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.

  • Letter
  • Published:

Protein quality control at the inner nuclear membrane

Abstract

The nuclear envelope is a double membrane that separates the nucleus from the cytoplasm. The inner nuclear membrane (INM) functions in essential nuclear processes including chromatin organization and regulation of gene expression1. The outer nuclear membrane is continuous with the endoplasmic reticulum and is the site of membrane protein synthesis. Protein homeostasis in this compartment is ensured by endoplasmic-reticulum-associated protein degradation (ERAD) pathways that in yeast involve the integral membrane E3 ubiquitin ligases Hrd1 and Doa10 operating with the E2 ubiquitin-conjugating enzymes Ubc6 and Ubc7 (refs 2, 3). However, little is known about protein quality control at the INM. Here we describe a protein degradation pathway at the INM in yeast (Saccharomyces cerevisiae) mediated by the Asi complex consisting of the RING domain proteins Asi1 and Asi3 (ref. 4). We report that the Asi complex functions together with the ubiquitin-conjugating enzymes Ubc6 and Ubc7 to degrade soluble and integral membrane proteins. Genetic evidence suggests that the Asi ubiquitin ligase defines a pathway distinct from, but complementary to, ERAD. Using unbiased screening with a novel genome-wide yeast library based on a tandem fluorescent protein timer5, we identify more than 50 substrates of the Asi, Hrd1 and Doa10 E3 ubiquitin ligases. We show that the Asi ubiquitin ligase is involved in degradation of mislocalized integral membrane proteins, thus acting to maintain and safeguard the identity of the INM.

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: The Asi complex is a Ubc6/Ubc7-dependent E3 ubiquitin ligase of the INM.
Figure 2: Functional overlap between Asi and ERAD E3 ubiquitin ligases.
Figure 3: Systematic identification of substrates for Asi and ERAD E3 ubiquitin ligases.

Similar content being viewed by others

References

  1. Mekhail, K. & Moazed, D. The nuclear envelope in genome organization, expression and stability. Nature Rev. Mol. Cell Biol. 11, 317–328 (2010)

    Article  CAS  Google Scholar 

  2. Zattas, D. & Hochstrasser, M. Ubiquitin-dependent protein degradation at the yeast endoplasmic reticulum and nuclear envelope. Crit. Rev. Biochem. Mol. Biol. http://dx.doi.org/10.3109/10409238.2014.959889 (2014)

  3. Ruggiano, A., Foresti, O. & Carvalho, P. Quality control: ER-associated degradation: protein quality control and beyond. J. Cell Biol. 204, 869–879 (2014)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Zargari, A. et al. Inner nuclear membrane proteins Asi1, Asi2, and Asi3 function in concert to maintain the latent properties of transcription factors Stp1 and Stp2. J. Biol. Chem. 282, 594–605 (2007)

    Article  CAS  PubMed  Google Scholar 

  5. Khmelinskii, A. et al. Tandem fluorescent protein timers for in vivo analysis of protein dynamics. Nature Biotechnol. 30, 708–714 (2012)

    Article  CAS  Google Scholar 

  6. Deng, M. & Hochstrasser, M. Spatially regulated ubiquitin ligation by an ER/nuclear membrane ligase. Nature 443, 827–831 (2006)

    Article  ADS  CAS  PubMed  Google Scholar 

  7. Boban, M., Pantazopoulou, M., Schick, A., Ljungdahl, P. O. & Foisner, R. A nuclear ubiquitin-proteasome pathway targets the inner nuclear membrane protein Asi2 for degradation. J. Cell Sci. 127, 3603–3613 (2014)

    CAS  PubMed  Google Scholar 

  8. Hu, C.-D., Chinenov, Y. & Kerppola, T. K. Visualization of interactions among bZIP and Rel family proteins in living cells using bimolecular fluorescence complementation. Mol. Cell 9, 789–798 (2002)

    Article  CAS  PubMed  Google Scholar 

  9. Forsberg, H., Hammar, M., Andréasson, C., Molinér, A. & Ljungdahl, P. O. Suppressors of ssy1 and ptr3 null mutations define novel amino acid sensor-independent genes in Saccharomyces cerevisiae. Genetics 158, 973–988 (2001)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Boban, M. et al. Asi1 is an inner nuclear membrane protein that restricts promoter access of two latent transcription factors. J. Cell Biol. 173, 695–707 (2006)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Omnus, D. J. & Ljungdahl, P. O. Latency of transcription factor Stp1 depends on a modular regulatory motif that functions as cytoplasmic retention determinant and nuclear degron. Mol. Biol. Cell 25, 3823–3833 (2014)

