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:

Specific recognition and ubiquitination of translating ribosomes by mammalian CCR4–NOT

Abstract

Translation affects messenger RNA stability and, in yeast, this is mediated by the Ccr4–Not deadenylation complex. The details of this process in mammals remain unclear. Here, we use cryogenic electron microscopy (cryo-EM) and crosslinking mass spectrometry to show that mammalian CCR4–NOT specifically recognizes ribosomes that are stalled during translation elongation in an in vitro reconstituted system with rabbit and human components. Similar to yeast, mammalian CCR4–NOT inserts a helical bundle of its CNOT3 subunit into the empty E site of the ribosome. Our cryo-EM structure shows that CNOT3 also locks the L1 stalk in an open conformation to inhibit further translation. CCR4–NOT is required for stable association of the nonconstitutive subunit CNOT4, which ubiquitinates the ribosome, likely to signal stalled translation elongation. Overall, our work shows that human CCR4–NOT not only detects but also enforces ribosomal stalling to couple translation and mRNA decay.

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: Human CCR4–NOT and CNOT4 recognize stalled ribosomes in an in vitro translation system.
Fig. 2: Structure of mammalian CNOT3-bound to the 80S ribosome.
Fig. 3: CNOT3 mutations in human disease.
Fig. 4: CCR4–NOT binds 80S ribosomes in the vicinity of the E site.
Fig. 5: The CNOT4 RRM and ZnF domains are required for association with the 80S ribosome and eS7 ubiquitination.
Fig. 6: Model for CCR4–NOT and CNOT4 recognition of mammalian ribosomes during slow elongation.

Similar content being viewed by others

Data availability

Cryo-EM maps and molecular models have been deposited in the Electron Microscopy Data Bank (accession code EMD-16052) and in the PDB (accession code 8BHF). We also analyzed previously reported structures (PDB 6TB3 and PDB 5LZV) and used PDB 6SGC for initial model building. EM micrographs are available from the EMPIAR database with accession code EMPIAR-11593. MS data have been deposited in jPOST (project ID JPST001798, PRIDE ID PXD035522). Cancer Hotspot mutations on CNOT3 were accessed from the COSMIC database (v.96). Source data are provided with this paper. All other data are available in the main text or as part of the Extended Data or supplementary materials. Original gels and blot images are provided in the source data. Correspondence and requests for materials should be addressed to L.A.P. or V.C. All unique materials are available upon request with completion of a standard Materials Transfer Agreement.

References

  1. Veltri, A. J. et al. Distinct elongation stalls during translation are linked with distinct pathways for mRNA degradation. eLife 11, e76038 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Hanson, G. & Coller, J. Codon optimality, bias and usage in translation and mRNA decay. Nat. Rev. Mol. Cell Biol. 19, 20–30 (2018).

    Article  CAS  PubMed  Google Scholar 

  3. Brandman, O. & Hegde, R. S. Ribosome-associated protein quality control. Nat. Struct. Mol. Biol. 23, 7–15 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Presnyak, V. et al. Codon optimality is a major determinant of mRNA stability. Cell 160, 1111–1124 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Forrest, M. E. et al. Codon and amino acid content are associated with mRNA stability in mammalian cells. PLoS ONE 15, e0228730 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Jeacock, L., Faria, J. & Horn, D. Codon usage bias controls mRNA and protein abundance in trypanosomatids. eLife 7, e32496 (2018).

  7. Harigaya, Y. & Parker, R. Analysis of the association between codon optimality and mRNA stability in Schizosaccharomyces pombe. BMC Genomics 17, 895 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  8. de Freitas Nascimento, J., Kelly, S., Sunter, J. & Carrington, M. Codon choice directs constitutive mRNA levels in trypanosomes. eLife 7, e32467 (2018).

  9. Burrow, D. A. et al. Attenuated codon optimality contributes to neural-specific mRNA decay in Drosophila. Cell Rep. 24, 1704–1712 (2018).

    Article  PubMed Central  Google Scholar 

  10. Mishima, Y. & Tomari, Y. Codon usage and 3′ UTR length determine maternal mRNA stability in zebrafish. Mol. Cell 61, 874–885 (2016).

    Article  CAS  PubMed  Google Scholar 

  11. Bazzini, A. A. et al. Codon identity regulates mRNA stability and translation efficiency during the maternal‐to‐zygotic transition. EMBO J. 35, 2087–2103 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Wu, Q. et al. Translation affects mRNA stability in a codon-dependent manner in human cells. eLife 8, e45396 (2019).

