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:

Reversible protein aggregation is a protective mechanism to ensure cell cycle restart after stress

Abstract

Protein aggregation is mostly viewed as deleterious and irreversible causing several pathologies. However, reversible protein aggregation has recently emerged as a novel concept for cellular regulation. Here, we characterize stress-induced, reversible aggregation of yeast pyruvate kinase, Cdc19. Aggregation of Cdc19 is regulated by oligomerization and binding to allosteric regulators. We identify a region of low compositional complexity (LCR) within Cdc19 as necessary and sufficient for reversible aggregation. During exponential growth, shielding the LCR within tetrameric Cdc19 or phosphorylation of the LCR prevents unscheduled aggregation, while its dephosphorylation is necessary for reversible aggregation during stress. Cdc19 aggregation triggers its localization to stress granules and modulates their formation and dissolution. Reversible aggregation protects Cdc19 from stress-induced degradation, thereby allowing cell cycle restart after stress. Several other enzymes necessary for G1 progression also contain LCRs and aggregate reversibly during stress, implying that reversible aggregation represents a conserved mechanism regulating cell growth and survival.

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: Cdc19 forms reversible, solid aggregates during stress.
Figure 2: Formation of monomeric Cdc19 is necessary for aggregation during stress.
Figure 3: Reversible aggregates co-localize with and modulate formation of stress granules.
Figure 4: Monomeric Cdc19 is required for the formation of amyloid-like aggregates in vitro.
Figure 5: Reversible aggregation depends on the presence of an LCR.
Figure 6: Phosphorylation of the LCR regulates aggregation.
Figure 7: Dephosphorylation of the Cdc19 LCR is necessary for reversible, amyloid-like aggregation in vivo.

Similar content being viewed by others

References

  1. Broach, J. R. Nutritional control of growth and development in yeast. Genetics 192, 73–105 (2012).

    Article  Google Scholar 

  2. De Virgilio, C. & Loewith, R. Cell growth control: little eukaryotes make big contributions. Oncogene 25, 6392–6415 (2006).

    Article  CAS  Google Scholar 

  3. Kroemer, G., Marino, G. & Levine, B. Autophagy and the integrated stress response. Mol. Cell 40, 280–293 (2010).

    Article  CAS  Google Scholar 

  4. Protter, D. S. & Parker, R. Principles and properties of stress granules. Trends Cell Biol. 26, 668–679 (2016).

    Article  CAS  Google Scholar 

  5. Buchan, J. R. mRNP granules. Assembly, function, and connections with disease. RNA Biol. 11, 1019–1030 (2014).

    Article  Google Scholar 

  6. Wippich, F. et al. Dual specificity kinase DYRK3 couples stress granule condensation/dissolution to mTORC1 signaling. Cell 152, 791–805 (2013).

    Article  CAS  Google Scholar 

  7. Mnaimneh, S. et al. Exploration of essential gene functions via titratable promoter alleles. Cell 118, 31–44 (2004).

    Article  CAS  Google Scholar 

  8. Narayanaswamy, R. et al. Widespread reorganization of metabolic enzymes into reversible assemblies upon nutrient starvation. Proc. Natl Acad. Sci. USA 106, 10147–10152 (2009).

    Article  CAS  Google Scholar 

  9. Saad, S., Peter, M. & Dechant, R. In scarcity and abundance: metabolic signals regulating cell growth. Physiology (Bethesda) 28, 298–309 (2013).

    CAS  Google Scholar 

  10. Kryndushkin, D., Pripuzova, N., Burnett, B. G. & Shewmaker, F. Non-targeted identification of prions and amyloid-forming proteins from yeast and mammalian cells. J. Biol. Chem. 288, 27100–27111 (2013).

    Article  CAS  Google Scholar 

  11. Wallace, E. W. et al. Reversible, specific, active aggregates of endogenous proteins assemble upon heat stress. Cell 162, 1286–1298 (2015).

    Article  CAS  Google Scholar 

  12. Alberti, S., Mateju, D., Mediani, L. & Carra, S. Granulostasis: protein quality control of RNP granules. Front. Mol. Neurosci. 10, 84 (2017).

