• No results found

Hsp70 nucleotide exchange factor Fes1 is essential for ubiquitin-dependent degradation of misfolded cytosolic proteins

N/A
N/A
Protected

Academic year: 2022

Share "Hsp70 nucleotide exchange factor Fes1 is essential for ubiquitin-dependent degradation of misfolded cytosolic proteins"

Copied!
19
0
0

Loading.... (view fulltext now)

Full text

(1)

Stockholm University

This is an accepted version of a paper published in Proceedings of the National Academy of Sciences of the United States of America. This paper has been peer- reviewed but does not include the final publisher proof-corrections or journal pagination.

Citation for the published paper:

Gowda, N., Kandasamy, G., Froehlich, M., Dohmen, R., Andréasson, C. (2013)

"Hsp70 nucleotide exchange factor Fes1 is essential for ubiquitin-dependent degradation of misfolded cytosolic proteins."

Proceedings of the National Academy of Sciences of the United States of America, 110(15): 5975-80

Access to the published version may require subscription.

Permanent link to this version:

http://urn.kb.se/resolve?urn=urn:nbn:se:su:diva-89195

http://su.diva-portal.org

(2)

Title:

Hsp70 nucleotide exchange factor Fes1 is essential for ubiquitin- dependent degradation of misfolded cytosolic proteins

Authors:

Naveen Kumar Chandappa Gowda (Stockholm University) Ganapathi Kandasamy (University of Cologne)

Marceli S. Froehlich (University of Cologne) R. Jürgen Dohmen (University of Cologne) Claes Andréasson (Stockholm University) Published:

Proc Natl Acad Sci U S A. 2013 Apr 9;110(15):5975-80. doi:

10.1073/pnas.1216778110. Epub 2013 Mar 25.

Content:

Final submitted author version (see PNAS for published version)

16/4 2013

(3)

Submission PDF

Hsp70 nucleotide exchange factor Fes1 is essential for ubiquitin-dependent degradation of misfolded

cytosolic proteins

Naveen Kumar Chandappa Gowda1, Ganapathi Kandasamy2, Marceli S. Fröhlich2,#, R. Jürgen Dohmen2, Claes Andréasson1

1Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, S-10691 Stockholm, Sweden.2Institute for Genetics, University of Cologne, Cologne Biocenter, Zülpicher Strasse 47a, D-50674 Cologne, Germany.#Present address: Grünenthal GmbH, Global Early Clinical Development, Zieglerstraße 6, D-52078 Aachen, Germany

Submitted to Proceedings of the National Academy of Sciences of the United States of America Protein quality control systems protect cells against the accumula-

tion of toxic misfolded proteins by promoting their selective degra- dation. Malfunctions of quality control systems are linked to aging and neurodegenerative disease. Folding of polypeptides is facili- tated by the association of Hsp70 molecular chaperones. If folding cannot be achieved, Hsp70 interacts with ubiquitylation enzymes that promote the proteasomal degradation of the misfolded pro- tein. However, the factors that direct Hsp70 substrates towards the degradation machinery have remained unknown. Here, we identify Fes1, an Hsp70 nucleotide exchange factor of hitherto unclear physiological function, as a cytosolic triaging factor that promotes proteasomal degradation of misfolded proteins. Fes1 selectively interacts with misfolded proteins bound by Hsp70 and triggers their release from the chaperone. In the absence of Fes1, misfolded proteins fail to undergo polyubiquitylation, aggregate and induce a strong heat shock response. Our findings reveal that Hsp70 direct proteins towards either folding or degradation by employing distinct nucleotide exchange factors.

Hsp70 | Molecular Chaperone | proteasome | Quality control | ubiq- uitin

Cells utilize quality control (QC) mechanisms to counteract the cytotoxic accumulation of misfolded proteins triggered by in- ternal damage and environmental stressors. QC mechanisms se- lectively recognize and target misfolded polypeptides to folding- promoting factors or remove them from the cell by degradation.

Malfunction of QC mechanisms results in deleterious conse- quences and is linked to many pressing human maladies including neurological disorders as well as to aging (1, 2).

In eukaryotes, misfolded cytosolic or nuclear proteins are de- graded by the ubiquitin-proteasome system (UPS) (3). Proteaso- mal degradation is induced by polyubiquitylation of the substrate, which labels the protein for degradation by the 26S proteasome.

In this process, a set of enzymes is required to activate (E1 enzyme) and selectively conjugate (E2 and E3 enzymes) ubiquitin to the misfolded substrate or its growing polyubiquitin chain (4).

E3 ligases involved in QC recognize structural characteristics of a variety of misfolded proteins. For example, the nuclear ubiq- uitin E3 ligase San1 targets misfolded proteins to proteasomal degradation by directly binding to the exposed hydrophobicity of misfolded proteins (5).

Molecular chaperones of the Hsp70 class promote protein folding and prevent aggregation by binding and shielding hy- drophobic peptide segments of client proteins (6, 7 ). The associ- ation of Hsp70 with substrate is controlled by its ATPase activity.

When the Hsp70 carries ATP, substrate can bind and release with fast kinetics. ATP hydrolysis is stimulated by co-chaperones of the Hsp40 class that directly associate with and deliver substrates to the Hsp70. Substrate remains associated with Hsp70 until ADP release is triggered by nucleotide exchange factors (NEFs) and ATP rebinds. As an outcome, NEFs trigger substrate release from Hsp70s.

Interaction of Hsp70 with substrates is coordinated with QC mechanisms so that misfolded proteins are triaged between fold- ing and degradation (8-10). For example, in metazoan cells, the Hsp70-interacting E3 ligase CHIP induces ubiquitin-proteasome- mediated degradation of misfolded proteins (11, 12). In yeast, a functional Hsp70 system is required for ubiquitylation and pro- teasomal degradation of many misfolded proteins (13, 14). Three E3 ligases, Hul5, San1 and Ubr1, have been shown to ubiquitylate misfolded proteins that depend on Hsp70 for their degradation, but how Hsp70 and the E3 ligases coordinate their interactions is not known (15-19). NEFs are candidates that potentially could coordinate QC by releasing misfolded proteins from Hsp70 but up till now their roles in this process have been undefined.

Here we report that the NEF Fes1 is a key triaging factor in protein QC that it is specifically required for targeting misfolded proteins recognized by Hsp70 to the UPS. Our results support a model in which Fes1 facilitates the selective release of misfolded proteins from the Hsp70 system thereby promoting their destruc- tion by the ubiquitin-proteasome machinery.

Results

Cells lacking Fes1 are hypersensitive to induced protein misfold- ing and display a strong and constitutive heat shock response

Fes1 is a biochemically well-characterized yeast Hsp70 NEF of the HspBP1-class with no assigned cell biological function (20, 21). Using a reverse genetics approach, we set out to investigate the involvement of Fes1 in QC. We constructed a fes1∆ strain and could confirm the previously reported sensitivity to high temperature (Fig. S1A) (22). Importantly, fes1∆ cells were also found to be hypersensitive to protein misfolding induced either by the toxic proline ring analogue azetidine-2-carboxylate (AzC) or the toxic arginine analogue canavanine (Can) (Fig. 1A). The sensitivity of fes1∆ cells to distinct conditions that induce protein misfolding implicate Fes1 in QC.

