EM through tilting. Nat. Methods 14, 793–796 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Pettersen, E. F. et al. UCSF Chimera—a visualization system for exploratory research and analysis. J. Comput. Chem. 25, 1605–1612 (2004).

    Article  CAS  PubMed  Google Scholar 

  67. Emsley, P., Lohkamp, B., Scott, W. G. & Cowtan, K. Features and development of Coot. Acta Crystallogr. D 66, 486–501 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Jumper, J. et al. Highly accurate protein structure prediction with AlphaFold. Nature 596, 583–589 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Holm, L. Using Dali for protein structure comparison. Struct. Bioinformatics 2112, 29–42 (2020).

    Article  CAS  Google Scholar 

  71. Waterhouse, A. M., Procter, J. B., Martin, D. M., Clamp, M. & Barton, G. J. Jalview Version 2—a multiple sequence alignment editor and analysis workbench. Bioinformatics 25, 1189–1191 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Goddard, T. D. et al. UCSF ChimeraX: meeting modern challenges in visualization and analysis. Protein Sci. 27, 14–25 (2018).

    Article  CAS  PubMed  Google Scholar 

  73. Baranovskiy, A. G. et al. Structural basis for inhibition of DNA replication by aphidicolin. Nucleic Acids Res. 42, 14013–14021 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Perera, R. L. et al. Mechanism for priming DNA synthesis by yeast DNA polymerase α. eLife 2, e00482 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by grants from NIH R35GM131901 and NSF MCB2016540 to J.F. and NIH R01GM071940 to Z.H.Z. We acknowledge the use of instruments at the Electron Imaging Center for Nanomachines supported by UCLA and instrumentation grants from NIH (1S10OD018111 and U24GM116792) and NSF (DBI-1338135 and DMR-1548924). The UCLA-DOE NMR core facility is supported in part by US Department of Energy grant DE-AC02-06CH11357 and NIH instrumentation grants S10OD016336 and S10OD025073.

Author information

Authors and Affiliations

Authors

Contributions

Y.H. and H.S. prepared and checked electron microscopy samples. Y.H. collected and analysed cryo-EM data. Y.H. and H.S. built the models. H.C. expressed and purified samples for NMR analysis. H.C., Y.W. and L.S. collected and analysed NMR data. H.S. and B.L. conducted activity assays. B.L. and Y.H. conducted EMSA assays. Z.H.Z. supervised cryo-EM data collection and processing. J.F. supervised all aspects of the project. Y.H. and J.F. made figures and wrote the manuscript, with input from H.S., Y.W. and B.L.

Corresponding author

Correspondence to Juli Feigon.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature thanks the anonymous reviewers for their contribution to the peer review of this work. Peer review 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 Cryo-EM data processing workflow of TtCST in telomerase holoenzyme and evaluations of the final reconstructions.

a, Data processing workflow (detailed in Methods). b, Euler angle distributions of particles used for the final 3.5 Å resolution reconstruction. c, Local resolution evaluation of the 3.5 Å resolution reconstruction shown for the overall map (upper) and for the TtCST–p50 region (lower). d, Plot of the Fourier shell correlation (FSC) as a function of the spatial frequency demonstrating the resolutions of final reconstructions. e, 3D FSC analysis65 of the 3.5 Å resolution reconstruction. Shown are the global FSC (red line), the spread of directional resolution values (area encompassed by the green dotted lines) and the histogram of directional resolutions evenly sampled over the 3D FSC (blue bars). A sphericity (0.5 threshold) of 0.899 indicates almost isotropic angular distribution of resolution (a value of 1 stands for completely isotropic angular distribution). f, FSC coefficient as a function of spatial frequency between model and cryo-EM density map. g, Representative cryo-EM densities (grey and mesh) encasing the related atomic models (coloured sticks and ribbons). h, Superimposition of cryo-EM densities (low-pass filtered to 5 Å) and model of Ctc1 OB-A in complex with p50 CBM.

Extended Data Fig. 2 Structural details of TtCST.

a, Domain organization of TtCST subunits. Invisible regions in the cryo-EM map are shown as dashed boxes. Intermolecular interactions are indicated as grey shading. b, Ribbon diagrams of TtCST subunits/domains with secondary-structure elements labelled. Unmodelled regions are shown as dashed lines. Crystal structure of Ten1–Stn1 OB23 (PDB 5DOI) is shown in grey and overlaid with the cryo-EM structure for comparison. c, Ribbon representation of TtCST structure with individual OB domain coloured as indicated. d, Close-up view of the interface between Ctc1 OB-C and Stn1 OB. Salt bridge and hydrogen-bonding interactions are shown as dashed yellow lines. e, Two arginine sidechains on Stn1 OB (ribbon) clamp D499 on Ctc1 OB-C (electrostatic surface). f, g, Detailed interactions between Stn1 OB and Ten1. h, Close-up views of the cryo-EM densities of the interfaces between CST subunits (Fig. 1e, Extended Data Fig. 2d–g). i, Overall view of the interface between TtCST and TERT–TER. j–l, Close-up views of interactions between TtCST and TERT–TER as indicated in dashed boxes in h. These interactions stabilize TtCST in the predominant conformation.

