1
|
Frachon N, Demaretz S, Seaayfan E, Chelbi L, Bakhos-Douaihy D, Laghmani K. AUP1 Regulates the Endoplasmic Reticulum-Associated Degradation and Polyubiquitination of NKCC2. Cells 2024; 13:389. [PMID: 38474353 PMCID: PMC10931229 DOI: 10.3390/cells13050389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 01/30/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Inactivating mutations of kidney Na-K-2Cl cotransporter NKCC2 lead to antenatal Bartter syndrome (BS) type 1, a life-threatening salt-losing tubulopathy. We previously reported that this serious inherited renal disease is linked to the endoplasmic reticulum-associated degradation (ERAD) pathway. The purpose of this work is to characterize further the ERAD machinery of NKCC2. Here, we report the identification of ancient ubiquitous protein 1 (AUP1) as a novel interactor of NKCC2 ER-resident form in renal cells. AUP1 is also an interactor of the ER lectin OS9, a key player in the ERAD of NKCC2. Similar to OS9, AUP1 co-expression decreased the amount of total NKCC2 protein by enhancing the ER retention and associated protein degradation of the cotransporter. Blocking the ERAD pathway with the proteasome inhibitor MG132 or the α-mannosidase inhibitor kifunensine fully abolished the AUP1 effect on NKCC2. Importantly, AUP1 knock-down or inhibition by overexpressing its dominant negative form strikingly decreased NKCC2 polyubiquitination and increased the protein level of the cotransporter. Interestingly, AUP1 co-expression produced a more profound impact on NKCC2 folding mutants. Moreover, AUP1 also interacted with the related kidney cotransporter NCC and downregulated its expression, strongly indicating that AUP1 is a common regulator of sodium-dependent chloride cotransporters. In conclusion, our data reveal the presence of an AUP1-mediated pathway enhancing the polyubiquitination and ERAD of NKCC2. The characterization and selective regulation of specific ERAD constituents of NKCC2 and its pathogenic mutants could open new avenues in the therapeutic strategies for type 1 BS treatment.
Collapse
Affiliation(s)
- Nadia Frachon
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France; (N.F.); (S.D.); (E.S.); (D.B.-D.)
- CNRS, ERL8228, F-75006 Paris, France
| | - Sylvie Demaretz
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France; (N.F.); (S.D.); (E.S.); (D.B.-D.)
- CNRS, ERL8228, F-75006 Paris, France
| | - Elie Seaayfan
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France; (N.F.); (S.D.); (E.S.); (D.B.-D.)
- CNRS, ERL8228, F-75006 Paris, France
| | - Lydia Chelbi
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France; (N.F.); (S.D.); (E.S.); (D.B.-D.)
- CNRS, ERL8228, F-75006 Paris, France
| | - Dalal Bakhos-Douaihy
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France; (N.F.); (S.D.); (E.S.); (D.B.-D.)
- CNRS, ERL8228, F-75006 Paris, France
| | - Kamel Laghmani
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France; (N.F.); (S.D.); (E.S.); (D.B.-D.)
- CNRS, ERL8228, F-75006 Paris, France
| |
Collapse
|
2
|
Cui Y, Liu D, Zhao Z, Zhang J, Li S, Liu Z. Transcriptome analysis and identification of genes associated with leaf crude protein content in foxtail millet [ Setaria italica (L.) P. Beauv.]. Front Genet 2023; 14:1122212. [PMID: 36741329 PMCID: PMC9895776 DOI: 10.3389/fgene.2023.1122212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/06/2023] [Indexed: 01/22/2023] Open
Abstract
Introduction: Spruce spider mite is a primary insect pest of Chinese chestnut in China and seriously influences its yield and quality. However, the current management against this mite is costly and poorly effective. In previous research, we bred several foxtail millet materials for interplanting with chestnut tree, and found that they had high levels of crude protein (CP) in leaves and attracted spruce spider mite to feed on the leaves, thereby reducing chestnut damage. Methods: In this study, four foxtail millet varieties with significant differences in leaf crude protein content were used for high-throughput sequencing and identification of genes associated with leaf crude protein content. Gene enrichment analyses were carried out to comprehend the functions of these genes and the biological processes in which they are involved. In addition, transcription factors (TFs) were evaluated. Results: 435 differentially expressed genes (DEGs) were identified, suggesting their potential role in crude protein accumulation. Some differentially expressed genes were found to be associated with nitrogen metabolism and ubiquitin-mediated proteolysis pathways. Moreover, we identified 40 TF genes categorized into 11 transcription factor families. Discussion: Our findings represent an important resource that clarifies the mechanisms of accumulation and control of leaf crude protein in foxtail millet, and provide an opportunity for suppression of spruce spider mite attack on Chinese chestnut by interplanting with foxtail millet varieties with high concentrations of leaf crude protein.
Collapse
Affiliation(s)
- Yanjiao Cui
- Department of Life Sciences, Tangshan Normal University, Tangshan, China
| | - Dan Liu
- Tianjin Key Laboratory of Crop Genetics and Breeding, Institute of Crop Sciences, Tianjin Academy of Agricultural Sciences, Tianjin, China
| | - Zilong Zhao
- Department of Life Sciences, Tangshan Normal University, Tangshan, China
| | - Jing Zhang
- Department of Life Sciences, Tangshan Normal University, Tangshan, China
| | - Suying Li
- Department of Life Sciences, Tangshan Normal University, Tangshan, China,*Correspondence: Suying Li, ; Zhengli Liu,
| | - Zhengli Liu
- Department of Life Sciences, Tangshan Normal University, Tangshan, China,*Correspondence: Suying Li, ; Zhengli Liu,
| |
Collapse
|
3
|
Smith CE, Tsai YC, Liang YH, Khago D, Mariano J, Li J, Tarasov SG, Gergel E, Tsai B, Villaneuva M, Clapp ME, Magidson V, Chari R, Byrd RA, Ji X, Weissman AM. A structurally conserved site in AUP1 binds the E2 enzyme UBE2G2 and is essential for ER-associated degradation. PLoS Biol 2021; 19:e3001474. [PMID: 34879065 PMCID: PMC8699718 DOI: 10.1371/journal.pbio.3001474] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 12/23/2021] [Accepted: 11/05/2021] [Indexed: 12/22/2022] Open
Abstract
Endoplasmic reticulum-associated degradation (ERAD) is a protein quality control pathway of fundamental importance to cellular homeostasis. Although multiple ERAD pathways exist for targeting topologically distinct substrates, all pathways require substrate ubiquitination. Here, we characterize a key role for the UBE2G2 Binding Region (G2BR) of the ERAD accessory protein ancient ubiquitous protein 1 (AUP1) in ERAD pathways. This 27-amino acid (aa) region of AUP1 binds with high specificity and low nanomolar affinity to the backside of the ERAD ubiquitin-conjugating enzyme (E2) UBE2G2. The structure of the AUP1 G2BR (G2BRAUP1) in complex with UBE2G2 reveals an interface that includes a network of salt bridges, hydrogen bonds, and hydrophobic interactions essential for AUP1 function in cells. The G2BRAUP1 shares significant structural conservation with the G2BR found in the E3 ubiquitin ligase gp78 and in vitro can similarly allosterically activate ubiquitination in conjunction with ERAD E3s. In cells, AUP1 is uniquely required to maintain normal levels of UBE2G2; this is due to G2BRAUP1 binding to the E2 and preventing its rapid degradation. In addition, the G2BRAUP1 is required for both ER membrane recruitment of UBE2G2 and for its activation at the ER membrane. Thus, by binding to the backside of a critical ERAD E2, G2BRAUP1 plays multiple critical roles in ERAD.