To further investigate the physiological role of Fes1, we ana- lyzed the overall protein profile of fes1∆ cells grown under non- stressful conditions (YPD, 25°C) by SDS-PAGE. Three protein species migrating at positions between the 64 and 97 kDa size standards were markedly induced in samples from fes1∆ cells

Reserved for Publication Footnotes 1

2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68

www.pnas.org --- --- PNAS Issue Date Volume Issue Number 1--??

69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136

(4)

Submission PDF

Fig. 1. Inactivation of FES1 results in hypersensitivity to induced protein misfolding and strong activation of the heat shock response. (A) fes1Δ strains are hypersensitive to the amino acid analogues azetidine-2-carboxylic acid (AzC, 0.1 μM) and canavanine (Can, 3.6 mM). (B) Total protein extracts from wild-type (WT) and fes1Δ cells grown in YPD at 25°C separated by SDS-PAGE and stained with Coomassie Brilliant Blue. The arrows mark the positions of induced Hsp70, Hsp90 and Hsp104 (see Results). (C) Western analysis of HA-epitope tagged Hsp70s Ssa1, Ssa2, Ssa3 and Ssa4 expressed from their endogenous loci in WT and fes1Δ cells. Protein extract gel loading was adjusted as indicated (%). (D) Hsf1-Myc hypo- and hyperphosphorylation status in WT and fes1Δ cells was monitored using a gel shift assay based on Phos-Tag. Heat shocked (HS) cells were transferred from 25°C to 37°C for 30 minutes. Standard SDS-PAGE (Right panel). (E) Heat shock induction in Hsp70 NEF single and double mutant strains analyzed as in (B) and growth phenotypes at different temperatures.

Fig. 2. Fes1 is required for UPS degradation of misfolded DHFR mutants. Two folding deficient variants of DHFR (DHFRMutC-Ura3, DHFRMutD-Ura3) fused to Ura3 were expressed from the PCUP1promoter (see Fig. S3) in isogenic wild-type (wt), fes1∆, sse1∆ and snl1∆ strains. A third substrate (Ub-V76-Ura3) is a non-cleavable ubiquitin fusion protein that is targeted by the UFD pathway. Following cycloheximide addition (CH) extracts were prepared by glass bead lysis of cells collected at the indicated time points and analyzed by quantitative Western blot. The data represent at least 3 experiments with independent transformants (error bars indicate standard deviation).

compared to the wild-type control (Fig. 1B). After purification, using denaturing anion exchange chromatography, these proteins were identified by mass spectrometry as Hsp70 (70 kDa), Hsp90 (81 kDa) and Hsp104 (102 kDa), a finding that was confirmed by Western analysis (Fig. S1B). Further analysis of Ssa class Hsp70 proteins in fes1Δ cells revealed an expression pattern that was strikingly similar to that resulting from a strong and constitutive heat shock response in wild-type cells (Fig. 1C). While SSA2 is a constitutively expressed gene that is unresponsive to stress, SSA1, SSA3 and SSA4 are all transcriptionally induced by heat shock factor (Hsf1) in response to cytoplasmic protein folding stress (23). This response is triggered by accumulation of mis- folded proteins in the cytoplasm via a mechanism that relies on hyperphosphorylation of Hsf1 (24). We directly tested if Hsf1 was constitutively activated in fes1Δ cells under non-stressful growth conditions by assessing the phosphorylation status of Hsf1 using phosphoprotein gel shift analysis. This assay revealed that Hsf1 was hyperphosphorylated in the fes1Δ mutant in comparison to wild-type cells (Fig. 1D). Taken together, our data clearly show

that the heat shock response is constitutively active in fes1Δ cells at non-stressful conditions, suggesting that the cells accumulate misfolded proteins in the cytoplasm.

As a supporting experiment to test whether fes1Δ mutants accumulate misfolded proteins, we employed an aggregation re- porter consisting of a fusion between highly stable GFP and thermolabile firefly luciferase that unfolds after heat shock and co-aggregates with available misfolded proteins (25). Before heat shock, both wild-type and fes1Δ cells grown at 25°C expressed Luciferase-GFP evenly in the cytoplasm (Fig. S1C), with 9.3%

and 25.0% of the cells containing Luciferase-GFP aggregates, respectively. After heat shock at 37°C for 30 minutes, 20.7% and 42.7% of the wild-type and fes1Δ cells, respectively, contained aggregates.

As Fes1 represents only one of the three eukaryotic classes of Hsp70 NEFs present in the cytoplasm of yeast, we asked whether the Hsp110-class members Sse1 and Sse2 or the BAG- class member Snl1 have a similar or overlapping role in pro- tecting cells against misfolded proteins (26, 27). Remarkably, we 137

138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204

205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272

(5)

Submission PDF

Fig. 3. Fes1 interacts with and is required for UPS degradation of Hsp70- associated misfolded protein Rpo41T920-L1217. (A) Growth on medium selec- tive for interaction of a yeast two-hybrid reporter strain expressing Fes1 or the Hsp70-binding mutant Fes1A79R, R195A as baits and Hsp70 (SSA4), Hsp40 (SIS1) or a fragment of mitochondrial RNA polymerase (RPO41) as preys. The isolated RPO41 expresses a truncated polypeptide (T920-L1217) that corresponds to an internal region of the C-terminal domain (CTD) of mitochondrial RNA polymerase (marked in black in the crystal structure of human homologue). (B) Western analysis of total lysate (T) and supernatant (S) and pellet (P) fractions following ultracentrifugation (541000×g). Pgk1 functions as a soluble control protein. (C) Degradation ofRpo41T920-L1217 in wild-type (WT) and fes1Δ cells after translation arrest by cycloheximide (CHX). (D) Degradation at restrictive temperature (37°C) as in (C) in strains carrying mutations in Hsp70 (ssa1-45 ssa2Δ ssa3Δ ssa4Δ), the proteasome (cim3-1) or the ubiquitin E3 ligase Ubr1 (ubr1Δ). (E) Degradation as in (C) in strains carrying Hsp70 binding (A79R, R195A) mutations in the chromoso- mal FES1 locus. (F) UPS degradation (CHX) of transcription factor Stp1-HA.