    Article  PubMed  PubMed Central  Google Scholar 

  12. Wienken, C. J., Baaske, P., Rothbauer, U., Braun, D. & Duhr, S. Protein-binding assays in biological liquids using microscale thermophoresis. Nature Commun. 1, 100 (2010)

    Article  ADS  CAS  Google Scholar 

  13. Kostova, Z., Mariano, J., Scholz, S., Koenig, C. & Weissman, A. M. A. Ubc7p-binding domain in Cue1p activates ER-associated protein degradation. J. Cell Sci. 122, 1374–1381 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Biederer, T., Volkwein, C. & Sommer, T. Role of Cue1p in ubiquitination and degradation at the ER surface. Science 278, 1806–1809 (1997)

    Article  ADS  CAS  PubMed  Google Scholar 

  15. Costanzo, M., Baryshnikova, A., Myers, C. L., Andrews, B. & Boone, C. Charting the genetic interaction map of a cell. Curr. Opin. Biotechnol. 22, 66–74 (2011)

    Article  CAS  PubMed  Google Scholar 

  16. Costanzo, M. et al. The genetic landscape of a cell. Science 327, 425–431 (2010)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  17. Friedlander, R., Jarosch, E., Urban, J., Volkwein, C. & Sommer, T. A regulatory link between ER-associated protein degradation and the unfolded-protein response. Nature Cell Biol. 2, 379–384 (2000)

    Article  CAS  PubMed  Google Scholar 

  18. Foresti, O., Rodriguez-Vaello, V., Funaya, C. & Carvalho, P. Quality control of inner nuclear membrane proteins by the Asi complex. Science 346, 751–755 (2014)

    Article  ADS  CAS  PubMed  Google Scholar 

  19. Khmelinskii, A., Meurer, M., Duishoev, N., Delhomme, N. & Knop, M. Seamless gene tagging by endonuclease-driven homologous recombination. PLoS ONE 6, e23794 (2011)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  20. Baryshnikova, A. et al. Synthetic genetic array (SGA) analysis in Saccharomyces cerevisiae and Schizosaccharomyces pombe. Methods Enzymol. 470, 145–179 (2010)

    Article  CAS  PubMed  Google Scholar 

  21. Zattas, D., Adle, D. J., Rubenstein, E. M. & Hochstrasser, M. N-terminal acetylation of the yeast Derlin Der1 is essential for Hrd1 ubiquitin-ligase activity toward luminal ER substrates. Mol. Biol. Cell 24, 890–900 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Uttenweiler, A., Schwarz, H., Neumann, H. & Mayer, A. The vacuolar transporter chaperone (VTC) complex is required for microautophagy. Mol. Biol. Cell 18, 166–175 (2007)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Sharpe, H. J., Stevens, T. J. & Munro, S. A comprehensive comparison of transmembrane domains reveals organelle-specific properties. Cell 142, 158–169 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Meinema, A. C., Poolman, B. & Veenhoff, L. M. The transport of integral membrane proteins across the nuclear pore complex. Nucleus 3, 322–329 (2012)

    Article  PubMed  Google Scholar 

  25. Ellenberg, J. et al. Nuclear membrane dynamics and reassembly in living cells: targeting of an inner nuclear membrane protein in interphase and mitosis. J. Cell Biol. 138, 1193–1206 (1997)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Soullam, B. & Worman, H. J. The amino-terminal domain of the lamin B receptor is a nuclear envelope targeting signal. J. Cell Biol. 120, 1093–1100 (1993)

    Article  CAS  PubMed  Google Scholar 

  27. Hinshaw, J. E., Carragher, B. O. & Milligan, R. A. Architecture and design of the nuclear pore complex. Cell 69, 1133–1141 (1992)

    Article  CAS  PubMed  Google Scholar 

  28. Beck, M., Lucić, V., Förster, F., Baumeister, W. & Medalia, O. Snapshots of nuclear pore complexes in action captured by cryo-electron tomography. Nature 449, 611–615 (2007)

