Browse > Article
http://dx.doi.org/10.4062/biomolther.2020.089

Endoplasmic Reticulum Stress-Mediated p62 Downregulation Inhibits Apoptosis via c-Jun Upregulation  

Yu, Wenjun (Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University)
Wang, Busong (Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University)
Zhou, Liang (Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University)
Xu, Guoqiang (Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University)
Publication Information
Biomolecules & Therapeutics / v.29, no.2, 2021 , pp. 195-204 More about this Journal
Abstract
Cereblon (CRBN), a substrate receptor of cullin 4-RING E3 ligase (CRL4) regulates the ubiquitination and degradation of c-Jun, mediating the lipopolysaccharide-induced cellular response. However, the upstream signaling pathway that regulates this process is unknown. In this study, we describe how endoplasmic reticulum (ER) stress reversely regulates sequestosome-1 (p62)and c-Jun protein levels. Furthermore, our study reveals that expression of p62 attenuates c-Jun protein levels through the ubiquitinproteasome system. Conversely, siRNA knockdown of p62 elevates c-Jun protein levels. Immunoprecipitation and immunoblotting experiments demonstrate that p62 interacts with c-Jun and CRBN to form a ternary protein complex. Moreover, we find that CRBN knockdown completely abolishes the inhibitory effect of p62 on c-Jun. Using brefeldin A as an inducer of ER stress, we demonstrate that the p62/c-Jun axis participates in the regulation of ER stress-induced apoptosis, and that CRBN is required for this regulation. In summary, we have identified an upstream signaling pathway, which regulates p62-mediated c-Jun degradation. Our findings elucidate the underlying molecular mechanism by which p62/c-Jun axis regulates the ER stress-induced apoptosis, and provide a new molecular connection between ER stress and apoptosis.
Keywords
p62; c-Jun; CRBN; Endoplasmic reticulum stress; Apoptosis; Ubiquitination;
Citations & Related Records
연도 인용수 순위
  • Reference
1 Tian, Z., Wang, C., Hu, C., Tian, Y., Liu, J. and Wang, X. (2014) Autophagic-lysosomal inhibition compromises ubiquitin-proteasome system performance in a p62 dependent manner in cardiomyocytes. PLoS ONE 9, e100715.   DOI
2 Tilija Pun, N., Jang, W. J. and Jeong, C. H. (2020) Role of autophagy in regulation of cancer cell death/apoptosis during anti-cancer therapy: focus on autophagy flux blockade. Arch. Pharm. Res. 43, 475-488.   DOI
3 Wang, C. and Wang, X. (2015) The interplay between autophagy and the ubiquitin-proteasome system in cardiac proteotoxicity. Biochim. Biophys. Acta 1852, 188-194.   DOI
4 Wang, D., Xu, Q., Yuan, Q., Jia, M., Niu, H., Liu, X., Zhang, J., Young, C. Y. and Yuan, H. (2019) Proteasome inhibition boosts autophagic degradation of ubiquitinated-AGR2 and enhances the antitumor efficiency of bevacizumab. Oncogene 38, 3458-3474.   DOI
5 Wu, D., Hao, Z., Ren, H. and Wang, G. (2018) Loss of VAPB regulates autophagy in a Beclin 1-dependent manner. Neurosci. Bull. 34, 1037-1046.   DOI
6 Yamaguchi, H. and Wang, H. G. (2004) CHOP is involved in endoplasmic reticulum stress-induced apoptosis by enhancing DR5 expression in human carcinoma cells. J. Biol. Chem. 279, 45495-45502.   DOI
