リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

大学・研究所にある論文を検索できる 「SEL1L degradation intermediates stimulate cytosolic aggregation of polyglutamine‐expanded protein」の論文概要。リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

コピーが完了しました

URLをコピーしました

論文の公開元へ論文の公開元へ
書き出し

SEL1L degradation intermediates stimulate cytosolic aggregation of polyglutamine‐expanded protein

Hattori, Tokuya Hanafusa, Ken Wada, Ikuo Hosokawa, Nobuko 京都大学 DOI:10.1111/febs.15761

2021.08

概要

Misfolded proteins in the endoplasmic reticulum (ER) are degraded by ER-associated degradation (ERAD). In mammalian cells, the HRD1–SEL1L membrane ubiquitin ligase complex plays a central role in this process. However, SEL1L is inherently unstable, and excess SEL1L is also degraded by ERAD. Accordingly, when proteasome activity is inhibited, multiple degradation intermediates of SEL1L appear in the cytosol. In this study, we searched for factors that inhibit SEL1L degradation and identified OS-9 and XTP3-B, two ER lectins that regulate glycoprotein ERAD. SEL1L degradation was characterized by a ladder of degradation products, and the C-terminal Pro-rich region of SEL1L was responsible for generation of this pattern. In the cytosol, these degradation intermediates stimulated aggregation of polyglutamine-expanded Huntingtin protein (Htt-polyQ-GFP) by interacting with aggregation-prone proteins, including Htt-polyQ-GFP. Collectively, our findings indicate that peptide fragments of ER proteins generated during ERAD may affect protein aggregation in the cytosol, revealing the interconnection of protein homeostasis across subcellular compartments.

この論文で使われている画像

参考文献

1. Hipp, M. S., Kasturi, P. & Hartl, F. U. (2019) The proteostasis network and its decline in

ageing, Nat Rev Mol Cell Biol. 20, 421-435.

2. Ellgaard, L., McCaul, N., Chatsisvili, A. & Braakman, I. (2016) Co- and Post-Translational

Protein Folding in the ER, Traffic. 17, 615-38.

3.

Preston, G. M. & Brodsky, J. L. (2017) The evolving role of ubiquitin modification in

endoplasmic reticulum-associated degradation, Biochem J. 474, 445-469.

4. Berner, N., Reutter, K. R. & Wolf, D. H. (2018) Protein Quality Control of the Endoplasmic

Reticulum and Ubiquitin-Proteasome-Triggered Degradation of Aberrant Proteins: Yeast Pioneers

the Path, Annu Rev Biochem.

5. Carvalho, P., Goder, V. & Rapoport, T. A. (2006) Distinct ubiquitin-ligase complexes define

convergent pathways for the degradation of ER proteins, Cell. 126, 361-73.

6. Wu, X., Siggel, M., Ovchinnikov, S., Mi, W., Svetlov, V., Nudler, E., Liao, M., Hummer, G.

& Rapoport, T. A. (2020) Structural basis of ER-associated protein degradation mediated by the

Hrd1 ubiquitin ligase complex, Science. 368.

7. Ruggiano, A., Foresti, O. & Carvalho, P. (2014) Quality control: ER-associated degradation:

protein quality control and beyond, J Cell Biol. 204, 869-79.

8. Hipp, M. S., Park, S. H. & Hartl, F. U. (2014) Proteostasis impairment in protein-misfolding

and -aggregation diseases, Trends Cell Biol. 24, 506-14.

9.

Chiti, F. & Dobson, C. M. (2017) Protein Misfolding, Amyloid Formation, and Human

Disease: A Summary of Progress Over the Last Decade, Annu Rev Biochem. 86, 27-68.

10. Labbadia, J. & Morimoto, R. I. (2015) The biology of proteostasis in aging and disease,

Annu Rev Biochem. 84, 435-64.

11.

Lieberman, A. P., Shakkottai, V. G. & Albin, R. L. (2019) Polyglutamine Repeats in

Neurodegenerative Diseases, Annu Rev Pathol. 14, 1-27.