Induced Hsp70-dependent ubiquitin-proteasome degradation of fructose- 1,6-bisphosphatase (Fbp1-HA) followed after addition of 2% glucose to YP- Ethanol medium.

found a strong constitutive heat shock response to be a specific property of fes1Δ cells, as none of the other mutants (sse1Δ, sse2Δ and snl1Δ) displayed high-level expression of Hsp70, Hsp90

Fig. 4. Fes1 is required for the release of misfolded proteins from Hsp70 and their targeting to the UPS (A) Western blot analysis (WB) of amounts of Hsp70 Ssa2-HA copurifying with DHFRMutC-FLAG immunoprecipitated (IP) from WT and fes1Δ protein extracts. (B) Western analysis of total polyu- biquitin levels (Ubi(n)) in wild-type (WT), fes1Δ and sse1Δ cells grown at 25°C or heat-shocked at 42°C (HS) for 30 minutes. Loading was normalized to amount of cells used for denaturing protein extraction. (C)Western blot analysis (WB) of amounts of Ubr1-HA copurifying with DHFRMutD-FLAG immunoprecipitated (IP) from WT and fes1Δ protein extracts. Quantification of the amount of Ubr1-HA copurifying with DHFRMutD-FLAG normalized to wild-type (100%) (error bars represent standard deviation of experiments performed with 5 independent transformants). (D) Growth of cells (fes1Δ) transformed with vector control (vc) or plasmids expressing Fes1, Snl1cat or Sse1. Heat shock response of each transformant was assessed by im- munoblotting (Hsp104). (E) UPS degradation after cycloheximide (CHX) of Rpo41T920-L1217followed in fes1Δ cells expressing Fes1, Snl1cator Sse1.

and Hsp104 (Fig. 1E). This is particularly striking in the light of the fact that the sse1Δ mutation causes a much more severe 273

274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340

Footline Author PNAS Issue Date Volume Issue Number 3

341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408

(6)

Submission PDF

Fig. 5. Model for Hsp70 substrate triage regulated by NEFs. For details, see Discussion.

growth defect than fes1Δ (Fig. 1E). Moreover, NEF double mu- tants accumulated Hsp70, Hsp90 and Hsp104 proteins to similar extents as fes1Δ single mutants, and none of the double mutations exacerbated the growth phenotypes of the individual mutations.

The strong heat shock response in fes1Δ cells prompted us to perform a series of supporting experiments to investigate the importance of the induction of heat shock proteins for fes1Δ cells (Fig. S2). First, we partially attenuated the activity of the essential Hsf1 by removal of the C-terminal activation domain (hsf1ΔCT) (28). When fes1Δ was combined with this mutation, the hsf1ΔCT mutation attenuated the induction of Hsp70 and Hsp90, which went along with a strong synthetic phenotype; the double mutant displayed extremely poor growth at 30°C (Fig.

S2A and B). Similarly, when fes1Δ was combined with deletions of genes encoding either cotranslationally acting Hsp70 proteins (ssb1Δ ssb2Δ), or the inducible Hsp70 system (ssa1Δ ssa3Δ ssa4Δ) (29), as well as a deletion of HSP104, which encodes the main cytoplasmic disaggregation machine in S. cerevisiae (30), all re- sulted in severe synthetic growth defects, in particular at higher temperature (Fig. S2C). Together, these findings demonstrate that fes1Δ cells depend on and induce multiple chaperone systems to cope with the defects associated with the lack of Fes1.

Fes1 is specifically required for UPS-dependent degradation of misfolded proteins

The observation that misfolded proteins accumulate in fes1∆

cells prompted us to ask if Fes1 is a general factor required for degradation of misfolded proteins associated with Hsp70.

Initially, we took advantage of two mutant versions of the globu- lar and compactly folded dihydrofolate reductase (DHFR) that are metabolically unstable at 30°C (Fig. S3). The basic char- acterization of these DHFR mutants is presented as SI in Fig.

S3-S8. Briefly, degradation of reporter proteins (DHFR-Ura3) carrying the DHFRmutCor DHFRmutDmodules was found to be dependent on the ubiquitin-proteasome pathway as their degra- dation was severely impaired by mutations in genes encoding UPS components (uba1-ts, ubc4∆ ubc5∆, ubr1∆, and ump1∆).

Moreover, degradation of these proteins (DHFR-Ura3) requires the Ssa-class of Hsp70 and Hsp40 (Figs. S4-S6). Pulldown experi- ments performed with DHFRmutC-FLAG demonstrated that this folding-deficient protein is associated with Ssa1 and Ydj1, which are representatives of the Hsp70 and Hsp40 chaperone classes, respectively (Figs. S3, S5 and 6). Failure to degrade DHFRmutC leads to its accumulation as intracellular aggregates (Fig. S7). We tested if Fes1 associates with DHFRmutCby performing immuno- precipitation. Fes1 copurified with DHFRmutC-FLAG but not with the natively folded control protein DHFR-FLAG (Fig. S8), indicating that Fes1 interacts specifically with misfolded DHFR.

We therefore asked whether Fes1 is required for degradation of DHFRmutC-Ura3 and DHFRmutD-Ura3 and found that the fes1Δ mutation caused a drastic stabilization of both proteins (Fig. 2),

which was also reflected by growth of the cells on media lacking uracil (Fig. S8).

Independent evidence for a general role for Fes1 in the degradation of misfolded proteins came from the analysis of an interactor identified in a Fes1 2‑hybrid screen. In this screen, we found Fes1 to interact with Hsp70 of both the Ssa class (9 isolates for Ssa1 and 2 for Ssa4) and the ribosome-associated Ssb class (1 Ssb1 isolate) (31). Apart from Hsp70 proteins, a number of other interactors were found, including Sis1, an Hsp40 that binds directly to Hsp70 (32, 33), and an internal fragment of the mitochondrial RNA polymerase (Rpo41T920-L1217). Im- portantly, two well-characterized mutations in Fes1 substituting residues specifically required for the association with Hsp70 (Fes1A79R, R195A) abolished the interaction with both Sis1 and Rpo41T920-L1217(Fig. 3A) (21). We conclude that Fes1 interaction with both these proteins depends on Hsp70, most likely via formation of ternary complexes. A comparison of the interact- ing stretch of Rpo41T920-L1217with the corresponding sequence (C897-I1179) in the crystal structure of human mitochondrial RNA polymerase (34) indicated that the identified segment does not represent a stable independently folding structural do- main (Fig. 3A). Rpo41T920-L1217 is an internal fragment of the C-terminal polymerase domain taken out of structural context wherein it is normally stabilized by numerous interactions with both the N- and C-terminal domains. Direct measurement of solubility by ultracentrifugation showed that the bulk of the Rpo41T920-L1217population was aggregated (Fig. 3B). These ob- servations indicated that Rpo41T920-L1217is a misfolded polypep- tide with a tendency to aggregate and therefore a likely target for ubiquitin-dependent QC mechanisms. We tested this hypoth- esis by monitoring the stability of Rpo41T920-L1217 in wild-type and fes1Δ cells. Strikingly, while Rpo41T920-L1217was degraded in wild-type cells, the protein was completely stable in fes1Δ cells over 90 minutes (Fig. 3C). Similar to what was observed for the misfolded DHFR deriviatives described above, Hsp70 function and the proteasome system was required for the degradation of Rpo41T920-L1217(Fig. 3D) (35). Inactivation of the E3 ubiquitin ligase Ubr1, which is required for the selective ubiquitylation of misfolded proteins, resulted in a stabilization of Rpo41T920-L1217 (16, 17). Cells expressing Fes1A79R, R195A that do not interact with Hsp70 did not support degradation of Rpo41T920-L1217(Fig.

