51
|
Chen X, Yang Q, Xiao L, Tang D, Dou QP, Liu J. Metal-based proteasomal deubiquitinase inhibitors as potential anticancer agents. Cancer Metastasis Rev 2018; 36:655-668. [PMID: 29039082 PMCID: PMC5721122 DOI: 10.1007/s10555-017-9701-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Deubiquitinases (DUBs) play an important role in protein quality control in eukaryotic cells due to their ability to specifically remove ubiquitin from substrate proteins. Therefore, recent findings have focused on the relevance of DUBs to cancer development, and pharmacological intervention on these enzymes has become a promising strategy for cancer therapy. In particular, several DUBs are physically and/or functionally associated with the proteasome and are attractive targets for the development of novel anticancer drugs. The successful clinical application of cisplatin in cancer treatment has prompted researchers to develop various metal-based anticancer agents with new properties. Recently, we have reported that several metal-based drugs, such as the antirheumatic gold agent auranofin (AF), the antifouling paint biocides copper pyrithione (CuPT) and zinc pyrithione (ZnPT), and also our two synthesized complexes platinum pyrithione (PtPT) and nickel pyrithione (NiPT), can target the proteasomal DUBs UCHL5 and USP14. In this review, we summarize the recently reported small molecule inhibitors of proteasomal DUBs, with a focus on discussion of the unique nature of metal-based proteasomal DUB inhibitors and their anticancer activity.
Collapse
Affiliation(s)
- Xin Chen
- Protein Modification and Degradation Lab, School of Basic Medical Sciences, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qianqian Yang
- Protein Modification and Degradation Lab, School of Basic Medical Sciences, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lu Xiao
- Protein Modification and Degradation Lab, School of Basic Medical Sciences, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou, China
| | - Daolin Tang
- Protein Modification and Degradation Lab, School of Basic Medical Sciences, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Surgery, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Q Ping Dou
- Protein Modification and Degradation Lab, School of Basic Medical Sciences, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou, China.,The Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Detroit, USA.,Department of Oncology, Pharmacology and Pathology, School of Medicine, Wayne State University, Detroit, MI, 48201-2013, USA
| | - Jinbao Liu
- Protein Modification and Degradation Lab, School of Basic Medical Sciences, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
52
|
Brunt L, Scholpp S. The function of endocytosis in Wnt signaling. Cell Mol Life Sci 2018; 75:785-795. [PMID: 28913633 PMCID: PMC5809524 DOI: 10.1007/s00018-017-2654-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 08/17/2017] [Accepted: 09/06/2017] [Indexed: 10/31/2022]
Abstract
Wnt growth factors regulate one of the most important signaling networks during development, tissue homeostasis and disease. Despite the biological importance of Wnt signaling, the mechanism of endocytosis during this process is ill described. Wnt molecules can act as paracrine signals, which are secreted from the producing cells and transported through neighboring tissue to activate signaling in target cells. Endocytosis of the ligand is important at several stages of action: One central function of endocytic trafficking in the Wnt pathway occurs in the source cell. Furthermore, the β-catenin-dependent Wnt ligands require endocytosis for signal activation and to regulate gene transcription in the responding cells. Alternatively, Wnt/β-catenin-independent signaling regulates endocytosis of cell adherence plaques to control cell migration. In this comparative review, we elucidate these three fundamental interconnected functions, which together regulate cellular fate and cellular behavior. Based on established hypotheses and recent findings, we develop a revised picture for the complex function of endocytosis in the Wnt signaling network.
Collapse
Affiliation(s)
- Lucy Brunt
- Living Systems Institute, School of Biosciences, College of Life and Environmental Science, University of Exeter, Exeter, EX4 4QD, UK
| | - Steffen Scholpp
- Living Systems Institute, School of Biosciences, College of Life and Environmental Science, University of Exeter, Exeter, EX4 4QD, UK.
- Institute of Toxicology and Genetics (ITG), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany.
| |
Collapse
|
53
|
Deubiquitinase inhibitor b-AP15 activates endoplasmic reticulum (ER) stress and inhibits Wnt/Notch1 signaling pathway leading to the reduction of cell survival in hepatocellular carcinoma cells. Eur J Pharmacol 2018; 825:10-18. [PMID: 29454609 DOI: 10.1016/j.ejphar.2018.02.020] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Revised: 02/01/2018] [Accepted: 02/14/2018] [Indexed: 12/11/2022]
Abstract
b-AP15, a potent and selective inhibitor of the ubiquitin-specific peptidase 14 (USP14), displays in vitro and in vivo antitumor abilities on some types of cancer cells. However, the mechanism underlying its action is not well elucidated. The purposes of the present study are to observe the potential impacts of b-AP15 on cell survival of hepatocellular carcinoma cells and to investigate whether and how this compound inhibits some survival-promoting signaling pathways. We found that b-AP15 significantly decreased cell viability and increased cell apoptosis in a dose-dependent manner in hepatocellular carcinoma cells, along with the perturbation of cell cycle and the decreased expressions of cell cycle-related proteins. We also demonstrated that the endoplasmic reticulum (ER) stress and the unfolded protein response (UPR) were enhanced by b-AP15 supplementation. The inhibition of ER stress/UPR only partly attenuated the cytotoxicity of b-AP15 on hepatocellular carcinoma cells. In addition, b-AP15 treatment inhibited Wnt/β-catenin and Notch1 signaling pathways, and suppressed phosphorylation of STAT3, Akt, and Erk1/2, which were not restored by the inhibition of ER stress/UPR. Furthermore, the expression levels of signaling molecules in Notch1 were reduced by specific inhibitor of Wnt/β-catenin pathway. Notably, either Wnt or Notch1 signaling inhibitor mitigated phosphorylation of STAT3, Akt, and Erk1/2, and mimicked the cytotoxicity of b-AP15 on hepatocellular carcinoma cells. These results clearly indicate that b-AP15 induced cytotoxic response to hepatocellular carcinoma cells by augmenting ER stress/UPR and inhibiting Wnt/Notch1 signaling pathways. This new finding provides a novel mechanism by which b-AP15 produces its antitumor therapeutic effects.
Collapse
|
54
|
Abstract
More than a decade after a Nobel Prize was awarded for the discovery of the ubiquitin-proteasome system and clinical approval of proteasome and ubiquitin E3 ligase inhibitors, first-generation deubiquitylating enzyme (DUB) inhibitors are now approaching clinical trials. However, although our knowledge of the physiological and pathophysiological roles of DUBs has evolved tremendously, the clinical development of selective DUB inhibitors has been challenging. In this Review, we discuss these issues and highlight recent advances in our understanding of DUB enzymology and biology as well as technological improvements that have contributed to the current interest in DUBs as therapeutic targets in diseases ranging from oncology to neurodegeneration.
Collapse
Affiliation(s)
- Jeanine A. Harrigan
- Mission Therapeutics Ltd, Moneta, Babraham Research Campus, Cambridge, CB22 3AT UK
| | - Xavier Jacq
- Mission Therapeutics Ltd, Moneta, Babraham Research Campus, Cambridge, CB22 3AT UK
| | - Niall M. Martin
- Mission Therapeutics Ltd, Moneta, Babraham Research Campus, Cambridge, CB22 3AT UK
- Present Address: and Department of Biochemistry, The Wellcome Trust and Cancer Research UK Gurdon Institute, Tennis Court Road, University of Cambridge, Cambridge, CB2 1QN UK
- Present address: Artios Pharmaceuticals Ltd, Maia, Babraham Research Campus, Cambridge CB22 3AT, UK,
| | - Stephen P. Jackson
- Mission Therapeutics Ltd, Moneta, Babraham Research Campus, Cambridge, CB22 3AT UK
- Present Address: and Department of Biochemistry, The Wellcome Trust and Cancer Research UK Gurdon Institute, Tennis Court Road, University of Cambridge, Cambridge, CB2 1QN UK
- Present address: Artios Pharmaceuticals Ltd, Maia, Babraham Research Campus, Cambridge CB22 3AT, UK,
| |
Collapse
|
55
|
Min Y, Lee S, Kim MJ, Chun E, Lee KY. Ubiquitin-Specific Protease 14 Negatively Regulates Toll-Like Receptor 4-Mediated Signaling and Autophagy Induction by Inhibiting Ubiquitination of TAK1-Binding Protein 2 and Beclin 1. Front Immunol 2017; 8:1827. [PMID: 29326710 PMCID: PMC5736539 DOI: 10.3389/fimmu.2017.01827] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 12/04/2017] [Indexed: 11/24/2022] Open
Abstract
Ubiquitin-specific protease 14 (USP14), one of three proteasome-associated deubiquitinating enzymes, has multifunctional roles in cellular context. Here, we report a novel molecular mechanism and function of USP14 in regulating autophagy induction and nuclear factor-kappa B (NF-κB) activation induced by toll-like receptor (TLR) 4 (TLR4). USP14 interacted with tumor necrosis factor (TNF) receptor-associated factor 6 (TRAF6) and interrupted the association of Beclin 1 with TRAF6, leading to inhibition of TRAF6-mediated ubiquitination of Beclin 1. Reduced expression of USP14 in USP14-knockdown (USP14KD) THP-1 cells enhanced autophagy induction upon TLR4 stimulation as shown by the increased conversion of cytosolic LC3-I to membrane-bound LC3-II. Moreover, USP14KD human breast carcinoma MDA-MB-231 cells and USP14KD human hepatic adenocarcinoma SK-HEP-1 cells showed increased cell migration and invasion, indicating that USP14 is negatively implicated in the cancer progression by the inhibition of autophagy induction. Furthermore, we found that USP14 interacted with TAK1-binding protein (TAB) 2 protein and induced deubiquitination of TAB 2, a key factor in the activation of NF-κB. Functionally, overexpression of USP14 suppressed TLR4-induced activation of NF-κB. In contrast, USP14KD THP-1 cells showed enhanced activation of NF-κB, NF-κB-dependent gene expression, and production of pro-inflammatory cytokines such as IL-6, IL-1β, and tumor necrosis factor-α. Taken together, our data demonstrate that USP14 can negatively regulate autophagy induction by inhibiting Beclin 1 ubiquitination, interrupting association between TRAF6 and Beclin 1, and affecting TLR4-induced activation of NF-κB through deubiquitination of TAB 2 protein.
