添加链接
link之家
链接快照平台
  • 输入网页链接,自动生成快照
  • 标签化管理网页链接
The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely. As a library, NLM provides access to scientific literature. Inclusion in an NLM database does not imply endorsement of, or agreement with, the contents by NLM or the National Institutes of Health.
Learn more: PMC Disclaimer ] strain was created by repeated passaging of WT YPH499 cells on YPD containing 3 mM GDN (guanidine HCl). Prion status was confirmed by transient expression of Rnq1-GFP and examination for isolates with soluble Rnq1-GFP by microscopy.

The SSA1-GFP strain was created by transforming strain YPH499 with BstEII (New England Biolabs) cut plasmid pRS405P ADH Ssa1GFP and selection on media lacking leucine.

The SIS1-GFP strain was created by transforming a PCR generated cassette using pFA6a-GFP(S65T)-KanMX6 108 as a template with primers 5 Sis1-GFP and 3 Sis1-GFP into strain YPH499. Transformants were selected by growth on media containing 300 μg/ml G418 and confirmed by genomic PCR.

The mCherry-NLS strains were created by transforming the SSA1-GFP or SIS1-GFP strains with BsmI (New England Biolabs) cut plasmid pRS306P GPD -GST-mCh-NLS and selection on media lacking uracil.

Growth and heat treatment

Yeast strains were cultured at 30 °C, unless stated otherwise. Heat treatments were conducted in a pre-warmed 37 °C or 39 °C incubator, whereas treatment at 50 °C was performed in a 50 °C shaking heat block. Cultures were grown in synthetic complete media lacking amino acids necessary for plasmid selection and supplemented with either 2% glucose or 2% raffinose and 2% galactose. Experimental comparisons were made between identical growth conditions unless indicated otherwise. Cultures were maintained for >18 h with dilution and harvested during log phase.

HEK293T and DnaJB6 knockout cells 62 (Supplementary Table 6 ) were cultured in DMEM supplemented with 10% Fetal Bovine Serum, Penicillin and Streptomycin in a 37 °C humidified incubator at 5% CO 2 . Passaging was performed twice weekly. Transient transfections were performed using 1 µg of DNA and PEI transfection reagent (1:6 ratio). Heat shock experiments were performed for 1 h at 43 °C in a water bath. Samples were collected after ~24 h recovery at 37 o C, unless otherwise indicated.

Preparation of cell extracts

Yeast pellets were resuspended in 500 μl lysis buffer (25 mM Tris pH 7.5, 50 mM KCl, 10 mM MgCl 2 , 1 mM EDTA, 5% glycerol, 0.5% Triton X-100) supplemented with protease inhibitors (cOmplete mini, EDTA-free Protease Inhibitor Cocktail, Roche). Cells were lysed by bead-milling with acid washed glass beads in a MP Beadbeater24 (2 × 20 s at 6.0 m/s with intermittent cooling on ice). Extracts were cleared from cell debris by centrifugation at 500 × g for 5 min at 4 °C. Protein levels were normalized using the Bio-Rad Protein Assay (Bio-Rad). Samples containing polyQ expanded Htt were first precipitated with 25% trichloroacetic acid (TCA) before dissolution with formic acid at 37 °C and preparation for SDS-PAGE 109 .

Mammalian cell pellets were resuspended in RIPA buffer (25 mM Tris pH 7.4, 150 mM NaCl, 1 mM MgCl 2 , 1% NP40, 1% sodium deoxycholate, 0.1% SDS) supplemented with protease inhibitors (Roche) and DENARASE (c-LEcta, 50 U/ml) and vortexed on ice for 1 h. Protein levels were normalized using the DC Protein Assay (Bio-Rad).

Cell fractionation

Total yeast cell lysates were separated into soluble and pellet fractions by centrifugation at 15,000 × g for 15 min at 4 °C. After collection of the soluble supernatant fraction, pellets were washed with lysis buffer (2 × 15,000 × g for 5 min at 4 °C).

SDS-PAGE

Protein samples were suspended in sample buffer (50 mM Tris pH 6.8, 8% glycerol, 2% SDS, 20% β-mercaptoethanol, containing bromophenol blue), heated to 95 °C for 3 min and separated by electrophoresis on 10 or 12% self-made Tris-glycine poly-acrylamide (Serva) or TGX FastCast acrylamide solution (Bio-Rad) gels using Laemmli (25 mM Tris, 192 mM glycine, 0.1% SDS) running buffer at 30 mA/gel.

Filter retardation assay

Filter retardation assays for the detection of total and SDS resistant polyQ aggregates were performed as previously described 26 , 37 , 45 . Cell lysates were incubated with 125 U benzonase (Novagen) or smDNase (Max Planck Institute for Biochemistry core facility) with rotation for 1 h at 4 °C. Samples were then incubated with or without SDS buffer (2% SDS, 50 mM DTT) before being loaded onto a pre-wetted (0.1% SDS) 0.2 μm pore size cellulose acetate membrane in a Hoefer slot-blot apparatus. Membranes were subsequently washed four times with 0.1% SDS before immunoblotting.

SDD-AGE

Semi-denaturing detergent agarose gel electrophoresis was performed as previously described 75 with a modified lysis buffer (25 mM Tris pH 7.5, 50 mM KCl, 10 mM MgCl 2 , 1 mM EDTA, 5% glycerol, 0.5% Triton X-100). Yeast extracts were incubated at 30 °C for 7 min in SDD-AGE sample buffer (50 mM Tris pH 6.8, 8% glycerol, 2% SDS, supplemented with bromophenol blue). Samples were then run on an agarose gel (1.5% agarose, 0.1% SDS, in 25 mM Tris, 192 mM glycine) in running buffer (25 mM Tris, 192 mM glycine, 0.1% SDS) at 160 V for 1 h at 4 °C.