Extended Data Fig. 3 NMR spectra and structural study of Ctc1 OB-A with p50 peptide.

a, Assigned 1H-15N HSQC spectrum of 15N-labelled Ctc1 OB-A (residues 1-183) in the presence of unlabelled p50 peptide (residues 228-250). Inset shows the expanded central region of the spectrum. b, Superimposed 1H-15N HSQC spectra of 15N-labelled Ctc1 OB-A in the presence (yellow) and absence (gray) of unlabelled p50 peptide. Signals from the same residues with chemical shift differences of more than 0.25 ppm are connected by dashed arrows. Signals from residues 68-70 that only appear in the presence of p50 peptide are labelled with asterisks. c, Chemical shift perturbation (CSP) index of 15N-labelled Ctc1 OB-A upon binding p50 peptide. Magenta box indicates the region that is shown in Fig. 2d. d, Chemical-shift-based secondary-structure score of Ctc1 OB-A in the absence (grey) and presence (yellow) of p50 peptide. The scores are determined using TALOS+ (ref. 54). Top and bottom edges of each bar represent the probabilities of each residue assigned to be α helix and β sheet, respectively. The secondary structure of Ctc1 OB-A observed in the cryo-EM structure is shown below for comparison. e, Plot of long range (greater than 5 residues) 1H-1H NOE restraints observed within Ctc1 OB-A. Residues with pairwise NOE restraint(s) are connected by a link. Links are colour coded as indicated based on the number of NOE restraints between the two connected residues.

Extended Data Fig. 4 Identifying the 'invisible' interface between Ctc1 OB-A and p50 peptide using NMR methods.

a, Schematic diagram of p50 and constructs of p50 peptide. The N-terminal 30 kDa and 25 kDa fragments of p50 are labelled as p50N30 and p50N25, respectively. Previous biochemical study showed that p50N30 could bind Ctc1, whereas p50N25 could not43. The cryo-EM structure of p50 ends at residue 208 (Fig. 2b). On the basis of these facts, a series of p50 peptides in the range of residues 213-255 were designed to explore additional interface between p50 and Ctc1 OB-A that are 'invisible' in the cryo-EM structure. b, NMR binding study of p50 peptides with Ctc1 OB-A. Two regions of 1H-15N HSQC spectra of 15N-labelled Ctc1 OB-A in the absence (apo) and presence of unlabelled p50 peptides were shown. Chemical shifts of T71 and A73 were chosen to illustrate the binding process in this and the following panels, c and d. p50228-250 peptide is determined to be the optimal construct and was used for other NMR studies presented here. c, Titration series of p50 peptide into 15N-labelled Ctc1 OB-A. The binding is in the slow exchange regime and saturated at 1:1 stoichiometry. d, Truncations of two unstructured loops (residues 38-49 and 131-146) of Ctc1 OB-A individually have no effect on its binding with p50 peptide. e, Secondary-structure score of p50228-250 in the presence of Ctc1 OB-A. f, CSP index of p50 peptide upon binding Ctc1 OB-A. 1H-15N HSQC spectra shown in Fig. 2c were used for the CSP calculation. g, Model of the interactions between Ctc1 OB-A and p50. CS-Rosetta models of p50228-250 are shown in the grey box with arrows pointing to the binding surface on Ctc1 OB-A. Unstructured linkers are shown as dashed lines.

Extended Data Fig. 5 Characterization of purified Tetrahymena PolαPrim samples and their assembly with telomerase holoenzyme.