7 Yang, J., Huang, M., Zhou, L., He, X., Jiang, X., Zhang, Y. and Xu, G. (2018) Cereblon suppresses lipopolysaccharide-induced inflammatory response through promoting the ubiquitination and degradation of c-Jun. J. Biol. Chem. 293, 10141-10157.   DOI
8 Zaffagnini, G., Savova, A., Danieli, A., Romanov, J., Tremel, S., Ebner, M., Peterbauer, T., Sztacho, M., Trapannone, R., Tarafder, A. K., Sachse, C. and Martens, S. (2018) p62 filaments capture and present ubiquitinated cargos for autophagy. EMBO J. 37, e98308.   DOI
9 Zhang, Y. B., Gong, J. L., Xing, T. Y., Zheng, S. P. and Ding, W. (2013) Autophagy protein p62/SQSTM1 is involved in HAMLET-induced cell death by modulating apotosis in U87MG cells. Cell Death Dis. 4, e550.   DOI
10 Bossy-Wetzel, E., Bakiri, L. and Yaniv, M. (1997) Induction of apoptosis by the transcription factor c-Jun. EMBO J. 16, 1695-1709.   DOI
11 Bustamante, H. A., Gonzalez, A. E., Cerda-Troncoso, C., Shaughnessy, R., Otth, C., Soza, A. and Burgos, P. V. (2018) Interplay between the autophagy-lysosomal pathway and the ubiquitin-proteasome system: a target for therapeutic development in Alzheimer's disease. Front. Cell. Neurosci. 12, 126.   DOI
12 Lamark, T., Kirkin, V., Dikic, I. and Johansen, T. (2009) NBR1 and p62 as cargo receptors for selective autophagy of ubiquitinated targets. Cell Cycle 8, 1986-1990.   DOI
13 Korolchuk, V. I., Mansilla, A., Menzies, F. M. and Rubinsztein, D. C. (2009) Autophagy inhibition compromises degradation of ubiquitin-proteasome pathway substrates. Mol. Cell 33, 517-527.   DOI
14 Korolchuk, V. I., Menzies, F. M. and Rubinsztein, D. C. (2010) Mechanisms of cross-talk between the ubiquitin-proteasome and autophagy-lysosome systems. FEBS Lett. 584, 1393-8139.   DOI
15 Kouzarides, T. and Ziff, E. (1988) The role of the leucine zipper in the fos-jun interaction. Nature 336, 646-651.   DOI
16 Levine, B. and Kroemer, G. (2008) Autophagy in the pathogenesis of disease. Cell 132, 27-42.   DOI
17 Lin, J. F., Lin, Y. C., Tsai, T. F., Chen, H. E., Chou, K. Y. and Hwang, T. I. S. (2017) Cisplatin induces protective autophagy through activation of BECN1 in human bladder cancer cells. Drug Des. Devel. Ther. 11, 1517-1533.   DOI
18 Lippai, M. and Low, P. (2014) The role of the selective adaptor p62 and ubiquitin-like proteins in autophagy. Biomed Res. Int. 2014, 832704.   DOI
19 Liu, W. J., Ye, L., Huang, W. F., Guo, L. J., Xu, Z. G., Wu, H. L., Yang, C. and Liu, H. F. (2016) p62 links the autophagy pathway and the ubiqutin-proteasome system upon ubiquitinated protein degradation. Cell. Mol. Biol. Lett. 21, 29.   DOI
20 Liu, Y., Kern, J. T., Walker, J. R., Johnson, J. A., Schultz, P. G. and Luesch, H. (2007) A genomic screen for activators of the antioxidant response element. Proc. Natl. Acad. Sci. U.S.A. 104, 5205-5210.   DOI
21 Ding, W. X., Ni, H. M., Gao, W., Hou, Y. F., Melan, M. A., Chen, X., Stolz, D. B., Shao, Z. M. and Yin, X. M. (2007) Differential effects of endoplasmic reticulum stress-induced autophagy on cell survival. J. Biol. Chem. 282, 4702-4710.   DOI
22 Ciechanover, A. (2005) Proteolysis: from the lysosome to ubiquitin and the proteasome. Nat. Rev. Mol. Cell Biol. 6, 79-87.   DOI
23 Del Prete, D., Rice, R. C., Rajadhyaksha, A. M. and D'Adamio, L. (2016) Amyloid precursor protein (APP) may act as a substrate and a recognition unit for CRL4CRBN and Stub1 E3 ligases facilitating ubiquitination of proteins involved in presynaptic functions and neurodegeneration. J. Biol. Chem. 291, 17209-17227.   DOI