Immunoblotting

Proteins were transferred from poly-acrylamide gels to nitrocellulose membranes in transfer buffer (25 mM Tris, 192 mM glycine, 20% methanol) at 110 V for 1 h using the Mini Trans-Blot Cell (Bio-Rad) or for 10 min using the Trans-Blot Turbo Transfer System (Bio-Rad). Proteins were transferred from agarose gels (SDD-AGE) to nitrocellulose membranes in transfer buffer containing 0.01% SDS at 7 V for 14 h using the Genie Blotter System (Research Products International). Membranes were washed with TBST (10 mM Tris-HCl pH7.5, 150 mM NaCl, 0.1% Tween-20), blocked with 4% powdered milk in TBST, and incubated with primary antibodies overnight at 4 °C. Information regarding the primary antibodies used in this study can be found in Supplementary Table 7 . After washing with TBST, membranes were incubated with horseradish peroxidase (HRP)-conjugated secondary antibodies for 1 h and washed again. Imaging was performed using Luminata Classico substrate and the ImageQuant LAS 4000 mini detector or Bio-Rad ChemiDoc imaging system. Densitometric analyses were performed using LICOR Image Studio Lite or Image Lab (Bio-Rad) software.

Immunoprecipitation after crosslinking

Yeast cells were pelleted and resuspended in crosslinking (XL) buffer (1.2 M sorbitol, 5 mM EDTA, 0.1 M KH 2 PO 4 /K 2 HPO 4 pH 7.5). DSP solution (50 mM dithiobis(succinimidyl propionate) in DMSO) was added to 1 mM final concentration and samples were incubated at 30 °C for 20 min before quenching by addition of 0.1 volume 1 M Tris pH 7.5 for 15 min. Lysates were prepared as above and soluble fractions isolated by centrifugation at 15,000 × g for 15 min at 4 °C. Thousand microgram of protein (determined by Bradford assay) were diluted in lysis buffer, total fractions removed, and remaining samples incubated with 50 μl Milltenyi μMACS magnetic anti-Myc beads for 1 h at 4 °C. Samples were passed over the magnetic columns and washed (4× lysis buffer with 1% Triton X-100 and 2 × 20 mM Tris-HCl pH 7.5) before elution with hot SDS sample buffer.

Confocal imaging

Mid-to-late log phase yeast cells (0.5–1 OD 600 ) were adhered to the chamber of a concanavalin A coated µ-Slide (Ibidi, 80626) for 5 min at room temperature. Cells were washed three times with culture media. Imaging was done with an Olympus (Tokyo, Japan) FV1000 confocal microscope setup equipped with an Olympus PLAPON 60×/NA1.42 oil immersion objective. The GFP fluorophore was detected using an excitation wavelength of 488 nm and emission of 505–540 nm. The mCherry fluorophore was detected using an excitation wavelength of 559 nm and emission of 575–675 nm. Image analysis was carried out in Fiji 110 .

Analysis of aggregate density

Cells were prepared as described above and imaged with an Olympus (Tokyo, Japan) FV1000 confocal microscope setup equipped with an Olympus PLAPON 60×/NA1.42 oil immersion objective using the same nonsaturating acquisition settings for all samples. Image analysis was carried out in Fiji 110 . To analyze the fluorescent density of aggregates, circular regions within the aggregate were selected and the average fluorescence signal was compared between samples. To obtain the total cellular fluorescence signal, the integrated fluorescence density over the whole area of the analyzed cells was calculated.

Analysis of Ssa1 enrichment

Cells were prepared as described above and imaged with an Olympus (Tokyo, Japan) FV1000 confocal microscope setup equipped with an Olympus PLAPON 60×/NA1.42 oil immersion objective using the same nonsaturating acquisition settings for all samples. Image analysis was carried out in Fiji 110 . Regions of interest were selected outlining the FLuc aggregate as well as a large area in the cell not containing any aggregate (soluble). The enrichment of Ssa1 in aggregates over soluble signal was calculated as the ratio between the average fluorescence of Ssa1 in these two regions. To obtain the total cellular fluorescence signal, the integrated fluorescence density over the whole area of the analyzed cells was calculated.

FRAP analysis

Samples were prepared as described above. In vivo fluorescence recovery after photobleaching (FRAP) experiments were carried out at MPIB Imaging Facility (Martinsried, Germany) on a ZEISS (Jena, Germany) LSM780 confocal laser scanning microscope equipped with a ZEISS Plan-APO 63×/NA1.46 oil immersion objective. Circular regions of constant size were bleached after 20 frames and monitored for at least 80 s (0.38 s/frame) for fluorescence recovery in a single focal plane. Image analysis was carried out in Fiji 110 . Fluorescence intensity data were corrected for photobleaching and normalized to the average fluorescence intensity before (1) and after (0) bleaching (relative fluorescence), except the experiment including soluble material, which was only normalized pre-bleach due to the rapid recovery (reported as fluorescence intensity).