12. Orr, H. T. & Zoghbi, H. Y. (2007) Trinucleotide repeat disorders, Annu Rev Neurosci. 30,

575-621.

13. Duennwald, M. L. & Lindquist, S. (2008) Impaired ERAD and ER stress are early and

specific events in polyglutamine toxicity, Genes Dev. 22, 3308-19.

14. Hosokawa, N. & Wada, I. (2016) Association of the SEL1L protein transmembrane domain

with HRD1 ubiquitin ligase regulates ERAD-L, Febs j. 283, 157-72.

15.

Iida, Y., Fujimori, T., Okawa, K., Nagata, K., Wada, I. & Hosokawa, N. (2011) SEL1L

protein critically determines the stability of the HRD1-SEL1L endoplasmic reticulum-associated

32

A Self-archived copy in

Kyoto University Research Information Repository

https://repository.kulib.kyoto-u.ac.jp

degradation (ERAD) complex to optimize the degradation kinetics of ERAD substrates, J Biol

Chem. 286, 16929-39.

16. Mittl, P. R. & Schneider-Brachert, W. (2007) Sel1-like repeat proteins in signal transduction,

Cell Signal. 19, 20-31.

17. Christianson, J. C., Shaler, T. A., Tyler, R. E. & Kopito, R. R. (2008) OS-9 and GRP94

deliver mutant alpha1-antitrypsin to the Hrd1-SEL1L ubiquitin ligase complex for ERAD, Nat

Cell Biol. 10, 272-82.

18. Kang, S. W., Rane, N. S., Kim, S. J., Garrison, J. L., Taunton, J. & Hegde, R. S. (2006)

Substrate-specific translocational attenuation during ER stress defines a pre-emptive quality

control pathway, Cell. 127, 999-1013.

19. Misaghi, S., Pacold, M. E., Blom, D., Ploegh, H. L. & Korbel, G. A. (2004) Using a small

molecule inhibitor of peptide: N-glycanase to probe its role in glycoprotein turnover, Chem Biol.

11, 1677-87.

20.

Suzuki, T. (2007) Cytoplasmic peptide:N-glycanase and catabolic pathway for free N-

glycans in the cytosol, Semin Cell Dev Biol. 18, 762-9.

21. Wiertz, E. J., Jones, T. R., Sun, L., Bogyo, M., Geuze, H. J. & Ploegh, H. L. (1996) The

human cytomegalovirus US11 gene product dislocates MHC class I heavy chains from the

endoplasmic reticulum to the cytosol, Cell. 84, 769-79.

22. Wolf, D. H. & Stolz, A. (2012) The Cdc48 machine in endoplasmic reticulum associated

protein degradation, Biochim Biophys Acta. 1823, 117-24.

23. Magnaghi, P., D'Alessio, R., Valsasina, B., Avanzi, N., Rizzi, S., Asa, D., Gasparri, F., Cozzi,

L., Cucchi, U., Orrenius, C., Polucci, P., Ballinari, D., Perrera, C., Leone, A., Cervi, G., Casale,

E., Xiao, Y., Wong, C., Anderson, D. J., Galvani, A., Donati, D., O'Brien, T., Jackson, P. K. &

Isacchi, A. (2013) Covalent and allosteric inhibitors of the ATPase VCP/p97 induce cancer cell

death, Nat Chem Biol. 9, 548-56.

24. Xu, C. & Ng, D. T. (2015) Glycosylation-directed quality control of protein folding, Nat

Rev Mol Cell Biol. 16, 742-52.

25. Cattaneo, M., Baronchelli, S., Schiffer, D., Mellai, M., Caldera, V., Saccani, G. J., Dalpra,

L., Daga, A., Orlandi, R., DeBlasio, P. & Biunno, I. (2014) Down-modulation of SEL1L, an

unfolded protein response and endoplasmic reticulum-associated degradation protein, sensitizes

glioma stem cells to the cytotoxic effect of valproic acid, J Biol Chem. 289, 2826-38.