Collapse
Affiliation(s)
- Christopher E. Smith
- Laboratory of Protein Dynamics and Signaling, Center for Cancer Research, NCI, National Institutes of Health, Frederick, Maryland, United States of America
| | - Yien Che Tsai
- Laboratory of Protein Dynamics and Signaling, Center for Cancer Research, NCI, National Institutes of Health, Frederick, Maryland, United States of America
| | - Yu-He Liang
- Center for Structural Biology, Center for Cancer Research, NCI, National Institutes of Health, Frederick, Maryland, United States of America
| | - Domarin Khago
- Center for Structural Biology, Center for Cancer Research, NCI, National Institutes of Health, Frederick, Maryland, United States of America
| | - Jennifer Mariano
- Laboratory of Protein Dynamics and Signaling, Center for Cancer Research, NCI, National Institutes of Health, Frederick, Maryland, United States of America
| | - Jess Li
- Center for Structural Biology, Center for Cancer Research, NCI, National Institutes of Health, Frederick, Maryland, United States of America
| | - Sergey G. Tarasov
- Center for Structural Biology, Center for Cancer Research, NCI, National Institutes of Health, Frederick, Maryland, United States of America
| | - Emma Gergel
- Laboratory of Protein Dynamics and Signaling, Center for Cancer Research, NCI, National Institutes of Health, Frederick, Maryland, United States of America
| | - Borong Tsai
- Laboratory of Protein Dynamics and Signaling, Center for Cancer Research, NCI, National Institutes of Health, Frederick, Maryland, United States of America
| | - Matthew Villaneuva
- Laboratory of Protein Dynamics and Signaling, Center for Cancer Research, NCI, National Institutes of Health, Frederick, Maryland, United States of America
| | - Michelle E. Clapp
- Genome Modification Core, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Valentin Magidson
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Raj Chari
- Genome Modification Core, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - R. Andrew Byrd
- Center for Structural Biology, Center for Cancer Research, NCI, National Institutes of Health, Frederick, Maryland, United States of America
| | - Xinhua Ji
- Center for Structural Biology, Center for Cancer Research, NCI, National Institutes of Health, Frederick, Maryland, United States of America
| | - Allan M. Weissman
- Laboratory of Protein Dynamics and Signaling, Center for Cancer Research, NCI, National Institutes of Health, Frederick, Maryland, United States of America
| |
Collapse
|
4
|
Wang S, Wang Y, Wang S, Tong H, Tang Z, Wang J, Zhang Y, Ou J, Quan Z. Long Non-coding RNA FIRRE Acts as a miR-520a-3p Sponge to Promote Gallbladder Cancer Progression via Mediating YOD1 Expression. Front Genet 2021; 12:674653. [PMID: 34168678 PMCID: PMC8219055 DOI: 10.3389/fgene.2021.674653] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/06/2021] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVES The role of lncRNAs in gallbladder cancer (GBC) remains poorly understood. In this study, we explored the function of functional intergenic repeating RNA element (FIRRE) in GBC. MATERIALS AND METHODS Whole transcriptome resequencing was performed in three pairs of GBC tissues and adjacent non-tumor tissues. lncRNA FIRRE expression was verified by real-time PCR. The function of FIRRE in GBC was evaluated by experiments in vitro and in vivo. The mechanism of FIRRE was investigated via fluorescent in situ hybridization, RNA pull-down, dual luciferase reporter assays, and RNA immunoprecipitation. RESULTS FIRRE level was dramatically increased in GBC tissues compared to that in the adjacent non-tumor tissues. High expression of FIRRE was closely related to clinical stage and poor prognosis in GBC patients. Moreover, FIRRE remarkably enhanced proliferation and migration, and inhibited apoptosis of GBC cells. Mechanistically, FIRRE modulated YOD1 expression by sponging miR-520a-3p, thus contributing to the development of GBC. CONCLUSION Our data revealed that FIRRE might act as a novel mediator in GBC progression by sponging miR-520a-3p and regulating YOD1. FIRRE might be regarded as a potential diagnostic marker or target for GBC treatment.
Collapse
Affiliation(s)
- Shuqing Wang
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yang Wang
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shouhua Wang
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huanjun Tong
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhaohui Tang
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiandong Wang
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongjie Zhang
- Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Jingmin Ou
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiwei Quan
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
5
|
The ER-embedded UBE2J1/RNF26 ubiquitylation complex exerts spatiotemporal control over the endolysosomal pathway. Cell Rep 2021; 34:108659. [PMID: 33472082 DOI: 10.1016/j.celrep.2020.108659] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 10/26/2020] [Accepted: 12/22/2020] [Indexed: 02/02/2023] Open
Abstract
The endolysosomal system fulfills a wide variety of cellular functions, many of which are modulated through interactions with other organelles. In particular, the ER exerts spatiotemporal constraints on the organization and motility of endosomes and lysosomes. We have recently described the ER transmembrane E3 ubiquitin ligase RNF26 as a regulator of endolysosomal perinuclear positioning and transport dynamics. Here, we report that the ubiquitin conjugating enzyme UBE2J1, also anchored in the ER membrane, partners with RNF26 in this context, and that the cellular activity of the resulting E2/E3 pair is localized in a perinuclear ER subdomain and supported by transmembrane interactions. Through modification of SQSTM1/p62 on lysine 435, the ER-embedded UBE2J1/RNF26 ubiquitylation complex recruits endosomal adaptors to immobilize their cognate vesicles in the perinuclear region of the cell. The resulting spatiotemporal compartmentalization promotes the trafficking of activated EGFR to lysosomes and facilitates the termination of EGF-induced AKT signaling.
Collapse
|
6
|
van de Weijer ML, Krshnan L, Liberatori S, Guerrero EN, Robson-Tull J, Hahn L, Lebbink RJ, Wiertz EJHJ, Fischer R, Ebner D, Carvalho P. Quality Control of ER Membrane Proteins by the RNF185/Membralin Ubiquitin Ligase Complex. Mol Cell 2020; 79:768-781.e7. [PMID: 32738194 PMCID: PMC7482433 DOI: 10.1016/j.molcel.2020.07.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 05/06/2020] [Accepted: 07/07/2020] [Indexed: 12/16/2022]
Abstract
Misfolded proteins in the endoplasmic reticulum (ER) are degraded by ER-associated degradation (ERAD). Although ERAD components involved in degradation of luminal substrates are well characterized, much less is known about quality control of membrane proteins. Here, we analyzed the degradation pathways of two short-lived ER membrane model proteins in mammalian cells. Using a CRISPR-Cas9 genome-wide library screen, we identified an ERAD branch required for quality control of a subset of membrane proteins. Using biochemical and mass spectrometry approaches, we showed that this ERAD branch is defined by an ER membrane complex consisting of the ubiquitin ligase RNF185, the ubiquitin-like domain containing proteins TMUB1/2 and TMEM259/Membralin, a poorly characterized protein. This complex cooperates with cytosolic ubiquitin ligase UBE3C and p97 ATPase in degrading their membrane substrates. Our data reveal that ERAD branches have remarkable specificity for their membrane substrates, suggesting that multiple, perhaps combinatorial, determinants are involved in substrate selection. The RNF185 ubiquitin ligase, Membralin, and TMUB1/2 assemble into an ERAD complex RNF185/Membralin complex targets membrane proteins, including CYP51A1 and TMUB2 RNF185/Membralin and TEB4 ERAD complexes recognize distinct substrate features TEB4 ERAD complex recognizes substrates through their transmembrane domain
Collapse
Affiliation(s)
- Michael L van de Weijer
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Logesvaran Krshnan
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Sabrina Liberatori
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Elena Navarro Guerrero
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford OX3 7FZ, UK
| | - Jacob Robson-Tull
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Lilli Hahn
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Robert Jan Lebbink
- Medical Microbiology, University Medical Center Utrecht, 3584 Utrecht, the Netherlands
| | - Emmanuel J H J Wiertz
- Medical Microbiology, University Medical Center Utrecht, 3584 Utrecht, the Netherlands
| | - Roman Fischer
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford OX3 7FZ, UK
| | - Daniel Ebner
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford OX3 7FZ, UK
| | - Pedro Carvalho
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK.
| |
Collapse
|
7
|
Lopata A, Kniss A, Löhr F, Rogov VV, Dötsch V. Ubiquitination in the ERAD Process. Int J Mol Sci 2020; 21:ijms21155369. [PMID: 32731622 PMCID: PMC7432864 DOI: 10.3390/ijms21155369] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/23/2020] [Accepted: 07/27/2020] [Indexed: 01/08/2023] Open
Abstract
In this review, we focus on the ubiquitination process within the endoplasmic reticulum associated protein degradation (ERAD) pathway. Approximately one third of all synthesized proteins in a cell are channeled into the endoplasmic reticulum (ER) lumen or are incorporated into the ER membrane. Since all newly synthesized proteins enter the ER in an unfolded manner, folding must occur within the ER lumen or co-translationally, rendering misfolding events a serious threat. To prevent the accumulation of misfolded protein in the ER, proteins that fail the quality control undergo retrotranslocation into the cytosol where they proceed with ubiquitination and degradation. The wide variety of misfolded targets requires on the one hand a promiscuity of the ubiquitination process and on the other hand a fast and highly processive mechanism. We present the various ERAD components involved in the ubiquitination process including the different E2 conjugating enzymes, E3 ligases, and E4 factors. The resulting K48-linked and K11-linked ubiquitin chains do not only represent a signal for degradation by the proteasome but are also recognized by the AAA+ ATPase Cdc48 and get in the process of retrotranslocation modified by enzymes bound to Cdc48. Lastly we discuss the conformations adopted in particular by K48-linked ubiquitin chains and their importance for degradation.