24 Dikic, I. (2017) Proteasomal and autophagic degradation systems. Annu. Rev. Biochem. 86, 193-224.   DOI
25 Zhu, X., Huang, L., Gong, J., Shi, C., Wang, Z., Ye, B., Xuan, A., He, X., Long, D., Zhu, X., Ma, N. and Leng, S. (2017) NF-κB pathway link with ER stress-induced autophagy and apoptosis in cervical tumor cells. Cell Death Dis. 3, 17059.   DOI
26 Mackman, N., Brand, K. and Edgington, T. S. (1991) Lipopolysaccharide-mediated transcriptional activation of the human tissue factor gene in THP-1 monocytic cells requires both activator protein 1 and nuclear factor kappa B binding sites. J. Exp. Med. 174, 1517-1526.   DOI
27 Zhao, P., Xiao, X., Kim, A. S., Leite, M. F., Xu, J., Zhu, X., Ren, J. and Li, J. (2008) c-Jun inhibits thapsigargin-induced ER stress through up-regulation of DSCR1/Adapt78. Exp. Biol. Med. (Maywood) 233, 1289-1300.   DOI
28 Zhou, L., Hao, Z., Wang, G. and Xu, G. (2018) Cereblon suppresses the formation of pathogenic protein aggregates in a p62-dependent manner. Hum. Mol. Genet. 27, 667-678.   DOI
29 Zhou, L., Wang, H., Chen, D., Gao, F., Ying, Z. and Wang, G. (2014) p62/Sequestosome 1 regulates aggresome formation of pathogenic ataxin-3 with expanded polyglutamine. Int. J. Mol. Sci. 15, 14997-15010.   DOI
30 Zhu, K., Dunner, K., Jr. and McConkey, D. J. (2010) Proteasome inhibitors activate autophagy as a cytoprotective response in human prostate cancer cells. Oncogene 29, 451-462.   DOI
31 Zhu, Y., Lei, Q., Li, D., Zhang, Y., Jiang, X., Hu, Z. and Xu, G. (2018) Proteomic and biochemical analyses reveal a novel mechanism for promoting protein ubiquitination and degradation by UFBP1, a key component of ufmylation. J. Proteome Res. 17, 1509-1520.   DOI
32 Zotti, T., Scudiero, I., Settembre, P., Ferravante, A., Mazzone, P., D'Andrea, L., Reale, C., Vito, P. and Stilo, R. (2014) TRAF6-mediated ubiquitination of NEMO requires p62/sequestosome-1. Mol. Immunol. 58, 27-31.   DOI
33 Grice, G. L. and Nathan, J. A. (2016) The recognition of ubiquitinated proteins by the proteasome. Cell. Mol. Life Sci. 73, 3497-3506.   DOI
34 Fuest, M., Willim, K., MacNelly, S., Fellner, N., Resch, G. P., Blum, H. E. and Hasselblatt, P. (2012) The transcription factor c-Jun protects against sustained hepatic endoplasmic reticulum stress thereby promoting hepatocyte survival. Hepatology 55, 408-418.   DOI
35 Gal, J., Strom, A. L., Kilty, R., Zhang, F. and Zhu, H. (2007) p62 accumulates and enhances aggregate formation in model systems of familial amyotrophic lateral sclerosis. J. Biol. Chem. 282, 11068-11077.   DOI
36 Geetha, T., Seibenhener, M. L., Chen, L., Madura, K. and Wooten, M. W. (2008) p62 serves as a shuttling factor for TrkA interaction with the proteasome. Biochem. Biophys. Res. Commun. 374, 33-37.   DOI
37 Milan, E., Perini, T., Resnati, M., Orfanelli, U., Oliva, L., Raimondi, A., Cascio, P., Bachi, A., Marcatti, M., Ciceri, F. and Cenci, S. (2015) A plastic SQSTM1/p62-dependent autophagic reserve maintains proteostasis and determines proteasome inhibitor susceptibility in multiple myeloma cells. Autophagy 11, 1161-1178.   DOI
38 Marino, S., Petrusca, D. N., Silberman, R., Toscani, D., Anderson, J. L., Giuliani, N., Xie, X. Q., Kurihara, N. and Roodman, G. D. (2017) Inhibition of p62-ZZ domain-mediated signaling overcomes bortezomib resistance in multiple myeloma cells independent of their p53 status. Blood 130, 4421.