β-Galactosidase activity measurements

LacZ expression was measured using a standard ß-galactosidase assay 103 . Briefly, cell pellets were resuspended in 700 µl of Z buffer (60 mM Na 2 HPO 4 , 40 mM NaH 2 PO 4 , 10 mM KCl, 1 mM MgSO 4 , 50 mM ß-mercaptoethanol) supplemented with 50 µl of 0.1% SDS and 50 µl of chloroform, vortexed and incubated for 5 min at 30 °C. Two hundred microliter of substrate (4 mg/ml ONPG (2-Nitrophenyl β- d -galactopyranoside, Sigma-Aldrich) in Z buffer) was added with further incubation (~3–13 min) before quenching with 350 µl 1 M Na 2 CO 3 , clearance by centrifugation at 500 × g , and spectrophotometric measurement at A 420 . Activity was calculated in standard Miller Units. 1 Miller Unit = 1000 × (A 420 / (O.D. 600 × ml × t min )).

mRNA sequencing

Cells were harvested in triplicate and sent to Novogene Co., Ltd. (Hong Kong) for RNA extraction, library preparation, Illumina paired-end sequencing, mapping, and bioinformatics analysis. Briefly, 1 µg RNA per sample was used for library generation using NEB Next Ultra RNA Library Prep Kit for Illumina (NEB). Library preparations were subjected to Illumina paired-end sequencing (150 bp, 10 million reads). Reads were indexed using Bowtie v2.2.3 and aligned to the reference genome using TopHat v2.0.12. Read numbers were counted using HTSeq v0.6.1 and gene expression was calculated using FPKM (Fragments per Kilobase of transcript sequence per Million base pairs sequenced) method. The differentially expressed genes between samples were calculated using the DESeq R package and the resulting p values were adjusted using the Benjamini and Hochberg false discovery rate approach. Differentially expressed genes are defined as having an adjusted p value less than 0.05. Gene ontology (GO) enrichment analysis of the differentially expressed genes was performed using the GOseq R package. Significance was corrected for gene length bias and adjusted p values less than 0.05 indicate a significantly enriched term.

Statistical analysis

Error bars represent standard deviation from at least three experiments. Significance was determined for two-sample comparisons using the unpaired t -test function with a threshold of two-tailed p values less than 0.05. Significance among multiple conditions were determined using ANOVA and Dunnett’s multiple comparisons test with a threshold of p < 0.05. Calculations were performed using Graphpad software.

Reporting summary

Further information on research design is available in the Nature Research Reporting Summary linked to this article.

Supplementary information

Supplementary Information (13M, pdf)
Peer Review File (242K, pdf)

Acknowledgements

We thank Johannes Buchner (Technical University of Munich), Elizabeth Craig (University of Wisconsin—Madison) and Ineke Braakman (University of Utrecht) for providing antibodies, Harm Kampinga (University Medical Center Groningen) for providing plasmids and cell lines, Eduardo de Mattos and Arun Thiruvalluvan (University Medical Center Groningen) for assistance with the DnaJB6 cell line, and Yury Kukushkin and Sae-Hun Park for preliminary data. We thank Lucas Cairo, Gopal Jayaraj, and Sae-Hun Park for critically reading the manuscript. FRAP experiments were performed at the Max Planck Institute of Biochemistry Imaging Core Facility. This research has received funding from the European Commission (FP7 GA ERC-2012-SyG_318987–ToPAG) and the Munich Cluster for Systems Neurology (SyNergy). C.L.K. acknowledges funding by the Alexander von Humboldt Foundation (Postdoctoral Fellowship 3.1-USA/1162753 HFST-P). M.H.M.G. was supported by a DFG fellowship through the Graduate School of Quantitative Biosciences Munich (QBM).

Source data

Source Data File (23M, xlsx)

Author contributions

C.L.K. planned and performed most experiments and analyzed the data. M.H.M.G. assisted with experiments and performed the imaging and FRAP analysis. F.U.H. conceived the project and participated in data interpretation together with the other authors. C.L.K and F.U.H. wrote the paper with contributions from M.S.H.

Funding

Open Access funding enabled and organized by Projekt DEAL.

Data availability

The transcriptome data discussed in this publication have been deposited in NCBI’s gene expression omnibus and are accessible through GEO series accession number {"type":"entrez-geo","attrs":{"text":"GSE151215","term_id":"151215"}} GSE151215 . The string database is available at https://string-db.org (v 11, organism S. cerevisiae). All other data supporting the findings of this study are available within the manuscript. Source data are provided with this paper.

Competing interests

The authors declare no competing interests.

Footnotes

Peer review information Nature Communications thanks the anonymous reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Supplementary information is available for this paper at 10.1038/s41467-020-20000-x.