Collapse
|
8
|
Regulation of CFTR Biogenesis by the Proteostatic Network and Pharmacological Modulators. Int J Mol Sci 2020; 21:ijms21020452. [PMID: 31936842 PMCID: PMC7013518 DOI: 10.3390/ijms21020452] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 01/06/2020] [Accepted: 01/08/2020] [Indexed: 12/14/2022] Open
Abstract
Cystic fibrosis (CF) is the most common lethal inherited disease among Caucasians in North America and a significant portion of Europe. The disease arises from one of many mutations in the gene encoding the cystic fibrosis transmembrane conductance regulator, or CFTR. The most common disease-associated allele, F508del, along with several other mutations affect the folding, transport, and stability of CFTR as it transits from the endoplasmic reticulum (ER) to the plasma membrane, where it functions primarily as a chloride channel. Early data demonstrated that F508del CFTR is selected for ER associated degradation (ERAD), a pathway in which misfolded proteins are recognized by ER-associated molecular chaperones, ubiquitinated, and delivered to the proteasome for degradation. Later studies showed that F508del CFTR that is rescued from ERAD and folds can alternatively be selected for enhanced endocytosis and lysosomal degradation. A number of other disease-causing mutations in CFTR also undergo these events. Fortunately, pharmacological modulators of CFTR biogenesis can repair CFTR, permitting its folding, escape from ERAD, and function at the cell surface. In this article, we review the many cellular checkpoints that monitor CFTR biogenesis, discuss the emergence of effective treatments for CF, and highlight future areas of research on the proteostatic control of CFTR.
Collapse
|
9
|
YOD1 attenuates neurogenic proteotoxicity through its deubiquitinating activity. Neurobiol Dis 2018; 112:14-23. [PMID: 29330040 DOI: 10.1016/j.nbd.2018.01.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 12/04/2017] [Accepted: 01/08/2018] [Indexed: 11/21/2022] Open
Abstract
Ubiquitination, a fundamental post-translational modification of intracellular proteins, is enzymatically reversed by deubiquitinase enzymes (deubiquitinases). >90 deubiquitinases have been identified. One of these enzymes, YOD1, possesses deubiquitinase activity and is similar to ovarian tumor domain-containing protein 1, which is associated with regulation of the endoplasmic reticulum (ER)-associated degradation pathway. Indeed, YOD1 is reported to be involved in the ER stress response induced by mislocalization of unfolded proteins in mammalian cells. However, it has remained unclear whether YOD1 is associated with pathophysiological conditions such as mitochondrial damage, impaired proteostasis, and neurodegeneration. We demonstrated that YOD1 possesses deubiquitinating activity and exhibits preference for K48- and K63-linked ubiquitin. Furthermore, YOD1 expression levels increased as a result of various stress conditions. We demonstrated that the neurogenic proteins that cause Huntington disease and Parkinson's disease induced upregulation of YOD1 level. We observed that YOD1 reduced disease cytotoxicity through efficient degradation of mutant proteins, whereas this activity was abolished by catalytically inactive YOD1. Additionally, YOD1 localized to Lewy bodies in Parkinson's disease patients. Collectively, these data suggest that the deubiquitinase YOD1 contributes to pathogenesis of neurodegenerative disease by decreasing ubiquitination of abnormal proteins and their subsequent degradation.
Collapse
|
10
|
Chen S, Tan Y, Deng H, Shen Z, Liu Y, Wu P, Tan C, Jiang Y. UBE2J2 promotes hepatocellular carcinoma cell epithelial-mesenchymal transition and invasion in vitro. Oncotarget 2017; 8:71736-71749. [PMID: 29069742 PMCID: PMC5641085 DOI: 10.18632/oncotarget.17601] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 04/12/2017] [Indexed: 11/25/2022] Open
Abstract
Ubiquitin-conjugating enzyme E2 J2 (UBE2J2) is an ubiquitin proteasome component that responds to proteotoxic stress. We found that UBE2J2 was highly expressed in cellular protrusions of HCCLM3 metastatic hepatocellular carcinoma (HC) cells. Immunohistochemical analyses showed that UBE2J2 was expressed at higher levels in HC patient tissues than in corresponding non-tumor tissues. Because cellular protrusions are important for cell invasion, we hypothesized that UBE2J2 promotes HC cell invasion. We used chip-based surface plasmon resonance (SPR) to assess possible mechanisms of UBE2J2-regulated HCCLM3 cell invasion. We found that p-EGFR interacted with UBE2J2, and this finding was confirmed by co-immunoprecipitation analysis. UBE2J2 overexpression activated endothelial-mesenchymal transition in the non-invasive SMMC7721 HC cell line, and promoted invasion. UBE2J2 silencing reduced HCCLM3 cell invasion and endocytosis, and downregulated p-EGFR expression. p-EGFR inhibition by lapatinib reduced UBE2J2-promoted cell invasion, suggesting p-EGFR is important for UBE2J2-mediated HCCLM3 cell invasion. These findings demonstrate that endocytosis by HC cells is closely related to invasion, and may provide new anti-HC therapeutic targets. UBE2J2 may also be a novel biomarker for clinical HC diagnosis.
Collapse
Affiliation(s)
- Shaopeng Chen
- Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China
| | - Ying Tan
- Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China
| | | | - Zhifa Shen
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Yanhong Liu
- Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China
| | - Pan Wu
- Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China
| | - Chunyan Tan
- Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China
| | - Yuyang Jiang
- Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China.,School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, P. R. China
| |
Collapse
|
11
|
Schimmack G, Schorpp K, Kutzner K, Gehring T, Brenke JK, Hadian K, Krappmann D. YOD1/TRAF6 association balances p62-dependent IL-1 signaling to NF-κB. eLife 2017; 6. [PMID: 28244869 PMCID: PMC5340530 DOI: 10.7554/elife.22416] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 02/26/2017] [Indexed: 12/25/2022] Open
Abstract
The ubiquitin ligase TRAF6 is a key regulator of canonical IκB kinase (IKK)/NF-κB signaling in response to interleukin-1 (IL-1) stimulation. Here, we identified the deubiquitinating enzyme YOD1 (OTUD2) as a novel interactor of TRAF6 in human cells. YOD1 binds to the C-terminal TRAF homology domain of TRAF6 that also serves as the interaction surface for the adaptor p62/Sequestosome-1, which is required for IL-1 signaling to NF-κB. We show that YOD1 competes with p62 for TRAF6 association and abolishes the sequestration of TRAF6 to cytosolic p62 aggregates by a non-catalytic mechanism. YOD1 associates with TRAF6 in unstimulated cells but is released upon IL-1β stimulation, thereby facilitating TRAF6 auto-ubiquitination as well as NEMO/IKKγ substrate ubiquitination. Further, IL-1 triggered IKK/NF-κB signaling and induction of target genes is decreased by YOD1 overexpression and augmented after YOD1 depletion. Hence, our data define that YOD1 antagonizes TRAF6/p62-dependent IL-1 signaling to NF-κB. DOI:http://dx.doi.org/10.7554/eLife.22416.001
Collapse
Affiliation(s)
- Gisela Schimmack
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Kenji Schorpp