3E). We conclude that, similar to degradation of the misfolded DHFR derivatives described above, the proteolytic targeting of the aberrant polypeptide Rpo41T920-L1217 by the UPS depends on the Ssa1-class of Hsp70 and its NEF Fes1. Together, our findings obtained with multiple distinct test proteins identify Fes1 as a factor essential for the degradation of misfolded cytosolic proteins.

We found that the role of Fes1 in ubiquitin-dependent pro- teolysis is specific for misfolded proteins because the degra- dation of several other known ubiquitin-dependent substrates was unaffected. Specifically, the artificial ubiquitin-fusion degra- dation (UFD) pathway substrate UbV76-Ura3 (36) is degraded normally in fes1Δ cells (Figs. 2C and S8). Similarly, the extremely fast degradation of two natural UPS substrates, the latent tran- scription factor Stp1 (37) and the glucose-triggered and Hsp70- dependent degradation of the gluconeogenetic enzyme fructose- 1,6-bisphosphatase (Fbp1) (38) proceeded at wild-type rates (Fig.

3F). In contrast to the specific effects of the fes1∆ mutation towards misfolded proteins, a severe reduction of the total NEF activity in the cytoplasm by the sse1Δ mutation (Sse1 is sevenfold more abundant than Fes1 (39)) resulted not only in an increased stability of folding-impaired substrates, but also of the UFD pathway substrate UbV76‑Ura3 (Figs. 2C and S8). Deletion of SNL1 had no effects on any of the substrates tested (Figs 2 and 409

410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476

477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544

(7)

Submission PDF

S8). Thus, in contrast to Sse1 or Snl1, Fes1 has a dedicated and specific role in misfolded protein degradation.

Fes1 releases misfolded proteins from Hsp70 thereby target- ing them for ubiquitylation

An established biochemical function of NEFs is to stimulate nucleotide exchange in Hsp70, a process that results in sub- strate release (22, 31). We tested if Fes1 promotes the release of misfolded proteins from Hsp70 by monitoring the amount of Hsp70 that binds misfolded DHFRMutCin fes1Δ cells and wild- type cells. We choose the Hsp70 Ssa2 for detection because it is expressed at the same constitutive level in both cell types (Fig. 1C). It is worthy to note that Ssa2 is competing with the higher induced levels of Ssa1, Ssa3 and Ssa4 in the fes1Δ strain.

Strikingly, when immunoprecipitating DHFRMutCfrom wild-type and fes1Δ cells and analyzing the amount of Ssa2 that copu- rified, we found that substantially more Ssa2 copurified with DHFRMutCimmunoprecipitated from fes1Δ cells than from wild- type cells (Fig. 4A) despite the higher levels of competing Ssa chaperones. Next, we investigated the involvement of Fes1 in heat shock-induced ubiquitylation (15). We predicted that misfolded proteins induced by heat treatment require Fes1 for efficient ubiquitylation. Confirming our prediction, heat shock-induced ubiquitylation was severely impaired in fes1Δ cells (Fig. 4B). In sse1Δ cells, in contrast, the levels of ubiquitylated proteins were already increased at the lower temperature and further increased upon heat stress. The increased amounts of ubiquitin modified proteins in this mutants even in the absence of external stress argue strongly against a specific function of Sse1 in promoting ubiquitylation of misfolded proteins and rather point to a gen- eral role of Sse1 in protein folding. Finally, we directly investi- gated if Fes1 promotes interactions between ubiquitin ligases and their misfolded protein substrates. The ubiquitin ligase Ubr1-HA specifically copurified misfolded DHFRMutDbut not with native DHFR (Fig. 4C). Importantly,∼5-fold more Ubr1-HA bound the misfolded protein in purifications from wild-type compared to fes1Δ extracts. We conclude that Fes1 has an important general role in releasing misfolded proteins from Hsp70 and targeting them for ubiquitylation and proteasomal degradation.

A structurally unrelated NEF but not Sse1 can partially replace Fes1 function

We tested if Fes1 function could be executed by a structurally unrelated Hsp70 NEF by expressing the soluble C-terminal cat- alytic BAG domain of Snl1 (Snl1Cat) (40) and Sse1. Expression of the Snl1Cat in fes1Δ cells resulted in partial suppression of the temperature-sensitive growth phenotype (Fig. 4D) as well as partial restoration of Rpo41T920-L1217 degradation (Fig. 4E).

In contrast, Sse1 overexpression exacerbated the temperature- sensitive growth phenotype and did not suppress the defect in Rpo41T920-L1217degradation. Expression of Snl1Cator Sse1 did not repress the induced heat shock response of fes1Δ cells (Fig. 4D).

The observation that Snl1Catpartially can replace the function of Fes1 suggests that Fes1 supports protein degradation by a simple Hsp70 release mechanism, while Sse1 functions in a competing pathway.

Discussion

Based on the findings reported here, we propose a model for the role of Fes1 in cytoplasmic protein QC that integrates all available data (Fig. 5). Abundant Hsp70 associates transiently with exposed hydrophophic patches of misfolded proteins and maintains these aggregation-prone proteins soluble. Hsp70 can refold misfolded proteins in ATPase-driven cycles catalyzed by the dominant NEFs of the Hsp110 class (Sse1/Sse2). Hsp110 contributes directly to the folding process by binding hydrophobic peptide stretches of the substrate protein. Misfolded proteins resilient to Hsp70- Hsp110 refolding will undergo repetitive and futile interactions

with Hsp70-Hsp110. Eventually the Hsp70-substrate complexes will encounter the NEF Fes1. Fes1 triggers nucleotide exchange of the Hsp70, which results in the release of the misfolded protein from Hsp70 and facilitates the interaction with downstream ubiq- uitin E3 ligases that target misfolded proteins for degradation by the UPS.

In our model, we propose that NEFs of the Hsp110 class mainly support protein folding activities of Hsp70s while Fes1 target Hsp70-associated misfolded proteins for degradation. This notion is supported by comparisons of the phenotypes of sse1Δ and fes1Δ strains (Fig. 1E) as well as by the observation that overexpression of the catalytic domain of Snl1 suppresses fes1Δ, whereas that of Sse1 does not (Fig. 4D and E). We find that sse1Δ cells, in contrast to fes1Δ cells, are very slow growing at all tem- peratures, likely because removal of this abundant NEF induces a general Hsp70 activity defect. Consistent with this understanding, sse1Δ mutants expressing folding-impaired Ura3 reporter remain uracil auxotrophs despite the observed stabilization of these re- porter proteins, indicating that they accumulate as non-natively folded protein (Fig. S8). Furthermore, sse1Δ cells do not exhibit a strong heat shock phenotype like fes1Δ cells (Fig. 1E) but do accu- mulate larger amounts of ubiquitylated proteins at normal growth temperature as well as under heat stress conditions (Fig. 4B). In conclusion, sse1Δ degradation phenotypes likely reflect general malfunction of the Hsp70 system, i.e. that misfolded proteins become stabilized in sse1Δ cells due to extensive aggregation or as a consequence of abundant production of misfolded polypeptides competing for the degradation pathways.