    Article  Google Scholar 

  13. Aguzzi, A. & Altmeyer, M. Phase separation: linking cellular compartmentalization to disease. Trends Cell Biol. 26, 547–558 (2016).

    Article  CAS  Google Scholar 

  14. Molliex, A. et al. Phase separation by low complexity domains promotes stress granule assembly and drives pathological fibrillization. Cell 163, 123–133 (2015).

    Article  CAS  Google Scholar 

  15. Patel, A. et al. A liquid-to-solid phase transition of the ALS protein FUS accelerated by disease mutation. Cell 162, 1066–1077 (2015).

    Article  CAS  Google Scholar 

  16. Kato, M. et al. Cell-free formation of RNA granules: low complexity sequence domains form dynamic fibers within hydrogels. Cell 149, 753–767 (2012).

    Article  CAS  Google Scholar 

  17. Xiang, S. et al. The LC domain of hnRNPA2 adopts similar conformations in hydrogel polymers, liquid-like droplets, and nuclei. Cell 163, 829–839 (2015).

    Article  CAS  Google Scholar 

  18. Letunic, I., Doerks, T. & Bork, P. SMART: recent updates, new developments and status in 2015. Nucleic Acids Res. 43, D257–D260 (2015).

    Article  CAS  Google Scholar 

  19. Jurica, M. S. et al. The allosteric regulation of pyruvate kinase by fructose-1,6-bisphosphate. Structure 6, 195–210 (1998).

    Article  CAS  Google Scholar 

  20. Anastasiou, D. et al. Pyruvate kinase M2 activators promote tetramer formation and suppress tumorigenesis. Nat. Chem. Biol. 8, 839–847 (2012).

    Article  CAS  Google Scholar 

  21. Merrins, M. J., Van Dyke, A. R., Mapp, A. K., Rizzo, M. A. & Satin, L. S. Direct measurements of oscillatory glycolysis in pancreatic islet beta-cells using novel fluorescence resonance energy transfer (FRET) biosensors for pyruvate kinase M2 activity. J. Biol. Chem. 288, 33312–33322 (2013).

    Article  CAS  Google Scholar 

  22. Yang, W. et al. PKM2 phosphorylates histone H3 and promotes gene transcription and tumorigenesis. Cell 150, 685–696 (2012).

    Article  CAS  Google Scholar 

  23. Yang, W. et al. Nuclear PKM2 regulates β-catenin transactivation upon EGFR activation. Nature 480, 118–122 (2011).

    Article  CAS  Google Scholar 

  24. Gao, X., Wang, H., Yang, J. J., Liu, X. & Liu, Z. R. Pyruvate kinase M2 regulates gene transcription by acting as a protein kinase. Mol. Cell 45, 598–609 (2012).

    Article  CAS  Google Scholar 

  25. Fenton, A. W. & Blair, J. B. Kinetic and allosteric consequences of mutations in the subunit and domain interfaces and the allosteric site of yeast pyruvate kinase. Arch. Biochem. Biophys. 397, 28–39 (2002).

    Article  CAS  Google Scholar 

  26. Kresnowati, M. T. A. P. et al. When transcriptome meets metabolome: fast cellular responses of yeast to sudden relief of glucose limitation. Mol. Syst. Biol. 2, 49 (2006).

    Article  CAS  Google Scholar 

  27. Boles, E. et al. Characterization of a glucose-repressed pyruvate kinase (Pyk2p) in Saccharomyces cerevisiae that is catalytically insensitive to fructose-1,6-bisphosphate. J. Bacteriol. 179, 2987–2993 (1997).

    Article  CAS  Google Scholar 

  28. Grüning, N.-M. et al. Pyruvate kinase triggers a metabolic feedback loop that controls redox metabolism in respiring cells. Cell Metab. 14, 415–427 (2011).

    Article  Google Scholar 

  29. Protter, D. S. & Parker, R. Principles and properties of stress granules. Trends Cell Biol. 26, 668–679 (2016).

    Article  CAS  Google Scholar 

  30. Buchan, J. R., Muhlrad, D. & Parker, R. P bodies promote stress granule assembly in Saccharomyces cerevisiae. J. Cell Biol. 183, 441–455 (2008).