    Article  ADS  CAS  PubMed  Google Scholar 

  29. Nakamura, N. The Role of the transmembrane RING finger proteins in cellular and organelle function. Membranes 1, 354–393 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Janke, C. et al. A versatile toolbox for PCR-based tagging of yeast genes: new fluorescent proteins, more markers and promoter substitution cassettes. Yeast 21, 947–962 (2004)

    Article  CAS  PubMed  Google Scholar 

  31. Andréasson, C. & Ljungdahl, P. O. The N-terminal regulatory domain of Stp1p is modular and, fused to an artificial transcription factor, confers full Ssy1p-Ptr3p-Ssy5p sensor control. Mol. Cell. Biol. 24, 7503–7513 (2004)

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Becuwe, M. et al. A molecular switch on an arrestin-like protein relays glucose signaling to transporter endocytosis. J. Cell Biol. 196, 247–259 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Léon, S., Erpapazoglou, Z. & Haguenauer-Tsapis, R. Ear1p and Ssh4p are new adaptors of the ubiquitin ligase Rsp5p for cargo ubiquitylation and sorting at multivesicular bodies. Mol. Biol. Cell 19, 2379–2388 (2008)

    Article  PubMed  PubMed Central  Google Scholar 

  34. Shyu, Y. J., Liu, H., Deng, X. & Hu, C. Identification of new fluorescent protein fragments for biomolecular fluorescence complementation analysis under physiological conditions. Biotechniques 40, 61 (2006)

    Article  CAS  PubMed  Google Scholar 

  35. Sommer, T. & Jentsch, S. A protein translocation defect linked to ubiquitin conjugation at the endoplasmic reticulum. Nature 365, 176–179 (1993)

    Article  ADS  CAS  PubMed  Google Scholar 

  36. Metzger, M. B. et al. A structurally unique E2-binding domain activates ubiquitination by the ERAD E2, Ubc7p, through multiple mechanisms. Mol. Cell 50, 516–527 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Sheff, M. A. & Thorn, K. S. Optimized cassettes for fluorescent protein tagging in Saccharomyces cerevisiae. Yeast 21, 661–670 (2004)

    Article  CAS  PubMed  Google Scholar 

  38. Laporte, D., Salin, B., Daignan-Fornier, B. & Sagot, I. Reversible cytoplasmic localization of the proteasome in quiescent yeast cells. J. Cell Biol. 181, 737–745 (2008)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Schneider, C. A., Rasband, W. S. & Eliceiri, K. W. NIH Image to ImageJ: 25 years of image analysis. Nature Methods 9, 671–675 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Knop, M. et al. Epitope tagging of yeast genes using a PCR-based strategy: more tags and improved practical routines. Yeast 15, 963–972 (1999)

    Article  CAS  PubMed  Google Scholar 

  41. Winzeler, E. A. et al. Functional characterization of the S. cerevisiae genome by gene deletion and parallel analysis. Science 285, 901–906 (1999)

    Article  CAS  PubMed  Google Scholar 

  42. Smyth, G. K. in Bioinformatics and Computational Biology Solutions Using R and Bioconductor (eds Gentleman, R., Carey, V., Dudoit, S., Irizarry, R. & Huber, W. ). 397–420 (Springer, 2005)

  43. Khmelinskii, A. & Knop, M. Analysis of protein dynamics with tandem fluorescent protein timers. Methods Mol. Biol. 1174, 195–210 (2014)

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank M. Lemberg, E. Schiebel and B. Bukau for support and discussions, A. Kaufmann, C.-T. Ho, A. Bartosik and B. Besenbeck for help with tFT library construction, K. Ryman for the qRT–PCR analysis of gene expression, M. Hochstrasser for strains, the GeneCore and the media kitchen facilities of the European Molecular Biology Laboratory (EMBL) and Donnelly Centre for support with infrastructure and media. This work was supported by the Sonderforschungsbereich 1036 (SFB1036, TP10) from the Deutsche Forschungsgemeinschaft (DFG) (M.K.), the Swedish Research Council grant VR2011-5925 (P.O.L.), INSERM and grants from ANR (ANR-12-JSV8-0003-001) and Biosit (G.R.), fellowships from the European Molecular Biology Organization (EMBO ALTF 1124-2010 and EMBO ASTF 546-2012) (A.K.) and fellowships from the Ministère de la Recherche et de l’Enseignement Supérieur and La Ligue Contre le Cancer (E.B.). M.K. received funds from the CellNetworks Cluster of Excellence (DFG) for support with tFT library construction. W.H. acknowledges funding from the EC Network of Excellence Systems Microscopy. C.B. was supported by funds from the Canadian Institute for Advanced Research (GNE-BOON-141871), National Institutes of Health (R01HG005853-01), Canadian Institute for Health Research (MOP-102629) and the National Science and Engineering Research Council (RGPIN 204899-6).