  13. Narula, A., Ellis, J., Taliaferro, J. M. & Rissland, O. S. Coding regions affect mRNA stability in human cells. RNA 25, 1751–1764 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Mishima, Y., Han, P., Ishibashi, K., Kimura, S. & Iwasaki, S. Ribosome slowdown triggers codon-mediated mRNA decay independently of ribosome quality control. EMBO J. 41, e109256 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Buschauer, R. et al. The Ccr4–Not complex monitors the translating ribosome for codon optimality. Science 368, eaay6912 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Passmore, L. A. & Coller, J. Roles of mRNA poly(A) tails in regulation of eukaryotic gene expression. Nat. Rev. Mol. Cell Biol. 23, 93–106 (2022).

    Article  CAS  PubMed  Google Scholar 

  17. Maillet, L., Tu, C., Hong, Y. K., Shuster, E. O. & Collart, M. A. The essential function of not1 lies within the Ccr4–not complex. J. Mol. Biol. 303, 131–143 (2000).

    Article  CAS  PubMed  Google Scholar 

  18. Chalabi Hagkarim, N. & Grand, R. J. The regulatory properties of the Ccr4–Not complex. Cells 9, 2379 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Bawankar, P., Loh, B., Wohlbold, L., Schmidt, S. & Izaurralde, E. NOT10 and C2orf29/NOT11 form a conserved module of the CCR4–NOT complex that docks onto the NOT1 N-terminal domain. RNA Biol. 10, 228–244 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Mauxion, F., Prève, B. & Séraphin, B. C2ORF29/CNOT11 and CNOT10 form a new module of the CCR4–NOT complex. RNA Biol. 10, 267–276 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Höpfler, M. et al. Mechanism of ribosome-associated mRNA degradation during tubulin autoregulation. Mol. Cell. https://doi.org/10.1016/j.molcel.2023.05.020 (2023).

  22. Mauxion, F. et al. The human CNOT1–CNOT10–CNOT11 complex forms a structural platform for protein–protein interactions. Cell Rep. 42, 111902 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Lau, N.-C. et al. Human Ccr4–Not complexes contain variable deadenylase subunits. Biochem. J. 422, 443–453 (2009).

    Article  CAS  PubMed  Google Scholar 

  24. Temme, C. et al. Subunits of the Drosophila CCR4–NOT complex and their roles in mRNA deadenylation. RNA 16, 1356–1370 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Keskeny, C. et al. A conserved CAF40-binding motif in metazoan NOT4 mediates association with the CCR4–NOT complex. Genes Dev. 33, 236–252 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Feng, Q. & Shao, S. In vitro reconstitution of translational arrest pathways. Methods 137, 20–36 (2018).

    Article  CAS  PubMed  Google Scholar 

  27. Frydman, J., Nimmesgern, E., Ohtsuka, K. & Hartl, F. U. Folding of nascent polypeptide chains in a high molecular mass assembly with molecular chaperones. Nature 370, 111–117 (1994).

    Article  CAS  PubMed  Google Scholar 

  28. Ingolia, N. T., Lareau, L. F. & Weissman, J. S. Ribosome profiling of mouse embryonic stem cells reveals the complexity and dynamics of mammalian proteomes. Cell 147, 789–802 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Shao, S., von der Malsburg, K. & Hegde, R. S. Listerin-dependent nascent protein ubiquitination relies on ribosome subunit dissociation. Mol. Cell 50, 637–648 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Weissmann, F. et al. biGBac enables rapid gene assembly for the expression of large multisubunit protein complexes. Proc. Natl Acad. Sci. USA 113, E2564–2569 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Wang, M., Herrmann, C. J., Simonovic, M., Szklarczyk, D. & von Mering, C. Version 4.0 of PaxDb: protein abundance data, integrated across model organisms, tissues, and cell-lines. Proteomics 15, 3163–3168 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Chandrasekaran, V. et al. Mechanism of ribosome stalling during translation of a poly(A) tail. Nat. Struct. Mol. Biol. 26, 1132–1140 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Fei, J., Kosuri, P., MacDougall, D. D. & Gonzalez, R. L. Coupling of ribosomal L1 stalk and tRNA dynamics during translation elongation. Mol. Cell 30, 348–359 (2008).