- Assay Development and Screening Platform, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Kerstin Kutzner
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Torben Gehring
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Jara Kerstin Brenke
- Assay Development and Screening Platform, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Kamyar Hadian
- Assay Development and Screening Platform, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Daniel Krappmann
- Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
12
|
Feng L, Zhang J, Zhu N, Ding Q, Zhang X, Yu J, Qiang W, Zhang Z, Ma Y, Huang D, Shen Y, Fang S, Yu Y, Wang H, Shen Y. Ubiquitin ligase SYVN1/HRD1 facilitates degradation of the SERPINA1 Z variant/α-1-antitrypsin Z variant via SQSTM1/p62-dependent selective autophagy. Autophagy 2017; 13:686-702. [PMID: 28121484 PMCID: PMC5388218 DOI: 10.1080/15548627.2017.1280207] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
SERPINA1/AAT/α-1-antitrypsin (serpin family A member 1) deficiency (SERPINA1/ AAT-D) is an autosomal recessive disorder characterized by the retention of misfolded SERPINA1/AAT in the endoplasmic reticulum (ER) of hepatocytes and a significant reduction of serum SERPINA1/AAT level. The Z variant of SERPINA1/AAT, containing a Glu342Lys (E342K) mutation (SERPINA1E342K/ATZ), the most common form of SERPINA1/AAT-D, is prone to misfolding and polymerization, which retains it in the ER of hepatocytes and leads to liver injury. Both proteasome and macroautophagy/autophagy pathways are responsible for disposal of SERPINA1E342K/ATZ after it accumulates in the ER. However, the mechanisms by which SERPINA1E342K/ATZ is selectively degraded by autophagy remain unknown. Here, we showed that ER membrane-spanning ubiquitin ligase (E3) SYVN1/HRD1 enhances the degradation of SERPINA1E342K/ATZ through the autophagy-lysosome pathway. We found that SYVN1 promoted SERPINA1E342K/ATZ, especially Triton X 100-insoluble SERPINA1E342K/ATZ clearance. However, the effect of SYVN1 in SERPINA1E342K/ATZ clearance was impaired after autophagy inhibition, as well as in autophagy-related 5 (atg5) knockout cells. On the contrary, autophagy induction enhanced SYVN1-mediated SERPINA1E342K/ATZ degradation. Further study showed that SYVN1 mediated SERPINA1E342K/ATZ ubiquitination, which is required for autophagic degradation of SERPINA1E342K/ATZ by promoting the interaction between SERPINA1E342K/ATZ and SQSTM1/p62 for formation of the autophagy complex. Interestingly, SYVN1-mediated lysine 48 (K48)-linked polyubiquitin chains that conjugated onto SERPINA1E342K/ATZ might predominantly bind to the ubiquitin-associated (UBA) domain of SQSTM1 and couple the ubiquitinated SERPINA1E342K/ATZ to the lysosome for degradation. In addition, autophagy inhibition attenuated the suppressive effect of SYVN1 on SERPINA1E342K/ATZ cytotoxicity, and the autophagy inducer rapamycin enhanced the suppressive effect of SYVN1 on SERPINA1E342K/ATZ-induced cell apoptosis. Therefore, this study proved that SYVN1 enhances SERPINA1E342K/ATZ degradation through SQSTM1-dependent autophagy and attenuates SERPINA1E342K/ATZ cytotoxicity.
Collapse
Affiliation(s)
- Lijie Feng
- a School of Basic Medical Sciences , Anhui Medical University , Hefei, Anhui , China.,b Institute of Biopharmaceuticals, Anhui Medical University , Hefei, Anhui , China
| | - Jin Zhang
- b Institute of Biopharmaceuticals, Anhui Medical University , Hefei, Anhui , China
| | - Na Zhu
- b Institute of Biopharmaceuticals, Anhui Medical University , Hefei, Anhui , China.,c The 4th Affiliated Hospital, Anhui Medical University , Hefei, Anhui , China
| | - Qian Ding
- a School of Basic Medical Sciences , Anhui Medical University , Hefei, Anhui , China.,b Institute of Biopharmaceuticals, Anhui Medical University , Hefei, Anhui , China
| | - Xiaojie Zhang
- a School of Basic Medical Sciences , Anhui Medical University , Hefei, Anhui , China.,b Institute of Biopharmaceuticals, Anhui Medical University , Hefei, Anhui , China
| | - Jishuang Yu
- a School of Basic Medical Sciences , Anhui Medical University , Hefei, Anhui , China.,b Institute of Biopharmaceuticals, Anhui Medical University , Hefei, Anhui , China
| | - Weimin Qiang
- a School of Basic Medical Sciences , Anhui Medical University , Hefei, Anhui , China.,b Institute of Biopharmaceuticals, Anhui Medical University , Hefei, Anhui , China
| | - Zhetao Zhang
- a School of Basic Medical Sciences , Anhui Medical University , Hefei, Anhui , China.,b Institute of Biopharmaceuticals, Anhui Medical University , Hefei, Anhui , China
| | - Yuyang Ma
- a School of Basic Medical Sciences , Anhui Medical University , Hefei, Anhui , China.,b Institute of Biopharmaceuticals, Anhui Medical University , Hefei, Anhui , China
| | - Dake Huang
- a School of Basic Medical Sciences , Anhui Medical University , Hefei, Anhui , China
| | - Yujun Shen
- a School of Basic Medical Sciences , Anhui Medical University , Hefei, Anhui , China.,b Institute of Biopharmaceuticals, Anhui Medical University , Hefei, Anhui , China
| | - Shengyun Fang
- a School of Basic Medical Sciences , Anhui Medical University , Hefei, Anhui , China.,b Institute of Biopharmaceuticals, Anhui Medical University , Hefei, Anhui , China.,d Center for Biomedical Engineering and Technology , University of Maryland , Baltimore , MD , USA
| | - Yifan Yu
- e Actuarial Science, School of Continuing Education , Columbia University , New York , NY , USA
| | - Haiping Wang
- a School of Basic Medical Sciences , Anhui Medical University , Hefei, Anhui , China.,b Institute of Biopharmaceuticals, Anhui Medical University , Hefei, Anhui , China
| | - Yuxian Shen
- a School of Basic Medical Sciences , Anhui Medical University , Hefei, Anhui , China.,b Institute of Biopharmaceuticals, Anhui Medical University , Hefei, Anhui , China
| |
Collapse
|
13
|
Rivera-Monroy J, Musiol L, Unthan-Fechner K, Farkas Á, Clancy A, Coy-Vergara J, Weill U, Gockel S, Lin SY, Corey DP, Kohl T, Ströbel P, Schuldiner M, Schwappach B, Vilardi F. Mice lacking WRB reveal differential biogenesis requirements of tail-anchored proteins in vivo. Sci Rep 2016; 6:39464. [PMID: 28000760 PMCID: PMC5175141 DOI: 10.1038/srep39464] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 11/22/2016] [Indexed: 02/06/2023] Open
Abstract
Tail-anchored (TA) proteins are post-translationally inserted into membranes. The TRC40 pathway targets TA proteins to the endoplasmic reticulum via a receptor comprised of WRB and CAML. TRC40 pathway clients have been identified using in vitro assays, however, the relevance of the TRC40 pathway in vivo remains unknown. We followed the fate of TA proteins in two tissue-specific WRB knockout mouse models and found that their dependence on the TRC40 pathway in vitro did not predict their reaction to receptor depletion in vivo. The SNARE syntaxin 5 (Stx5) was extremely sensitive to disruption of the TRC40 pathway. Screening yeast TA proteins with mammalian homologues, we show that the particular sensitivity of Stx5 is conserved, possibly due to aggregation propensity of its cytoplasmic domain. We establish that Stx5 is an autophagy target that is inefficiently membrane-targeted by alternative pathways. Our results highlight an intimate relationship between the TRC40 pathway and cellular proteostasis.
