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 architecture of the spliceosomal U4/U6.U5 tri-snRNP

Abstract

U4/U6.U5 tri-snRNP is a 1.5-megadalton pre-assembled spliceosomal complex comprising U5 small nuclear RNA (snRNA), extensively base-paired U4/U6 snRNAs and more than 30 proteins, including the key components Prp8, Brr2 and Snu114. The tri-snRNP combines with a precursor messenger RNA substrate bound to U1 and U2 small nuclear ribonucleoprotein particles (snRNPs), and transforms into a catalytically active spliceosome after extensive compositional and conformational changes triggered by unwinding of the U4 and U6 (U4/U6) snRNAs. Here we use cryo-electron microscopy single-particle reconstruction of Saccharomyces cerevisiae tri-snRNP at 5.9 Å resolution to reveal the essentially complete organization of its RNA and protein components. The single-stranded region of U4 snRNA between its 3′ stem–loop and the U4/U6 snRNA stem I is loaded into the Brr2 helicase active site ready for unwinding. Snu114 and the amino-terminal domain of Prp8 position U5 snRNA to insert its loop I, which aligns the exons for splicing, into the Prp8 active site cavity. The structure provides crucial insights into the activation process and the active site of the spliceosome.

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: Overview of the U4/U6.U5 tri-snRNP structure with its protein and RNA components modelled into cryo-EM density.
Figure 2: Prp8 in tri-snRNP.
Figure 3: The snRNA components of U4/U6.U5 tri-snRNP.
Figure 4: Structure of Snu114 in tri-snRNP.
Figure 5: Brr2 mode of unwinding.
Figure 6: Insights into activation mechanism and the active site of the spliceosome.

Similar content being viewed by others

Accession codes

Primary accessions

Electron Microscopy Data Bank

Data deposits

3D cryo-EM density maps of yeast U4/U6.U5 tri-snRNP have been deposited with the Electron Microscopy Database under accession number EMD-2966.

References

  1. Will, C. L. & Lührmann, R. Spliceosome structure and function. Cold Spring Harb. Perspect. Biol. 3, a003707 (2011)

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Chan, S. P. & Cheng, S. C. The Prp19-associated complex is required for specifying interactions of U5 and U6 with pre-mRNA during spliceosome activation. J. Biol. Chem. 280, 31190–31199 (2005)

    CAS  PubMed  Google Scholar 

  3. Fabrizio, P. et al. The evolutionarily conserved core design of the catalytic activation step of the yeast spliceosome. Mol. Cell 36, 593–608 (2009)

    CAS  PubMed  Google Scholar 

  4. Fica, S. M. et al. RNA catalyses nuclear pre-mRNA splicing. Nature 503, 229–234 (2013)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  5. Newman, A. J. & Norman, C. U5 snRNA interacts with exon sequences at 5′ and 3′ splice sites. Cell 68, 743–754 (1992)

    CAS  PubMed  Google Scholar 

  6. Sontheimer, E. J. & Steitz, J. A. The U5 and U6 small nuclear RNAs as active site components of the spliceosome. Science 262, 1989–1996 (1993)

    ADS  CAS  PubMed  Google Scholar 

  7. Stevens, S. W. et al. Biochemical and genetic analyses of the U5, U6, and U4/U6.U5 small nuclear ribonucleoproteins from Saccharomyces cerevisiae. RNA 7, 1543–1553 (2001)

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Gottschalk, A. et al. Identification by mass spectrometry and functional analysis of novel proteins of the yeast [U4/U6.U5] tri-snRNP. EMBO J. 18, 4535–4548 (1999)

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Turner, I. A., Norman, C. M., Churcher, M. J. & Newman, A. J. Dissection of Prp8 protein defines multiple interactions with crucial RNA sequences in the catalytic core of the spliceosome. RNA 12, 375–386 (2006)

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Galej, W. P., Oubridge, C., Newman, A. J. & Nagai, K. Crystal structure of Prp8 reveals active site cavity of the spliceosome. Nature 493, 638–643 (2013)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  11. Fabrizio. P., Laggerbauer, B., Lauber, J., Lane, W. S. & Lührmann, R. An evolutionarily conserved U5 snRNP-specific protein is a GTP-binding factor closely related to the ribosomal translocase EF-2. EMBO J. 16, 4092–4106 (1997)