Collapse
Affiliation(s)
- Yoon Min
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Sena Lee
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Mi-Jeong Kim
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Eunyoung Chun
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, United States.,The Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Ki-Young Lee
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, South Korea.,Samsung Medical Center, Seoul, South Korea.,Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Samsung Medical Center, Sungkyunkwan University, Seoul, South Korea
| |
Collapse
|
56
|
de Poot SAH, Tian G, Finley D. Meddling with Fate: The Proteasomal Deubiquitinating Enzymes. J Mol Biol 2017; 429:3525-3545. [PMID: 28988953 DOI: 10.1016/j.jmb.2017.09.015] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 09/20/2017] [Accepted: 09/26/2017] [Indexed: 01/06/2023]
Abstract
Three deubiquitinating enzymes-Rpn11, Usp14, and Uch37-are associated with the proteasome regulatory particle. These enzymes allow proteasomes to remove ubiquitin from substrates before they are translocated into the core particle to be degraded. Although the translocation channel is too narrow for folded proteins, the force of translocation unfolds them mechanically. As translocation proceeds, ubiquitin chains bound to substrate are drawn to the channel's entry port, where they can impede further translocation. Rpn11, situated over the port, can remove these chains without compromising degradation because substrates must be irreversibly committed to degradation before Rpn11 acts. This coupling between deubiquitination and substrate degradation is ensured by the Ins-1 loop of Rpn11, which controls ubiquitin access to its catalytic site. In contrast to Rpn11, Usp14 and Uch37 can rescue substrates from degradation by promoting substrate dissociation from the proteasome prior to the commitment step. Uch37 is unique in being a component of both the proteasome and a second multisubunit assembly, the INO80 complex. However, only recruitment into the proteasome activates Uch37. Recruitment to the proteasome likewise activates Usp14. However, the influence of Usp14 on the proteasome depends on the substrate, due to its marked preference for proteins that carry multiple ubiquitin chains. Usp14 exerts complex control over the proteasome, suppressing proteasome activity even when inactive in deubiquitination. A major challenge for the field will be to elucidate the specificities of Rpn11, Usp14, and Uch37 in greater depth, employing not only model in vitro substrates but also their endogenous targets.
Collapse
Affiliation(s)
- Stefanie A H de Poot
- Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA
| | - Geng Tian
- Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA
| | - Daniel Finley
- Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
57
|
Boselli M, Lee BH, Robert J, Prado MA, Min SW, Cheng C, Silva MC, Seong C, Elsasser S, Hatle KM, Gahman TC, Gygi SP, Haggarty SJ, Gan L, King RW, Finley D. An inhibitor of the proteasomal deubiquitinating enzyme USP14 induces tau elimination in cultured neurons. J Biol Chem 2017; 292:19209-19225. [PMID: 28972160 DOI: 10.1074/jbc.m117.815126] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Indexed: 11/06/2022] Open
Abstract
The ubiquitin-proteasome system (UPS) is responsible for most selective protein degradation in eukaryotes and regulates numerous cellular processes, including cell cycle control and protein quality control. A component of this system, the deubiquitinating enzyme USP14, associates with the proteasome where it can rescue substrates from degradation by removal of the ubiquitin tag. We previously found that a small-molecule inhibitor of USP14, known as IU1, can increase the rate of degradation of a subset of proteasome substrates. We report here the synthesis and characterization of 87 variants of IU1, which resulted in the identification of a 10-fold more potent USP14 inhibitor that retains specificity for USP14. The capacity of this compound, IU1-47, to enhance protein degradation in cells was tested using as a reporter the microtubule-associated protein tau, which has been implicated in many neurodegenerative diseases. Using primary neuronal cultures, IU1-47 was found to accelerate the rate of degradation of wild-type tau, the pathological tau mutants P301L and P301S, and the A152T tau variant. We also report that a specific residue in tau, lysine 174, is critical for the IU1-47-mediated tau degradation by the proteasome. Finally, we show that IU1-47 stimulates autophagic flux in primary neurons. In summary, these findings provide a powerful research tool for investigating the complex biology of USP14.
Collapse
Affiliation(s)
- Monica Boselli
- From the Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115
| | - Byung-Hoon Lee
- From the Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115.,the Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, 42988 Daegu, Korea
| | - Jessica Robert
- From the Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115
| | - Miguel A Prado
- From the Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115
| | - Sang-Won Min
- the Department of Neurology, Gladstone Institute of Neurological Diseases, University of California, San Francisco, California 94158
| | - Chialin Cheng
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114
| | - M Catarina Silva
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114
| | - Changhyun Seong
- From the Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115.,Regeneron Pharmaceuticals, Tarrytown, New York 10591, and
| | - Suzanne Elsasser
- From the Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115
| | - Ketki M Hatle
- From the Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115
| | - Timothy C Gahman
- Small Molecule Discovery Program, Ludwig Institute for Cancer Research, La Jolla, California 92093
| | - Steven P Gygi
- From the Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115
| | - Stephen J Haggarty
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114
| | - Li Gan
- the Department of Neurology, Gladstone Institute of Neurological Diseases, University of California, San Francisco, California 94158
| | - Randall W King
- From the Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115,
| | - Daniel Finley
- From the Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115,
| |
Collapse
|
58
|
Wei J, Dong S, Bowser RK, Khoo A, Zhang L, Jacko AM, Zhao Y, Zhao J. Regulation of the ubiquitylation and deubiquitylation of CREB-binding protein modulates histone acetylation and lung inflammation. Sci Signal 2017; 10:10/483/eaak9660. [PMID: 28611184 DOI: 10.1126/scisignal.aak9660] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Cyclic adenosine monophosphate (cAMP) response element-binding protein (CREB)-binding protein (CBP) is a histone acetyltransferase that plays a pivotal role in the control of histone modification and the expression of cytokine-encoding genes in inflammatory diseases, including sepsis and lung injury. We found that the E3 ubiquitin ligase subunit FBXL19 targeted CBP for site-specific ubiquitylation and proteasomal degradation. The ubiquitylation-dependent degradation of CBP reduced the extent of lipopolysaccharide (LPS)-dependent histone acetylation and cytokine release in mouse lung epithelial cells and in a mouse model of sepsis. Furthermore, we demonstrated that the deubiquitylating enzyme USP14 (ubiquitin-specific peptidase 14) stabilized CBP by reducing its ubiquitylation. LPS increased the stability of CBP by reducing the association between CBP and FBXL19 and by activating USP14. Inhibition of USP14 reduced CBP protein abundance and attenuated LPS-stimulated histone acetylation and cytokine release. Together, our findings delineate the molecular mechanisms through which CBP stability is regulated by FBXL19 and USP14, which results in the modulation of chromatin remodeling and the expression of cytokine-encoding genes.