Collapse
Affiliation(s)
- Jhon Rivera-Monroy
- Department of Molecular Biology, Universitätsmedizin Göttingen, D-37073 Göttingen, Germany
| | - Lena Musiol
- Department of Molecular Biology, Universitätsmedizin Göttingen, D-37073 Göttingen, Germany
| | - Kirsten Unthan-Fechner
- Department of Molecular Biology, Universitätsmedizin Göttingen, D-37073 Göttingen, Germany
| | - Ákos Farkas
- Department of Molecular Biology, Universitätsmedizin Göttingen, D-37073 Göttingen, Germany
| | - Anne Clancy
- Department of Molecular Biology, Universitätsmedizin Göttingen, D-37073 Göttingen, Germany
| | - Javier Coy-Vergara
- Department of Molecular Biology, Universitätsmedizin Göttingen, D-37073 Göttingen, Germany
| | - Uri Weill
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Sarah Gockel
- Department of Cardiology &Pulmonology, Universitätsmedizin Göttingen, D-37075 Göttingen, Germany
| | - Shuh-Yow Lin
- Howard Hughes Medical Institute and Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - David P Corey
- Howard Hughes Medical Institute and Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Tobias Kohl
- Department of Cardiology &Pulmonology, Universitätsmedizin Göttingen, D-37075 Göttingen, Germany
| | - Philipp Ströbel
- Institute of Pathology, Universitätsmedizin Göttingen, D-37073 Göttingen, Germany
| | - Maya Schuldiner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Blanche Schwappach
- Department of Molecular Biology, Universitätsmedizin Göttingen, D-37073 Göttingen, Germany.,Max-Planck Institute for Biophysical Chemistry, D-37077, Göttingen, Germany
| | - Fabio Vilardi
- Department of Molecular Biology, Universitätsmedizin Göttingen, D-37073 Göttingen, Germany
| |
Collapse
|
14
|
Hagiwara M, Ling J, Koenig PA, Ploegh HL. Posttranscriptional Regulation of Glycoprotein Quality Control in the Endoplasmic Reticulum Is Controlled by the E2 Ub-Conjugating Enzyme UBC6e. Mol Cell 2016; 63:753-67. [PMID: 27570074 DOI: 10.1016/j.molcel.2016.07.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 06/20/2016] [Accepted: 07/19/2016] [Indexed: 12/11/2022]
Abstract
ER-associated degradation (ERAD) is essential for protein quality control in the ER, not only when the ER is stressed, but also at steady state. We report a new layer of homeostatic control, in which ERAD activity itself is regulated posttranscriptionally and independently of the unfolded protein response by adjusting the endogenous levels of EDEM1, OS-9, and SEL1L (ERAD enhancers). Functional UBC6e requires its precise location in the ER to form a supramolecular complex with Derlin2. This complex targets ERAD enhancers for degradation, a function that depends on UBC6e's enzymatic activity. Ablation of UBC6e causes upregulation of active ERAD enhancers and so increases clearance not only of terminally misfolded substrates, but also of wild-type glycoproteins that fold comparatively slowly in vitro and in vivo. The levels of proteins that comprise the ERAD machinery are thus carefully tuned and adjusted to prevailing needs.
Collapse
Affiliation(s)
| | - Jingjing Ling
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | | | - Hidde L Ploegh
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.
| |
Collapse
|
15
|
Sasset L, Petris G, Cesaratto F, Burrone OR. The VCP/p97 and YOD1 Proteins Have Different Substrate-dependent Activities in Endoplasmic Reticulum-associated Degradation (ERAD). J Biol Chem 2015; 290:28175-28188. [PMID: 26463207 DOI: 10.1074/jbc.m115.656660] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Indexed: 11/06/2022] Open
Abstract
Endoplasmic reticulum-associated degradation (ERAD) is an essential quality control mechanism of the folding state of proteins in the secretory pathway that targets unfolded/misfolded polypeptides for proteasomal degradation. The cytosolic p97/valosin-containing protein is an essential ATPase for degradation of ERAD substrates. It has been considered necessary during retro-translocation to extract proteins from the endoplasmic reticulum that are otherwise supposed to accumulate in the endoplasmic reticulum lumen. The activity of the p97-associated deubiquitinylase YOD1 is also required for substrate disposal. We used the in vivo biotinylation retro-translocation assay in mammalian cells under conditions of impaired p97 or YOD1 activity to directly discriminate their requirements and diverse functions in ERAD. Using different ERAD substrates, we found that both proteins participate in two distinct retro-translocation steps. For CD4 and MHC-Iα, which are induced to degradation by the HIV-1 protein Vpu and by the CMV immunoevasins US2 and US11, respectively, p97 and YOD1 have a retro-translocation-triggering role. In contrast, for three other spontaneous ERAD model substrates (NS1, NHK-α1AT, and BST-2/Tetherin), p97 and YOD1 are required in the downstream events of substrate deglycosylation and proteasomal degradation.
Collapse
Affiliation(s)
- Linda Sasset
- International Centre for Genetic Engineering and Biotechnology, Padriciano 99, 34149 Trieste, Italy
| | - Gianluca Petris
- CIBIO, University of Trento, Via delle Regole 101, 38123 Mattarello, Italy.
| | - Francesca Cesaratto
- International Centre for Genetic Engineering and Biotechnology, Padriciano 99, 34149 Trieste, Italy
| | - Oscar R Burrone
- International Centre for Genetic Engineering and Biotechnology, Padriciano 99, 34149 Trieste, Italy
| |
Collapse
|
16
|
Aseervatham J, Tran L, Machaca K, Boudker O. The Role of Flexible Loops in Folding, Trafficking and Activity of Equilibrative Nucleoside Transporters. PLoS One 2015; 10:e0136779. [PMID: 26406980 PMCID: PMC4583308 DOI: 10.1371/journal.pone.0136779] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 08/07/2015] [Indexed: 11/25/2022] Open
Abstract
Equilibrative nucleoside transporters (ENTs) are integral membrane proteins, which reside in plasma membranes of all eukaryotic cells and mediate thermodynamically downhill transport of nucleosides. This process is essential for nucleoside recycling, and also plays a key role in terminating adenosine-mediated cellular signaling. Furthermore, ENTs mediate the uptake of many drugs, including anticancer and antiviral nucleoside analogues. The structure and mechanism, by which ENTs catalyze trans-membrane transport of their substrates, remain unknown. To identify the core of the transporter needed for stability, activity, and for its correct trafficking to the plasma membrane, we have expressed human ENT deletion mutants in Xenopus laevis oocytes and determined their localization, transport properties and susceptibility to inhibition. We found that the carboxyl terminal trans-membrane segments are essential for correct protein folding and trafficking. In contrast, the soluble extracellular and intracellular loops appear to be dispensable, and must be involved in the fine-tuning of transport regulation.
Collapse
Affiliation(s)
- Jaya Aseervatham
- Department of Physiology and Biophysics, Weill Cornell Medical College in Qatar, Qatar foundation, Education City, Doha, Qatar
| | - Lucky Tran
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, United States of America
| | - Khaled Machaca
- Department of Physiology and Biophysics, Weill Cornell Medical College in Qatar, Qatar foundation, Education City, Doha, Qatar
| | - Olga Boudker
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, United States of America
- * E-mail:
| |
Collapse
|
17
|
Intracellular expression of camelid single-domain antibodies specific for influenza virus nucleoprotein uncovers distinct features of its nuclear localization. J Virol 2014; 89:2792-800. [PMID: 25540369 DOI: 10.1128/jvi.02693-14] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Perturbation of protein-protein interactions relies mostly on genetic approaches or on chemical inhibition. Small RNA viruses, such as influenza A virus, do not easily lend themselves to the former approach, while chemical inhibition requires that the target protein be druggable. A lack of tools thus constrains the functional analysis of influenza virus-encoded proteins. We generated a panel of camelid-derived single-domain antibody fragments (VHHs) against influenza virus nucleoprotein (NP), a viral protein essential for nuclear trafficking and packaging of the influenza virus genome. We show that these VHHs can target NP in living cells and perturb NP's function during infection. Cytosolic expression of NP-specific VHHs (αNP-VHHs) disrupts virus replication at an early stage of the life cycle. Based on their specificity, these VHHs fall into two distinct groups. Both prevent nuclear import of the viral ribonucleoprotein (vRNP) complex without disrupting nuclear import of NP alone. Different stages of the virus life cycle thus rely on distinct nuclear localization motifs of NP. Their molecular characterization may afford new means of intervention in the virus life cycle. IMPORTANCE Many proteins encoded by RNA viruses are refractory to manipulation due to their essential role in replication. Thus, studying their function and determining how to disrupt said function through pharmaceutical intervention are difficult. We present a novel method based on single-domain-antibody technology that permits specific targeting and disruption of an essential influenza virus protein in the absence of genetic manipulation of influenza virus itself. Characterization of such interactions may help identify new targets for pharmaceutical intervention. This approach can be extended to study proteins encoded by other viral pathogens.