Biochemical differences between Hsp110 and Fes1 might explain how these two NEFs can induce so diverse fates of Hsp70 substrates as folding and degradation. In contrast to Fes1, Hsp110 is a chaperone that recently has been shown to directly bind hy- drophobic peptides (41-43). Thus, direct binding of hydrophobic patches might help to maintain the misfolded protein soluble, help the protein to fold and shield it from interactions with the ubiquitylation machinery. In support of the latter concept, the E3 ligase San1 directly interacts with hydrophobic patches of its misfolded protein substrates (5). Indeed, Sse1 inhibits San1- dependent ubiquitylation in vitro corroborating the notion of direct competition for substrate binding (14). Further support for the hypothesis of qualitative activity differences between Hsp110 and Fes1 is provided by the observation that purified Hsp70- Hsp40-Sse1 can extract and refold luciferase from aggregates wheras Hsp70-Hsp40-Fes1 cannot (42). When all these data are considered, a simple hypothesis emerges: Sse1 has substrate bind- ing activities and promotes protein folding probably by mediating rebinding to Hsp70, while Fes1 is devoid of the same activities and therefore triggers release of Hsp70 substrates that are to be ubiquitylated and degraded. Our model provides the conceptual foundation for how distinct classes of NEFs regulate Hsp70 func- tion in protein folding and degradation.

Materials and Methods

Strains, plasmids and growth media

Details on constructions of strains and plasmids are available upon request. Plasmid inserts expressing various DHFR-derived test proteins used in this study are depicted in Fig. S1A. Constructs expressing DHFR-Ura3 and DHFRmutC-Ura3 were generated and kindly provided by Jörg Höckendorff (to be published elsewhere). Standard media include YPD medium and ammonia-based synthetic complete dextrose (SC).

Heat shock protein purification and mass spectrometry analysis Strains were expanded in YPD medium at 25°C and harvested at loga- rithmic growth phase by centrifugation. Cells were lysed in Denaturating lysis buffer (40 mM Tris-HCl pH 7.5 1 mM EDTA 8 M urea) by bead beating. After preclearing by centrifugation (21100×g) the supernatant was applied onto a gravity-flow Q-Sepharose (GE Healthcare Life Sciences) column. The column was extensively washed with Denaturating lysis buffer and bound proteins were eluted using a NaCl step gradient (50 mM increments up to 600 mM NaCl) prepared in Denaturating lysis buffer. Eluted proteins were separated by SDS-PAGE followed by Coomassie Brilliant Blue staining. Protein bands were excised and subjected to tryptic extraction and MS analysis.

545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612

Footline Author PNAS Issue Date Volume Issue Number 5

613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680

(8)

Submission PDF

Yeast two-hybrid assay

A genomic yeast plasmid library (pJG4-5) was screened for interactors (growth on SC-galactose media lacking lysine and leucine) with full length Fes1 fused to the N-terminus of the DNA binding domain of LexA in strain CAY235 (44).

Analytical ultracentrifugation of Rpo41T920-L1217

Cells expressing Rpo41T920-L1217-HA were grown at 30°C to logarithmic growth phase, harvested by centrifugation and lysed in 50 mM Tris-HCl pH 7.5, 5 mM MgCl2, 10 mM DTT, 1 mM PMSF, 1 mM N-ethylmaleimide by bead beating. Lysates were cleared from cell debris by low speed centrifugation (1500×g) and detergents were added to the following final concentrations, 1% NP40, 1% Tween 20 and 0.1 % SB3-14. The solubilized lysates were subjected to 30 minutes of ultracentrifugation in a TLA 100.3 rotor (Beckman) at 100000 rpm (541000×g) and analyzed by Western blotting.

Western blot analysis and determination of protein stability

Cells were grown at 30°C to logarithmic growth phase and cyclohex- imide was added to the cultures at a final concentration of 100 mg/L.

Strains carrying temperature sensitive alleles were expanded at permissive 25°C and incubated at restrictive 37°C for 30 minutes prior to cycloheximide addition. Protein extracts were prepared from samples taken at the time points indicated either by glass bead lysis in ice cold Native lysis buffer (50 mM Na-HEPES pH 7.5, 150 mM NaCl, 5 mM EDTA, 1% Triton-X100 containing Complete™ Protease Inhibitor Cocktail (Roche)) or by NaOH and trichloroacetic acid (TCA) as described previously (45). Equal amounts of proteins were separated by SDS-PAGE and analyzed by quantitative Western

blotting (46) or by chemiluminescence detection (SuperSignal West Dura Extended-Duration Substrate; Pierce, Rockford, IL) and quantified using an LAS1000 system (Fuji Photo Film Co., Tokyo, Japan).

Co‑immunoprecipitation of mutant DHFR and associated proteins Cells were grown at 30°C to logarithmic growth phase in SC medium supplemented with 100 or 400 μM CuSO4to induce the expression of the DHFR constructs. After harvest by centrifugation, cells were broken by glass bead lysis in either LWB150 (40 mm Hepes-KOH, pH 7.4, 150 mm KCl, 5 mm MgCl2, 5% (v/v) glycerol), 0.1% (v/v) Triton X-100) or Native lysis buffer (see above). The lysates, precleared by centrifugation, were incubated with ANTI- FLAG® M2 Affinity Gel (Sigma) for 60 minutes. After washing extensively with buffer, bound proteins were eluted with sample buffer or Native lysis buffer containing 3X FLAG‑Peptide (200 mg/L). Co-immunoprecipitated proteins were detected by Western blotting.

Acknowledgments.

We thank Andreas Bachmair, Stefan Jentsch, Erica Johnson, Nils Johns- son, Jörg Höckendorff, Mark Hochstrasser, Kiran Madura, Thomas Sommer, and Alexander Varshavsky for yeast strains and plasmids. This work was sup- ported by a grant from the Deutsche Forschungsgemeinschaft (Do 649/2) to R.J.D. and grants from the Swedish Science Council, Carl Tryggers stiftelse för vetenskaplig forskning and Jeanssons Stiftelser to C.A. Author Contributions N.K.C.G. and G.K. designed, performed and analyzed the experiments. M.S.F.

characterized the degradation pathway of misfolded DHFR mutants. R.J.D.

and C.A. supervised the project and provided conceptual context to the data.

1. Ross CA & Poirier MA (2005) Opinion: What is the role of protein aggregation in neurode- generation? Nat Rev Mol Cell Biol 6(11):891-898.

2. Hartl FU, Bracher A, & Hayer-Hartl M (2011) Molecular chaperones in protein folding and proteostasis. Nature 475(7356):324-332.