    Article  CAS  Google Scholar 

  31. Brengues, M. & Parker, R. Accumulation of polyadenylated mRNA, Pab1p, eIF4E, and eIF4G with P-bodies in Saccharomyces cerevisiae. Mol. Biol. Cell 18, 2592–2602 (2007).

    Article  CAS  Google Scholar 

  32. Beckmann, B. M. et al. The RNA-binding proteomes from yeast to man harbour conserved enigmRBPs. Nat. Commun. 6, 10127 (2015).

    Article  CAS  Google Scholar 

  33. Kramer, K. et al. Photo-cross-linking and high-resolution mass spectrometry for assignment of RNA-binding sites in RNA-binding proteins. Nat. Methods 11, 1064–1070 (2014).

    Article  CAS  Google Scholar 

  34. Berchowitz, L. E. et al. Regulated formation of an amyloid-like translational repressor governs gametogenesis. Cell 163, 406–418 (2015).

    Article  CAS  Google Scholar 

  35. Han, T. W. et al. Cell-free formation of RNA granules: bound RNAs identify features and components of cellular assemblies. Cell 149, 768–779 (2012).

    Article  CAS  Google Scholar 

  36. Kwon, I. et al. Phosphorylation-regulated binding of RNA polymerase II to fibrous polymers of low-complexity domains. Cell 155, 1049–1060 (2013).

    Article  CAS  Google Scholar 

  37. Wu, R. et al. A large-scale method to measure absolute protein phosphorylation stoichiometries. Nat. Methods 8, 677–683 (2011).

    Article  CAS  Google Scholar 

  38. Jorgensen, P. et al. A dynamic transcriptional network communicates growth potential to ribosome synthesis and critical cell size. Genes Dev. 18, 2491–2505 (2004).

    Article  CAS  Google Scholar 

  39. Dechant, R., Saad, S., Ibanez, A. J. & Peter, M. Cytosolic pH regulates cell growth through distinct GTPases, Arf1 and Gtr1, to promote Ras/PKA and TORC1 activity. Mol. Cell 55, 409–421 (2014).

    Article  CAS  Google Scholar 

  40. Urban, J. et al. Sch9 is a major target of TORC1 in Saccharomyces cerevisiae. Mol. Cell 26, 663–674 (2007).

    Article  CAS  Google Scholar 

  41. Quintas, A., Vaz, D. C., Cardoso, I., Saraiva, M. J. & Brito, R. M. Tetramer dissociation and monomer partial unfolding precedes protofibril formation in amyloidogenic transthyretin variants. J. Biol. Chem. 276, 27207–27213 (2001).

    Article  CAS  Google Scholar 

  42. Westermark, G. T. & Westermark, P. Prion-like aggregates: infectious agents in human disease. Trends Mol. Med. 16, 501–507 (2010).

    Article  CAS  Google Scholar 

  43. Castello, A., Hentze, M. W. & Preiss, T. Metabolic enzymes enjoying new partnerships as RNA-binding proteins. Trends Endocrinol. Metab. 26, 746–757 (2015).

    Article  CAS  Google Scholar 

  44. Dechant, R. et al. Cytosolic pH is a second messenger for glucose and regulates the PKA pathway through V-ATPase. EMBO J. 29, 2515–2526 (2010).

    Article  CAS  Google Scholar 

  45. Pelet, S., Dechant, R., Lee, S. S., van Drogen, F. & Peter, M. An integrated image analysis platform to quantify signal transduction in single cells. Integr. Biol. 4, 1274–1282 (2012).