Collapse
|
18
|
Lam SY, Murphy C, Foley LA, Ross SA, Wang TC, Fleming JV. The human ubiquitin conjugating enzyme UBE2J2 (Ubc6) is a substrate for proteasomal degradation. Biochem Biophys Res Commun 2014; 451:361-6. [DOI: 10.1016/j.bbrc.2014.07.099] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 07/22/2014] [Indexed: 11/15/2022]
|
19
|
Nakamura N, Harada K, Kato M, Hirose S. Ubiquitin-specific protease 19 regulates the stability of the E3 ubiquitin ligase MARCH6. Exp Cell Res 2014; 328:207-216. [PMID: 25088257 DOI: 10.1016/j.yexcr.2014.07.025] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Revised: 07/20/2014] [Accepted: 07/22/2014] [Indexed: 10/25/2022]
Abstract
Ubiquitin-specific protease (USP)19 is a recently identified deubiquitinating enzyme (DUB) having multiple splice variants and cellular functions. One variant encodes an endoplasmic reticulum (ER)-anchored DUB that rescues misfolded transmembrane proteins from ER-associated degradation (ERAD), but the underlying mechanism remains to be elucidated. Here, we show that USP19 interacts with the ERAD-associated E3 ubiquitin ligase MARCH6. Overexpression of USP19 delayed the degradation of MARCH6, leading to an increase in its protein level. In contrast, USP19 depletion resulted in decreased expression of MARCH6. We also show that USP19 overexpression reduced ubiquitination of MARCH6, while its knockdown had the opposite effect. In particular, USP19 was found to protect MARCH6 by deubiquitination from the p97-dependent proteasomal degradation. In addition, USP19 knockdown leads to increased expression of mutant ABCB11, an ERAD substrate of MARCH6. Moreover, USP19 is itself subjected to endoproteolytic processing by DUB activity, and the processing cleaves off an N-terminal cytoplasmic region of unknown function. However, elimination of this processing had no evident effect on MARCH6 stabilization. These results suggest that USP19 is involved in the regulation of ERAD by controlling the stability of MARCH6 via deubiquitination.
Collapse
Affiliation(s)
- Nobuhiro Nakamura
- Department of Biological Sciences, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan.
| | - Kumi Harada
- Department of Biological Sciences, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Masako Kato
- Department of Biological Sciences, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Shigehisa Hirose
- Department of Biological Sciences, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| |
Collapse
|
20
|
Feng YX, Sokol ES, Del Vecchio CA, Sanduja S, Claessen JHL, Proia TA, Jin DX, Reinhardt F, Ploegh HL, Wang Q, Gupta PB. Epithelial-to-mesenchymal transition activates PERK-eIF2α and sensitizes cells to endoplasmic reticulum stress. Cancer Discov 2014; 4:702-15. [PMID: 24705811 DOI: 10.1158/2159-8290.cd-13-0945] [Citation(s) in RCA: 243] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
UNLABELLED Epithelial-to-mesenchymal transition (EMT) promotes both tumor progression and drug resistance, yet few vulnerabilities of this state have been identified. Using selective small molecules as cellular probes, we show that induction of EMT greatly sensitizes cells to agents that perturb endoplasmic reticulum (ER) function. This sensitivity to ER perturbations is caused by the synthesis and secretion of large quantities of extracellular matrix (ECM) proteins by EMT cells. Consistent with their increased secretory output, EMT cells display a branched ER morphology and constitutively activate the PERK-eIF2α axis of the unfolded protein response (UPR). Protein kinase RNA-like ER kinase (PERK) activation is also required for EMT cells to invade and metastasize. In human tumor tissues, EMT gene expression correlates strongly with both ECM and PERK-eIF2α genes, but not with other branches of the UPR. Taken together, our findings identify a novel vulnerability of EMT cells, and demonstrate that the PERK branch of the UPR is required for their malignancy. SIGNIFICANCE EMT drives tumor metastasis and drug resistance, highlighting the need for therapies that target this malignant subpopulation. Our findings identify a previously unrecognized vulnerability of cancer cells that have undergone an EMT: sensitivity to ER stress. We also find that PERK-eIF2α signaling, which is required to maintain ER homeostasis, is also indispensable for EMT cells to invade and metastasize.
Collapse
Affiliation(s)
- Yu-Xiong Feng
- Whitehead Institute for Biomedical Research; Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Ethan S Sokol
- Whitehead Institute for Biomedical Research; Department of Biology, Massachusetts Institute of Technology; Koch Institute for Integrative Cancer Research; Harvard Stem Cell Institute; Broad Institute, Cambridge, Massachusetts; and Department of Chemistry, Duke University, Durham, North CarolinaAuthors' Affiliations:Whitehead Institute for Biomedical Research; Department of Biology, Massachusetts Institute of Technology; Koch Institute for Integrative Cancer Research; Harvard Stem Cell Institute; Broad Institute, Cambridge, Massachusetts; and Department of Chemistry, Duke University, Durham, North Carolina
| | - Catherine A Del Vecchio
- Whitehead Institute for Biomedical Research; Department of Biology, Massachusetts Institute of Technology; Koch Institute for Integrative Cancer Research; Harvard Stem Cell Institute; Broad Institute, Cambridge, Massachusetts; and Department of Chemistry, Duke University, Durham, North Carolina
| | - Sandhya Sanduja
- Whitehead Institute for Biomedical Research; Department of Biology, Massachusetts Institute of Technology; Koch Institute for Integrative Cancer Research; Harvard Stem Cell Institute; Broad Institute, Cambridge, Massachusetts; and Department of Chemistry, Duke University, Durham, North Carolina
| | - Jasper H L Claessen
- Whitehead Institute for Biomedical Research; Department of Biology, Massachusetts Institute of Technology; Koch Institute for Integrative Cancer Research; Harvard Stem Cell Institute; Broad Institute, Cambridge, Massachusetts; and Department of Chemistry, Duke University, Durham, North Carolina
| | - Theresa A Proia
- Whitehead Institute for Biomedical Research; Department of Biology, Massachusetts Institute of Technology; Koch Institute for Integrative Cancer Research; Harvard Stem Cell Institute; Broad Institute, Cambridge, Massachusetts; and Department of Chemistry, Duke University, Durham, North Carolina
| | - Dexter X Jin
- Whitehead Institute for Biomedical Research; Department of Biology, Massachusetts Institute of Technology; Koch Institute for Integrative Cancer Research; Harvard Stem Cell Institute; Broad Institute, Cambridge, Massachusetts; and Department of Chemistry, Duke University, Durham, North CarolinaAuthors' Affiliations:Whitehead Institute for Biomedical Research; Department of Biology, Massachusetts Institute of Technology; Koch Institute for Integrative Cancer Research; Harvard Stem Cell Institute; Broad Institute, Cambridge, Massachusetts; and Department of Chemistry, Duke University, Durham, North Carolina
| | - Ferenc Reinhardt
- Whitehead Institute for Biomedical Research; Department of Biology, Massachusetts Institute of Technology; Koch Institute for Integrative Cancer Research; Harvard Stem Cell Institute; Broad Institute, Cambridge, Massachusetts; and Department of Chemistry, Duke University, Durham, North Carolina
| | - Hidde L Ploegh
- Whitehead Institute for Biomedical Research; Department of Biology, Massachusetts Institute of Technology; Koch Institute for Integrative Cancer Research; Harvard Stem Cell Institute; Broad Institute, Cambridge, Massachusetts; and Department of Chemistry, Duke University, Durham, North CarolinaAuthors' Affiliations:Whitehead Institute for Biomedical Research; Department of Biology, Massachusetts Institute of Technology; Koch Institute for Integrative Cancer Research; Harvard Stem Cell Institute; Broad Institute, Cambridge, Massachusetts; and Department of Chemistry, Duke University, Durham, North Carolina
| | - Qiu Wang
- Whitehead Institute for Biomedical Research; Department of Biology, Massachusetts Institute of Technology; Koch Institute for Integrative Cancer Research; Harvard Stem Cell Institute; Broad Institute, Cambridge, Massachusetts; and Department of Chemistry, Duke University, Durham, North Carolina
| | - Piyush B Gupta
- Whitehead Institute for Biomedical Research; Department of Biology, Massachusetts Institute of Technology; Koch Institute for Integrative Cancer Research; Harvard Stem Cell Institute; Broad Institute, Cambridge, Massachusetts; and Department of Chemistry, Duke University, Durham, North CarolinaAuthors' Affiliations:Whitehead Institute for Biomedical Research; Department of Biology, Massachusetts Institute of Technology; Koch Institute for Integrative Cancer Research; Harvard Stem Cell Institute; Broad Institute, Cambridge, Massachusetts
| |
Collapse
|
21
|
Bianchini E, Fanin M, Mamchaoui K, Betto R, Sandonà D. Unveiling the degradative route of the V247M α-sarcoglycan mutant responsible for LGMD-2D. Hum Mol Genet 2014; 23:3746-58. [PMID: 24565866 PMCID: PMC4065151 DOI: 10.1093/hmg/ddu088] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Many membrane and secretory proteins that fail to pass quality control in the endoplasmic reticulum (ER) are dislocated into the cytosol and degraded by the proteasome. In applying rigid rules, however, quality control sometimes discharges proteins that, even though defective, retain their function. The unnecessary removal of such proteins represents the pathogenetic hallmark of diverse genetic diseases, in the case of ΔF508 mutant of cystic fibrosis transmembrane conductance regulator being probably the best known example. Recently, the inappropriate proteasomal degradation of skeletal muscle sarcoglycans (α, β, γ and δ) with missense mutation has been proposed to be at the bases of mild-to-severe forms of limb girdle muscular dystrophy (LGMD) known as type 2D, 2E, 2C and 2F, respectively. The quality control pathway responsible for sarcoglycan mutant disposal, however, is so far unexplored. Here we reveal key components of the degradative route of V247M α-sarcoglycan mutant, the second most frequently reported mutation in LGMD-2D. The disclosure of the pathway, which is led by the E3 ligases HRD1 and RFP2, permits to identify new potential druggable targets of a disease for which no effective therapy is at present available. Notably, we show that the pharmacological inhibition of HRD1 activity rescues the expression of V247-α-sarcoglycan both in a heterologous cell model and in myotubes derived from a LGMD-2D patient carrying the L31P/V247M mutations. This represents the first evidence that the activity of E3 ligases, the enzymes in charge of mutant fate, can be eligible for drug interventions to treat sarcoglycanopathy.