3. Hochstrasser M (2009) Origin and function of ubiquitin-like proteins. Nature 458(7237):422- 429.

4. Komander D & Rape M (2012) The ubiquitin code. Annu Rev Biochem 81:203-229.

5. Fredrickson EK, Rosenbaum JC, Locke MN, Milac TI, & Gardner RG (2011) Exposed hydrophobicity is a key determinant of nuclear quality control degradation. Mol Biol Cell 22(13):2384-2395.

6. Goloubinoff P & De Los Rios P (2007) The mechanism of Hsp70 chaperones: (entropic) pulling the models together. Trends Biochem Sci 32(8):372-380.

7. Mayer MP & Bukau B (2005) Hsp70 chaperones: cellular functions and molecular mecha- nism. Cell Mol Life Sci 62(6):670-684.

8. Arndt V, Rogon C, & Hohfeld J (2007) To be, or not to be--molecular chaperones in protein degradation. Cell Mol Life Sci 64(19-20):2525-2541.

9. Kettern N, Dreiseidler M, Tawo R, & Hohfeld J (2010) Chaperone-assisted degradation:

multiple paths to destruction. Biol Chem 391(5):481-489.

10. McClellan AJ, Tam S, Kaganovich D, & Frydman J (2005) Protein quality control: chaperones culling corrupt conformations. Nat Cell Biol 7(8):736-741.

11. Jiang J, et al. (2001) CHIP is a U-box-dependent E3 ubiquitin ligase: identification of Hsc70 as a target for ubiquitylation. J Biol Chem 276(46):42938-42944.

12. Qian SB, McDonough H, Boellmann F, Cyr DM, & Patterson C (2006) CHIP-mediated stress recovery by sequential ubiquitination of substrates and Hsp70. Nature 440(7083):551-555.

13. McClellan AJ, Scott MD, & Frydman J (2005) Folding and quality control of the VHL tumor suppressor proceed through distinct chaperone pathways. Cell 121(5):739-748.

14. Park SH, et al. (2007) The cytoplasmic Hsp70 chaperone machinery subjects misfolded and endoplasmic reticulum import-incompetent proteins to degradation via the ubiquitin- proteasome system. Mol Biol Cell 18(1):153-165.

15. Fang NN, Ng AH, Measday V, & Mayor T (2011) Hul5 HECT ubiquitin ligase plays a major role in the ubiquitylation and turnover of cytosolic misfolded proteins. Nat Cell Biol 13(11):1344-1352.

16. Heck JW, Cheung SK, & Hampton RY (2010) Cytoplasmic protein quality control degrada- tion mediated by parallel actions of the E3 ubiquitin ligases Ubr1 and San1. Proc Natl Acad Sci U S A 107(3):1106-1111.

17. Eisele F & Wolf DH (2008) Degradation of misfolded protein in the cytoplasm is mediated by the ubiquitin ligase Ubr1. FEBS Lett 582(30):4143-4146.

18. Prasad R, Kawaguchi S, & Ng DT (2010) A nucleus-based quality control mechanism for cytosolic proteins. Mol Biol Cell 21(13):2117-2127.

19. Nillegoda NB, et al. (2010) Ubr1 and Ubr2 function in a quality control pathway for degradation of unfolded cytosolic proteins. Mol Biol Cell 21(13):2102-2116.

20. Andreasson C, Fiaux J, Rampelt H, Druffel-Augustin S, & Bukau B (2008) Insights into the structural dynamics of the Hsp110-Hsp70 interaction reveal the mechanism for nucleotide exchange activity. Proc Natl Acad Sci U S A 105(43):16519-16524.

21. Shomura Y, et al. (2005) Regulation of Hsp70 function by HspBP1: structural analysis reveals an alternate mechanism for Hsp70 nucleotide exchange. Mol Cell 17(3):367-379.

22. Kabani M, Beckerich JM, & Brodsky JL (2002) Nucleotide exchange factor for the yeast Hsp70 molecular chaperone Ssa1p. Mol Cell Biol 22(13):4677-4689.

23. Ingolia TD, Slater MR, & Craig EA (1982) Saccharomyces cerevisiae contains a complex multigene family related to the major heat shock-inducible gene of Drosophila. Mol Cell Biol 2(11):1388-1398.

24. Sorger PK & Pelham HR (1988) Yeast heat shock factor is an essential DNA-binding protein that exhibits temperature-dependent phosphorylation. Cell 54(6):855-864.

25. Gupta R, et al. (2011) Firefly luciferase mutants as sensors of proteome stress. Nat Methods

8(10):879-884.

26. Dragovic Z, Broadley SA, Shomura Y, Bracher A, & Hartl FU (2006) Molecular chaperones of the Hsp110 family act as nucleotide exchange factors of Hsp70s. EMBO J 25(11):2519- 2528.

27. Raviol H, Sadlish H, Rodriguez F, Mayer MP, & Bukau B (2006) Chaperone network in the yeast cytosol: Hsp110 is revealed as an Hsp70 nucleotide exchange factor. EMBO J 25(11):2510-2518.

28. Sorger PK (1990) Yeast heat shock factor contains separable transient and sustained response transcriptional activators. Cell 62(4):793-805.

29. Nelson RJ, Ziegelhoffer T, Nicolet C, Werner-Washburne M, & Craig EA (1992) The translation machinery and 70 kd heat shock protein cooperate in protein synthesis. Cell 71(1):97-105.

30. Parsell DA, Kowal AS, Singer MA, & Lindquist S (1994) Protein disaggregation mediated by heat-shock protein Hsp104. Nature 372(6505):475-478.

31. Dragovic Z, Shomura Y, Tzvetkov N, Hartl FU, & Bracher A (2006) Fes1p acts as a nucleotide exchange factor for the ribosome-associated molecular chaperone Ssb1p. Biol Chem 387(12):1593-1600.

32. Aron R, Lopez N, Walter W, Craig EA, & Johnson J (2005) In vivo bipartite interaction between the Hsp40 Sis1 and Hsp70 in Saccharomyces cerevisiae. Genetics 169(4):1873-1882.

33. Li J, Wu Y, Qian X, & Sha B (2006) Crystal structure of yeast Sis1 peptide-binding fragment and Hsp70 Ssa1 C-terminal complex. Biochem J 398(3):353-360.

34. Ringel R, et al. (2011) Structure of human mitochondrial RNA polymerase. Nature 478(7368):269-273.

35. Becker J, Walter W, Yan W, & Craig EA (1996) Functional interaction of cytosolic hsp70 and a DnaJ-related protein, Ydj1p, in protein translocation in vivo. Mol Cell Biol 16(8):4378-4386.

36. Johnson ES, Ma PC, Ota IM, & Varshavsky A (1995) A proteolytic pathway that recognizes ubiquitin as a degradation signal. J Biol Chem 270(29):17442-17456.