    Article  CAS  Google Scholar 

  46. Feng, Y. et al. Global analysis of protein structural changes in complex proteomes. Nat. Biotechnol. 32, 1036–1044 (2014).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank S. Alberti (Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany), M. Ralser (Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK) and K. Weis (Institute of Biochemistry, Department of Biology, ETH Zürich, Zürich, Switzerland) for providing reagents; S. McKnight, D. Frantz and L. Berchowitz for help with b-isox experiments; A. Timofiiva for help with microscopy; and T. Mayor, A. Smith, P. Kimmig and members of the Peter laboratory for helpful discussions and comments on the manuscript. This work was funded by the Swiss National Science Foundation (SNF, project grants 31003A_166513 to R.D. and 310030B_160312 to M.P.), the European Research Council (ERC, Rubinet) and ETH Zürich.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: S.S., M.P. and R.D.; investigation: S.S., G.C. and Y.F.; formal analysis: R.D.; writing—original draft preparation: S.S. and R.D.; writing—review and editing: S.S., G.C., Y.F., P.P., M.P. and R.D.; visualization: S.S. and R.D.; supervision: P.P., M.P. and R.D.; funding acquisition: M.P. and R.D.

Corresponding authors

Correspondence to Matthias Peter or Reinhard Dechant.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Cdc19 forms reversible aggregates upon stress and tetrameric Cdc19ΔPEP is unable to support cell growth in yeast.

(a,b) Major protein degradation pathways do not significantly contribute to reversibility of Cdc19 aggregates. Cells expressing Cdc19-GFP and the indicated mutations were grown to stationary phase, loaded into a microfluidic chip and reversibility of aggregates was scored upon glucose readdition in the presence of (a) PMSF (prevents vacuolar degradation) or (b) MG132 to inhibit proteasomal degradation. Note that expression of the Pdr1-Cyc8 fusion necessary for efficient inhibition of proteasomal activity by MG132. The reason for faster solubilisation of aggregates in this strain is unclear. Arrowheads indicate frames with near-complete solubilisation of detectable aggregates. Time relative to glucose readdition is indicated. Data shown are representative of 3 independent experiments. (c) Cdc19-aggregates in stationary phase are solid-like. Cells were grown to stationary phase and Cdc19-GFP aggregates were subjected to FRAP analysis as described in Fig. 1. Quantification of fluorescence recovery for all analyzed cells is shown as the mean of the relative fluorescence intensity as a function of time (red line) together with the individual cell traces (light and dark gray) (n = 3 independent experiments). (d) Cells harboring the tetO7-CDC19 allele expressing wild-type or mutant Cdc19 or an empty vector were grown in the presence or absence of doxycycline (10 μg/ml, added at t = 0h) and growth was analyzed using a BioLector (m2p-labs). Accumulation of biomass is shown as a function of time. Data shown are representative of 2 independent experiments. (e) Cytoplasmic acidification is not required for the formation of Cdc19 aggregates. Cells were grown as in Fig. 1a and representative images of Cdc19-GFP upon glucose starvation in C-starvation media (pH 4.6 or pH 7.4) or PBS (pH 7.4) from 3 independent experiments are shown. In all images, the scale bar represents 3 μm. Statistical source data for c can be found in Supplementary Table 4.

Supplementary Figure 2 Cdc19 colocalizes with RNA containing stress granules and failure of recruitment renders Cdc19 unstable.

(a) Cdc19 aggregates colocalize with the stress granule marker Pub1. Cells expressing Cdc19-GFP and Pub1-RFP were starved and localization of Cdc19 aggregates relative to Pub1 was analyzed. Data shown are representative of 2 independent experiments. (b) Cdc19 aggregates colocalize with the stress granule marker Pab1, but not with the P-body marker Edc3. Cells expressing Cdc19-GFP, Pab1-CFP and Edc3-RFP were grown as in Fig. 1 and timing of the appearance of P-bodies and stress granules and their localization was analyzed relative to Cdc19 aggregation. Data shown are representative of 3 independent experiments. Time relative to glucose readdition is indicated. Note that Cdc19-GFP was imaged using YFP filter sets to avoid any spectral mixing in cells expressing Pab1-CFP. (c) Cells expressing wild-type Cdc19-TAP or Cdc19ΔPEP-TAP and Hxk2-TAP as control were grown in SD media and UV treated. Cross-linked protein-mRNA complexes were isolated using poly-dT and analyzed by western-blot. Data shown are representative of 3 independent experiments. (d) Cells expressing wild-type or mutant Cdc19-GFP expressing Pab1-CFP were starved for glucose and localization of Cdc19 and Pab1 was analyzed at the indicated time-points. Note that Cdc19-GFP was imaged using YFP filter sets to avoid any spectral mixing in cells expressing Pab1-CFP. Data shown are representative of 3 independent experiments. (e) Cells expressing wild-type or mutant Cdc19 were grown as in Fig. 3g and relative abundance of Cdc19 was determined by mass-spectrometry and plotted as the mean ± S.E.M. of three technical replicates. In all images, the scale bar represents 3 μm. Uncropped blot is shown in Supplementary Fig. 5.