Collapse
Affiliation(s)
| | - Marina Fanin
- Department of Neurosciences, University of Padova, Padova 35131, Italy
| | - Kamel Mamchaoui
- Institut de Myologie, UPMC UM76, INSERM U974, CNRS UMR 7215, Paris 6, France
| | - Romeo Betto
- Institute of Neuroscience, Consiglio Nazionale delle Ricerche, Padova 35131, Italy
| | | |
Collapse
|
22
|
Kuijpers M, van Dis V, Haasdijk ED, Harterink M, Vocking K, Post JA, Scheper W, Hoogenraad CC, Jaarsma D. Amyotrophic lateral sclerosis (ALS)-associated VAPB-P56S inclusions represent an ER quality control compartment. Acta Neuropathol Commun 2013; 1:24. [PMID: 24252306 PMCID: PMC3893532 DOI: 10.1186/2051-5960-1-24] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 06/01/2013] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Protein aggregation and the formation of intracellular inclusions are a central feature of many neurodegenerative disorders, but precise knowledge about their pathogenic role is lacking in most instances. Here we have characterized inclusions formed in transgenic mice carrying the P56S mutant form of VAPB that causes various motor neuron syndromes including ALS8. RESULTS Inclusions in motor neurons of VAPB-P56S transgenic mice are characterized by the presence of smooth ER-like tubular profiles, and are immunoreactive for factors that operate in the ER associated degradation (ERAD) pathway, including p97/VCP, Derlin-1, and the ER membrane chaperone BAP31. The presence of these inclusions does not correlate with signs of axonal and neuronal degeneration, and axotomy leads to their gradual disappearance, indicating that they represent reversible structures. Inhibition of the proteasome and knockdown of the ER membrane chaperone BAP31 increased the size of mutant VAPB inclusions in primary neuron cultures, while knockdown of TEB4, an ERAD ubiquitin-protein ligase, reduced their size. Mutant VAPB did not codistribute with mutant forms of seipin that are associated with an autosomal dominant motor neuron disease, and accumulate in a protective ER derived compartment termed ERPO (ER protective organelle) in neurons. CONCLUSIONS The data indicate that the VAPB-P56S inclusions represent a novel reversible ER quality control compartment that is formed when the amount of mutant VAPB exceeds the capacity of the ERAD pathway and that isolates misfolded and aggregated VAPB from the rest of the ER. The presence of this quality control compartment reveals an additional level of flexibility of neurons to cope with misfolded protein stress in the ER.
Collapse
|
23
|
Precise timing of ATPase activation drives targeting of tail-anchored proteins. Proc Natl Acad Sci U S A 2013; 110:7666-71. [PMID: 23610396 DOI: 10.1073/pnas.1222054110] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The localization of tail-anchored (TA) proteins, whose transmembrane domain resides at the extreme C terminus, presents major challenges to cellular protein targeting machineries. In eukaryotic cells, the highly conserved ATPase, guided entry of tail-anchored protein 3 (Get3), coordinates the delivery of TA proteins to the endoplasmic reticulum. How Get3 uses its ATPase cycle to drive this fundamental process remains unclear. Here, we establish a quantitative framework for the Get3 ATPase cycle and show that ATP specifically induces multiple conformational changes in Get3 that culminate in its ATPase activation through tetramerization. Further, upstream and downstream components actively regulate the Get3 ATPase cycle to ensure the precise timing of ATP hydrolysis in the pathway: the Get4/5 TA loading complex locks Get3 in the ATP-bound state and primes it for TA protein capture, whereas the TA substrate induces tetramerization of Get3 and activates its ATPase reaction 100-fold. Our results establish a precise model for how Get3 harnesses the energy from ATP to drive the membrane localization of TA proteins and illustrate how dimerization-activated nucleotide hydrolases regulate diverse cellular processes.
Collapse
|
24
|
Sanyal S, Claessen JHL, Ploegh HL. A viral deubiquitylating enzyme restores dislocation of substrates from the endoplasmic reticulum (ER) in semi-intact cells. J Biol Chem 2012; 287:23594-603. [PMID: 22619172 DOI: 10.1074/jbc.m112.365312] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Terminally misfolded glycoproteins are ejected from the endoplasmic reticulum (ER) to the cytosol and are destroyed by the ubiquitin proteasome system. A dominant negative version of the deubiquitylating enzyme Yod1 (Yod1C160S) causes accumulation of dislocation substrates in the ER. Failure to remove ubiquitin from the dislocation substrate might therefore stall the reaction at the exit site from the ER. We hypothesized that addition of a promiscuous deubiquitylase should overcome this blockade and restore dislocation. We monitored ER-to-cytosol transport of misfolded proteins in cells permeabilized at high cell density by perfringolysin O, a pore-forming cytolysin. This method allows ready access of otherwise impermeant reagents to the intracellular milieu with minimal dilution of cytoplasmic components. We show that addition of the purified Epstein-Barr virus deubiquitylase to semi-intact cells indeed initiates dislocation of a stalled substrate intermediate, resulting in stabilization of substrates in the cytosol. Our data provide new mechanistic insight in the dislocation reaction and support a model where failure to deubiquitylate an ER-resident protein occludes the dislocon and causes upstream misfolded intermediates to accumulate.
Collapse
Affiliation(s)
- Sumana Sanyal
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | | | | |
Collapse
|
25
|
Fang L, Kaake RM, Patel VR, Yang Y, Baldi P, Huang L. Mapping the protein interaction network of the human COP9 signalosome complex using a label-free QTAX strategy. Mol Cell Proteomics 2012; 11:138-47. [PMID: 22474085 DOI: 10.1074/mcp.m111.016352] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The COP9 signalosome (CSN) is a multi-subunit protein complex that performs critical roles in controlling diverse cellular and developmental processes. Aberrant regulation of the CSN complex has been shown to lead to tumorigenesis. Despite its biological significance, our current knowledge of the function and regulation of the CSN complex is very limited. To explore CSN biology, we have developed and employed a new version of the tag team-based QTAX strategy (quantitative analysis of tandem affinity purified in vivo cross-linked (X) protein complexes) by incorporating a label-free MS method for quantitation. Coupled with protein interaction network analysis, this strategy produced a comprehensive and detailed assessment of the protein interaction network of the human CSN complex. In total, we quantitatively characterized 825 putative CSN-interacting proteins, with 270 classified as core interactors (captured by all three bait purifications). Biochemical validation further confirms the validity of selected identified interactors. This work presents the most complete analysis of the CSN interaction network to date, providing an inclusive set of physical interaction data consistent with physiological roles for the CSN. Moreover, the methodology described here is a general proteomic tool for the comprehensive study of protein interaction networks.