37. Pfirrmann T, Heessen S, Omnus DJ, Andreasson C, & Ljungdahl PO (2010) The prodomain of Ssy5 protease controls receptor-activated proteolysis of transcription factor Stp1. Mol Cell Biol 30(13):3299-3309.

38. Juretschke J, Menssen R, Sickmann A, & Wolf DH (2010) The Hsp70 chaperone Ssa1 is essential for catabolite induced degradation of the gluconeogenic enzyme fructose-1,6- bisphosphatase. Biochem Biophys Res Commun 397(3):447-452.

39. Ghaemmaghami S, et al. (2003) Global analysis of protein expression in yeast. Nature 425(6959):737-741.

40. Sondermann H, et al. (2002) Prediction of novel Bag-1 homologs based on structure/function analysis identifies Snl1p as an Hsp70 co-chaperone in Saccharomyces cerevisiae. J Biol Chem 277(36):33220-33227.

41. Goeckeler JL, et al. (2008) The yeast Hsp110, Sse1p, exhibits high-affinity peptide binding.

FEBS Lett 582(16):2393-2396.

42. Shorter J (2011) The mammalian disaggregase machinery: Hsp110 synergizes with Hsp70 and Hsp40 to catalyze protein disaggregation and reactivation in a cell-free system. PLoS One 6(10):e26319.

43. Xu X, et al. (2011) Unique peptide substrate binding properties of 110-kDa heat-shock protein (Hsp110) determine its distinct chaperone activity. J Biol Chem 287(8):5661-5672.

44. Omnus DJ, Pfirrmann T, Andréasson C, & Ljungdahl PO (2011) A phosphodegron con- trols nutrient-induced proteasomal activation of the signaling protease Ssy5. Mol Biol Cell 22(15):2754-2765.

45. Silve S, et al. (1991) Membrane insertion of uracil permease, a polytopic yeast plasma membrane protein. Mol Cell Biol 11(2):1114-1124.

46. Kurian L, Palanimurugan R, Godderz D, & Dohmen RJ (2011) Polyamine sensing by nascent ornithine decarboxylase antizyme stimulates decoding of its mRNA. Nature 477(7365):490- 494.

681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748

749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806 807 808 809 810 811 812 813 814 815 816

(9)

Supplementary Information

Fig. S1

FES1 mutant cells are hypersensitive to conditions inducing protein misfolding conditions and accumulate protein aggregates. (A) fes1 Δ strains are hypersensitive to elevated growth temperature. 10-fold dilutions series of cell suspensions were spotted onto YPD medium and were incubated at the indicated temperatures. (B) Total protein extracts from wild-type (WT) and fes1 Δ cells grown in YPD at 25°C separated by SDS-PAGE and stained with Coomassie Brilliant Blue or analyzed by Western blotting with antibodies specific for Hsp70 (Ssa1/2 rabbit polyclonal serum), Hsp90 (monoclonal antibody K41220A) and Hsp104 (rabbit polyclonal serum). (C) Images of GFP fluorescence microcopy of fixed wild-type (WT) and fes1 Δ cells expressing the aggregation reporter Luciferase-GFP grown at 25 °C or heat shocked (HS) at 37 °C for 30 minutes. The arrow shows an aggregate. The aggregation data was quantified by counting the fraction of cells (n=30) that contained detectable GFP- aggregates. The error bars indicate the standard deviation of three datasets from independent biological replicates. (D) Western analysis of Luciferase-GFP expression levels in the same strains under identical growth conditions as in (C).

Fig. S2

Cells lacking FES1 are hypersensitive to mutations that impair heat shock factor Hsf1 and chaperones Hsp70 and Hsp104. (A) Genetic interactions scored by growth phenotypes between fes1 Δ and a mutation that impair Hsf1 activity due to truncation of the C-terminal activation domain (hsf1 ΔCT). (B). Heat shock induction (Hsp70, Hsp90 and Hsp104) in the indicated strains was assessed by SDS-PAGE. (C) Genetic interactions between fes1 Δ and the genetic deletion of the three heat shock inducible Hsp70s (ssa1/3/4 Δ), of the ribosome-associated Hsp70s (ssb1/2Δ) or of the AAA+ ATPase and disaggregase Hsp104 (hsp104 Δ).

Fig. S3

Folding-deficient model substrate (A) Shown are the DHFR constructs used in this study, all of which were expressed from centromeric plasmids. (B) Cycloheximide chase assay of yeast cells expressing wild-type or the mutant DHFR-ha-Ura3 constructs. Proteins were detected by anti-HA western blotting. A cross-reactive band is indicated by an asterisk. (C) Growth assay of yeast cells expressing the same constructs as in (B) on minimal media lacking uracil. (D) Illustration of the crystal structure of mouse DHFR. The α helix (residues 116-128) deleted in DHFR

mutD

is highlighted in purple. The two residues (T39A, E173D) that are mutated in DHFR

mutC

are highlighted in magenta.

Fig S4

Folding-deficient proteins are degraded by the ubiquitin/proteasome system (UPS) (A) Folding-deficient DHFR is stabilized in the proteasome-deficient ump1∆

mutant (1). Cycloheximide chase assay of DHFR

mutD

-Ura3 stability in wild-type and

ump1∆ strains. A cross-reactive band is indicated by an asterisk. (B) A functional

ubiquitin-activating enzyme is required for the degradation of folding-deficient

DHFR. Cycloheximide chase assay of DHFR

mutD

-Ura3 in wild-type and uba1-ts26, a

strain expressing a temperature-sensitive ubiquitin-activating enzyme (2). Cells were

shifted to the restrictive temperature (37°C) 20 min before the addition of

(10)

cycloheximide. A cross-reactive band is indicated by an asterisk. (C) The redundant ubiquitin-conjugating enzymes Ubc4 or Ubc5 are required for degradation of folding- deficient DHFR (3). (D) DHFR

mutD

-Ura3 degradation is attenuated in the ubr1∆

mutant lacking the ubiquitin ligase Ubr1 (4). (E) Folding-deficient DHFR is ubiquitylated in vivo. Protein extracts from wild-type yeast cells expressing FLAG- tagged versions of wild-type DHFR or its mutant derivatives were subjected to a pulldown experiment using anti-FLAG resin. Bound material was analyzed by anti- ubiquitin and, after stripping, by anti-FLAG western blotting.

Fig. S5

Ssa class Hsp70 chaperones are required specifically for degradation of folding- deficient proteins. (A) Folding-deficient DHFR is stabilized in the ssa1-45 mutant (5). Cycloheximide chase assay of DHFR

mutC

-HA-Ura3 and DHFR

mutD

-HA-Ura3 stability in wild-type and ssa1-45 cells. Cells were shifted to the restrictive temperature (37°C) 20 min before the addition of cycloheximide. (B) The Hsp70 chaperone Ssa1 is associated with folding-deficient DHFR. Protein extracts from a yeast strain expressing a 3xHA tagged Ssa1 and FLAG-tagged versions of wild-type DHFR or its mutant derivative (mutC) were subjected to a pulldown experiment using anti-FLAG resin. Bound material was analyzed by anti-ha and, after stripping, by anti-FLAG western blotting. The control strain expressed Ssa1-3xHA but no FLAG- tagged substrate. (C) Ssa class chaperones are not required for the degradation of the UFD pathway substrate Ub

V76

-HA-Ura3. Shown is a cycloheximide chase assay with the same strains as in (A) but with the UFD substrate. Cross-reactive bands are indicated by asterisks.