39 Masud, A., Mohapatra, A., Lakhani, S. A., Ferrandino, A., Hakem, R. and Flavell, R. A. (2007) Endoplasmic reticulum stress-induced death of mouse embryonic fibroblasts requires the intrinsic pathway of apoptosis. J. Biol. Chem. 282, 14132-14139.   DOI
40 Meng, Q. and Xia, Y. (2011) c-Jun, at the crossroad of the signaling network. Protein Cell 2, 889-898.   DOI
41 Moon, J. L., Kim, S. Y., Shin, S. W. and Park, J. W. (2012) Regulation of brefeldin A-induced ER stress and apoptosis by mitochondrial NADP+-dependent isocitrate dehydrogenase. Biochem. Biophys. Res. Commun. 417, 760-764.   DOI
42 Hou, X. O., Si, J. M., Ren, H. G., Chen, D., Wang, H. F., Ying, Z., Hu, Q. S., Gao, F. and Wang, G. H. (2015) Parkin represses 6-hydroxydopamine-induced apoptosis via stabilizing scaffold protein p62 in PC12 cells. Acta Pharmacol. Sin. 36, 1300-1307.   DOI
43 Halazonetis, T. D., Georgopoulos, K., Greenberg, M. E. and Leder, P. (1988) c-Jun dimerizes with itself and with c-Fos, forming complexes of different DNA binding affinities. Cell 55, 917-924.   DOI
44 Hettinger, K., Vikhanskaya, F., Poh, M. K., Lee, M. K., de Belle, I., Zhang, J. T., Reddy, S. A. G. and Sabapathy, K. (2006) c-Jun promotes cellular survival by suppression of PTEN. Cell Death Differ. 14, 218-229.   DOI
45 Hewitt, G., Carroll, B., Sarallah, R., Correia-Melo, C., Ogrodnik, M., Nelson, G., Otten, E. G., Manni, D., Antrobus, R., Morgan, B. A., von Zglinicki, T., Jurk, D., Seluanov, A., Gorbunova, V., Johansen, T., Passos, J. F. and Korolchuk, V. I. (2016) SQSTM1/p62 mediates crosstalk between autophagy and the UPS in DNA repair. Autophagy 12, 1917-1930.   DOI
46 Rubinsztein, D. C. (2006) The roles of intracellular protein-degradation pathways in neurodegeneration. Nature 443, 780-786.   DOI
47 Narendra, D., Kane, L. A., Hauser, D. N., Fearnley, I. M. and Youle, R. J. (2010) p62/SQSTM1 is required for Parkin-induced mitochondrial clustering but not mitophagy; VDAC1 is dispensable for both. Autophagy 6, 1090-1106.   DOI
48 Ogata, M., Hino, S., Saito, A., Morikawa, K., Kondo, S., Kanemoto, S., Murakami, T., Taniguchi, M., Tanii, I., Yoshinaga, K., Shiosaka, S., Hammarback, J. A., Urano, F. and Imaizumi, K. (2006) Autophagy is activated for cell survival after endoplasmic reticulum stress. Mol. Cell. Biol. 26, 9220-9231.   DOI
49 Huang, X., Wang, X. N., Yuan, X. D., Wu, W. Y., Lobie, P. E. and Wu, Z. (2018) XIAP facilitates breast and colon carcinoma growth via promotion of p62 depletion through ubiquitination-dependent proteasomal degradation. Oncogene 38, 1448-1460.   DOI
50 Pankiv, S., Clausen, T. H., Lamark, T., Brech, A., Bruun, J. A., Outzen, H., Overvatn, A., Bjorkoy, G. and Johansen, T. (2007) p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J. Biol. Chem. 282, 24131-24145.   DOI
51 Sano, R. and Reed, J. C. (2013) ER stress-induced cell death mechanisms. Biochim. Biophys. Acta 1833, 3460-3470.   DOI