Author information

Authors and Affiliations

Authors

Contributions

G.R. designed the BiFC, microscale thermophoresis and ubiquitin pulldown experiments that were performed by E.B., G.L.D. and A.B. M.P., D.J.O. and A.G. contributed the biochemical analysis of Asi-dependent ubiquitylation. M.K. and A.K. designed and coordinated the tFT project. A.K. and M.M. designed and constructed the tFT library and performed the screens with help from D.K. and C.B. B.F. and J.D.B. developed the screen analysis methods, with input from A.K., M.K., W.H. and C.B. M.K., A.K., G.R. and P.O.L. prepared the figures and wrote the paper with input from all authors.

Corresponding authors

Correspondence to Gwenaël Rabut, Per O. Ljungdahl or Michael Knop.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Identification of Ubc6 and Ubc7 ubiquitin-conjugating enzymes as functional interacting partners of Asi1 and Asi3.

a, Quantification of BiFC signals in cells expressing VC–Ubc6 and all tested E3 ubiquitin ligases. BiFC signals were measured in the cytoplasm and nucleus of individual cells (n as shown). Whiskers extend from the tenth to the ninetieth percentiles. The same representation is used in c and d. b, Immunoblot showing expression levels of VC-tagged E2 ubiquitin-conjugating enzymes. Ubc11–VC could not be detected in the growth condition of the BiFC assay. c, Quantification of BiFC signals in cells co-expressing VC-tagged E2 ubiquitin-conjugating enzymes and Asi1–VN or Asi3–VN (n as shown). d, Detection of a significant BiFC signal between Asi1–VN and Ubc4–VC in cells lacking UBC6 (n as shown). e, Coomassie-stained gels of recombinant proteins used in microscale thermophoresis experiments. f, mRNA levels of AGP1 and GNP1 measured with qRT–PCR in the indicated strains (mean ± s.d., n = 3 clones). The signal was normalized to wild type (dashed line). g, Ubiquitylation of Stp1–HA or Stp1-RI17–33–HA (Stp1 variant in which amino acid residues 2–64 were replaced with Stp1 residues 17–33 flanked by minimal linker sequences) (left) and Stp2–HA or Stp2Δ2–13–HA (Stp2 variant lacking amino acid residues 2–13) (right) in strains expressing 6×His–ubiquitin. Stp1-RI17–33 and Stp2Δ2–13 variants exhibit compromised cytoplasmic retention and enhanced Asi-dependent degradation, whereas full-length Stp1 is degraded primarily in the cytoplasm in a SCFGrr1-dependent manner11. Total cell extracts (T), flow-through (F) and ubiquitin conjugates (E) eluted after immobilized-metal affinity chromatography were separated by SDS–PAGE followed by immunoblotting with antibodies against the HA-tag, Pgk1 and the His-tag. Representative immunoblots from three technical replicates. *P < 10−4 (a, c and d; one-way ANOVA with Bonferroni correction for multiple testing), and *P < 0.05, **P < 0.1 (f; two-tailed t-test).

Extended Data Figure 2 Lack of genetic interaction between ASI1 and HRD1 or DOA10 at 37 °C.

Tenfold serial dilutions of strains grown on synthetic complete medium for 2 days at 30 or 37 °C.