Collapse
Affiliation(s)
- Jianxin Wei
- Department of Medicine, Acute Lung Injury Center of Excellence, Vascular Medical Institute, and Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Su Dong
- Department of Medicine, Acute Lung Injury Center of Excellence, Vascular Medical Institute, and Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.,Department of Anesthesia, First Hospital of Jilin University, Changchun, China
| | - Rachel K Bowser
- Department of Medicine, Acute Lung Injury Center of Excellence, Vascular Medical Institute, and Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Andrew Khoo
- Department of Medicine, Acute Lung Injury Center of Excellence, Vascular Medical Institute, and Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Lina Zhang
- Department of Medicine, Acute Lung Injury Center of Excellence, Vascular Medical Institute, and Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.,Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Anastasia M Jacko
- Department of Medicine, Acute Lung Injury Center of Excellence, Vascular Medical Institute, and Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Yutong Zhao
- Department of Medicine, Acute Lung Injury Center of Excellence, Vascular Medical Institute, and Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| | - Jing Zhao
- Department of Medicine, Acute Lung Injury Center of Excellence, Vascular Medical Institute, and Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| |
Collapse
|
59
|
Xu D, Shan B, Sun H, Xiao J, Zhu K, Xie X, Li X, Liang W, Lu X, Qian L, Yuan J. USP14 regulates autophagy by suppressing K63 ubiquitination of Beclin 1. Genes Dev 2017; 30:1718-30. [PMID: 27542828 PMCID: PMC5002977 DOI: 10.1101/gad.285122.116] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 07/25/2016] [Indexed: 11/24/2022]
Abstract
Xu et al. show that USP14 regulates autophagy by controlling K63 ubiquitination of Beclin 1. Activation of USP14 by Akt-mediated phosphorylation provides a mechanism for Akt to negatively regulate autophagy by promoting K63 deubiquitination. The ubiquitin–proteasome system (UPS) and autophagy are two major intracellular degradative mechanisms that mediate the turnover of complementary repertoires of intracellular proteomes. Simultaneously activating both UPS and autophagy might provide a powerful strategy for the clearance of misfolded proteins. However, it is not clear whether UPS and autophagy can be controlled by a common regulatory mechanism. K48 deubiquitination by USP14 is known to inhibit UPS. Here we show that USP14 regulates autophagy by negatively controlling K63 ubiquitination of Beclin 1. Furthermore, we show that activation of USP14 by Akt-mediated phosphorylation provides a mechanism for Akt to negatively regulate autophagy by promoting K63 deubiquitination. Our study suggests that Akt-regulated USP14 activity modulates both proteasomal degradation and autophagy through controlling K48 and K63 ubiquitination, respectively. Therefore, regulation of USP14 provides a mechanism for Akt to control both proteasomal and autophagic degradation. We propose that inhibition of USP14 may provide a strategy to promote both UPS and autophagy for developing novel therapeutics targeting neurodegenerative diseases.
Collapse
Affiliation(s)
- Daichao Xu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Bing Shan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Huawang Sun
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Juan Xiao
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Kezhou Zhu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Xingxing Xie
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Xingyan Li
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Wei Liang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Xiaojuan Lu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Lihui Qian
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Junying Yuan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China; Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
60
|
Qiu GZ, Sun W, Jin MZ, Lin J, Lu PG, Jin WL. The bad seed gardener: Deubiquitinases in the cancer stem-cell signaling network and therapeutic resistance. Pharmacol Ther 2017; 172:127-138. [DOI: 10.1016/j.pharmthera.2016.12.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
61
|
Liu N, Kong T, Chen X, Hu H, Gu H, Liu S, Chen X, Yang Q, Li A, Xiong X, Zhang Z. Ubiquitin-specific protease 14 regulates LPS-induced inflammation by increasing ERK1/2 phosphorylation and NF-κB activation. Mol Cell Biochem 2017; 431:87-96. [PMID: 28364380 DOI: 10.1007/s11010-017-2978-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 02/24/2017] [Indexed: 12/19/2022]
Abstract
Persistent activation of nuclear factor B (NF-κB) is very important in the modulation of macrophages cellular response to microbial infections. The deubiquitinase USP14, which is critical for ubiquitin-mediated proteasomal degradation of proteins, is known to be involved in cancer, neurological diseases, and aging. However, the mechanism by which USP14 regulates inflammation remains unclear. Here, we demonstrated that decreasing the deubiquitinase activity of USP14 resulted in reduced lipopolysaccharides (LPS)-mediated tumor necrosis factor-α (TNF-α) and interleukin (IL)-6 release in THP-1 and RAW264.7 cells. Meanwhile, USP14 knockdown by siRNA showed the same effects, with no cytotoxicity in THP-1 cells. Moreover, inhibiting the deubiquitinase activity of USP14 or USP14 knockdown resulted in decreased ERK1/2 and IκBα phosphorylation, increased amounts of the NF-κB inhibitor IκBα, and reduced NF-κB p65 transport from the cytoplasm into nucleus. These findings suggested that USP14 induces NF-κB activity and ERK1/2 phosphorylation triggered by microbial infection.
Collapse
Affiliation(s)
- Ningning Liu
- Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Tianyu Kong
- Department of Critical Care Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Xiaohua Chen
- Department of Critical Care Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Huan Hu
- Department of Critical Care Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Hongjiao Gu
- Department of Critical Care Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Shiming Liu
- Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Xiaohui Chen
- Department of Emergency, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Qilin Yang
- Department of Critical Care Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Aiqun Li
- Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China
| | - Xuming Xiong
- Department of Critical Care Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China.
| | - Zhenhui Zhang
- Department of Critical Care Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, Guangdong, People's Republic of China.
| |
Collapse
|
62
|
Ubiquitin C-terminal hydrolase37 regulates Tcf7 DNA binding for the activation of Wnt signalling. Sci Rep 2017; 7:42590. [PMID: 28198400 PMCID: PMC5309806 DOI: 10.1038/srep42590] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 01/11/2017] [Indexed: 12/18/2022] Open
Abstract
The Tcf/Lef family of transcription factors mediates the Wnt/β-catenin pathway that is involved in a wide range of biological processes, including vertebrate embryogenesis and diverse pathogenesis. Post-translational modifications, including phosphorylation, sumoylation and acetylation, are known to be important for the regulation of Tcf/Lef proteins. However, the importance of ubiquitination and ubiquitin-mediated regulatory mechanisms for Tcf/Lef activity are still unclear. Here, we newly show that ubiquitin C-terminal hydrolase 37 (Uch37), a deubiquitinase, interacts with Tcf7 (formerly named Tcf1) to activate Wnt signalling. Biochemical analyses demonstrated that deubiquitinating activity of Uch37 is not involved in Tcf7 protein stability but is required for the association of Tcf7 to target gene promoter in both Xenopus embryo and human liver cancer cells. In vivo analyses further revealed that Uch37 functions as a positive regulator of the Wnt/β-catenin pathway downstream of β-catenin stabilization that is required for the expression of ventrolateral mesoderm genes during Xenopus gastrulation. Our study provides a new mechanism for chromatin occupancy of Tcf7 and uncovers the physiological significance of Uch37 during early vertebrate development by regulating the Wnt/β-catenin pathway.
Collapse
|
63
|
Chen X, Wu J, Chen Y, Ye D, Lei H, Xu H, Yang L, Wu Y, Gu W. Ubiquitin-specific protease 14 regulates cell proliferation and apoptosis in oral squamous cell carcinoma. Int J Biochem Cell Biol 2016; 79:350-359. [PMID: 27592452 DOI: 10.1016/j.biocel.2016.08.038] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 08/10/2016] [Accepted: 08/29/2016] [Indexed: 01/26/2023]
Abstract
Ubiquitin-specific protease 14, a deubiquitinating enzyme, has been implicated in the tumorigenesis and progression of several cancers, but its role in oral squamous cell carcinoma remains to be elucidated. The aim of this study was to explore the expression pattern and roles of Ubiquitin-specific protease 14 in the occurrence and development of oral squamous cell carcinoma. Interestingly, Ubiquitin-specific protease 14 was overexpressed in oral cancer tissues and cell lines at both mRNA and protein levels. b-AP15, a specific inhibitor of Ubiquitin-specific protease 14, significantly inhibited the growth of cancer cells and increased cell apoptosis in a dose-dependent manner. Moreover, knockdown of Ubiquitin-specific protease 14 by shRNA significantly inhibited the proliferation and migration of cancer cells in vitro. Finally, using a xenograft mouse model of oral squamous cell carcinoma, knockdown of Ubiquitin-specific protease 14 markedly inhibited tumor growth and triggered the cancer cell apoptosis in vivo, supporting previous results. In conclusion, for the first time we have demonstrated the expression pattern of Ubiquitin-specific protease 14 in oral squamous cell carcinoma and verified a relationship with tumor growth and metastasis. These results may highlight new therapeutic strategies for tumor treatment, application of Ubiquitin-specific protease 14 selective inhibitor, such as b-AP15, or knockdown by shRNA. Collectively, Ubiquitin-specific protease 14 could be a potential therapeutic target for oral squamous cell carcinoma patients.