26. Biunno, I., Cattaneo, M., Orlandi, R., Canton, C., Biagiotti, L., Ferrero, S., Barberis, M.,

Pupa, S. M., Scarpa, A. & Menard, S. (2006) SEL1L a multifaceted protein playing a role in tumor

progression, J Cell Physiol. 208, 23-38.

27. Anbanandam, A., Albarado, D. C., Tirziu, D. C., Simons, M. & Veeraraghavan, S. (2008)

Molecular basis for proline- and arginine-rich peptide inhibition of proteasome, J Mol Biol. 384,

33

A Self-archived copy in

Kyoto University Research Information Repository

https://repository.kulib.kyoto-u.ac.jp

219-27.

28. Sikorski, R. S., Boguski, M. S., Goebl, M. & Hieter, P. (1990) A repeating amino acid motif

in CDC23 defines a family of proteins and a new relationship among genes required for mitosis

and RNA synthesis, Cell. 60, 307-17.

29.

Kim, S., Nollen, E. A., Kitagawa, K., Bindokas, V. P. & Morimoto, R. I. (2002)

Polyglutamine protein aggregates are dynamic, Nat Cell Biol. 4, 826-31.

30.

Matsumoto, G., Kim, S. & Morimoto, R. I. (2006) Huntingtin and mutant SOD1 form

aggregate structures with distinct molecular properties in human cells, J Biol Chem. 281, 447785.

31. Gillis, J., Schipper-Krom, S., Juenemann, K., Gruber, A., Coolen, S., van den Nieuwendijk,

R., van Veen, H., Overkleeft, H., Goedhart, J., Kampinga, H. H. & Reits, E. A. (2013) The

DNAJB6 and DNAJB8 protein chaperones prevent intracellular aggregation of polyglutamine

peptides, J Biol Chem. 288, 17225-37.

32. Jeong, H., Sim, H. J., Song, E. K., Lee, H., Ha, S. C., Jun, Y., Park, T. J. & Lee, C. (2016)

Crystal structure of SEL1L: Insight into the roles of SLR motifs in ERAD pathway, Sci Rep. 6,

20261.

33. van der Goot, A. T., Pearce, M. M. P., Leto, D. E., Shaler, T. A. & Kopito, R. R. (2018)

Redundant and Antagonistic Roles of XTP3B and OS9 in Decoding Glycan and Non-glycan

Degrons in ER-Associated Degradation, Mol Cell. 70, 516-530 e6.

34. Choi, J. Y., Ryu, J. H., Kim, H. S., Park, S. G., Bae, K. H., Kang, S., Myung, P. K., Cho, S.,

Park, B. C. & Lee, D. H. (2007) Co-chaperone CHIP promotes aggregation of ataxin-1, Mol Cell

Neurosci. 34, 69-79.

35. Mueller, B., Lilley, B. N. & Ploegh, H. L. (2006) SEL1L, the homologue of yeast Hrd3p, is

involved in protein dislocation from the mammalian ER, J Cell Biol. 175, 261-70.

36. Neal, S., Syau, D., Nejatfard, A., Nadeau, S. & Hampton, R. Y. (2020) HRD Complex SelfRemodeling Enables a Novel Route of Membrane Protein Retrotranslocation, iScience. 23,

101493.

37. Hamdan, N., Kritsiligkou, P. & Grant, C. M. (2017) ER stress causes widespread protein

aggregation and prion formation, J Cell Biol. 216, 2295-2304.

38. Kitamura, A., Kubota, H., Pack, C. G., Matsumoto, G., Hirayama, S., Takahashi, Y., Kimura,

H., Kinjo, M., Morimoto, R. I. & Nagata, K. (2006) Cytosolic chaperonin prevents polyglutamine

toxicity with altering the aggregation state, Nat Cell Biol. 8, 1163-70.

39. Hirao, K., Natsuka, Y., Tamura, T., Wada, I., Morito, D., Natsuka, S., Romero, P., Sleno, B.,

Tremblay, L. O., Herscovics, A., Nagata, K. & Hosokawa, N. (2006) EDEM3, a soluble EDEM

homolog, enhances glycoprotein endoplasmic reticulum-associated degradation and mannose

trimming, J Biol Chem. 281, 9650-8.