References

1. Balch WE, Morimoto RI, Dillin A, Kelly JW. Adapting proteostasis for disease intervention. Science. 2008; 319 :916–919. doi: 10.1126/science.1141448. [ PubMed ] [ CrossRef ] [ Google Scholar ]
2. Hipp MS, Kasturi P, Hartl FU. The proteostasis network and its decline in ageing. Nat. Rev. Mol. Cell Biol. 2019; 20 :421–435. doi: 10.1038/s41580-019-0101-y. [ PubMed ] [ CrossRef ] [ Google Scholar ]
3. Klaips CL, Jayaraj GG, Hartl FU. Pathways of cellular proteostasis in aging and disease. J. Cell Biol. 2018; 217 :51–63. doi: 10.1083/jcb.201709072. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
4. Sontag EM, Samant RS, Frydman J. Mechanisms and functions of spatial protein quality control. Annu. Rev. Biochem. 2017; 86 :97–122. doi: 10.1146/annurev-biochem-060815-014616. [ PubMed ] [ CrossRef ] [ Google Scholar ]
5. Walter P, Ron D. The unfolded protein response: from stress pathway to homeostatic regulation. Science. 2011; 334 :1081–1086. doi: 10.1126/science.1209038. [ PubMed ] [ CrossRef ] [ Google Scholar ]
6. Jayaraj, G. G., Hipp, M. S. & Hartl, F. U. Functional modules of the proteostasis network. Cold Spring Harb. Perspect. Biol . 12 , a033951 (2020). [ PMC free article ] [ PubMed ]
7. Lindquist S, Craig EA. The heat-shock proteins. Annu. Rev. Genet. 1988; 22 :631–677. doi: 10.1146/annurev.ge.22.120188.003215. [ PubMed ] [ CrossRef ] [ Google Scholar ]
8. Morimoto RI. Regulation of the heat shock transcriptional response: cross talk between a family of heat shock factors, molecular chaperones, and negative regulators. Genes Dev. 1998; 12 :3788–3796. doi: 10.1101/gad.12.24.3788. [ PubMed ] [ CrossRef ] [ Google Scholar ]
9. Baler R, Welch WJ, Voellmy R. Heat shock gene regulation by nascent polypeptides and denatured proteins: hsp70 as a potential autoregulatory factor. J. Cell Biol. 1992; 117 :1151–1159. doi: 10.1083/jcb.117.6.1151. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
10. Krakowiak J, et al. Hsf1 and Hsp70 constitute a two-component feedback loop that regulates the yeast heat shock response. eLife. 2018; 7 :e31668. doi: 10.7554/eLife.31668. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
11. Zheng X, et al. Dynamic control of Hsf1 during heat shock by a chaperone switch and phosphorylation. eLife. 2016; 5 :e18638. doi: 10.7554/eLife.18638. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
12. Anckar J, Sistonen L. Regulation of HSF1 function in the heat stress response: implications in aging and disease. Annu. Rev. Biochem. 2011; 80 :1089–1115. doi: 10.1146/annurev-biochem-060809-095203. [ PubMed ] [ CrossRef ] [ Google Scholar ]
13. Veri, A. O., Robbins, N. & Cowen, L. E. Regulation of the heat shock transcription factor Hsf1 in fungi: implications for temperature-dependent virulence traits. FEMS Yeast Res . 18 , foy041 (2018). [ PMC free article ] [ PubMed ]
14. Masser AE, et al. Cytoplasmic protein misfolding titrates Hsp70 to activate nuclear Hsf1. eLife. 2019; 8 :e47791. doi: 10.7554/eLife.47791. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
15. Gomez-Pastor R, Burchfiel ET, Thiele DJ. Regulation of heat shock transcription factors and their roles in physiology and disease. Nat. Rev. Mol. Cell Biol. 2018; 19 :4–19. doi: 10.1038/nrm.2017.73. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
16. Pincus D. Regulation of Hsf1 and the heat shock response. Adv. Exp. Med. Biol. 2020; 1243 :41–50. doi: 10.1007/978-3-030-40204-4_3. [ PubMed ] [ CrossRef ] [ Google Scholar ]
17. Lamech LT, Haynes CM. The unpredictability of prolonged activation of stress response pathways. J. Cell Biol. 2015; 209 :781–787. doi: 10.1083/jcb.201503107. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
18. Roth DM, et al. Modulation of the maladaptive stress response to manage diseases of protein folding. PLoS Biol. 2014; 12 :e1001998. doi: 10.1371/journal.pbio.1001998. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
19. Labbadia J, Morimoto RI. Repression of the heat shock response Is a programmed event at the onset of reproduction. Mol. Cell. 2015; 59 :639–650. doi: 10.1016/j.molcel.2015.06.027. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
20. Cates J, et al. Sensing the heat stress by mammalian cells. BMC Biophys. 2011; 4 :16. doi: 10.1186/2046-1682-4-16. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
21. Triandafillou, C. G., Katanski, C. D., Dinner, A. R. & Drummond, D. A. Transient intracellular acidification regulates the core transcriptional heat shock response. bioRxiv 414706 (2019). [ PMC free article ] [ PubMed ]
22. Dobson, C. M., Knowles, T. P. J. & Vendruscolo, M. The amyloid phenomenon and its significance in biology and medicine. Cold Spring Harb. Perspect. Biol . 12 , ao33878 (2020). [ PMC free article ] [ PubMed ]
23. Taylor, R. C. & Dillin, A. Aging as an event of proteostasis collapse. Cold Spring Harb. Perspect. Biol . 3 , a004440 (2011). [ PMC free article ] [ PubMed ]
24. Morimoto, R. I. Cell-nonautonomous regulation of proteostasis in aging and disease. Cold Spring Harb. Perspect. Biol . 12 , a034074 (2020). [ PMC free article ] [ PubMed ]