Collapse
Affiliation(s)
- Xiangyun Chen
- Department of Clinical Laboratory, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Jingjing Wu
- Department of Clinical Laboratory, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yitian Chen
- Department of Clinical Laboratory, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Dongxia Ye
- Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Hu Lei
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hanzhang Xu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Li Yang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yingli Wu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Wenli Gu
- Department of Clinical Laboratory, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| |
Collapse
|
64
|
Kim JH, Kim E, Choi WH, Lee J, Lee JH, Lee H, Kim DE, Suh YH, Lee MJ. Inhibitory RNA Aptamers of Tau Oligomerization and Their Neuroprotective Roles against Proteotoxic Stress. Mol Pharm 2016; 13:2039-48. [PMID: 27120117 DOI: 10.1021/acs.molpharmaceut.6b00165] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tau is a cytosolic protein that functions in the assembly and stabilization of axonal microtubule networks. Its oligomerization may be the rate-limiting step of insoluble aggregate formation, which is a neuropathological hallmark of Alzheimer's disease (AD) and a number of other tauopathies. Recent evidence indicates that soluble tau oligomers are the toxic species for tau-mediated pathology during AD progression. Herein, we describe novel RNA aptamers that target human tau and were identified through an in vitro selection process. These aptamers significantly inhibited the oligomerization propensity of tau both in vitro and in cultured cell models of tauopathy without affecting the half-life of tau. Tauopathy model cells treated with the aptamers were less sensitized to proteotoxic stress induced by tau overexpression. Moreover, the tau aptamers significantly alleviated synthetic tau oligomer-mediated neurotoxicity and dendritic spine loss in primary hippocampal neurons. Thus, our study demonstrates that delaying tau assembly with RNA aptamers is an effective strategy for protecting cells under various neurodegenerative stresses originating from pathogenic tau oligomerization.
Collapse
Affiliation(s)
- Ji Hyeon Kim
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine , Seoul 03080, Korea.,Department of Biomedical Sciences, Seoul National University Graduate School , Seoul 03080, Korea
| | - Eunkyoung Kim
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine , Seoul 03080, Korea
| | - Won Hoon Choi
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine , Seoul 03080, Korea.,Department of Biomedical Sciences, Seoul National University Graduate School , Seoul 03080, Korea
| | - Jeeyoung Lee
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine , Seoul 03080, Korea.,Department of Biomedical Sciences, Seoul National University Graduate School , Seoul 03080, Korea
| | - Jung Hoon Lee
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine , Seoul 03080, Korea
| | - Hyojin Lee
- Department of Biomedical Sciences, Seoul National University Graduate School , Seoul 03080, Korea.,Neuroscience Research Institute, Seoul National University College of Medicine , Seoul 03080, Korea
| | - Dong-Eun Kim
- Department of Bioscience and Biotechnology, Konkuk University , Seoul 05029, Korea
| | - Young Ho Suh
- Department of Biomedical Sciences, Seoul National University Graduate School , Seoul 03080, Korea.,Neuroscience Research Institute, Seoul National University College of Medicine , Seoul 03080, Korea
| | - Min Jae Lee
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine , Seoul 03080, Korea.,Department of Biomedical Sciences, Seoul National University Graduate School , Seoul 03080, Korea.,Biomedical Research Institutue, Seoul National University Hospital , Seoul 03080, Korea
| |
Collapse
|
65
|
McClurg UL, Robson CN. Deubiquitinating enzymes as oncotargets. Oncotarget 2016; 6:9657-68. [PMID: 25962961 PMCID: PMC4496387 DOI: 10.18632/oncotarget.3922] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 04/08/2015] [Indexed: 12/19/2022] Open
Abstract
Carcinogenesis is a complex process tightly regulated at multiple levels by post-translational modifications. Epigenetics plays a major role in cancer development, all stable changes to the gene expression process that are not a result of a direct change in the DNA code are described as epigenetics. Epigenetic processes are regulated by post-translational modifications including ubiquitination which can directly affect either histones or transcription factors or may target their co-factors and interacting partners exerting an indirect effect. Deubiquitination of these target proteins is equally important and alterations in this pathway can also lead to cancer development, progression and metastasis. Only the correct, unaltered balance between ubiquitination and deubiquitination ensures healthy cellular homeostasis. In this review we focus on the role of deubiquitinating (DUB) enzymes in various aspects of epigenetics including the regulation of transcription factors, histone modifications, DNA damage repair pathways and cell cycle regulation. We discuss the impact of those processes on tumourigenesis and potential therapeutic applications of DUBs for cancer treatment.
Collapse
Affiliation(s)
- Urszula L McClurg
- Solid Tumour Target Discovery Laboratory, Newcastle Cancer Centre, Northern Institute for Cancer Research, Medical School, Newcastle University, Newcastle upon Tyne, UK
| | - Craig N Robson
- Solid Tumour Target Discovery Laboratory, Newcastle Cancer Centre, Northern Institute for Cancer Research, Medical School, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
66
|
Abstract
Deubiquitinases are deubiquitinating enzymes (DUBs), which remove ubiquitin from proteins, thus regulating their proteasomal degradation, localization and activity. Here, we discuss DUBs as anti-cancer drug targets.
Collapse
|
67
|
Downregulation of ubiquitin-specific protease 14 (USP14) inhibits breast cancer cell proliferation and metastasis, but promotes apoptosis. J Mol Histol 2015; 47:69-80. [PMID: 26712154 DOI: 10.1007/s10735-015-9650-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 12/20/2015] [Indexed: 02/07/2023]
Abstract
Breast cancer is the second leading cause of cancer-related death in women. Previously, evidence suggested that ubiquitin-specific protease 14 (USP14) was associated with various signal transduction pathways and tumourigenesis. In this study, we demonstrate that USP14 is a novel therapeutic target in breast cancer. A Western blot analysis of USP14 was performed using seven breast cancer tissues and paired adjacent normal tissues and showed that the expression of USP14 was increased in the breast cancer tissues. Immunohistochemistry was conducted on formalin-fixed paraffin-embedded sections of breast cancer samples from 100 cases. Using Pearson's χ(2) test, it was demonstrated that USP14 expression was associated with the histological grade, lymph node status and Ki-67 expression in the tumour. The Kaplan-Meier analysis revealed that increased USP14 expression in patients with breast cancer was associated with a poorer prognosis. In in vitro experiments, the highly migratory MDA-MB-231 cells that were treated with USP14-shRNA (shUSP14) exhibited decreased motility using Transwell migration assays. Next, we employed a starvation and re-feeding assay, and the CCK-8 assay demonstrated that USP14 regulated breast cancer cell proliferation. Furthermore, we used flow cytometry to analyse cellular apoptosis following USP14 knockdown. Taken together, our results suggested that USP14 was involved in the progression of breast cancer.
Collapse
|
68
|
Mund T, Graeb M, Mieszczanek J, Gammons M, Pelham HRB, Bienz M. Disinhibition of the HECT E3 ubiquitin ligase WWP2 by polymerized Dishevelled. Open Biol 2015; 5:150185. [PMID: 26701932 PMCID: PMC4703060 DOI: 10.1098/rsob.150185] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 11/23/2015] [Indexed: 12/13/2022] Open
Abstract
Dishevelled is a pivot in Wnt signal transduction, controlling both β-catenin-dependent transcription to specify proliferative cell fates, and cell polarity and other non-nuclear events in post-mitotic cells. In response to Wnt signals, or when present at high levels, Dishevelled forms signalosomes by dynamic polymerization. Its levels are controlled by ubiquitylation, mediated by various ubiquitin ligases, including NEDD4 family members that bind to a conserved PPxY motif in Dishevelled (mammalian Dvl1-3). Here, we show that Dvl2 binds to the ubiquitin ligase WWP2 and unlocks its ligase activity from autoinhibition. This disinhibition of WWP2 depends on several features of Dvl2 including its PPxY motif and to a lesser extent its DEP domain, but crucially on the ability of Dvl2 to polymerize, indicating that WWP2 is activated in Wnt signalosomes. We show that Notch intracellular domains are substrates for Dvl-activated WWP2 and their transcriptional activity is consequently reduced, providing a molecular mechanism for cross-talk between Wnt and Notch signalling. These regulatory interactions are conserved in Drosophila whose WWP2 orthologue, Suppressor-of-deltex, downregulates Notch signalling upon activation by Dishevelled in developing wing tissue. Attentuation of Notch signalling by Dishevelled signalosomes could be important during the transition of cells from the proliferative to the post-mitotic state.