34

A Self-archived copy in

Kyoto University Research Information Repository

https://repository.kulib.kyoto-u.ac.jp

40. Hosokawa, N., Kamiya, Y., Kamiya, D., Kato, K. & Nagata, K. (2009) Human OS-9, a lectin

required for glycoprotein endoplasmic reticulum-associated degradation, recognizes mannosetrimmed N-glycans, J Biol Chem. 284, 17061-8.

41. Hosokawa, N., Wada, I., Nagasawa, K., Moriyama, T., Okawa, K. & Nagata, K. (2008)

Human XTP3-B Forms an Endoplasmic Reticulum Quality Control Scaffold with the HRD1SEL1L Ubiquitin Ligase Complex and BiP, J Biol Chem. 283, 20914-24.

42. Sakurai, C., Itakura, M., Kinoshita, D., Arai, S., Hashimoto, H., Wada, I. & Hatsuzawa, K.

(2018) Phosphorylation of SNAP-23 at Ser95 causes a structural alteration and negatively

regulates Fc receptor-mediated phagosome formation and maturation in macrophages, Mol Biol

Cell. 29, 1753-1762.

43. Wada, I., Kai, M., Imai, S., Sakane, F. & Kanoh, H. (1997) Promotion of transferrin folding

by cyclic interactions with calnexin and calreticulin, EMBO J. 16, 5420-32.

44. Wanker, E. E., Scherzinger, E., Heiser, V., Sittler, A., Eickhoff, H. & Lehrach, H. (1999)

Membrane filter assay for detection of amyloid-like polyglutamine-containing protein aggregates,

Methods Enzymol. 309, 375-86.

35

A Self-archived copy in

Kyoto University Research Information Repository

https://repository.kulib.kyoto-u.ac.jp

Figure legends

Figure 1. SEL1L degradation intermediates can be detected in cells treated with proteasome

inhibitor.

(A) Schematic representation of the domain organization of SEL1L protein. FN type II,

fibronectin type II domain; TM, transmembrane.

(B) HEK 293 cells were transfected with wild-type SEL1L, S-SEL1L, or SEL1L-HA, and then

treated with MG132 for 6 h. Cells were extracted in a buffer containing 1% NP-40, and

supernatant was collected. After separation by 10% SDS-PAGE, specific signals were detected

by Western blotting. Blue, black, and gray arrows indicate endogenous SEL1L, full-length SSEL1L, and full-length SEL1L-HA, respectively. Angle brackets denote the SEL1L degradation

intermediates. Asterisks show signals non-specifically detected by the antibodies. Lane numbers

in parentheses indicate the same sample loaded in the left panel.

(C) Same as in A, except that cells were transfected with FolA-S-HA, which was detected with

anti-HA-tag antibody. Arrow indicates full-length FolA-S-HA.

(D) HEK 293 cells were transfected with WT SEL1L and HA-Ubiquitin, and SEL1L protein was

immunoprecipitated with anti-SEL1L antibody. The bracket indicates ubiquitinated SEL1L

protein detected by the anti-HA antibody. ** indicates rabbit IgG contained in the anti-sera used

36

A Self-archived copy in

Kyoto University Research Information Repository

https://repository.kulib.kyoto-u.ac.jp

for immunoprecipitation.

(E) Pulse-chase analysis of S-SEL1L expressed in HEK 293 cells. Cells were treated with MG132

for 3 h prior to metabolic labeling for 15 min (MG132 +) and chased for the indicated periods.

Cell lysates were immunoprecipitated using anti–S-tag or anti-SEL1L antibodies. The arrow

indicates full-length S-SEL1L, and the blue arrow indicates endogenous SEL1L. Angle brackets

denote degradation intermediates of S-SEL1L. Asterisk indicates a non-specific signal in MG132treated cells. Full-length S-SEL1L was quantified, and results are shown as means ± SD from

three independent experiments. **, P < 0.01; ***, P < 0.001 (two-tailed Student’s t-test, compared

with cells without MG132 treatment).