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Small, E. C., Leggett, S. R., Winans, A. A. & Staley, J. P. The EF-G-like GTPase Snu114p regulates spliceosome dynamics mediated by Brr2p, a DExD/H box ATPase. Mol. Cell 23, 389–399 (2006)

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Bartels. C., Urlaub, H., Lührmann, R. & Fabrizio P. Mutagenesis suggests several roles of Snu114p in pre-mRNA splicing. J. Biol. Chem. 278, 28324–28334 (2003)

    CAS  PubMed  Google Scholar 

  14. Raghunathan, P. L. & Guthrie, C. RNA unwinding in U4/U6 snRNPs requires ATP hydrolysis and the DEIH-box splicing factor Brr2. Curr. Biol. 8, 847–855 (1998)

    CAS  PubMed  Google Scholar 

  15. Laggerbauer, B., Achsel, T. & Lührmann, R. The human U5–200kD DEXH-box protein unwinds U4/U6 RNA duplices in vitro. Proc. Natl Acad. Sci. USA 95, 4188–4192 (1998)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  16. Liu, S., Rauhut, R., Vornlocher, H. P. & Lührmann, R. The network of protein-protein interactions within the human U4/U6.U5 tri-snRNP. RNA 12, 1418–1430 (2006)

    CAS  PubMed  PubMed Central  Google Scholar 

  17. van Nues, R. W. & Beggs, J. D. Functional contacts with a range of splicing proteins suggest a central role for Brr2p in the dynamic control of the order of events in spliceosomes of Saccharomyces cerevisiae. Genetics 157, 1451–1467 (2001)

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Sander, B. et al. Organization of core spliceosomal components U5 snRNA loop I and U4/U6 Di-snRNP within U4/U6.U5 Tri-snRNP as revealed by electron cryomicroscopy. Mol. Cell 24, 267–278 (2006)

    CAS  PubMed  Google Scholar 

  19. Häcker, I. et al. Localization of Prp8, Brr2, Snu114 and U4/U6 proteins in the yeast tri-snRNP by electron microscopy. Nature Struct. Mol. Biol. 15, 1206–1212 (2008)

    Google Scholar 

  20. McMullan, G. et al. Experimental observation of the improvement in MTF from backthinning a CMOS direct electron detector. Ultramicroscopy 109, 1144–1147 (2009)

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Li, X. et al. Electron counting and beam-induced motion correction enable near-atomic-resolution single-particle cryo-EM. Nature Methods 10, 584–590 (2013)

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Scheres, S. H. A Bayesian view on cryo-EM structure determination. J. Mol. Biol. 415, 406–418 (2012)

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Bai, X. C., McMullan, G. & Scheres, S. H. How cryo-EM is revolutionizing structural biology. Trends Biochem. Sci. 40, 49–57 (2015)

    CAS  PubMed  Google Scholar 

  24. Scheres, S. H. Beam-induced motion correction for sub-megadalton cryo-EM particles. Elife 3, e03665 (2014)

    PubMed  PubMed Central  Google Scholar 

  25. Scheres, S. H. & Chen, S. Prevention of overfitting in cryo-EM structure determination. Nature Methods 9, 853–854 (2012)

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Jørgensen, R. et al. Two crystal structures demonstrate large conformational changes in the eukaryotic ribosomal translocase. Nature Struct. Biol. 10, 379–385 (2003)

    PubMed  Google Scholar 

  27. Grainger, R. J., Barrass, J. D., Jacquier, A., Rain, J. C. & Beggs, J. D. Physical and genetic interactions of yeast Cwc21p, an ortholog of human SRm300/SRRM2, suggest a role at the catalytic center of the spliceosome. RNA 15, 2161–2173 (2009)

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Dix, I., Russell, C. S., O'Keefe, R. T., Newman, A. J. & Beggs, J. D. Protein-RNA interactions in the U5 snRNP of Saccharomyces cerevisiae. RNA 4, 1675–1686 (1998)

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Reuter, K., Nottrott, S., Fabrizio, P., Lührmann, R. & Ficner, R. Identification, characterization and crystal structure analysis of the human spliceosomal U5 snRNP-specific 15 kD protein. J. Mol. Biol. 294, 515–525 (1999)