Collapse
Affiliation(s)
- Thomas Mund
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Michael Graeb
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Juliusz Mieszczanek
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Melissa Gammons
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Hugh R B Pelham
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Mariann Bienz
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK
| |
Collapse
|
69
|
Huang G, Li L, Zhou W. USP14 activation promotes tumor progression in hepatocellular carcinoma. Oncol Rep 2015; 34:2917-24. [PMID: 26397990 DOI: 10.3892/or.2015.4296] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Accepted: 07/06/2015] [Indexed: 11/06/2022] Open
Abstract
To elucidate the molecular mechanisms underlying the pathogenesis and treatment of human primary hepatocellular carcinoma (HCC), it is important to explore novel HCC-associated genes. In the present study, we examined the expression of ubiquitin-specific peptidase 14 (USP14) in patients with HCC using quantitative PCR and immunohistochemical techniques. The expression of USP14 in tumor tissues of patients with HCC was significantly higher than that in adjacent non-cancerous and normal liver tissues. It was also determined whether the expression profile of USP14 was associated with the clinical characteristics of HCC. Increased USP14 expression was associated with some clinicopatho-logical variables, such as advancing tumor stage. A Kaplan-Meier curve analysis demonstrated that patients with HCC having a high USP14 expression had a significantly poorer prognosis after surgery than patients with lower USP14 expression levels. Knockdown of USP14 with the lentiviral vector delivery of shRNA in human hepatocarcinoma SMMC7721 cells suppressed cell proliferation, altered the cell cycle and induced cell apoptosis. Additionally, the Wnt/β-catenin pathway was activated in HCC patients with USP14 overexpression. These findings strongly suggested that USP14 activation plays an oncogenic role in promoting tumor progression in HCC. Thus, our findings suggested that USP14 is involved in the progression of HCC and may be a useful therapeutic target in HCC. These findings likely reflect the key role that USP14 plays in the pathogenesis of HCC. Therefore, the identification of USP14 and USP14-driven genes may promote the investigation of its functional role to develop more effective therapies for HCC, especially advanced HCC.
Collapse
Affiliation(s)
- Gang Huang
- Department of Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, P.R. China
| | - Limei Li
- Clinical and Translational Research Center Shanghai East Hospital, Key Laboratory of Arrhythmias of Ministry of Education, Shanghai, P.R. China
| | - Weiping Zhou
- Department of Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, P.R. China
| |
Collapse
|
70
|
D'Arcy P, Linder S. Molecular pathways: translational potential of deubiquitinases as drug targets. Clin Cancer Res 2015; 20:3908-14. [PMID: 25085788 DOI: 10.1158/1078-0432.ccr-14-0568] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The ubiquitin proteasome system (UPS) is the main system for controlled protein degradation and a key regulator of fundamental cellular processes. The dependency of cancer cells on a functioning UPS coupled with the clinical success of bortezomib for the treatment of multiple myeloma have made the UPS an obvious target for drug development. Deubiquitinases (DUB) are components of the UPS that encompass a diverse family of ubiquitin isopeptidases that catalyze the removal of ubiquitin moieties from target proteins or from polyubiquitin chains, resulting in altered signaling or changes in protein stability. Increasing evidence has implicated deregulation of DUB activity in the initiation and progression of cancer. The altered pattern of DUB expression observed in many tumors can potentially serve as a clinical marker for predicting disease outcome and therapy response. The finding of DUB overexpression in tumor cells suggests that they may serve as novel targets for the development of anticancer therapies. Several specific and broad-spectrum DUB inhibitors are shown to have antitumor activity in preclinical in vivo models with low levels of systemic toxicity. Future studies will hopefully establish the clinical potential for DUB inhibitors as a strategy to treat cancer.
Collapse
Affiliation(s)
- Pádraig D'Arcy
- Department of Oncology-Pathology, Karolinska Institute, Stockholm and
| | - Stig Linder
- Department of Oncology-Pathology, Karolinska Institute, Stockholm and Department of Medical Sciences, Division of Clinical Pharmacology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
71
|
Pham GH, Strieter ER. Peeling away the layers of ubiquitin signaling complexities with synthetic ubiquitin-protein conjugates. Curr Opin Chem Biol 2015; 28:57-65. [PMID: 26093241 DOI: 10.1016/j.cbpa.2015.06.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 05/29/2015] [Accepted: 06/02/2015] [Indexed: 10/23/2022]
Abstract
Covalent attachment of ubiquitin, a process termed ubiquitination, affects the location, function, and stability of modified proteins. Significant advances have been made in building synthetic ubiquitin-protein conjugates that can be used to investigate how ubiquitin regulates diverse biological processes. Herein we describe recent advances and discuss how chemical methods have been implemented to address the molecular underpinnings of ubiquitin-dependent cellular signaling.
Collapse
Affiliation(s)
- Grace H Pham
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, United States
| | - Eric R Strieter
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, United States.
| |
Collapse
|
72
|
Lee JH, Shin SK, Jiang Y, Choi WH, Hong C, Kim DE, Lee MJ. Facilitated Tau Degradation by USP14 Aptamers via Enhanced Proteasome Activity. Sci Rep 2015; 5:10757. [PMID: 26041011 PMCID: PMC4455164 DOI: 10.1038/srep10757] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 04/28/2015] [Indexed: 12/26/2022] Open
Abstract
The ubiquitin-proteasome system (UPS) is the primary mechanism by which intracellular proteins, transcription factors, and many proteotoxic proteins with aggregation-prone structures are degraded. The UPS is reportedly downregulated in various neurodegenerative disorders, with increased proteasome activity shown to be beneficial in many related disease models. Proteasomes function under tonic inhibitory conditions, possibly via the ubiquitin chain-trimming function of USP14, a proteasome-associated deubiquitinating enzyme (DUB). We identified three specific RNA aptamers of USP14 (USP14-1, USP14-2, and USP14-3) that inhibited its deubiquitinating activity. The nucleotide sequences of these non-cytotoxic USP14 aptamers contained conserved GGAGG motifs, with G-rich regions upstream, and similar secondary structures. They efficiently elevated proteasomal activity, as determined by the increased degradation of small fluorogenic peptide substrates and physiological polyubiquitinated Sic1 proteins. Additionally, proteasomal degradation of tau proteins was facilitated in the presence of the UPS14 aptamers in vitro. Our results indicate that these novel inhibitory UPS14 aptamers can be used to enhance proteasome activity, and to facilitate the degradation of proteotoxic proteins, thereby protecting cells from various neurodegenerative stressors.
Collapse
Affiliation(s)
- Jung Hoon Lee
- Department of Biochemistry and Molecular Biology, Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea
| | - Seung Kyun Shin
- Department of Applied Chemistry, College of Applied Sciences, Kyung Hee University, Yongin 446-701, Republic of Korea
| | - Yanxialei Jiang
- Department of Applied Chemistry, College of Applied Sciences, Kyung Hee University, Yongin 446-701, Republic of Korea
| | - Won Hoon Choi
- Department of Biochemistry and Molecular Biology, Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea
| | - Chaesun Hong
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 143-701, Republic of Korea
| | - Dong-Eun Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 143-701, Republic of Korea
| | - Min Jae Lee
- Department of Biochemistry and Molecular Biology, Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 110-799, Republic of Korea
| |
Collapse
|
73
|
Liu N, Huang H, Dou QP, Liu J. Inhibition of 19S proteasome-associated deubiquitinases by metal-containing compounds. Oncoscience 2015; 2:457-66. [PMID: 26097878 PMCID: PMC4468331 DOI: 10.18632/oncoscience.167] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 05/24/2015] [Indexed: 12/31/2022] Open
Abstract
Copper and gold complexes have clinical activity in several diseases including cancer. Recently, we have reported that the anti-cancer activity of copper (II) pyrithione CuPT and gold (I) complex auranofin is associated with targeting the 19S proteasome-associated deubiquitinases (DUBs), UCHL5 and USP14. Here we discuss metal DUB inhibitors in treating cancer and other diseases. (from Editor). Several copper and gold complexes have clinical activity in treating some human diseases including cancer. Recently, we have reported that the anti-cancer activity of copper (II) pyrithione CuPT and gold (I) complex auranofin is tightly associated with their ability to target and inhibit the 19S proteasome-associated deubiquitinases (DUBs), UCHL5 and USP14. In this article we review small molecule inhibitors of DUBs and 19S proteasome-associated DUBs. We then describe and discuss the ubique nature of CuPT and auranofin, which is inhibition of 19S proteasome-associated UCHL5 and USP14. We finally suggest the potential to develop novel, specific metal-based DUB inhibitors for treating cancer and other diseases.