Figure 2. SEL1L degradation intermediates accumulate in the cytosol.

(A–C) HEK 293 cells expressing SEL1L-HA or S-SEL1L were fractionated into cytosol (CE),

membrane (ME), and nuclear extracts (NE), and separated by 10% SDS-PAGE. Specific signals

were detected by Western blot using anti-SEL1L (A) or anti–S-tag antibodies (B), or antibodies

against ER (calnexin [CNX]), and cytosolic proteins (actin, HSC70/HSP70) (C). Blue brackets

and arrows indicate endogenous SEL1L and full-length S-SEL1L, respectively. Angle brackets

denote SEL1L degradation intermediates. Lane numbers in parenthesis in (B) indicate the same

samples loaded in (A) and (C).

37

A Self-archived copy in

Kyoto University Research Information Repository

https://repository.kulib.kyoto-u.ac.jp

(D, E) HEK 293 cells were homogenized by passing them through a 30-G needle in an isotonic

buffer containing 250 mM sucrose, and then cytosol (S: supernatant) and microsome (P: pellet)

fractions were separated by ultracentrifugation at 100,000 × g for 1 h. Specific signals were

detected by Western blotting using anti-SEL1L (D) or anti–S-tag antibodies (E). Blue brackets,

arrows and angle brackets are the same as in (A). Lane numbers in parenthesis in (E) indicate the

same samples loaded in (D).

Figure 3. SEL1L degradation intermediates are reduced and deglycosylated.

(A) Extracts of HEK 293 cells expressing the indicated plasmids were separated by 10% SDSPAGE under reducing (DTT +) or non-reducing (DTT -) conditions, and specific signals were

detected by Western blot analysis using anti-SEL1L antibody. Blue, gray, and black arrows

indicate endogenous SEL1L, full-length SEL1L-HA, and full-length S-SEL1L, respectively.

Angle brackets denote SEL1L degradation intermediates.

(B) Western blot analysis of SEL1L-HA and DssSEL1L-HA expressed in HEK 293 cells. Arrow

and arrowhead indicate full-length SEL1L-HA and DssSEL1L-HA, respectively. Angle brackets

denote SEL1L degradation intermediates. Asterisks indicate non-specific signals detected by antiHA antibody.

(C) HEK 293 cells expressing SEL1L-HA were treated with the PNGase inhibitor Z-VAD-FMK,

38

A Self-archived copy in

Kyoto University Research Information Repository

https://repository.kulib.kyoto-u.ac.jp

and cell lysates were analyzed by Western blot. Red angle brackets indicate SEL1L-HA

degradation intermediates, whose migration was delayed by Z-VAD-FMK treatment.

(D) Two-dimensional isoelectric focusing (IEF) electrophoresis of SEL1L and DssSEL1L-HA.

Cells extracted in a buffer containing 9.5 M urea were separated by IEF containing Ampholine

(pH 3–10 and 4–7) in a tube gel, and then by 10% SDS-PAGE. After blotting onto PVDF

membranes, specific signals were detected by Western blotting using anti-HA antibody (left

panel). Double-headed arrows indicate full-length SEL1L-HA. Signals exhibiting different

mobility in IEF are indicated by arrows (red in SEL1L-HA and black in DssSEL1L-HA). The

same membranes were blotted sequentially with anti-actin (1), anti-BiP (2), and anti-calnexin (3,

3’) antibodies to confirm comparable IEF separation on both membranes (right panel). Asterisks

indicate non-specific signals detected by the antibodies.

(E) Same as in E, except that p97/VCP inhibitor MNS-873 was used.

Figure 4. Accumulation of SEL1L degradation intermediates is inhibited by co-expression of OS9 and XTP3-B

(A–C) HEK 293 cells co-transfected with S-SEL1L and NHK-QQQ, OS-9v2-HA, or XTP3-BHA were treated with proteasome inhibitor (MG132 +). Cell lysates were separated by 10% SDSPAGE, and specific signals were detected by Western blotting with anti-SEL1L (A, upper panel),

39

A Self-archived copy in

Kyoto University Research Information Repository

https://repository.kulib.kyoto-u.ac.jp

anti-S-tag (A, lower panel), or indicated antibodies (B). Blue and black arrows indicate

endogenous SEL1L and full-length S-SEL1L, respectively. Angle brackets denote SEL1L

degradation intermediates.