25. Olzscha H, et al. Amyloid-like aggregates sequester numerous metastable proteins with essential cellular functions. Cell. 2011; 144 :67–78. doi: 10.1016/j.cell.2010.11.050. [ PubMed ] [ CrossRef ] [ Google Scholar ]
26. Park SH, et al. PolyQ proteins interfere with nuclear degradation of cytosolic proteins by sequestering the Sis1p chaperone. Cell. 2013; 154 :134–145. doi: 10.1016/j.cell.2013.06.003. [ PubMed ] [ CrossRef ] [ Google Scholar ]
27. Woerner AC, et al. Cytoplasmic protein aggregates interfere with nucleocytoplasmic transport of protein and RNA. Science. 2016; 351 :173–176. doi: 10.1126/science.aad2033. [ PubMed ] [ CrossRef ] [ Google Scholar ]
28. Kim YE, et al. Soluble oligomers of polyQ-expanded huntingtin target a multiplicity of key cellular factors. Mol. Cell. 2016; 63 :951–964. doi: 10.1016/j.molcel.2016.07.022. [ PubMed ] [ CrossRef ] [ Google Scholar ]
29. Yu A, et al. Tau protein aggregates inhibit the protein-folding and vesicular trafficking arms of the cellular proteostasis network. J. Biol. Chem. 2019; 294 :7917–7930. doi: 10.1074/jbc.RA119.007527. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
30. Ashkenazi A, et al. Polyglutamine tracts regulate beclin 1-dependent autophagy. Nature. 2017; 545 :108–111. doi: 10.1038/nature22078. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
31. Deriziotis P, et al. Misfolded PrP impairs the UPS by interaction with the 20S proteasome and inhibition of substrate entry. EMBO J. 2011; 30 :3065–3077. doi: 10.1038/emboj.2011.224. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
32. Holmes WM, Klaips CL, Serio TR. Defining the limits: protein aggregation and toxicity in vivo. Crit. Rev. Biochem. Mol. Biol. 2014; 49 :294–303. doi: 10.3109/10409238.2014.914151. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
33. Gil, R. S. et al. Neurodegenerative disease-associated protein aggregates are poor inducers of the heat shock response in neuronal-like cells. bioRxiv, 10.1101/2020.01.06.896654 (2020). [ PubMed ]
34. Gomez-Pastor R, et al. Abnormal degradation of the neuronal stress-protective transcription factor HSF1 in Huntington’s disease. Nat. Commun. 2017; 8 :14405. doi: 10.1038/ncomms14405. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
35. Kitamura A, et al. Cytosolic chaperonin prevents polyglutamine toxicity with altering the aggregation state. Nat. Cell Biol. 2006; 8 :1163–1169. doi: 10.1038/ncb1478. [ PubMed ] [ CrossRef ] [ Google Scholar ]
36. Hageman J, et al. A DNAJB chaperone subfamily with HDAC-dependent activities suppresses toxic protein aggregation. Mol. Cell. 2010; 37 :355–369. doi: 10.1016/j.molcel.2010.01.001. [ PubMed ] [ CrossRef ] [ Google Scholar ]
37. Muchowski PJ, et al. Hsp70 and hsp40 chaperones can inhibit self-assembly of polyglutamine proteins into amyloid-like fibrils. Proc. Natl Acad. Sci. USA. 2000; 97 :7841–7846. doi: 10.1073/pnas.140202897. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
38. Wolfe KJ, Ren HY, Trepte P, Cyr DM. The Hsp70/90 cochaperone, Sti1, suppresses proteotoxicity by regulating spatial quality control of amyloid-like proteins. Mol. Biol. Cell. 2013; 24 :3588–3602. doi: 10.1091/mbc.e13-06-0315. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
39. Aprile FA, et al. The molecular chaperones DNAJB6 and Hsp70 cooperate to suppress α-synuclein aggregation. Sci. Rep. 2017; 7 :9039. doi: 10.1038/s41598-017-08324-z. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
40. Chen S, Brown IR. Neuronal expression of constitutive heat shock proteins: implications for neurodegenerative diseases. Cell Stress Chaperones. 2007; 12 :51–58. doi: 10.1379/CSC-236R.1. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
41. Sittler A, et al. Geldanamycin activates a heat shock response and inhibits huntingtin aggregation in a cell culture model of Huntington’s disease. Hum. Mol. Genet. 2001; 10 :1307–1315. doi: 10.1093/hmg/10.12.1307. [ PubMed ] [ CrossRef ] [ Google Scholar ]
42. Calamini, B. et al. Probe Reports from the NIH Molecular Libraries Program (National Center for Biotechnology Information, Bethesda, 2010). [ PubMed ]
43. Duennwald ML, Jagadish S, Muchowski PJ, Lindquist S. Flanking sequences profoundly alter polyglutamine toxicity in yeast. Proc. Natl Acad. Sci. USA. 2006; 103 :11045–11050. doi: 10.1073/pnas.0604547103. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
44. Krobitsch S, Lindquist S. Aggregation of huntingtin in yeast varies with the length of the polyglutamine expansion and the expression of chaperone proteins. Proc. Natl Acad. Sci. USA. 2000; 97 :1589–1594. doi: 10.1073/pnas.97.4.1589. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
45. Wanker, E. E. et al. Methods in Enzymology Vol. 309, 375–386 (Academic Press, Cambridge, 1999). [ PubMed ]
46. Gruber A, et al. Molecular and structural architecture of polyQ aggregates in yeast. Proc. Natl Acad. Sci. USA. 2018; 115 :E3446–E3453. doi: 10.1073/pnas.1717978115. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
47. Peskett TR, et al. A liquid to solid phase transition underlying pathological huntingtin exon1 aggregation. Mol. Cell. 2018; 70 :588–601.e586. doi: 10.1016/j.molcel.2018.04.007. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