Collapse
Affiliation(s)
- Ningning Liu
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China ; Guangzhou Research Institute of Cardiovascular Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Hongbiao Huang
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China
| | - Q Ping Dou
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China ; The Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, and Departments of Oncology, Pharmacology and Pathology, School of Medicine, Wayne State University, Detroit, Michigan, USA
| | - Jinbao Liu
- State Key Lab of Respiratory Disease, Protein Modification and Degradation Lab, Department of Pathophysiology, Guangzhou Medical University, Guangdong, China
| |
Collapse
|
74
|
Ubiquitination of the Dishevelled DIX domain blocks its head-to-tail polymerization. Nat Commun 2015; 6:6718. [PMID: 25907794 PMCID: PMC4423210 DOI: 10.1038/ncomms7718] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 02/17/2015] [Indexed: 02/06/2023] Open
Abstract
Dishevelled relays Wnt signals from the plasma membrane to different cytoplasmic effectors. Its signalling activity depends on its DIX domain, which undergoes head-to-tail polymerization to assemble signalosomes. The DIX domain is ubiquitinated in vivo at multiple lysines, which can be antagonized by various deubiquitinases (DUBs) including the CYLD tumour suppressor that attenuates Wnt signalling. Here, we generate milligram quantities of pure human Dvl2 DIX domain mono-ubiquitinated at two lysines (K54 and K58) by genetically encoded orthogonal protection with activated ligation (GOPAL), to investigate their effect on DIX polymerization. We show that the ubiquitination of DIX at K54 blocks its polymerization in solution, whereas DIX58-Ub remains oligomerization-competent. DUB profiling identified 28 DUBs that cleave DIX-ubiquitin conjugates, half of which prefer, or are specific for, DIX54-Ub, including Cezanne and CYLD. These DUBs thus have the potential to promote Dvl polymerization and signalosome formation, rather than antagonize it as previously thought for CYLD.
Collapse
|
75
|
Abstract
The post-translational modification of proteins with ubiquitin represents a complex signalling system that co-ordinates essential cellular functions, including proteolysis, DNA repair, receptor signalling and cell communication. DUBs (deubiquitinases), the enzymes that disassemble ubiquitin chains and remove ubiquitin from proteins, are central to this system. Reflecting the complexity and versatility of ubiquitin signalling, DUB activity is controlled in multiple ways. Although several lines of evidence indicate that aberrant DUB function may promote human disease, the underlying molecular mechanisms are often unclear. Notwithstanding, considerable interest in DUBs as potential drug targets has emerged over the past years. The future success of DUB-based therapy development will require connecting the basic science of DUB function and enzymology with drug discovery. In the present review, we discuss new insights into DUB activity regulation and their links to disease, focusing on the role of DUBs as regulators of cell identity and differentiation, and discuss their potential as emerging drug targets.
Collapse
|
76
|
Bunn K, Daniel P, Rösken H, O’Neill A, Cameron-Christie S, Morgan T, Brunner H, Lai A, Kunst H, Markie D, Robertson S. Mutations in DVL1 cause an osteosclerotic form of Robinow syndrome. Am J Hum Genet 2015; 96:623-30. [PMID: 25817014 DOI: 10.1016/j.ajhg.2015.02.010] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 02/13/2015] [Indexed: 12/23/2022] Open
Abstract
Robinow syndrome (RS) is a phenotypically and genetically heterogeneous condition that can be caused by mutations in genes encoding components of the non-canonical Wnt signaling pathway. In contrast, germline mutations that act to increase canonical Wnt signaling lead to distinctive osteosclerotic phenotypes. Here, we identified de novo frameshift mutations in DVL1, a mediator of both canonical and non-canonical Wnt signaling, as the cause of RS-OS, an RS subtype involving osteosclerosis, in three unrelated individuals. The mutations all delete the DVL1 C terminus and replace it, in each instance, with a novel, highly basic sequence. We showed the presence of mutant transcript in fibroblasts from one individual with RS-OS and demonstrated unimpaired protein stability with transfected GFP-tagged constructs bearing a frameshift mutation. In vitro TOPFlash assays, in apparent contradiction to the osteosclerotic phenotype, revealed that the mutant allele was less active than the wild-type allele in the canonical Wnt signaling pathway. However, when the mutant and wild-type alleles were co-expressed, canonical Wnt activity was 2-fold higher than that in the wild-type construct alone. This work establishes that DVL1 mutations cause a specific RS subtype, RS-OS, and that the osteosclerosis associated with this subtype might be the result of an interaction between the wild-type and mutant alleles and thus lead to elevated canonical Wnt signaling.
Collapse
|
77
|
Abstract
Dishevelled (DVL) proteins, three of which have been identified in humans, are highly conserved components of canonical and noncanonical Wnt signaling pathways. These multifunctional proteins, originally discovered in the fruit fly, through their different domains mediate complex signal transduction: DIX (dishevelled, axin) and PDZ (postsynaptic density 95, discs large, zonula occludens-1) domains serve for canonical beta-catenin signaling, while PDZ and DEP (dishevelled, Egl-10, pleckstrin) domains serve for non-canonical signaling. In canonical or beta-catenin signaling, DVL forms large molecular supercomplexes at the plasma membrane consisting of Wnt-Fz-LRP5/6-DVL-AXIN. This promotes the disassembly of the beta-catenin destruction machinery, beta-catenin accumulation, and consequent activation of Wnt signaling. Therefore, DVLs are considered to be key regulators that rescue cytoplasmic beta-catenin from degradation. The potential medical importance of DVLs is in both human degenerative disease and cancer. The overexpression of DVL has been shown to potentiate the activation of Wnt signaling and it is now apparent that up-regulation of DVLs is involved in several types of cancer.
Collapse
Affiliation(s)
| | | | - Nives Pećina-Šlaus
- Nives Pećina-Šlaus, Laboratory of Neuro-oncology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Šalata 3, HR-10000 Zagreb, Croatia,
| |
Collapse
|
78
|
Chang B, Tessneer KL, McManus J, Liu X, Hahn S, Pasula S, Wu H, Song H, Chen Y, Cai X, Dong Y, Brophy ML, Rahman R, Ma JX, Xia L, Chen H. Epsin is required for Dishevelled stability and Wnt signalling activation in colon cancer development. Nat Commun 2015; 6:6380. [PMID: 25871009 PMCID: PMC4397653 DOI: 10.1038/ncomms7380] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 01/26/2015] [Indexed: 02/08/2023] Open
Abstract
Uncontrolled canonical Wnt signalling supports colon epithelial tumour expansion and malignant transformation. Understanding the regulatory mechanisms involved is crucial for elucidating the pathogenesis of and will provide new therapeutic targets for colon cancer. Epsins are ubiquitin-binding adaptor proteins upregulated in several human cancers; however, the involvement of epsins in colon cancer is unknown. Here we show that loss of intestinal epithelial epsins protects against colon cancer by significantly reducing the stability of the crucial Wnt signalling effector, dishevelled (Dvl2), and impairing Wnt signalling. Consistently, epsins and Dvl2 are correspondingly upregulated in colon cancer. Mechanistically, epsin binds Dvl2 via its epsin N-terminal homology domain and ubiquitin-interacting motifs and prohibits Dvl2 polyubiquitination and degradation. Our findings reveal an unconventional role for epsins in stabilizing Dvl2 and potentiating Wnt signalling in colon cancer cells to ensure robust colon cancer progression. The pro-carcinogenic role of Epsins suggests that they are potential therapeutic targets to combat colon cancer.