(C) HEK 293 cells were co-transfected with WT SEL1L and the indicated ER proteins, and cell

lysates were subjected to Western blotting with the indicated specific antibodies. MG132 was

added from 6 h prior to harvest. Bracket indicates endogenous SEL1L and transfected full-length

SEL1L, and angle brackets indicate SEL1L degradation intermediates.

(D–G) Western blot analysis of HEK 293 cells transfected with the indicated siRNAs for 48 h

(D). Levels of endogenous SEL1L (E), OS-9 (F, including both OS-9v1 and v2), and XTP3-B (G)

in D were quantified relative to cells treated with negative control siRNA (left-most lane). Results

are shown as means ± SD from three independent experiments. ns, P > 0.05; **, P < 0.01; ***, P

< 0.001 (two-tailed Student’s t-test, compared with cells transfected with negative control siRNA).

(H) Pulse-chase analysis of NHK-QQQ degradation. HEK 293 cells co-transfected with NHKQQQ, SEL1L, and OS-9v2-HA were metabolically labeled for 15 min and chased for the

indicated period. NHK-QQQ was immunoprecipitated and separated by 10% SDS-PAGE.

Radioisotope incorporation into NHK-QQQ is quantified in the graph. Error bars indicate means

± SD from three independent experiments. *, P < 0.05; ***, P < 0.001 (two-tailed Student’s t-

test, compared with cells co-transfected with SEL1L).

40

A Self-archived copy in

Kyoto University Research Information Repository

https://repository.kulib.kyoto-u.ac.jp

(I) Pulse-chase analysis of S-SEL1L degradation. Same as in A, except cells were transfected

with S-SEL1L and OS-9v2-HA and immunoprecipitated using anti–S-tag or anti-HA antibodies.

Radioactive incorporation to S-SEL1L was quantified in the graph. Error bars indicate means ±

SD from three independent experiments. *, P < 0.05; **, P < 0.01 (two-tailed Student’s t-test).

Figure 5. Degradation intermediates of endogenous SEL1L are detected in HEK 293 cells

exposed to ER stress

(A) Western blot analysis of endogenous SEL1L upon treatement with thapsigargin (Tg) or

tunicamycin (Tm). For clarity, the obtained images are shown in grayscale (top) or using the “fire”

lookup table of ImageJ. Blue brackets indicate full-length endogenous SEL1L. Angle brackets

denote SEL1L degradation intermediates. Double angle brackets indicate deglycosylated SEL1L.

(B) Western blot analysis of endogenous SEL1L in HEK 293 cells transfected with indicated

siRNAs. Thirty hours after transfection of siRNAs, cells were treated with MG132 for 6 h

(MG132 +). The same image is shown using the “fire” lookup table of ImageJ. The asterisk

indicates a non-specific signal detected by anti–OS-9 antibody.

(C) Immunoprecipitation analysis of S-SEL1L and HRD1-myc co-expressed in HEK 293 cells.

Cells were lysed in a buffer containing 1% NP-40 (NP-40) or 3% digitonin (Dig), and

immunoprecipitated using anti-S-tag or anti-c-myc antibodies. ** indicates mouse IgG used for

41

A Self-archived copy in

Kyoto University Research Information Repository

https://repository.kulib.kyoto-u.ac.jp

immunoprecipitation.

(D) Alkaline extraction of SEL1L-HA, S-SEL1L and HRD1-myc expressed in HEK 293 cells.

The microsomal fraction was prepared as in Fig. 2C and D from cells co-expressing SEL1L-HA

and HRD1-myc (left panel) or S-SEL1L and HRD1-myc (right panel). After incubation in an

alkaline solution containing 0.1 M Na2CO3 or in a buffer containing 1% NP-40, a supernatant

(Sup) and pellet (Ppt) were fractionated by ultracentrifugation at 100,000 × g.