    Article  CAS  PubMed  Google Scholar 

  34. Varadi, M. et al. AlphaFold Protein Structure Database: massively expanding the structural coverage of protein-sequence space with high-accuracy models. Nucleic Acids Res. 50, D439–D444 (2022).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Martin, R. et al. De novo variants in CNOT3 cause a variable neurodevelopmental disorder. Eur. J. Hum. Genet. 27, 1677–1682 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Tate, J. G. et al. COSMIC: the Catalogue Of Somatic Mutations In Cancer. Nucleic Acids Res. 47, D941–D947 (2019).

    Article  CAS  PubMed  Google Scholar 

  38. Boland, A. et al. Structure and assembly of the NOT module of the human CCR4–NOT complex. Nat. Struct. Mol. Biol. 20, 1289–1297 (2013).

    Article  CAS  PubMed  Google Scholar 

  39. Albert, T. K. et al. Identification of a ubiquitin–protein ligase subunit within the CCR4–NOT transcription repressor complex. EMBO J. 21, 355–364 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Brito Querido, J. et al. Structure of a human 48S translational initiation complex. Science 369, 1220–1227 (2020).

    Article  CAS  PubMed  Google Scholar 

  41. Querido, J. B. et al. The structure of a human translation initiation complex reveals two independent roles for the helicase eIF4A. Preprint at bioRXiv https://doi.org/10.1101/2022.12.07.519490 (2022).

  42. Ikeuchi, K. et al. Molecular basis for recognition and deubiquitination of 40S ribosomes by Otu2. Nat. Commun. 14, 2730 (2023).

  43. Hill, C. H. et al. Activation of the endonuclease that defines mRNA 3’ ends requires incorporation into an 8-subunit core cleavage and polyadenylation factor complex. Mol. Cell 73, 1217–1231.e11 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Sharma, A., Mariappan, M., Appathurai, S. & Hegde, R. S. In vitro dissection of protein translocation into the mammalian endoplasmic reticulum. Methods Mol. Biol. 619, 339–363 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. 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 

  46. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. 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  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  Google Scholar 

  50. The PyMOL Molecular Graphics System v.1.8. (Schrodinger, 2015).

  51. Sievers, F. et al. Fast, scalable generation of high-quality protein multiple sequence alignments using Clustal Omega. Mol. Syst. Biol. 7, 539 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Goujon, M. et al. A new bioinformatics analysis tools framework at EMBL-EBI. Nucleic Acids Res. 38, W695–699 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Waterhouse, A. M., Procter, J. B., Martin, D. M. A., 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 

  54. Graham, M., Combe, C., Kolbowski, L. & Rappsilber, J. xiView: a common platform for the downstream analysis of crosslinking mass spectrometry data. Preprint at bioRXiv https://doi.org/10.1101/561829 (2019).

  55. Rafiee, M.-R. et al. Protease-resistant streptavidin for interaction proteomics. Mol. Syst. Biol. 16, e9370 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Rappsilber, J., Mann, M. & Ishihama, Y. Protocol for micro-purification, enrichment, pre-fractionation and storage of peptides for proteomics using StageTips. Nat. Protoc. 2, 1896–1906 (2007).

    Article  CAS  PubMed  Google Scholar 

  57. Mendes, M. L. et al. An integrated workflow for crosslinking mass spectrometry. Mol. Syst. Biol. 15, e8994 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We are grateful to members of the Passmore, Ramakrishnan and Rappsilber laboratories for assistance and advice; F. Schildhauer for assistance with modified Streptavidin bead preparation; R. Hegde for the stall mRNA plasmid template; the Medical Research Council (MRC) LMB Electron Microscopy Facility for access and support of EM sample preparation and data collection; J. Grimmett and T. Darling (LMB scientific computation) and J.G. Shi (baculovirus) for support. We acknowledge Diamond Light Source for access to eBIC (proposal no. BI23268) funded by the Wellcome Trust, MRC and Biotechnology and Biological Sciences Research Council. This work was supported by the MRC, as part of United Kingdom Research and Innovation (also known as UK Research and Innovation), MRC file reference number MC_U105192715 (L.A.P.); a postdoctoral fellowship of the Deutsche Forschungsgemeinschaft (the German Research Foundation, project no. 429892960) to E.A and the European Union’s Horizon 2020 research and innovation program (European Research Council Consolidator grant agreement no. 725685) (to L.A.P.). V.C. was supported by V. Ramakrishnan whose funding was from the MRC (grant no. MC_U105184332), the Wellcome Trust (grant no. WT096570), the Agouron Institute, and the Louis-Jeantet Foundation. The Wellcome Centre for Cell Biology (J.R.) is supported by core funding from the Wellcome Trust (grant no. 203149). For the purpose of open access, the MRC-LMB has applied a CC BY public copyright licence to any Author Accepted Manuscript version arising.