Collapse
Affiliation(s)
- Baojun Chang
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA
| | - Kandice L Tessneer
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA
| | - John McManus
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA
| | - Xiaolei Liu
- 1] Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA [2] Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Scott Hahn
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA
| | - Satish Pasula
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA
| | - Hao Wu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA
| | - Hoogeun Song
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA
| | - Yiyuan Chen
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA
| | - Xiaofeng Cai
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA
| | - Yunzhou Dong
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA
| | - Megan L Brophy
- 1] Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA [2] Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Ruby Rahman
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA
| | - Jian-Xing Ma
- Department of Endocrinology and Diabetes, Harold Hamm Oklahoma Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Lijun Xia
- 1] Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA [2] Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| | - Hong Chen
- 1] Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA [2] Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, USA
| |
Collapse
|
79
|
Pharmacological folding chaperones act as allosteric ligands of Frizzled4. Nat Chem Biol 2015; 11:280-6. [DOI: 10.1038/nchembio.1770] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 02/10/2015] [Indexed: 02/01/2023]
|
80
|
Vaden JH, Bhattacharyya BJ, Chen PC, Watson JA, Marshall AG, Phillips SE, Wilson JA, King GD, Miller RJ, Wilson SM. Ubiquitin-specific protease 14 regulates c-Jun N-terminal kinase signaling at the neuromuscular junction. Mol Neurodegener 2015; 10:3. [PMID: 25575639 PMCID: PMC4417291 DOI: 10.1186/1750-1326-10-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 12/17/2014] [Indexed: 01/13/2023] Open
Abstract
Background Ubiquitin-specific protease 14 (USP14) is one of three proteasome-associated deubiquitinating enzymes that remove ubiquitin from proteasomal substrates prior to their degradation. In vitro evidence suggests that inhibiting USP14’s catalytic activity alters the turnover of ubiquitinated proteins by the proteasome, although whether protein degradation is accelerated or delayed seems to be cell-type and substrate specific. For example, combined inhibition of USP14 and the proteasomal deubiquitinating enzyme UCH37 halts protein degradation and promotes apoptosis in multiple myeloma cells, whereas USP14 inhibition alone accelerates the degradation of aggregate-prone proteins in immortalized cell lines. These findings have prompted interest in USP14 as a therapeutic target both inside and outside of the nervous system. However, loss of USP14 in the spontaneously occurring ataxia mouse mutant leads to a dramatic neuromuscular phenotype and early perinatal lethality, suggesting that USP14 inhibition may have adverse consequences in the nervous system. We therefore expressed a catalytically inactive USP14 mutant in the mouse nervous system to determine whether USP14’s catalytic activity is required for neuromuscular junction (NMJ) structure and function. Results Mice expressing catalytically inactive USP14 in the nervous system exhibited motor deficits, altered NMJ structure, and synaptic transmission deficits that were similar to what is observed in the USP14-deficient ataxia mice. Acute pharmacological inhibition of USP14 in wild type mice also reduced NMJ synaptic transmission. However, there was no evidence of altered proteasome activity when USP14 was inhibited either genetically or pharmacologically. Instead, these manipulations increased the levels of non-proteasome targeting ubiquitin conjugates. Specifically, we observed enhanced proteasome-independent ubiquitination of mixed lineage kinase 3 (MLK3). Consistent with the direct activation of MLK3 by ubiquitination, we also observed increased activation of its downstrea targets MAP kinase kinase 4 (MKK4) and c-Jun N-terminal kinase (JNK). In vivo inhibition of JNK improved motor function and synapse structure in the USP14 catalytic mutant mice. Conclusions USP14’s catalytic activity is required for nervous system structure and function and has an ongoing role in NMJ synaptic transmission. By regulating the ubiquitination status of protein kinases, USP14 can coordinate the activity of intracellular signaling pathways that control the development and activity of the NMJ. Electronic supplementary material The online version of this article (doi:10.1186/1750-1326-10-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jada H Vaden
- Department of Neurobiology, University of Alabama at Birmingham, Civitan International Research Center, Evelyn F. McKnight Brain Institute, 1825 University Blvd, Birmingham, AL, 35294, USA.
| | - Bula J Bhattacharyya
- Department of Molecular Pharmacology and Biological Chemistry, Northwestern University, 303 East Chicago Ave, Chicago, IL, 60611, USA.
| | - Ping-Chung Chen
- Department of Structural Biology, St. Jude Children's Research Hospital, Danny Thomas Place, Memphis, TN, 38105-3678, USA.
| | - Jennifer A Watson
- Department of Neurobiology, University of Alabama at Birmingham, Civitan International Research Center, Evelyn F. McKnight Brain Institute, 1825 University Blvd, Birmingham, AL, 35294, USA.
| | - Andrea G Marshall
- Department of Physiology, Neurobiology and Behavior, University of California, Davis, California, CA, USA.
| | - Scott E Phillips
- Department of Neurobiology, University of Alabama at Birmingham, Civitan International Research Center, Evelyn F. McKnight Brain Institute, 1825 University Blvd, Birmingham, AL, 35294, USA.
| | - Julie A Wilson
- Department of Neurobiology, University of Alabama at Birmingham, Civitan International Research Center, Evelyn F. McKnight Brain Institute, 1825 University Blvd, Birmingham, AL, 35294, USA.
| | - Gwendalyn D King
- Department of Neurobiology, University of Alabama at Birmingham, Civitan International Research Center, Evelyn F. McKnight Brain Institute, 1825 University Blvd, Birmingham, AL, 35294, USA.
| | - Richard J Miller
- Department of Molecular Pharmacology and Biological Chemistry, Northwestern University, 303 East Chicago Ave, Chicago, IL, 60611, USA.
| | - Scott M Wilson
- Department of Neurobiology, University of Alabama at Birmingham, Civitan International Research Center, Evelyn F. McKnight Brain Institute, 1825 University Blvd, Birmingham, AL, 35294, USA.
| |
Collapse
|
81
|
Mansour W, Nakasone MA, von Delbrück M, Yu Z, Krutauz D, Reis N, Kleifeld O, Sommer T, Fushman D, Glickman MH. Disassembly of Lys11 and mixed linkage polyubiquitin conjugates provides insights into function of proteasomal deubiquitinases Rpn11 and Ubp6. J Biol Chem 2014; 290:4688-4704. [PMID: 25389291 DOI: 10.1074/jbc.m114.568295] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Protein homeostasis is largely dependent on proteolysis by the ubiquitin-proteasome system. Diverse polyubiquitin modifications are reported to target cellular proteins to the proteasome. At the proteasome, deubiquitination is an essential preprocessing event that contributes to degradation efficiency. We characterized the specificities of two proteasome-associated deubiquitinases (DUBs), Rpn11 and Ubp6, and explored their impact on overall proteasome DUB activity. This was accomplished by constructing a panel of well defined ubiquitin (Ub) conjugates, including homogeneous linkages of varying lengths as well as a heterogeneously modified target. Rpn11 and Ubp6 processed Lys(11) and Lys(63) linkages with comparable efficiencies that increased with chain length. In contrast, processing of Lys(48) linkages by proteasome was inversely correlated to chain length. Fluorescently labeled tetra-Ub chains revealed endo-chain preference for Ubp6 acting on Lys(48) and random action for Rpn11. Proteasomes were more efficient at deconjugating identical substrates than their constituent DUBs by roughly 2 orders of magnitude. Incorporation into proteasomes significantly enhanced enzymatic efficiency of Rpn11, due in part to alleviation of the autoinhibitory role of its C terminus. The broad specificity of Rpn11 could explain how proteasomes were more effective at disassembling a heterogeneously modified conjugate compared with homogeneous Lys(48)-linked chains. The reduced ability to disassemble homogeneous Lys(48)-linked chains longer than 4 Ub units may prolong residency time on the proteasome.
Collapse
Affiliation(s)
- Wissam Mansour
- From the Department of Biology, Technion-Israel Institute of Technology, 32000 Haifa, Israel
| | - Mark A Nakasone
- From the Department of Biology, Technion-Israel Institute of Technology, 32000 Haifa, Israel,; the Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Organization, University of Maryland, College Park, Maryland 20742
| | - Maximilian von Delbrück
- the Max-Delbrück-Zentrum für Molekulare Medizin, Robert-Rössle-Strasse 10, 13125 Berlin, Germany, and
| | - Zanlin Yu
- From the Department of Biology, Technion-Israel Institute of Technology, 32000 Haifa, Israel
| | - Daria Krutauz
- From the Department of Biology, Technion-Israel Institute of Technology, 32000 Haifa, Israel
| | - Noa Reis
- From the Department of Biology, Technion-Israel Institute of Technology, 32000 Haifa, Israel
| | - Oded Kleifeld
- the Department of Biochemistry and Molecular Biology, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne 3800, Australia
| | - Thomas Sommer
- the Max-Delbrück-Zentrum für Molekulare Medizin, Robert-Rössle-Strasse 10, 13125 Berlin, Germany, and
| | - David Fushman
- the Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Organization, University of Maryland, College Park, Maryland 20742
| | - Michael H Glickman
- From the Department of Biology, Technion-Israel Institute of Technology, 32000 Haifa, Israel,.
| |
Collapse
|
82
|
The ubiquitin ligase RNF220 enhances canonical Wnt signaling through USP7-mediated deubiquitination of β-catenin. Mol Cell Biol 2014; 34:4355-66. [PMID: 25266658 DOI: 10.1128/mcb.00731-14] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Wnt/β-catenin signaling plays critical roles in embryonic development and disease. Here, we identify RNF220, a RING domain E3 ubiquitin ligase, as a new regulator of β-catenin. RNF220 physically interacts with β-catenin, but instead of promoting its ubiquitination and proteasomal degradation, it stabilizes β-catenin and promotes canonical Wnt signaling. Our analysis showed that RNF220 interacts with USP7, a ubiquitin-specific peptidase, which is required for RNF220 to stabilize β-catenin. The RNF220/USP7 complex deubiquitinates β-catenin and enhances canonical Wnt signaling. Interestingly, the stability of RNF220 itself is negatively regulated by Gsk3β, which is a key component of the β-catenin destruction complex and is inhibited upon Wnt stimulation. Accordingly, the RNF220/USP7 complex works as a positive feedback regulator of β-catenin signaling. In colon cancer cells with stimulated Wnt signaling, knockdown of RNF220 or USP7 impairs Wnt signaling and expression of Wnt target genes, suggesting a potentially novel role of RNF220 in Wnt-related tumorigenesis.