(E) Pulse-chase analysis of S-SEL1L co-expressed with or without HRD1-myc in HEK 293 cells.

Cells were pulse-labeled for 15 min and chased for the indicated periods. Cell lysates were

immunoprecipitated using anti–S-tag or anti-SEL1L antibodies. The bracket indicates full-length

S-SEL1L. The blue arrow indicates endogenous SEL1L. Full-length S-SEL1L was quantified.

Results are shown as means ± SD from three independent experiments. *, P < 0.05; **, P < 0.01;

***, P < 0.001 (two-tailed Student’s t-test, compared with cells without co-expression of HRD1myc).

(F) Cycloheximide-chase analysis of NHK degradation. HEK 293 cells co-transfected with NHK,

HRD1-myc, and S-SEL1L were chased for the indicated period after addition of cycloheximide.

The cell lysate was separated by 10% SDS-PAGE and analyzed by Western blotting. The signal

intensity of NHK is quantified in the graph. Error bars indicate means ± SD from three

independent experiments. *, P < 0.05; **, P < 0.01 (two-tailed Student’s t-test, compared with

42

A Self-archived copy in

Kyoto University Research Information Repository

https://repository.kulib.kyoto-u.ac.jp

cells co-transfected with HRD1-myc).

Figure 6. Deletion of SEL1L proline-rich region promotes SEL1L degradation.

(A) Western blot analysis of HEK 293, HeLa, HepG2 (HG2), RD, and Capan2 cells (upper panel),

and Coomassie Brilliant Blue (CBB) staining of the same membrane (lower panel). Twenty

micrograms of each cell lysate was separated by SDS-PAGE and blotted with anti-SEL1L

antibody.

(B) Cells were transfected with SEL1L-HA and treated with MG12 for 6 h (MG132 +). Arrows

indicate full-length SEL1L-HA, and angle brackets denote SEL1L-HA degradation intermediates.

Asterisks indicate non-specific signal detected by anti-HA antibody.

(C) Western blot analysis of SEL1L-HA and SEL1L-DPR-HA expressed in HEK 293 cells. Red

arrow and angle brackets denote full-length SEL1L-DPR-HA and degradation intermediates,

respectively.

(D) Pulse-chase analysis of NHK-QQQ and SEL1L-DPR-HA degradation. HEK 293 cells cotransfected with NHK-QQQ and SEL1L-HA or SEL1L-DPR-HA were metabolically labeled for

15 min and chased for the indicated period. Radioisotope incorporation into NHK-QQQ and

SEL1L-HA or SEL1L-DPR-HA is quantified in the graph. Error bars indicate means ± SD from

three independent experiments. *, P < 0.05; **, P < 0.01 (two-tailed Student’s t-test).

43

A Self-archived copy in

Kyoto University Research Information Repository

https://repository.kulib.kyoto-u.ac.jp

Figure 7. Htt-polyQ76-GFP aggregates form in HEK293 cells.

(A) HEK 293 cells were transfected with Htt-polyQ76-GFP and SEL1L-HA or DssSEL1L-HA.

Twenty-four hours after transfection, nuclei were stained with Hoechst 33342 and analyzed by

fluorescence microscopy. MG132 was added 6 h prior to microscopic observation (MG132 +).

Scale bars, 50 µm.

(B) Cells in A containing Htt-polyQ76-GFP aggregates were counted, and the percentage of GFPpositive cells (~300–500 cells) with aggregates was calculated. Error bars indicate means ± SD

from three independent experiments. ns, P > 0.05; *, P < 0.05; **, P < 0.01; ***, P < 0.001 (twotailed Student’s t-test, compared with mock-transfected cells (MG132 -), or between the samples

indicated by the bracket).