Supplementary Figure 3 Cdc19 forms Thioflavin-T positive aggregates in vitro and LCRs might generally contribute to aggregation.

(a) Wild-type and mutant Cdc19 incubated at the indicated temperatures were mixed with ThT as in Fig. 4e and emission spectra were recorded. Data shown are representative of 3 independent experiments. (b) Sbp1 and Ssa2 colocalize with stress granules upon glucose starvation. Cells expressing Sbp1-GFP or Ssa2-GFP and Pab1-CFP were grown as in Fig. 1 and scored for localization of the indicated proteins. Data shown are representative of 3 independent experiments. Note that Sbp1-GFP and Ssa2-GFP were imaged using YFP filter sets to avoid any spectral mixing in cells expressing Pab1-CFP. The scale bar represents 3 μm.

Supplementary Figure 4 Dephosphorylation of the Cdc19 LCR is required for reversible aggregation.

(a) Schematic representation of the MS-workflow to determine the fraction of phosphorylated peptides. Unphosphorylated peptides were measured with or without phosphatase treatment (PPase) and abundance relative to a control peptide was determined. (b) The LCR of Cdc19 is part of the subunit interface in Cdc19 tetramers. Crystal structure of Cdc19 showing a part of the subunit interface of Cdc19 tetramers. Blue: ribbon representation of Cdc19 with the region corresponding to the LCR shown in yellow. Thr372, Thr376, Ser377 and Ser385 are shown in space filling representation. Other subunits of the Cdc19 tetramer (light gray, dark-gray, red) are shown as surface representations. Note that Ser385 forms intersubunit contacts (hydrogen bond via side chain-OH), strongly suggesting that phosphorylation at this site would be incompatible with phosphorylation. (c) Cells expressing wild-type or mutant Cdc19-GFP expressing Pab1-CFP were starved for glucose and localization of Cdc19 and Pab1 was analyzed at the indicated time-points. Note that Cdc19-GFP was imaged using YFP filter sets to avoid any spectral mixing in cells expressing Pab1-CFP. Data shown are representative of 3 independent experiments. (d) Cells as in Fig. 7a were grown and analyzed for aggregate formation upon heat shock (30 min, 42 °C) and after recovery at 30 °C for 60 min. Representative images of 3 independent experiments for the indicated conditions are shown. In all images, the scale bar represents 3 μm.

Supplementary Figure 5 Unprocessed scans of Coomassie-stained gels and western blots.

Original scans of coomasie-stained gels from figs 3d and 4c. Original scans of western blots using a Fusion Fx (Vilber Lourmat) imager from Figs 1d, 2a, 3d, g, 4d, 5b, 7b, f and Supplementary Fig. 2c.

Supplementary information

Supplementary Information

Supplementary Information (PDF 4512 kb)

Life Sciences Reporting Summary (PDF 83 kb)

Supplementary Table 1

Supplementary Information (XLSX 30 kb)

Supplementary Table 2

Supplementary Information (XLSX 28 kb)

Supplementary Table 3

Supplementary Information (XLSX 44 kb)

Supplementary Table 4

Supplementary Information (XLSX 145 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Saad, S., Cereghetti, G., Feng, Y. et al. Reversible protein aggregation is a protective mechanism to ensure cell cycle restart after stress. Nat Cell Biol 19, 1202–1213 (2017). https://doi.org/10.1038/ncb3600

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3600

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