48. Ripaud L, et al. Overexpression of Q-rich prion-like proteins suppresses polyQ cytotoxicity and alters the polyQ interactome. Proc. Natl Acad. Sci. USA. 2014; 111 :18219–18224. doi: 10.1073/pnas.1421313111. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
49. Liu XD, Liu PC, Santoro N, Thiele DJ. Conservation of a stress response: human heat shock transcription factors functionally substitute for yeast HSF. EMBO J. 1997; 16 :6466–6477. doi: 10.1093/emboj/16.21.6466. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
50. Chafekar SM, Duennwald ML. Impaired heat shock response in cells expressing full-length polyglutamine-expanded Huntingtin. PLoS ONE. 2012; 7 :e37929. doi: 10.1371/journal.pone.0037929. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
51. Kayatekin C, et al. Prion-like proteins sequester and suppress the toxicity of huntingtin exon 1. Proc. Natl Acad. Sci. USA. 2014; 111 :12085–12090. doi: 10.1073/pnas.1412504111. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
52. Gokhale KC, Newnam GP, Sherman MY, Chernoff YO. Modulation of prion-dependent polyglutamine aggregation and toxicity by chaperone proteins in the yeast model. J. Biol. Chem. 2005; 280 :22809–22818. doi: 10.1074/jbc.M500390200. [ PubMed ] [ CrossRef ] [ Google Scholar ]
53. Sondheimer N, Lopez N, Craig EA, Lindquist S. The role of Sis1 in the maintenance of the [RNQ+] prion. EMBO J. 2001; 20 :2435–2442. doi: 10.1093/emboj/20.10.2435. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
54. Hines JK, Higurashi T, Srinivasan M, Craig EA. Influence of prion variant and yeast strain variation on prion-molecular chaperone requirements. Prion. 2011; 5 :238–244. doi: 10.4161/pri.17818. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
55. Meriin AB, et al. Huntington toxicity in yeast model depends on polyglutamine aggregation mediated by a prion-like protein Rnq1. J. Cell Biol. 2002; 157 :997–1004. doi: 10.1083/jcb.200112104. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
56. Rosenzweig R, Nillegoda NB, Mayer MP, Bukau B. The Hsp70 chaperone network. Nat. Rev. Mol. Cell Biol. 2019; 20 :665–680. doi: 10.1038/s41580-019-0133-3. [ PubMed ] [ CrossRef ] [ Google Scholar ]
57. Clerico EM, et al. Hsp70 molecular chaperones: multifunctional allosteric holding and unfolding machines. Biochem. J. 2019; 476 :1653–1677. doi: 10.1042/BCJ20170380. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
58. Yu HY, Ziegelhoffer T, Craig EA. Functionality of class A and class B J-protein co-chaperones with Hsp70. FEBS Lett. 2015; 589 :2825–2830. doi: 10.1016/j.febslet.2015.07.040. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
59. Kampinga HH, Craig EA. The HSP70 chaperone machinery: J proteins as drivers of functional specificity. Nat. Rev. Mol. Cell Biol. 2010; 11 :579–592. doi: 10.1038/nrm2941. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
60. Gillis J, et al. The DNAJB6 and DNAJB8 protein chaperones prevent intracellular aggregation of polyglutamine peptides. J. Biol. Chem. 2013; 288 :17225–17237. doi: 10.1074/jbc.M112.421685. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
61. Kakkar V, et al. The S/T-rich motif in the DNAJB6 chaperone delays polyglutamine aggregation and the onset of disease in a mouse model. Mol. Cell. 2016; 62 :272–283. doi: 10.1016/j.molcel.2016.03.017. [ PubMed ] [ CrossRef ] [ Google Scholar ]
62. Thiruvalluvan A, et al. DNAJB6, a key factor in neuronal sensitivity to amyloidogenesis. Mol. Cell. 2020; 78 :346–358.e349. doi: 10.1016/j.molcel.2020.02.022. [ PubMed ] [ CrossRef ] [ Google Scholar ]
63. Meng E, Shevde LA, Samant RS. Emerging roles and underlying molecular mechanisms of DNAJB6 in cancer. Oncotarget. 2016; 7 :53984–53996. doi: 10.18632/oncotarget.9803. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ] Retracted
64. Sugars KL, Rubinsztein DC. Transcriptional abnormalities in Huntington disease. Trends Genet. 2003; 19 :233–238. doi: 10.1016/S0168-9525(03)00074-X. [ PubMed ] [ CrossRef ] [ Google Scholar ]
65. Tsoi H, Lau TC-K, Tsang S-Y, Lau K-F, Chan HYE. CAG expansion induces nucleolar stress in polyglutamine diseases. Proc. Natl Acad. Sci. USA. 2012; 109 :13428–13433. doi: 10.1073/pnas.1204089109. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
66. Pincus D, et al. Genetic and epigenetic determinants establish a continuum of Hsf1 occupancy and activity across the yeast genome. Mol. Biol. Cell. 2018; 29 :3168–3182. doi: 10.1091/mbc.E18-06-0353. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
67. Solís E J, et al. Defining the essential function of yeast Hsf1 reveals a compact transcriptional program for maintaining eukaryotic proteostasis. Mol. Cell. 2016; 63 :60–71. doi: 10.1016/j.molcel.2016.05.014. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
68. Gong Y, et al. An atlas of chaperone-protein interactions in Saccharomyces cerevisiae: implications to protein folding pathways in the cell. Mol. Syst. Biol. 2009; 5 :275–275. doi: 10.1038/msb.2009.26. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