a, Size-exclusion chromatography (SEC) profile (left) and SDS-PAGE gel (right) of Polα. b, Representative 2D-class averages of Polα particles obtained from negative-stain EM images. c, SEC profile (left) and SDS-PAGE gel (right) of PolαPrim. d, Representative 2D-class averages of PolαPrim particles obtained from negative-stain EM images. e, Silver-stained SDS-PAGE gel of affinity purified telomerase–Polα (lane 2) shows that Polα can bind telomerase in the absence of Primase. Telomerase (lane 1) and Polα (lane 3) samples were loaded on the same gel for comparison. f, Silver-stained SDS-PAGE gel of affinity purified telomerase–PolαPrim samples shows assembly of the complex with or without sstDNA. g, Representative 2D-class averages of affinity purified telomerase–PolαPrim obtained from negative-stain EM images. Densities are assigned on the basis of the cryo-EM structure (Fig. 3a) obtained with the same batch of sample. Smeared densities (red arrows) are observed near POLA1core in several classes, so we were able to assign them to the PolαPrim platform, which comprises POLA2, POLA1CTD, PRIM2N, and PRIM1. h, Representative 2D-class averages of telomerase particles shown for comparison with g. i, Activity assays of telomerase-PolαPrim (lanes 1-4) and PolαPrim alone (lane 5). Direct telomerase activity assays were conducted for G-strand synthesis alone in the presence of dTTP and dGTP (lanes 1 and 3). PolαPrim activity assays were conducted for C-strand synthesis alone in the presence of ATP, CTP, dATP and dCTP (lanes 2, 4 and 5). 32P-dGTP and 32P-dCTP were used to label the G-strand and C-strand products, respectively. A longer exposure is shown for lane 5 so that products can be seen. RC, recovery control. All lanes are from a single gel. j, Activity assays of C-strand synthesis (lane 1) relative to that in 50 mM LiCl (lane 2), 50 mM NaCl (lane 3), or 50 mM KCl (lane 4). For lanes 2-4, the DNA templates were incubated in assay buffer containing 50 mM of the indicated cations on ice for 30 min before the reaction.

Extended Data Fig. 6 Cryo-EM structure determination of Tetrahymena telomerase–PolαPrim complex.

a, Data-processing workflow (detailed in Methods). b, Resolution of final reconstructions determined by gold-standard FSC at the 0.143 criterion. c, Particle distribution (upper) and local resolution evaluation (lower) of the 2.9 Å resolution reconstruction of telomerase core. d, Particle distribution (upper) and local resolution evaluation (lower) of the 4.2 Å resolution reconstruction of TtCST–POLA1core. e, 3D FSC analysis65 of the 2.9 Å resolution reconstruction of telomerase core (left) and the 4.2 Å resolution reconstruction of TtCST–POLA1core (right). For each reconstruction, the global FSC (red line), the spread of directional resolution values (area encompassed by the green dotted lines) and the histogram of directional resolutions evenly sampled over the 3D FSC (blue bars) are shown. A sphericity of 0.958 was determined for telomerase core (left), indicating almost isotropic angular distribution of resolution. A sphericity of 0.736 was determined for TtCST-POLA1core (right), suggesting slightly anisotropic angular distribution of resolution. f, FSC curves for refined models versus the corresponding cryo-EM density maps. g-j, Representative cryo-EM densities (transparency surface) encasing the related atomic models (colour sticks and ribbons) for telomerase-core RNP (g), CST (h), Stn1 WH-WH (i) and POLA1core (j).

Extended Data Fig. 7 Cryo-EM structure determination of PolαPrim.

a, Data processing workflow (detailed in Methods). b, Representative 2D-class averages of PolαPrim particles obtained from cryo-EM images. The two classes shown in Fig. 3e are labelled with red boxes. c, Resolution of final reconstructions determined by gold-standard FSC at the 0.143 criterion. d, Euler angle distributions of particles used for the final reconstructions. e, Local resolution evaluations of the final reconstructions. f, FSC coefficients as a function of spatial frequency between model and cryo-EM density maps. The composite map is generated using Phenix69 with two focused refined maps (detailed in Methods). For the full map and the composite map, the complete model was used to calculate the FSCs. For the two focused refined maps, only corresponding regions of the model were used to calculate the FSCs. g, Representative cryo-EM densities (grey mesh) encasing the related atomic models (coloured sticks and ribbons).

Extended Data Fig. 8 Structural conservation of Tetrahymena PolαPrim.