Collapse
Affiliation(s)
- Lei Fang
- Departments of Physiology & Biophysics and Developmental & Cell Biology, University of California, Irvine, California 92697, USA
| | | | | | | | | | | |
Collapse
|
26
|
Suzuki M, Otsuka T, Ohsaki Y, Cheng J, Taniguchi T, Hashimoto H, Taniguchi H, Fujimoto T. Derlin-1 and UBXD8 are engaged in dislocation and degradation of lipidated ApoB-100 at lipid droplets. Mol Biol Cell 2012; 23:800-10. [PMID: 22238364 PMCID: PMC3290640 DOI: 10.1091/mbc.e11-11-0950] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Apolipoprotein B-100 after lipidation is dislocated from the ER lumen to the cytoplasmic surface of lipid droplets for proteasomal degradation. UBXD8 in lipid droplets and Derlin-1 in the ER membrane interact with each other and with ApoB and are engaged in the pre- and postdislocation steps, respectively. Apolipoprotein B-100 (ApoB) is the principal component of very low density lipoprotein. Poorly lipidated nascent ApoB is extracted from the Sec61 translocon and degraded by proteasomes. ApoB lipidated in the endoplasmic reticulum (ER) lumen is also subjected to proteasomal degradation, but where and how it dislocates to the cytoplasm remain unknown. In the present study, we demonstrate that ApoB after lipidation is dislocated to the cytoplasmic surface of lipid droplets (LDs) and accumulates as ubiquitinated ApoB in Huh7 cells. Depletion of UBXD8, which is almost confined to LDs in this cell type, decreases recruitment of p97 to LDs and causes an increase of both ubiquitinated ApoB on the LD surface and lipidated ApoB in the ER lumen. In contrast, abrogation of Derlin-1 function induces an accumulation of lipidated ApoB in the ER lumen but does not increase ubiquitinated ApoB on the LD surface. UBXD8 and Derlin-1 bind with each other and with lipidated ApoB and show colocalization around LDs. These results indicate that ApoB after lipidation is dislocated from the ER lumen to the LD surface for proteasomal degradation and that Derlin-1 and UBXD8 are engaged in the predislocation and postdislocation steps, respectively.
Collapse
Affiliation(s)
- Michitaka Suzuki
- Department of Anatomy and Molecular Cell Biology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa, Nagoya 466-8550, Japan
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Claessen JHL, Ploegh HL. BAT3 guides misfolded glycoproteins out of the endoplasmic reticulum. PLoS One 2011; 6:e28542. [PMID: 22174835 PMCID: PMC3234288 DOI: 10.1371/journal.pone.0028542] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Accepted: 11/10/2011] [Indexed: 01/06/2023] Open
Abstract
Secretory and membrane proteins that fail to acquire their native conformation within the lumen of the Endoplasmic Reticulum (ER) are usually targeted for ubiquitin-dependent degradation by the proteasome. How partially folded polypeptides are kept from aggregation once ejected from the ER into the cytosol is not known. We show that BAT3, a cytosolic chaperone, is recruited to the site of dislocation through its interaction with Derlin2. Furthermore, we observe cytoplasmic BAT3 in a complex with a polypeptide that originates in the ER as a glycoprotein, an interaction that depends on the cytosolic disposition of both, visualized even in the absence of proteasomal inhibition. Cells depleted of BAT3 fail to degrade an established dislocation substrate. We thus implicate a cytosolic chaperone as an active participant in the dislocation of ER glycoproteins.
Collapse
Affiliation(s)
- Jasper H. L. Claessen
- Whitehead Institute for Biomedical Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Hidde L. Ploegh
- Whitehead Institute for Biomedical Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts
- * E-mail:
| |
Collapse
|
28
|
Abstract
The endoplasmic reticulum (ER) uses an elaborate surveillance system called the ER quality control (ERQC) system. The ERQC facilitates folding and modification of secretory and membrane proteins and eliminates terminally misfolded polypeptides through ER-associated degradation (ERAD) or autophagic degradation. This mechanism of ER protein surveillance is closely linked to redox and calcium homeostasis in the ER, whose balance is presumed to be regulated by a specific cellular compartment. The potential to modulate proteostasis and metabolism with chemical compounds or targeted siRNAs may offer an ideal option for the treatment of disease.
Collapse
|
29
|
The ubiquitin-selective segregase VCP/p97 orchestrates the response to DNA double-strand breaks. Nat Cell Biol 2011; 13:1376-82. [PMID: 22020440 DOI: 10.1038/ncb2367] [Citation(s) in RCA: 221] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Accepted: 09/23/2011] [Indexed: 12/12/2022]
Abstract
Unrepaired DNA double-strand breaks (DSBs) cause genetic instability that leads to malignant transformation or cell death. Cells respond to DSBs with the ordered recruitment of signalling and repair proteins to the site of lesion. Protein modification with ubiquitin is crucial for the signalling cascade, but how ubiquitylation coordinates the dynamic assembly of these complexes is poorly understood. Here, we show that the human ubiquitin-selective protein segregase p97 (also known as VCP; valosin-containing protein) cooperates with the ubiquitin ligase RNF8 to orchestrate assembly of signalling complexes and efficient DSB repair after exposure to ionizing radiation. p97 is recruited to DNA lesions by its ubiquitin adaptor UFD1-NPL4 and Lys-48-linked ubiquitin (K48-Ub) chains, whose formation is regulated by RNF8. p97 subsequently removes K48-Ub conjugates from sites of DNA damage to orchestrate proper association of 53BP1, BRCA1 and RAD51, three factors critical for DNA repair and genome surveillance mechanisms. Impairment of p97 activity decreases the level of DSB repair and cell survival after exposure to ionizing radiation. These findings identify the p97-UFD1-NPL4 complex as an essential factor in ubiquitin-governed DNA-damage response, highlighting its importance in guarding genome stability.
Collapse
|
30
|
Spandl J, Lohmann D, Kuerschner L, Moessinger C, Thiele C. Ancient ubiquitous protein 1 (AUP1) localizes to lipid droplets and binds the E2 ubiquitin conjugase G2 (Ube2g2) via its G2 binding region. J Biol Chem 2011; 286:5599-606. [PMID: 21127063 PMCID: PMC3037673 DOI: 10.1074/jbc.m110.190785] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Revised: 11/25/2010] [Indexed: 01/03/2023] Open
Abstract
Lipid droplets (LDs), the major intracellular storage sites for neutral lipids, consist of a neutral lipid core surrounded by a phospholipid monolayer membrane. In addition to their function in lipid storage, LDs participate in lipid biosynthesis and recently were implicated in proteasomal protein degradation and autophagy. To identify components of the protein degradation machinery on LDs, we studied several candidates identified in previous LD proteome analyses. Here, we demonstrate that the highly conserved and broadly expressed ancient ubiquitous protein 1 (AUP1) localizes to LDs, where it integrates into the LD surface in a monotopic fashion with both termini facing the cytosol. AUP1 contains a C-terminal domain with strong homology to a domain known as G2BR, which binds E2 ubiquitin conjugases. We show that AUP1, by means of its G2BR domain, binds to Ube2g2. This binding is abolished by deletion or mutation of the G2BR domain, although the LD localization of AUP1 is not affected. The presence of the AUP1-Ube2g2 complex at LDs provides a direct molecular link between LDs and the cellular ubiquitination machinery.
Collapse
Affiliation(s)
- Johanna Spandl
- From the Life and Medical Sciences Institute, University of Bonn, Carl-Troll-Strasse 31, 53115 Bonn, Germany
| | - Daniel Lohmann
- From the Life and Medical Sciences Institute, University of Bonn, Carl-Troll-Strasse 31, 53115 Bonn, Germany
| | - Lars Kuerschner
- From the Life and Medical Sciences Institute, University of Bonn, Carl-Troll-Strasse 31, 53115 Bonn, Germany
| | - Christine Moessinger
- From the Life and Medical Sciences Institute, University of Bonn, Carl-Troll-Strasse 31, 53115 Bonn, Germany
| | - Christoph Thiele
- From the Life and Medical Sciences Institute, University of Bonn, Carl-Troll-Strasse 31, 53115 Bonn, Germany
| |
Collapse
|