Extended Data Figure 3 tFT screens for substrates of Asi and ERAD E3 ubiquitin ligases.

a, Tagging approach used to construct the tFT library in a strain carrying the I-SceI meganuclease under an inducible promoter. First, a module for seamless C-terminal protein tagging with the mCherry-sfGFP timer is integrated into a genomic locus of interest using conventional PCR targeting. Subsequent I-SceI expression leads to excision of the heterologous terminator and the URA3 selection marker, followed by repair of the double-strand break by homologous recombination between the mCherry and mCherryΔN sequences. A tFT fusion protein is expressed under control of endogenous promoter and terminator in the final strain. b, Workflow of screens for substrates of E3 ubiquitin ligases involved in protein degradation. Each tFT query strain is crossed to an array of mutants carrying different gene deletion alleles. The resulting strains are imaged with a fluorescence plate reader to identify proteins with altered stability in each mutant. c, Volcano plots of the screens for proteins with altered stability in the indicated mutants. Plots show z-scores for changes in protein stability on the x axis and the negative logarithm of P values adjusted for multiple testing on the y axis. The number of proteins with increased (red) or decreased (blue) stability at 1% false discovery rate is indicated. d, Fraction of proteins in the tFT library and in the three clusters in Fig. 3b mapped to the full yeast slim set of component GO terms. Note that the GO term cytoplasm contains all cellular contents except the nucleus and the plasma membrane. e, The three clusters in Fig. 3b are enriched for proteins in the indicated component GO terms. Bar plot shows −log10-transformed P values of significant enrichments.

Extended Data Figure 4 Analysis of integral membrane protein substrates of the Asi E3 ubiquitin ligase.

a, Differences in the log10mCherry/sfGFP intensity ratio between the indicated mutants and the wild type (mean ± s.d., n = 4) for tFT-tagged proteins from the Asi cluster in Fig. 3b. b, Quantification of BiFC signals in strains co-expressing VC–Ubc6 and Asi3–VN (top). BiFC signals were measured in the cytoplasm and nucleus of individual cells (n as shown). Whiskers extend from tenth to ninetieth percentiles. A substantial BiFC signal is retained in the asi2Δ mutant, despite reduced expression of Asi3 (immunoblot, bottom). c, Quantification of sfGFP signals in strains expressing tFT-tagged proteins from the Asi cluster in Fig. 3b. Fluorescence microscopy examples representative of five fields of view (top). Scale bar, 5 μm. sfGFP intensities were measured in individual cells (middle) and at the nuclear rim (bottom). For each protein, measurements were normalized to the mean of the respective wild type. Whiskers extend from minimum to maximum values. *P < 0.05 (a and c; two-tailed t-test) and *P < 10−4 (b; one-way ANOVA with Bonferroni correction for multiple testing).

Extended Data Figure 5 Cycloheximide chase experiments with substrates of the Asi E3 ubiquitin ligase.

Degradation of 3×HA-tagged proteins after blocking translation with cycloheximide. Whole-cell extracts were separated by SDS–PAGE followed by immunoblotting with antibodies against the HA tag and Pgk1 as loading control. Representative immunoblots from two technical replicates. Left, wild-type and asi1Δ immunoblots are reproduced in Fig. 3f.

Extended Data Figure 6 Influence of tagging and expression levels on localization of Vtc1 and Vtc4.

Fluorescence microscopy of strains expressing Vtc1 or Vtc4 tagged endogenously with monomeric yeast codon-optimized enhanced GFP (myeGFP) at the C terminus or tagged with sfGFP at the N terminus and expressed under control of endogenous or TEF1 promoters. Representative deconvolved images of five fields of view with 100 cells each. Arrowheads indicate nuclear rim localization. Scale bar, 5 μm.

Supplementary information

Supplementary Information

This file contains Supplementary Methods, Supplementary Notes 1 and 2, a description of Supplementary Tables 1, 2 and 3 and Supplementary Tables 4 and 5. (PDF 495 kb)

Supplementary Data

This file contains Supplementary Table 1 – see Supplementary Information document for full description. (XLSX 671 kb)

Supplementary Data

This file contains Supplementary Table 2 – see Supplementary Information document for full description. (XLSX 181 kb)

Supplementary Data

This file contains Supplementary Table 3 – see Supplementary Information document for full description. (XLSX 775 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Khmelinskii, A., Blaszczak, E., Pantazopoulou, M. et al. Protein quality control at the inner nuclear membrane. Nature 516, 410–413 (2014). https://doi.org/10.1038/nature14096

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature14096

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