    CAS  PubMed  Google Scholar 

  30. Maeder, C., Kutach, A. K. & Guthrie, C. ATP-dependent unwinding of U4/U6 snRNAs by the Brr2 helicase requires the C terminus of Prp8. Nature Struct. Mol. Biol. 16, 42–48 (2009)

    CAS  Google Scholar 

  31. Nguyen, T. H. D. et al. Structural basis of Brr2-Prp8 interactions and implications for U5 snRNP biogenesis and the spliceosome active site. Structure 21, 910–919 (2013)

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Mozaffari-Jovin, S. et al. Inhibition of RNA helicase Brr2 by the C-terminal tail of the spliceosomal protein Prp8. Science 341, 80–84 (2013)

    ADS  CAS  PubMed  Google Scholar 

  33. Liu, S. et al. Binding of the human Prp31 Nop domain to a composite RNA-protein platform in U4 snRNP. Science 316, 115–120 (2007)

    ADS  CAS  PubMed  Google Scholar 

  34. Schultz, A., Nottrott, S., Hartmuth, K. & Lührmann, R. RNA structural requirements for the association of the spliceosomal hPrp31 protein with the U4 and U4atac small nuclear ribonucleoproteins. J. Biol. Chem. 281, 28278–28286 (2006)

    CAS  PubMed  Google Scholar 

  35. Ayadi, L. et al. Functional and structural characterization of the Prp3 binding domain of the yeast Prp4 splicing factor. J. Mol. Biol. 284, 673–687 (1998)

    CAS  PubMed  Google Scholar 

  36. Korneta, I., Magnus, M. & Bujnicki, J. M. Structural bioinformatics of the human spliceosomal proteome. Nucleic Acids Res. 40, 7046–7065 (2012)

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Nottrott, S., Urlaub, H. & Lührmann, R. Hierarchical, clustered protein interactions with U4/U6 snRNA: a biochemical role for U4/U6 proteins. EMBO J. 21, 5527–5538 (2002)

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Galisson, F. & Legrain, P. The biochemical defects of prp4–1 and prp6–1 yeast splicing mutants reveal that the PRP6 protein is required for the accumulation of the [U4/U6.U5] tri-snRNP. Nucleic Acids Res. 21, 1555–1562 (1993)

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Makarov, E. M., Makarova, O. V., Achsel, T. & Lührmann, R. The human homologue of the yeast splicing factor prp6p contains multiple TPR elements and is stably associated with the U5 snRNP via protein-protein interactions. J. Mol. Biol. 298, 567–575 (2000)

    CAS  PubMed  Google Scholar 

  40. Boon, K. L. et al. Prp8 mutations that cause human retinitis pigmentosa lead to a U5 snRNP maturation defect in yeast. Nature Struct. Mol. Biol. 14, 1077–1083 (2007)

    CAS  Google Scholar 

  41. Mozaffari-Jovin, S. et al. The Prp8 RNase H-like domain inhibits Brr2-mediated U4/U6 snRNA unwinding by blocking Brr2 loading onto the U4 snRNA. Genes Dev. 26, 2422–2434 (2012)

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Hahn, D., Kudla, G., Tollervey, D. & Beggs, J. D. Brr2p-mediated conformational rearrangements in the spliceosome during activation and substrate repositioning. Genes Dev. 26, 2408–2421 (2012)

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Büttner, K., Nehring, S. & Hofner, K. P. Structural basis for DNA duplex separation by a superfamily‐2 helicase. Nature Struct. Mol. Biol. 14, 647–652 (2007)

    Google Scholar 

  44. Tourigny, D. S., Fernández, I. S., Kelley, A. C. & Ramakrishnan, V. Elongation factor G bound to the ribosome in an intermediate state of translocation. Science 340, 1235490 (2013)

    PubMed  Google Scholar 

  45. Lin. J, Gagnon, M. G., Bulkley, D. & Steitz, T. A. Conformational changes of elongation factor G on the ribosome during tRNA translocation. Cell 160, 219–227 (2015)

    PubMed  PubMed Central  Google Scholar 

  46. Kuhn, A. N. & Brow, D. A. Suppressors of a cold-sensitive mutation in yeast U4 RNA define five domains in the splicing factor Prp8 that influence spliceosome activation. Genetics 155, 1667–1682 (2000)