69. Duennwald ML, Lindquist S. Impaired ERAD and ER stress are early and specific events in polyglutamine toxicity. Genes Dev. 2008; 22 :3308–3319. doi: 10.1101/gad.1673408. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
70. Leitman J, Ulrich Hartl F, Lederkremer GZ. Soluble forms of polyQ-expanded huntingtin rather than large aggregates cause endoplasmic reticulum stress. Nat. Commun. 2013; 4 :2753. doi: 10.1038/ncomms3753. [ PubMed ] [ CrossRef ] [ Google Scholar ]
71. Travers KJ, et al. Functional and genomic analyses reveal an essential coordination between the unfolded protein response and ER-associated degradation. Cell. 2000; 101 :249–258. doi: 10.1016/S0092-8674(00)80835-1. [ PubMed ] [ CrossRef ] [ Google Scholar ]
72. Cox JS, Walter P. A novel mechanism for regulating activity of a transcription factor that controls the unfolded protein response. Cell. 1996; 87 :391–404. doi: 10.1016/S0092-8674(00)81360-4. [ PubMed ] [ CrossRef ] [ Google Scholar ]
73. Klaips CL, Hochstrasser ML, Langlois CR, Serio TR. Spatial quality control bypasses cell-based limitations on proteostasis to promote prion curing. Elife. 2014; 3 :e04288. doi: 10.7554/eLife.04288. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
74. Morley JF, Brignull HR, Weyers JJ, Morimoto RI. The threshold for polyglutamine-expansion protein aggregation and cellular toxicity is dynamic and influenced by aging in Caenorhabditis elegans. Proc. Natl Acad. Sci. USA. 2002; 99 :10417–10422. doi: 10.1073/pnas.152161099. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
75. Kryndushkin DS, Alexandrov IM, Ter-Avanesyan MD, Kushnirov VV. Yeast [PSI+] prion aggregates are formed by small Sup35 polymers fragmented by Hsp104. J. Biol. Chem. 2003; 278 :49636–49643. doi: 10.1074/jbc.M307996200. [ PubMed ] [ CrossRef ] [ Google Scholar ]
76. Horton LE, James P, Craig EA, Hensold JO. The yeast hsp70 homologue Ssa is required for translation and interacts with Sis1 and Pab1 on translating ribosomes. J. Biol. Chem. 2001; 276 :14426–14433. doi: 10.1074/jbc.M100266200. [ PubMed ] [ CrossRef ] [ Google Scholar ]
77. Tsai J, Douglas MG. A conserved HPD sequence of the J-domain is necessary for YDJ1 stimulation of Hsp70 ATPase activity at a site distinct from substrate binding. J. Biol. Chem. 1996; 271 :9347–9354. doi: 10.1074/jbc.271.16.9347. [ PubMed ] [ CrossRef ] [ Google Scholar ]
78. Yan W, Craig EA. The glycine-phenylalanine-rich region determines the specificity of the yeast Hsp40 Sis1. Mol. Cell. Biol. 1999; 19 :7751–7758. doi: 10.1128/MCB.19.11.7751. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
79. Borges J, et al. Identification of regions involved in substrate binding and dimer stabilization within the central domains of yeast Hsp40 Sis1. PLoS ONE. 2012; 7 :e50927. doi: 10.1371/journal.pone.0050927. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
80. Peffer S, Goncalves D, Morano KA. Regulation of the Hsf1-dependent transcriptome via conserved bipartite contacts with Hsp70 promotes survival in yeast. J. Biol. Chem. 2019; 294 :12191–12202. doi: 10.1074/jbc.RA119.008822. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
81. Mosser DD, Duchaine J, Massie B. The DNA-binding activity of the human heat shock transcription factor is regulated in vivo by hsp70. Mol. Cell. Biol. 1993; 13 :5427–5438. doi: 10.1128/MCB.13.9.5427. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
82. Ghaemmaghami S, et al. Global analysis of protein expression in yeast. Nature. 2003; 425 :737–741. doi: 10.1038/nature02046. [ PubMed ] [ CrossRef ] [ Google Scholar ]
83. Leung TK, Rajendran MY, Monfries C, Hall C, Lim L. The human heat-shock protein family. Expression of a novel heat-inducible HSP70 (HSP70B’) and isolation of its cDNA and genomic DNA. Biochem. J. 1990; 267 :125–132. doi: 10.1042/bj2670125. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
84. Raychaudhuri S, et al. Interplay of acetyltransferase EP300 and the proteasome system in regulating heat shock transcription factor 1. Cell. 2014; 156 :975–985. doi: 10.1016/j.cell.2014.01.055. [ PubMed ] [ CrossRef ] [ Google Scholar ]
85. Gupta R, et al. Firefly luciferase mutants as sensors of proteome stress. Nat. Methods. 2011; 8 :879–884. doi: 10.1038/nmeth.1697. [ PubMed ] [ CrossRef ] [ Google Scholar ]
86. Kroschwald S, et al. Different material states of Pub1 condensates define distinct modes of stress adaptation and recovery. Cell Rep. 2018; 23 :3327–3339. doi: 10.1016/j.celrep.2018.05.041. [ PubMed ] [ CrossRef ] [ Google Scholar ]
87. Kroschwald S, et al. Promiscuous interactions and protein disaggregases determine the material state of stress-inducible RNP granules. eLife. 2015; 4 :e06807. doi: 10.7554/eLife.06807. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
88. Banani SF, Lee HO, Hyman AA, Rosen MK. Biomolecular condensates: organizers of cellular biochemistry. Nat. Rev. Mol. Cell. Biol. 2017; 18 :285–298. doi: 10.1038/nrm.2017.7. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
89. Alberti S, Carra S. Quality control of membraneless organelles. J. Mol. Biol. 2018; 430 :4711–4729. doi: 10.1016/j.jmb.2018.05.013. [ PubMed ] [ CrossRef ] [ Google Scholar ]