52 Seibenhener, M. L., Babu, J. R., Geetha, T., Wong, H. C., Krishna, N. R. and Wooten, M. W. (2004) Sequestosome 1/p62 is a polyubiquitin chain binding protein involved in ubiquitin proteasome degradation. Mol. Cell. Biol. 24, 8055-8068.   DOI
53 Shaulian, E. and Karin, M. (2002) AP-1 as a regulator of cell life and death. Nat. Cell Biol. 4, E131-E136.   DOI
54 Shin, W. H., Park, J. H. and Chung, K. C. (2020) The central regulator p62 between ubiquitin proteasome system and autophagy and its role in the mitophagy and Parkinson's disease. BMB Rep. 53, 56-63.   DOI
55 Jian, Y., Gao, W., Geng, C., Zhou, H., Leng, Y., Li, Y. and Chen, W. (2017) Arsenic trioxide potentiates sensitivity of multiple myeloma cells to lenalidomide by upregulating cereblon expression levels. Oncol. Lett. 14, 3243-3248.   DOI
56 Ichimura, Y., Waguri, S., Sou, Y. S., Kageyama, S., Hasegawa, J., Ishimura, R., Saito, T., Yang, Y., Kouno, T., Fukutomi, T., Hoshii, T., Hirao, A., Takagi, K., Mizushima, T., Motohashi, H., Lee, M. S., Yoshimori, T., Tanaka, K., Yamamoto, M. and Komatsu, M. (2013) Phosphorylation of p62 activates the Keap1-Nrf2 pathway during selective autophagy. Mol. Cell 51, 618-631.   DOI
57 Iurlaro, R. and Munoz-Pinedo, C. (2016) Cell death induced by endoplasmic reticulum stress. FEBS J. 283, 2640-2652.   DOI
58 Jain, A., Lamark, T., Sjottem, E., Larsen, K. B., Awuh, J. A., Overvatn, A., McMahon, M., Hayes, J. D. and Johansen, T. (2010) p62/SQSTM1 is a target gene for transcription factor NRF2 and creates a positive feedback loop by inducing antioxidant response element-driven gene transcription. J. Biol. Chem. 285, 22576-22591.   DOI
59 Jin, Z., Li, Y., Pitti, R., Lawrence, D., Pham, V. C., Lill, J. R. and Ashkenazi, A. (2009) Cullin 3-based polyubiquitination and p62-dependent aggregation of caspase-8 mediate extrinsic apoptosis signaling. Cell 137, 721-735.   DOI
60 Komatsu, M., Kurokawa, H., Waguri, S., Taguchi, K., Kobayashi, A., Ichimura, Y., Sou, Y. S., Ueno, I., Sakamoto, A., Tong, K. I., Kim, M., Nishito, Y., Iemura, S., Natsume, T., Ueno, T., Kominami, E., Motohashi, H., Tanaka, K. and Yamamoto, M. (2010) The selective autophagy substrate p62 activates the stress responsive transcription factor Nrf2 through inactivation of Keap1. Nat. Cell Biol. 12, 213-223.   DOI
61 Tang, D., Kang, R., Berghe, T. V., Vandenabeele, P. and Kroemer, G. (2019) The molecular machinery of regulated cell death. Cell Res. 29, 347-364.   DOI
62 Shvets, E., Fass, E., Scherz-Shouval, R. and Elazar, Z. (2008) The N-terminus and Phe52 residue of LC3 recruit p62/SQSTM1 into autophagosomes. J. Cell Sci. 121, 2685-2695.   DOI
63 Song, P., Li, S., Wu, H., Gao, R., Rao, G., Wang, D., Chen, Z., Ma, B., Wang, H., Sui, N., Deng, H., Zhang, Z., Tang, T., Tan, Z., Han, Z., Lu, T., Zhu, Y. and Chen, Q. (2016) Parkin promotes proteasomal degradation of p62: implication of selective vulnerability of neuronal cells in the pathogenesis of Parkinson's disease. Protein Cell 7, 114-129.   DOI