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Li, Z. & Brow, D. A. A spontaneous duplication in U6 spliceosomal RNA uncouples the early and late functions of the ACAGA element in vivo. RNA 2, 879–894 (1996)

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Toor, N., Keating, K. S., Taylor, S. D. & Pyle, A. M. Crystal structure of a self-spliced group II intron. Science 320, 77–82 (2008)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  49. Fica, S. M., Mefford, M. A., Piccirilli, J. A. & Staley, J. P. Evidence for a group II intron-like catalytic triplex in the spliceosome. Nature Struct. Mol. Biol. 21, 464–471 (2013)

    Google Scholar 

  50. Sharp, P. A. Five easy pieces. Science 254, 663 (1991)

    ADS  CAS  PubMed  Google Scholar 

  51. Schreieck, A. et al. RNA polymerase II termination involves C-terminal domain tyrosine dephosphorylation by CPF subunit Glc7. Nature Struct. Mol. Biol. 21, 175–179 (2014)

    CAS  Google Scholar 

  52. Mindell, J. A. & Grigorieff, N. Accurate determination of local defocus and specimen tilt in electron microscopy. J. Struct. Biol. 142, 334–347 (2003)

    PubMed  Google Scholar 

  53. Elmlund, H., Elmlund, D. & Bengio, S. PRIME: probabilistic initial 3D model generation for single-particle cryo-electron microscopy. Structure 21, 1299–1306 (2013)

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  55. Chen, S. et al. High-resolution noise substitution to measure overfitting and validate resolution in 3D structure determination by single particle electron cryomicroscopy. Ultramicroscopy 135, 24–35 (2013)

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Scheres, S. H. W., Nuñez-Ramirez, R., Sorzano, C. O. S., Carazo, J. M. & Marabini, R. Image processing for electron microscopy single-particle analysis using Xmipp. Nature Protocols 3, 977–990 (2008)

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Tang, G. et al. EMAN2: an extensive image processing suite for electron microscopy. J. Struct. Biol. 157, 38–46 (2007)

    CAS  PubMed  Google Scholar 

  58. Goddard, T. D., Huang, C. C. & Ferrin, T. E. Visualizing density maps with UCSF Chimera. J. Struct. Biol. 157, 281–287 (2007)

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Zhang, Y. I-TASSER server for protein 3D structure prediction. BMC Bioinformatics 9, 40 (2008)

    PubMed  PubMed Central  Google Scholar 

  61. Malmström, L. et al. Superfamily assignments for the yeast proteome through integration of structure prediction with the gene ontology. PLoS Biol. 5, e76 (2007)

    PubMed  PubMed Central  Google Scholar 

  62. Wu, X. H., Chen, R. C., Gao, Y. & Wu, Y. D. The effect of Asp-His-Ser/Thr-Trp tetrad on the thermostability of WD40-repeat proteins. Biochemistry 49, 10237–10245 (2010)

    CAS  PubMed  Google Scholar 

  63. Rother, M. et al. ModeRNA server: an online tool for modeling RNA 3D structures. Bioinformatics 27, 2441–2442 (2011)

    CAS  PubMed  Google Scholar 

  64. Dobbyn, H. C. et al. Analysis of pre-mRNA and pre-rRNA processing factor Snu13p structure and mutants. Biochem. Biophys. Res. Commun. 360, 857–862 (2007)

    CAS  PubMed  Google Scholar 

  65. Leung, A. K., Nagai, K. & Li, J. Structure of the spliceosomal U4 snRNP core domain and its implication for snRNP biogenesis. Nature 473, 536–539 (2011)

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  66. Chanfreau, G., Elela, S. A., Ares, M., Jr & Guthrie, C. Alternative 3′-end processing of U5 snRNA by RNase III. Genes Dev. 11, 2741–2751 (1997)

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Zhou, L. et al. Crystal structures of the Lsm complex bound to the 3′end sequence of U6 small nuclear RNA. Nature 506, 116–120 (2014)

    ADS  CAS  PubMed  Google Scholar 

  68. Query, C. C. & Konarska, M. M. Suppression of multiple substrate mutations by spliceosomal prp8 alleles suggests functional correlations with ribosomal ambiguity mutants. Mol. Cell 14, 343–354 (2004)