Author information

Authors and Affiliations

Authors

Contributions

E.A. designed CNOT4 mutants and truncations and purified all proteins (CCR4–NOT and CNOT4) used in this study. J.A.W.S. initially designed the CCR4–NOT expression and purification scheme. V.C. and E.A. designed and performed in vitro translation assays, cryo-EM sample preparation, cryo-EM data collection, processing and analysis. E.A. produced crosslinking MS sample and F.J.O’R. measured the crosslinked sample and analyzed it. L.A.P. and J.R. supervised the research. E.A., V.C. and L.A.P. wrote the manuscript with contributions from all the authors.

Corresponding authors

Correspondence to Viswanathan Chandrasekaran or Lori A. Passmore.

Ethics declarations

Competing interests

L.A.P. is an inventor on a patent filed by the MRC for all-gold EM supports (PCT/GB2014/051896), licensed to Quantifoil under the trademark UltrAuFoil. The other authors declare no competing interests.

Peer review

Peer review information

Nature Structural & Molecular Biology thanks Mark Bartlam, Ariel Bazzini, and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available. Sara Osman and Dimitris Typas were the primary editors on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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 In vitro reconstitution of ribosome stalling during elongation, CCR4-NOT/CNOT4 binding and eS7 mono-ubiquitination.

(a) Autoradiogram of a sucrose gradient fractionation of stall mRNA in RRL shows that the majority of the radiolabeled 3x FLAG-β-VHP-(UUA)3 is on stalled monosomes (fractions 5-8; 200 µl gradient). * indicates a small amount of full-length protein product, released from the ribosome at the stop codon and therefore migrating in the supernatant (fractions 1-3). The band below the full-length protein product corresponds to released, shortened protein product, resulting from ribosomal stalling on the three UUA leucine codons of the stall mRNA. We note that unlike in the previous cryoEM structure of yeast Ccr4-Not bound to stalled RNCs, we observed mainly stalled monosomes. Multiple rounds of initiation are disfavored in the RRL and only <5% of mRNAs have two or more ribosomes translating on a single mRNA (compare signals in monosome fractions 6–8 and polysome fractions 9–11). Data are representative of experiments performed twice. (b) Autoradiogram visualizing that full-length protein (*) accumulates in the supernatant when stall mRNA is translated in RRL supplemented with total tRNA from pig liver. Data are representative of experiments performed twice. (c) Immunoblot of the 80S peak fractions from a sucrose gradient of a sample containing CCR4-NOT, CNOT4 and stall mRNA using antibodies against CNOT1, CNOT3, CNOT4, CNOT6, CNOT11 and CNOT9 (12 ml gradient). Data are representative of experiments performed twice. (d-f) Sucrose gradient fractions of non-radiolabeled translation reactions of stall mRNA in the presence of the indicated CCR4-NOT and/or CNOT4 proteins immunoblotted using antibodies against CNOT3, CNOT4 and eS7 (12 ml gradient). The mono-ubiquitinated eS7 band is also labeled. Data are representative of experiments performed six (d,e) or four (f) times. (g-i) Same experiments as in panels (d-f) but performed on ribosomes elongating on native α- and β-globin mRNA in RRL not treated with nuclease (12 ml gradient). Data are representative of experiments performed twice. In d-i, asterisks denote cross-reacting bands. We note that in previous studies in yeast, eS7 is mono-ubiquitinated in monosome fractions and poly-ubiquitinated in polysome fractions. We observe eS7 mono-ubiquitination in the monosome peak but no poly-ubiquitination. We conclude that poly-ubiquitination is not required for stable binding of CCR4-NOT and CNOT4.

Source data

Extended Data Fig. 2 RELION 3.1 processing workflow.

Two datasets were collected, processed separately and merged at the end to yield a final reconstruction at 3.1 Å resolution.

Extended Data Fig. 3 CryoEM analysis of 80S ribosomes bound to CNOT3.