a, Superposition of Tetrahymena (Tt), human and yeast POLA1core structures45,46,47,73,74 shown in an overall view. b, Structural comparison of PolαPrim platform of Tt and human45,46. The structures were superposed based on POLA2–POLA1CTD (left) or PRIM1 (right). Arrows indicate dynamics of the unaligned regions. PRIM2C in human structures are shown as transparent ribbons. PRIM2C is not observed in the Tt structure. c, Structures of PolαPrim in an autoinhibited conformation (left, modelled on the basis of PDB 5EXR45) and an active conformation (right, modelled on the basis of a low-resolution cryo-EM map in Extended Data Fig. 7a). The DNA-DNA duplexes on POLA1core were modelled on the basis of PDB 5IUD47. In the autoinhibited conformation (left), the active site on POLA1core is sterically blocked by POLA1CTD and POLA2 for DNA entry. In the active conformation (right), dynamics of subunits are indicated with arrows. d-e, Superposition of Tt, human and yeast POLA1core structures for the regions that are on the interface with TtCST. Conserved domains/motifs are labeled as indicated. The β11-β12 hairpin in Tt POLA1core is longer than those in human and yeast (d). The α19 is structured only in Tt POLA1 when binding TtCST (e). f, Close-up views of the interface between Tt POLA1core α19 (ribbon) and TtCST (surface/electrostatic surface). In the lower panel, locations of positively and negatively charged residues on α19 are indicated using blue and red balls, respectively. g, Sequence-conservation analysis of the β11-β12 hairpin and α19 of POLA1. Charged residues on α19 are indicated with black arrows. h, Close-up view of the interface between POLA1core and TtCST with sstDNA. Cryo-EM densities are shown as transparent surface. The template entry port formed by POLA1core NTD and Exo and Ctc1 OB-C is indicated by a cycle. i, Path of sstDNA in the cryo-EM structure of Tt telomerase–PolαPrim. The sstDNA binds in the C-shape cleft of Ctc1 OB-C with its 5' side, while its 3' side passes through the template entry port to the active site of POLA1core (left). A G-quadruplex (GQ) formed by four Tt telomere repeats (modelled on the basis of PDB 7JKU48) is observed on a positively charged DNA binding surface of POLA1core between the palm and thumb (right). j, Superimposition of the GQ structure and cryo-EM density. Weak density of sstDNA can be observed connecting the sstDNA on Ctc1 OB-C to the GQ.

Extended Data Fig. 9 Comparison of TtCST and human CST.

a, Domain diagrams of TtCST and human CST. b, Structural homology analysis of individual OB domains of TtCtc1 (OB-A to -C) and human CTC1 (OB-A to -G) using the Dali sever70. On the basis of the resulted pairwise Z-scores, TtCtc1 OB-B and OB-C are identified as homologues of human CTC1 OB-F and OB-G, respectively. c, Structural comparison of individual domains from TtCST (colour) with corresponding domains from human CST (grey). Structures of WH-WH domains of Tt Stn1 and human STN1 were superposed based on WH1 or WH2 domain. The relative orientation of the two WH domains is different between Tt and human. d, The interface between Stn1 WH-WH and Ctc1 in the cryo-EM structure of Tt telomerase–PolαPrim. Cryo-EM densities are shown as transparent surfaces. An previously unstructured loop of Ctc1 OB-C (Extended Data Fig. 2b) partially forms an α helix (α520-540) and contributes to the interface with Stn1 WH-WH. e, Comparison of DNA binding sites on TtCtc1 (colour) and human CTC1 (grey). Conserved residues located on the DNA binding interface are shown as sticks. Cryo-EM densities of TtCtc1 are shown as transparent surfaces. In the decameric structure of human CST19 (PDB 6W6W), sstDNA primarily binds on CTC1 OB-F. However, in TtCST, the equivalent sstDNA binding site on OB-B is partially occluded by a helix (α6) that is part of an unstructured loop in hCTC1 OB-F. The helix α6 abuts TERT in TtCST without PolαPrim (Extended Data Fig. 2k) and Stn1 WH2 when PolαPrim is bound (as shown in d). f, Sequence conservation analysis of Ctc1 residues on the DNA binding interface. Residues shown in e are indicated with black arrows. Conserved cysteines in the zinc ribbon motifs are indicated with pink arrows. g, SEC profile and SDS-PAGE gel of TtCST–p50 co-expressed in Sf9 cells. Gel samples are from the peak fractions of the SEC profile as indicated. h, EMSA of purified wild-type TtCST with d(GTTGGG)n, where n = 3, 5 or 10. i, Substitutions of TtCtc1 OB-B conserved residues K303E/K306E/F308A and F264A/Y268A substantially decrease d(GTTGGG)5 binding, as indicated by EMSAs. These results suggest that the binding site on TtCtc1 OB-B might be accessible to sstDNA in free TtCST where neither TERT nor Stn1 WH-WH stabilize helix α6. Wedges indicate two-fold dilution of TtCST. The first lane of each gel is a TtCST-free control. j, Quantifications of fraction of bound DNA for all the independent EMSA experiments with TtCST WT and variants as indicated (n = 3 biological replicates). KD values were determined as described in Methods. k, Effect of TtCST residue substitutions on d(GTTGGG)5 binding. Data are mean ± s.d. from n = 3 biological replicates shown in j. *P = 0.04, **P = 0.009, ****P < 0.0001; one-tailed unpaired t-tests.

Extended Data Table 1 Cryo-EM data collection, refinement and validation statistics

Supplementary information

Supplementary Fig. 1

Source images for all data obtained from electrophoretic gels.

Reporting Summary

Peer Review File

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

He, Y., Song, H., Chan, H. et al. Structure of Tetrahymena telomerase-bound CST with polymerase α-primase. Nature 608, 813–818 (2022). https://doi.org/10.1038/s41586-022-04931-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-022-04931-7

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