    CAS  PubMed  Google Scholar 

  69. Umen, J. G. & Guthrie, C. Mutagenesis of the yeast gene PRP8 reveals domains governing the specificity and fidelity of 3′ splice site selection. Genetics 143, 723–739 (1996)

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Liu, L., Query, C. C. & Konarska, M. M. Opposing classes of prp8 alleles modulate the transition between the catalytic steps of pre-mRNA splicing. Nature Struct. Mol. Biol. 14, 519–526 (2007)

    CAS  Google Scholar 

  71. Dagher, S. F. & Fu, X. D. Evidence for a role of Sky1p-mediated phosphorylation in 3′ splice site recognition involving both Prp8 and Prp17/Slu4. RNA 7, 1284–1297 (2001)

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Ben-Yehuda, S. et al. Extensive genetic interactions between PRP8 and PRP17/CDC40, two yeast genes involved in pre-mRNA splicing and cell cycle progression. Genetics 154, 61–71 (2000)

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Collins, C. A. & Guthrie, C. Allele-specific genetic interactions between Prp8 and RNA active site residues suggest a function for Prp8 at the catalytic core of the spliceosome. Genes Dev. 13, 1970–1982 (1999)

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Siatecka, M., Reyes, J. L. & Konarska, M. M. Functional interactions of Prp8 with both splice sites at the spliceosomal catalytic center. Genes Dev. 13, 1983–1993 (1999)

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Kuhn, A. N., Li, Z. & Brow, D. A. Splicing factor Prp8 governs U4/U6 RNA unwinding during activation of the spliceosome. Mol. Cell 3, 65–75 (1999)

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank S. Chen, G. McMullan, J. Grimmett and T. Darling for smooth running of the EM and computing facilities; P. da Fonseca, N. Unwin, I. Sanchez Fernandez, A. Amunts, P. Emsley, G. Murshudov and A. Brown for advice; A. Easter and L. Passmore for reagents; M. Skehel for mass spectrometry; and J. Li, Y. Kondo and the members of the spliceosome group for help and advice throughout the project. We are grateful to R. Henderson, D. Barford, S. Fica, P.-C. Lin and L. Strittmatter for critical reading of the manuscript. We thank V. Ramakrishnan, J. Löwe and R. Henderson for their continuing support and encouragements. T.H.D.N. was supported in part by a Herchel Smith Research Studentship. X.-c.B. was supported by a European Union Marie Curie Fellowship. The project was supported by the Medical Research Council (MC_U105184330 to K.N. and MC_UP_A025_1013 to S.H.W.S.).

Author information

Authors and Affiliations

Authors

Contributions

T.H.D.N. developed the purification procedure for yeast tri-snRNP, prepared EM grids, collected all EM images, processed data, calculated the maps and built and fitted most of the components into the map. W.P.G. built most of the unknown components and made essential contributions to sequence analysis, homology modelling and model fitting. X.-c.B. helped T.H.D.N. with image processing and map calculation. C.G.S. guided T.H.D.N. with EM sample preparation and data collection. A.J.N. produced the Brr2 TAPS-tagged strain and contributed to the project through his knowledge of yeast spliceosome. T.H.D.N. and W.P.G prepared all illustrations. T.H.D.N prepared the video. S.H.W.S. carried out multi-body refinement and oversaw the EM analysis. K.N. initiated and orchestrated the project. T.H.D.N., W.P.G., A.J.N. and K.N. interpreted the results and wrote the paper with crucial contribution from all other authors.

Corresponding authors

Correspondence to Thi Hoang Duong Nguyen or Kiyoshi Nagai.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 U4/U6.U5 tri-snRNP sample used for this study.

a, Coomassie-blue-stained SDS–PAGE gel showing protein composition of the purified tri-snRNP. U5-, U4/U6- and tri-snRNP-specific proteins are labelled in blue, red and teal, respectively. Sm proteins present in both U5 and U4/U6 are in black. b, Toluidine-blue-stained denaturing acrylamide (9%) gel showing RNA compositions. c, Electron cryo-micrograph of tri-snRNP where the carbon-coated grid was discharged in N-amylamine. d, e, Reference-free two-dimensional class averages of a data set collected on a grid discharged in air and N-amylamine, respectively.