(a) Representative micrograph used for single particle analysis. Scale bar: 50 nm. A total of 32,762 micrographs were used for this study. (b) Hand-picked two-dimensional class averages of 80S ribosomes (Table 1). (c) Gold-standard Fourier shell correlation (FSC) curve (blue) of the final map illustrating an overall resolution of 3.1 Å. The phase-randomized, masked FSC curve (orange) is also shown. (d) Final map segmented to show 60S (cyan), 40S (yellow), CNOT3 (red-orange) and the P-site tRNA (green). (e) Map from panel (d) colored according to the local resolution (cyan - high resolution; magenta - low resolution).

Extended Data Fig. 4 Details around the P-site tRNA in the cryoEM map of 80S CNOT3.

(a-b) The cryoEM map and atomic model are shown for the nascent chain (a) and the codon-anticodon in the P site (b). This map was calculated from two-fold downscaled particles (to a pixel size of 1.66 Å/pix) to improve interpretability. The P-site tRNA and nascent chain are contoured at 10 r.m.s.d. (c) Packing interactions between CNOT3 helical bundle a (red-orange) and the P-site tRNA (green). (d) Packing interactions between CNOT3 helical bundle a (red-orange) and the 28S rRNA (cyan). Salt-bridges are indicated with black, dashed lines.

Extended Data Fig. 5 Evolutionary conservation of the interaction between CNOT3 and the 80S ribosome.

(a) Multiple sequence alignments of the two helical bundles across representative eukaryotes (bundle a: 1-111, bundle b: 115-236). Numbering is according to the human sequence and frequently mutated residues in developmental disorders and cancer are indicated with red and orange asterisks, respectively. Helical bundle a has a sequence identity of 54.5% between yeast and human; helical bundle b (residues 120-213 in yeast Not5 and 115-236 in human CNOT3) has a sequence identity of 29.8%. High conservation, dark purple; lower conservation, lighter purple. (b) Superposition of budding yeast (Sc) Not5 helical bundle a (from PDB 6TB3) and helical bundle b (Alphafold2 model, residues 120-213) on human (Hs) CNOT3 helical bundles a and b (residues 1-236). Black lines indicate the extended helices in the human structure (residues 147-169), which are missing in yeast. Note that the cryoEM structure of the yeast complex only contains bundle a. Oc, Oryctolagus cuniculus. (c) Surface charge of helical bundle b of Hs CNOT3. (d) Surface charge of the AlphaFold2 prediction of helical bundle b of Sc Not5. Red- negative surface charge, blue- positive surface charge.

Extended Data Fig. 6 Crosslinking mass spectrometry of CCR4-NOT- and CNOT4-bound stalled 80S ribosomes.

(a-b) Circle diagrams of observed crosslinks (a) between selected CCR4-NOT subunits or CNOT4 and 80S ribosomal proteins or (b) within CCR4-NOT and CNOT4. Dashed lines indicate crosslinks between proteins that were not visible in the structure and therefore not modelled on the structure. In panels (a) and (b), the indicated subunits are highlighted. (c) Diagrams of observed crosslinks of CNOT3 to other CCR4-NOT subunits and CNOT4 when bound to 80S. CNOT3 is drawn to a larger scale than the other proteins.

Extended Data Fig. 7 Repetitions of experiments in Fig. 5.

The experimental repeats of data shown in Fig. 5 are shown. Note that sample 9 (*) is missing in the anti-CNOT4 Western blot of panel b.

Source data

Supplementary information

Reporting Summary

Peer Review File

Supplementary Video 1

Cryo-EM structure of a stalled ribosome bound to CNOT3. The atomic model of the cryo-EM structure is shown in cartoon form with 40S (yellow), 60S (cyan), L1 stalk (blue), P site tRNA (green), mRNA (salmon) and CNOT3 (red) indicated. The electrostatic surface charge of CNOT3 is also shown.

Supplementary Table 1

Table of oligonucleotides, antibodies and reagents.

Source data

Source Data Fig. 1

Unprocessed gels and western blots.

Source Data Fig. 5

Unprocessed gels and western blots.

Source Data Extended Data Fig. 1

Unprocessed gels and western blots.

Source Data Extended Data Fig. 7

Unprocessed gels and western blots.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Absmeier, E., Chandrasekaran, V., O’Reilly, F.J. et al. Specific recognition and ubiquitination of translating ribosomes by mammalian CCR4–NOT. Nat Struct Mol Biol 30, 1314–1322 (2023). https://doi.org/10.1038/s41594-023-01075-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-023-01075-8

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