90. Dormann, D. FG-nucleoporins caught in the act of liquid–liquid phase separation. J. Cell Biol . 219 , e201910211 (2019). [ PMC free article ] [ PubMed ]
91. Schmidt HB, Görlich D. Transport selectivity of nuclear pores, phase separation, and membraneless organelles. Trends Biochem. Sci. 2016; 41 :46–61. doi: 10.1016/j.tibs.2015.11.001. [ PubMed ] [ CrossRef ] [ Google Scholar ]
92. Lopez N, Aron R, Craig EA. Specificity of class II Hsp40 Sis1 in maintenance of yeast prion [RNQ+] Mol. Biol. Cell. 2003; 14 :1172–1181. doi: 10.1091/mbc.e02-09-0593. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
93. Aron R, Lopez N, Walter W, Craig EA, Johnson J. In vivo bipartite interaction between the Hsp40 Sis1 and Hsp70 in Saccharomyces cerevisiae. Genetics. 2005; 169 :1873–1882. doi: 10.1534/genetics.104.037242. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
94. Sarparanta J, et al. Mutations affecting the cytoplasmic functions of the co-chaperone DNAJB6 cause limb-girdle muscular dystrophy. Nat. Genet. 2012; 44 :450–455. doi: 10.1038/ng.1103. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
95. Escusa-Toret S, Vonk WIM, Frydman J. Spatial sequestration of misfolded proteins by a dynamic chaperone pathway enhances cellular fitness during stress. Nat. Cell Biol. 2013; 15 :1231–1243. doi: 10.1038/ncb2838. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
96. Lu Z, Cyr DM. Protein folding activity of Hsp70 Is modified differentially by the Hsp40 co-chaperones Sis1 and Ydj1. J. Biol. Chem. 1998; 273 :27824–27830. doi: 10.1074/jbc.273.43.27824. [ PubMed ] [ CrossRef ] [ Google Scholar ]
97. Feder, Z. A. et al. Subcellular localization of the J-protein Sis1 regulates the heat shock response. bioRxiv 10.1101/2020.04.02.022491 (2020). [ PMC free article ] [ PubMed ]
98. Alford, B. D., Valiant, G. & Brandman, O. Genome-wide, time-sensitive interrogation of the heat shock response under diverse stressors via ReporterSeq. bioRxiv 10.1101/2020.03.29.014845 (2020).
99. Frottin F, et al. The nucleolus functions as a phase-separated protein quality control compartment. Science. 2019; 365 :342–347. doi: 10.1126/science.aaw9157. [ PubMed ] [ CrossRef ] [ Google Scholar ]
100. Liebman SW, Chernoff YO. Prions in yeast. Genetics. 2012; 191 :1041–1072. doi: 10.1534/genetics.111.137760. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
101. Smith C, D’Mello SR. Cell and context-dependent effects of the heat shock protein DNAJB6 on neuronal survival. Mol. Neurobiol. 2016; 53 :5628–5639. doi: 10.1007/s12035-015-9452-3. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
102. Mumberg D, Müller R, Funk M. Yeast vectors for the controlled expression of heterologous proteins in different genetic backgrounds. Gene. 1995; 156 :119–122. doi: 10.1016/0378-1119(95)00037-7. [ PubMed ] [ CrossRef ] [ Google Scholar ]
103. Holmes WM, Mannakee BK, Gutenkunst RN, Serio TR. Loss of amino-terminal acetylation suppresses a prion phenotype by modulating global protein folding. Nat. Commun. 2014; 5 :4383. doi: 10.1038/ncomms5383. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
104. Sikorski RS, Hieter P. A system of shuttle vectors and yeast host strains designed for efficient manipulation of DNA in Saccharomyces cerevisiae. Genetics. 1989; 122 :19–27. [ PMC free article ] [ PubMed ] [ Google Scholar ]
105. Douglas PM, et al. Chaperone-dependent amyloid assembly protects cells from prion toxicity. Proc. Natl Acad. Sci. USA. 2008; 105 :7206–7211. doi: 10.1073/pnas.0802593105. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
106. Harding RJ, et al. Design and characterization of mutant and wildtype huntingtin proteins produced from a toolkit of scalable eukaryotic expression systems. J. Biol. Chem. 2019; 294 :6986–7001. doi: 10.1074/jbc.RA118.007204. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]
107. Garí E, Piedrafita L, Aldea M, Herrero E. A set of vectors with a tetracycline-regulatable promoter system for modulated gene expression in Saccharomyces cerevisiae. Yeast. 1997; 13 :837–848. doi: 10.1002/(SICI)1097-0061(199707)13:9<837::AID-YEA145>3.0.CO;2-T. [ PubMed ] [ CrossRef ] [ Google Scholar ]
108. Bähler J, et al. Heterologous modules for efficient and versatile PCR-based gene targeting in Schizosaccharomyces pombe. Yeast. 1998; 14 :943–951. doi: 10.1002/(SICI)1097-0061(199807)14:10<943::AID-YEA292>3.0.CO;2-Y. [ PubMed ] [ CrossRef ] [ Google Scholar ]
109. Hazeki N, Tukamoto T, Goto J, Kanazawa I. Formic acid dissolves aggregates of an N-terminal huntingtin fragment containing an expanded polyglutamine tract: applying to quantification of protein components of the aggregates. Biochem. Biophys. Res. Commun. 2000; 277 :386–393. doi: 10.1006/bbrc.2000.3682. [ PubMed ] [ CrossRef ] [ Google Scholar ]
110. Schindelin J, et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods. 2012; 9 :676–682. doi: 10.1038/nmeth.2019. [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]

Articles from Nature Communications are provided here courtesy of Nature Publishing Group