Extended Data Figure 2 Classification and refinement procedures used in this study.

A total of 367,327 particles were subjected to reference-free 2D classification. A subset of 347,241 particles from good 2D classes was selected for 3D classification using an initial model obtained from SIMPLE-PRIME53, which was low-pass filtered to 60 Å. The data were divided into four 3D classes, two of which (a total of 179,079 particles) showed better features and were combined for refinement. This resulted in a 7.6 Å reconstruction. To further improve the reconstruction, these particles were subjected to beam-induced motion correction (particle polishing)24. Refinement of these polished particles with a soft mask around the rigid part of the map (as indicated by the red envelope) yielded a 5.9 Å reconstruction while refinement with a mask around the whole map yielded a 6.4 Å reconstruction. The polished particles were also subject to further 3D classification with a finer angular sampling of 1.8°. The most populated class (47,674 particles), which also has the best rotational accuracy, was refined with a soft mask around the whole density. This resulted in a 7.0 Å reconstruction. In this study, the 5.9 Å reconstruction was used for subsequent biological interpretation. All steps were performed in RELION22 unless otherwise stated.

Extended Data Figure 3 CryoEM maps and tilt-pair validation.

a, CryoEM density of the whole tri-snRNP at 5.9 Å resolution by ‘gold standard’ Fourier shell correlation (FSC) of 0.143 criterion at two different contour levels. The high contour map (gold) shows well-resolved densities for protein and RNA helices and flat densities for β-sheets. The low contour map (silver) shows densities for the more flexible head and arm. The map was sharpened by a B-factor of −214 Å2 and low-pass filtered to 5.9 Å as determined by RELION. b, The unsharpened full map of tri-snRNP. c, The map resulting from multi-body refinement, in which tri-snRNP is divided into four parts: the head, body, arm and foot. This resulted in better density for the arm domain (indicated by red circles), which is at 20 Å resolution. d, Tilt-pair validation plot for tri-snRNP. This was obtained from 1,196 particles from 32 micrograph pairs, imaged at 0° and 10° tilt angles. The position of each dot represents the direction and the amount of tilting for a particle pair in polar coordinates. Blue dots correspond to in-plane tilt transformations; red and purple dots correspond to out-of-plane tilt transformations. Blue dots cluster in the same region of the plot at a tilt angle of approximately 10° as indicated by the red circle.

Extended Data Figure 4 Resolution estimation of tri-snRNP map.

a, Local resolution of the tri-snRNP map estimated by ResMap using the colour scheme shown in panel c. b, Local resolution of the tri-snRNP map calculated by ‘gold-standard’ FSC. For each component of the map that we modelled protein/RNA components, a soft mask (with a 30-pixel soft edge) surrounding the region of interest was prepared and used for FSC calculations. Convolution effects of the masks on the FSC curves were corrected using high-resolution noise substitution55. Resolution was estimated at FSC = 0.143. Local resolution for the unmodelled region of the map (in red) was not estimated. c, Local resolution of model versus map. The map of each modelled component was extracted from the map using a soft mask (with a 5-pixel soft edge) surrounding the component. The model was converted into density by EMAN57. FSC of model versus map was calculated using Xmipp56. The map is coloured according to resolution estimates based on a FSC threshold of 0.25. The lower resolution estimates from the FSC of model versus map compared to the estimates from ResMap and the gold-standard FSCs are explained by the nature of our models. Because of the limited resolution of our map, we did not perform full atomic refinement, but placed known crystal structures and homology models as rigid bodies in the map. d, Gold-standard FSC curves for the whole tri-snRNP map and some of its components calculated as described in b. e, FSC curves of model versus map for the whole model and some of the components. f, The full tri-snRNP map in which portions of the structure produced from crystal structures, homology modelling and de novo building or unmodelled are coloured as indicated.