(C) Immunocytochemistry of HeLa cells transfected with SEL1L-HA or DssSEL1L-HA, treated

with or without MG132. Cells were fixed and stained with anti-HA and anti-calreticulin (CRT,

ER marker, left panel) or anti-HA and anti-HSP70/HSC70 (cytosolic marker) antibodies (right

panel). SEL1L-HA and DssSEL1L-HA were visualized with Alexa Fluor 594, CRT and

HSP70/HSC70 were stained with Alexa Fluor 488, and images were acquired by confocal

microscopy. Scale bars, 10 µm.

44

A Self-archived copy in

Kyoto University Research Information Repository

https://repository.kulib.kyoto-u.ac.jp

Figure 8. Immunostaining of polyQ inclusions.

HEK 293 cells (A, B, C, E, and F) and HeLa cells (D) were transfected with Htt-polyQ76-GFP

and SEL1L-HA (A, B and D), Htt-polyQ76-GFP and DssSEL1L-HA (C, D), Htt-polyQ23 and

SEL1L-HA (E), or Htt-polyQ23 and DssSEL1L-HA (F). Twenty-four hours after transfection,

cells were fixed and stained with anti-GFP and anti-HA antibodies. GFP was visualized with

Alexa Fluor 488, HA was stained with Alexa Fluor 596, and images were analyzed by confocal

microscopy. MG132 was added 6 h prior to microscopic observation (MG132 +). Nuclei were

counterstained with DAPI. Scale bars, 10 µm (upper panel) and 5 µm (magnified).

Figure 9. SEL1L SLR expressed in the cytosol promotes polyQ aggregation.

(A, B) Htt-polyQ76-GFP aggregation formation analyzed by fluorescence microscopy. Same as

in Fig. 7A and B, except that cells were co-transfected with SEL1L-R5-9-HA.

(C) Immunostaining of Htt-polyQ76-GFP. Same as in Fig. 8B, except that cells were cotransfected with SEL1L-R5-9-HA.

(D, E) Filter-trap assays of Htt-polyQ76-GFP co-transfected with SEL1L-HA, Htt-polyQ23-GFP

(D), and Htt-polyQ76-GFP co-transfected with mock, DssSEL1L-HA, or SEL1L-R5-9-HA (E). Cell

lysates were serially diluted, and the indicated amounts of protein were applied to the filter.

(F) Model of HRD1–SEL1L complex formation. SEL1L and HRD1 form a stable complex in the

45

A Self-archived copy in

Kyoto University Research Information Repository

https://repository.kulib.kyoto-u.ac.jp

ER membrane, but SEL1L expressed in excess is rapidly degraded by ERAD (left). OS-9 and

XTP3-B associate with and stabilize SEL1L until the HRD1–SEL1L complex is formed.

Alternatively, SEL1L–lectin complex surveils the ER for misfolded proteins and dynamically

forms a HRD1-containing membrane complex that is competent for retrotranslocation (right).

46

A Self-archived copy in

Kyoto University Research Information Repository

https://repository.kulib.kyoto-u.ac.jp

Figure 1

A Self-archived copy in

Kyoto University Research Information Repository

https://repository.kulib.kyoto-u.ac.jp

Figure 2

A Self-archived copy in

Kyoto University Research Information Repository

https://repository.kulib.kyoto-u.ac.jp

Figure 3

A Self-archived copy in

Kyoto University Research Information Repository

https://repository.kulib.kyoto-u.ac.jp

Figure 4

A Self-archived copy in

Kyoto University Research Information Repository

https://repository.kulib.kyoto-u.ac.jp

Figure 5

A Self-archived copy in

Kyoto University Research Information Repository

https://repository.kulib.kyoto-u.ac.jp

Figure 6

A Self-archived copy in

Kyoto University Research Information Repository

https://repository.kulib.kyoto-u.ac.jp

Figure 7

A Self-archived copy in

Kyoto University Research Information Repository

https://repository.kulib.kyoto-u.ac.jp

Figure 8

A Self-archived copy in

Kyoto University Research Information Repository

https://repository.kulib.kyoto-u.ac.jp

Figure 9

...

参考文献をもっと見る

全国の大学の
卒論・修論・学位論文

一発検索!

この論文の関連論文を見る