Fig. S6

Hsp40 Ydj1 is required specifically for degradation of folding-deficient proteins.

(A) Folding-deficient DHFR is stabilized in the ydj1-151 mutant. Cycloheximide chase assay of DHFR

mutC

-HA-Ura3 and DHFR

mutD

-HA-Ura3 stability in wild-type and ydj1-151 cells (6, 7). Cells were shifted to the restrictive temperature (37°C) 20 min before the addition of cycloheximide. A crossreactive band is indicated by an asterisk. (B) The Hsp40 co-chaperone Ydj1 is associated with folding-deficient DHFR. Protein extracts from a yeast strain expressing a 3xha tagged Ydj1 and FLAG-tagged versions of wild-type DHFR or its mutant derivative (mutC) were subjected to a pulldown experiment using anti-FLAG resin. The control strain expressed Ydj1-3xHA but no FLAG-tagged substrate. Bound material was analyzed by anti-ha and, after stripping, by anti-FLAG western blotting. (C) Cycloheximide chase assay showing degradation of the UFD pathway substrate Ub

V76

-HA-Ura3 in wild-type (wt) and ydj1-151 cells. Control, extract of wt cells with an empty vector.

Cross-reactive bands are indicated by asterisks.

Fig. S7

Formation of intracellular aggregates of folding-deficient proteins. Cells with the indicated genotypes expressing DHFR

mutC

-GFP were grown at 30°C and shifted for 20 min to 37°C before inspection in the fluorescence microscope (left panels). Right panels, differential interference contrast (DIC) images of the same frames.

Fig. S8

Fes1 binds to misfolded proteins and promotes their degradation. (A) Fes1 is

associated with folding-deficient DHFR. Protein extracts from a yeast strain

(11)

expressing a 3xHA tagged Fes1 and FLAG-tagged versions of wild-type DHFR or its mutant derivative (mutC) were subjected to a pulldown experiment using anti-FLAG resin. The control strain expressed Fes1-3xHA but no FLAG-tagged substrate. Bound material was analyzed by anti-ha and, after stripping, by anti-FLAG western blotting.

(B) Fes1∆ cells are impaired in the degradation of folding-deficient DHFR

mutD

-Ura3 protein. Two transformants each of wild-type, fes1∆, sse1∆, snl1∆, ump1∆ and ubr1∆

cells expressing the indicated proteins were grown to mid-log phase and spotted at a concentration of 10 OD600 per ml onto selective media lacking the indicated ingredients. Plates were incubated for 3 days at 30°C.

References

1. Ramos PC, Hockendorff J, Johnson ES, Varshavsky A, & Dohmen RJ (1998) Ump1p is required for proper maturation of the 20S proteasome and becomes its substrate upon completion of the assembly. Cell 92(4):489-499.

2. Palanimurugan R, Scheel H, Hofmann K, & Dohmen RJ (2004) Polyamines regulate their synthesis by inducing expression and blocking degradation of ODC antizyme. EMBO J 23(24):4857-4867.

3. Seufert W & Jentsch S (1990) Ubiquitin-conjugating enzymes UBC4 and UBC5 mediate selective degradation of short-lived and abnormal proteins.

EMBO J 9(2):543-550.

4. Bartel B, Wunning I, & Varshavsky A (1990) The recognition component of the N-end rule pathway. EMBO J 9(10):3179-3189.

5. Kim S, Schilke B, Craig EA, & Horwich AL (1998) Folding in vivo of a newly translated yeast cytosolic enzyme is mediated by the SSA class of cytosolic yeast Hsp70 proteins. Proc Natl Acad Sci U S A 95(22):12860- 12865.

6. Caplan AJ, Cyr DM, & Douglas MG (1992) YDJ1p facilitates polypeptide translocation across different intracellular membranes by a conserved mechanism. Cell 71(7):1143-1155.

7. Lee DH, Sherman MY, & Goldberg AL (1996) Involvement of the molecular

chaperone Ydj1 in the ubiquitin-dependent degradation of short-lived and

abnormal proteins in Saccharomyces cerevisiae. Mol Cell Biol 16(9):4773-

4781.

(12)

Luciferase-GFP

25°C

HS

fes1 6 WT

Pgk1

25°C HS

Luciferase-GFP

6

fes1 6

C

D

WT fes1 6

WT fes1 6

WT fes1 6

25°C 37°C 39°C

A

Fig. S1

10 20 30 40 50

WT

25°C HS

Cells w/ Luci-GFP foci

(%)

fes1 6 WT fes16

B

kDa

64

51

_-Hsp104 _-Hsp90

_-Hsp70

WT fes1 6

WT fes1 6

WT fes1 6

WT fes1

6

(13)

$ %

&

:7 IHV

6

ƒ&

ƒ&

KVI

6 &7 IHV

6 KVI

6 &7







N'D

:7 IHV

6 KVI

6 &7 IHV

6 KVI

6 &7

+VS

+VS

+VS

ƒ&

ƒ&

:7 IHV

6

:7 IHV

6

:7 IHV

6 VVD

6

VVE

6

KVS

VVD 6 6

IHV

6

IHV

6VVE

6

IHV

6 KVS

6

)LJ6

(14)

$ % &

'

)LJ6

(15)

)LJ6

$

%

&

'

(

(16)

)LJ6

$ %

&

(17)

)LJ6

%

&

$

(18)

)LJ6

(19)

)LJ6

$

%

References

Related documents

Therefore, we aim to conduct a study on the motivation factors influencing success connected to rural areas of Sweden using three entrepreneurs from Sollefteå to answer the

The bias factor of an edge dislocation can be calculated by solving the dif- fusion equation with a drift term if the interaction energy between the dislocation and a mobile defect

Given different perceptions at the national level between Germany and India identified by GLOBE research project [2.3], and differences from an organizational culture standpoint

The Framework for Strategic Sustainable Development (see Section 1) was developed with a whole-systems perspective precisely for this reason. The framework allows for

In summary, then, under the present politico-economic structure of the Sudanese society, corruption (surrounding the state as an axis of financial gains) has become a mode of

Karin C arlsson Tissue F actor in C omplex - Studies of interactions between blood coagulation proteins

During initiation, the translation initiation region (TIR) of mRNA and the initiator tRNA (fMet-tRNA f Met ) are selected by the 30S subunit to form a ternary complex (Gualerzi and

Two commonly used methods for estimating large autoregressive models are dimensional reduction by projection onto latent factors and regularised VAR estimation by penalisation of