Extended Data Figure 5 Fitting of protein components into tri-snRNP map.

a, Prp8(885–2,413) crystal structure10 (PDB 4I43, green) and additional helices built de novo assigned to the N terminus of Prp8 (blue). b, Brr2–Jab1/MPN complex31 (PDB 4BGD). c, Snu114 homology model based on EF2 (ref. 26). d, The Prp6 TPR motifs built into the tri-snRNP map. e, U5 Sm proteins (grey) with Sm site (blue) based on the human U4 Sm structure (PDB 4WZJ). f, Dib1 (ref. 29) (PDB 1QGV). g, (i) Prp31. (ii), Comparison between the crystal structure of human Prp31(78–333) (ref. 33) (PDB 2OZB, grey) and that in tri-snRNP (yellow and blue). The coiled-coil domain (yellow) rotates by 60° in tri-snRNP with respect to the Nop domain (grey). Additional helices (blue) that extend from the N and C termini were built. h, U4 Sm proteins with part of U4 snRNA (blue) based on the human U4 Sm structure. i, Prp3 model. The ferredoxin-like domain was obtained from homology modelling while the extra helices were built de novo. j, Prp4 WD40 homology model with the extra helices built de novo. k, Snu13 (ref. 64) (PDB 2ALE). l, U6 LSm proteins67 (PDB 4M77).

Extended Data Figure 6 Fitting of the RNA components in tri-snRNP map.

a, c, The sequences and predicted secondary structures of U4/U6 snRNA and the long version of U5 snRNA, respectively. b, d, The maps of the fitted parts of U4/U6 snRNA and U5 snRNA, respectively. Unmodelled density assigned to U5 snRNA is also shown in d.

Extended Data Figure 7 Sequence alignment of yeast and human Snu114 with yeast and human elongation factor 2 (EF-2).

The secondary structures of our homology model for yeast Snu114 and the yeast EF-2 (ref. 26) (PDB 1N0V) are shown on the top and bottom of the alignment, respectively. Important sequence elements are also shown. The greyscale shading indicates the level of sequence conservation. A higher level of conservation is shown in a darker shade.

Extended Data Figure 8 The effect of ATP on Brr2-TAPS purified tri-snRNP.

a, Ethidium-bromide-stained native agarose gel (0.5%) showing the effects of ATP addition to Brr2-TAPS purified tri-snRNP used in this study. Upon ATP addition either without or with GTP/GDP, tri-snRNP fell apart (lanes 1–4). Under the same conditions, the addition of ADP or the non-hydrolysable ATP-analogue, AMPPNP, had no effects on the complex (lanes 5, 6). b, c, The effect of ATP addition observed by negative stain microscopy. When ATP was not present, tri-snRNP particles could be observed. When ATP was added to the sample before grid preparations, tri-snRNP particles fell apart as observed by many small components on the micrograph rather than tri-snRNP particles. d, Tri-snRNP model where U4/U6 snRNP proteins are not shown. In tri-snRNP, Brr2–Prp8Jab complex is loosely associated to the remaining U5 snRNP components including Prp8large, Prp8RNaseH, Prp8Nterm, Snu114, Dib1, U5 Sm proteins and U5 snRNA. After U4/U6 snRNA unwinding by Brr2, Brr2–Prp8Jab could be repositioned within the spliceosome. e, A schematic showing the arrangement of tri-snRNP protein and RNA components.

Extended Data Table 1 Components and modelling of yeast U4/U6.U5 tri-snRNP
Extended Data Table 2 U4-cs1 suppressors

Supplementary information

The architecture of the spliceosomal U4/U6.U5 tri-snRNP

The video sequences showing the cryoEM density at two different contour levels; tri-snRNP map with all modeled components; fitting of available crystal structures into the cryoEM density: Brr2-Jab1/MPN (Prp8) complex31, Prp8 RNase H and large domains10, U4 and U5 Sm core domains65, Lsm core domain67 fitted into the multi-body map, Snu13 (ref. 64), human Prp31 (ref. 33) with remodeling, human Dib1 (ref. 29); fitting of homology models: Snu114 based translation factor EF2 (ref. 26), WD40 domain of Prp4, ferredoxin-like domain of Prp3 (ref. 36), TPR domain of Prp6; fitting of double helical RNA of the U4/U6 snRNA duplex and U5 snRNA; fitting of α-helices attributed to the N-terminal domain Prp8, Prp3 and Prp4; near complete pseudo-atomic structure of the yeast U4/U6.U5 tri-snRNP. (MOV 42926 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nguyen, T., Galej, W., Bai, Xc. et al. The architecture of the spliceosomal U4/U6.U5 tri-snRNP. Nature 523, 47–52 (2015). https://doi.org/10.1038/nature14548

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature14548

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