Collapse
|
83
|
Ristic G, Tsou WL, Todi SV. An optimal ubiquitin-proteasome pathway in the nervous system: the role of deubiquitinating enzymes. Front Mol Neurosci 2014; 7:72. [PMID: 25191222 PMCID: PMC4137239 DOI: 10.3389/fnmol.2014.00072] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 07/10/2014] [Indexed: 01/18/2023] Open
Abstract
The Ubiquitin-Proteasome Pathway (UPP), which is critical for normal function in the nervous system and is implicated in various neurological diseases, requires the small modifier protein ubiquitin to accomplish its duty of selectively degrading short-lived, abnormal or misfolded proteins. Over the past decade, a large class of proteases collectively known as deubiquitinating enzymes (DUBs) has increasingly gained attention in all manners related to ubiquitin. By cleaving ubiquitin from another protein, DUBs ensure that the UPP functions properly. DUBs accomplish this task by processing newly translated ubiquitin so that it can be used for conjugation to substrate proteins, by regulating the "where, when, and why" of UPP substrate ubiquitination and subsequent degradation, and by recycling ubiquitin for re-use by the UPP. Because of the reliance of the UPP on DUB activities, it is not surprising that these proteases play important roles in the normal activities of the nervous system and in neurodegenerative diseases. In this review, we summarize recent advances in understanding the functions of DUBs in the nervous system. We focus on their role in the UPP, and make the argument that understanding the UPP from the perspective of DUBs can yield new insight into diseases that result from anomalous intra-cellular processes or inter-cellular networks. Lastly, we discuss the relevance of DUBs as therapeutic options for disorders of the nervous system.
Collapse
Affiliation(s)
- Gorica Ristic
- Department of Pharmacology, Wayne State University School of Medicine Detroit, MI, USA
| | - Wei-Ling Tsou
- Department of Pharmacology, Wayne State University School of Medicine Detroit, MI, USA ; Department of Neurology, Wayne State University School of Medicine Detroit, MI, USA
| | - Sokol V Todi
- Department of Pharmacology, Wayne State University School of Medicine Detroit, MI, USA ; Department of Neurology, Wayne State University School of Medicine Detroit, MI, USA
| |
Collapse
|
84
|
Hagemann AIH, Kurz J, Kauffeld S, Chen Q, Reeves PM, Weber S, Schindler S, Davidson G, Kirchhausen T, Scholpp S. In vivo analysis of formation and endocytosis of the Wnt/β-catenin signaling complex in zebrafish embryos. J Cell Sci 2014; 127:3970-82. [PMID: 25074807 PMCID: PMC4163645 DOI: 10.1242/jcs.148767] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
After activation by Wnt/β-Catenin ligands, a multi-protein complex assembles at the clustering membrane-bound receptors and intracellular signal transducers into the so-called Lrp6-signalosome. However, the mechanism of signalosome formation and dissolution is yet not clear. Our imaging studies of live zebrafish embryos show that the signalosome is a highly dynamic structure. It is continuously assembled by Dvl2-mediated recruitment of the transducer complex to the activated receptors and partially disassembled by endocytosis. We find that, after internalization, the ligand-receptor complex and the transducer complex take separate routes. The Wnt–Fz–Lrp6 complex follows a Rab-positive endocytic path. However, when still bound to the transducer complex, Dvl2 forms intracellular aggregates. We show that this endocytic process is not only essential for ligand-receptor internalization but also for signaling. The μ2-subunit of the endocytic Clathrin adaptor Ap2 interacts with Dvl2 to maintain its stability during endocytosis. Blockage of Ap2μ2 function leads to Dvl2 degradation, inhibiton of signalosome formation at the plasma membrane and, consequently, reduction of signaling. We conclude that Ap2μ2-mediated endocytosis is important to maintain Wnt/β-catenin signaling in vertebrates.
Collapse
Affiliation(s)
- Anja I H Hagemann
- Karlsruhe Institute of Technology (KIT), Institute of Toxicology and Genetics (ITG), 76021 Karsruhe, Germany
| | - Jennifer Kurz
- Karlsruhe Institute of Technology (KIT), Institute of Toxicology and Genetics (ITG), 76021 Karsruhe, Germany
| | - Silke Kauffeld
- Karlsruhe Institute of Technology (KIT), Institute of Toxicology and Genetics (ITG), 76021 Karsruhe, Germany
| | - Qing Chen
- Karlsruhe Institute of Technology (KIT), Institute of Toxicology and Genetics (ITG), 76021 Karsruhe, Germany
| | - Patrick M Reeves
- Departments of Cell Biology and Pediatrics, Harvard Medical School and Program in Cellular and Molecular Medicine at Boston Children's Hospital, Boston, 02115 MA, USA
| | - Sabrina Weber
- Karlsruhe Institute of Technology (KIT), Institute of Toxicology and Genetics (ITG), 76021 Karsruhe, Germany
| | - Simone Schindler
- Karlsruhe Institute of Technology (KIT), Institute of Toxicology and Genetics (ITG), 76021 Karsruhe, Germany
| | - Gary Davidson
- Karlsruhe Institute of Technology (KIT), Institute of Toxicology and Genetics (ITG), 76021 Karsruhe, Germany
| | - Tomas Kirchhausen
- Departments of Cell Biology and Pediatrics, Harvard Medical School and Program in Cellular and Molecular Medicine at Boston Children's Hospital, Boston, 02115 MA, USA
| | - Steffen Scholpp
- Karlsruhe Institute of Technology (KIT), Institute of Toxicology and Genetics (ITG), 76021 Karsruhe, Germany
| |
Collapse
|
85
|
de Groot REA, Ganji RS, Bernatik O, Lloyd-Lewis B, Seipel K, Šedová K, Zdráhal Z, Dhople VM, Dale TC, Korswagen HC, Bryja V. Huwe1-mediated ubiquitylation of dishevelled defines a negative feedback loop in the Wnt signaling pathway. Sci Signal 2014; 7:ra26. [PMID: 24643799 DOI: 10.1126/scisignal.2004985] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2024]
Abstract
Wnt signaling plays a central role in development, adult tissue homeostasis, and cancer. Several steps in the canonical Wnt/β-catenin signaling cascade are regulated by ubiquitylation, a protein modification that influences the stability, subcellular localization, or interactions of target proteins. To identify regulators of the Wnt/β-catenin pathway, we performed an RNA interference screen in Caenorhabditis elegans and identified the HECT domain-containing ubiquitin ligase EEL-1 as an inhibitor of Wnt signaling. In human embryonic kidney 293T cells, knockdown of the EEL-1 homolog Huwe1 enhanced the activity of a Wnt reporter in cells stimulated with Wnt3a or in cells that overexpressed casein kinase 1 (CK1) or a constitutively active mutant of the Wnt co-receptor low-density lipoprotein receptor-related protein 6 (LRP6). However, knockdown of Huwe1 had no effect on reporter gene expression in cells expressing constitutively active β-catenin, suggesting that Huwe1 inhibited Wnt signaling upstream of β-catenin and downstream of CK1 and LRP6. Huwe1 bound to and ubiquitylated the cytoplasmic Wnt pathway component Dishevelled (Dvl) in a Wnt3a- and CK1ε-dependent manner. Mass spectrometric analysis showed that Huwe1 promoted K63-linked, but not K48-linked, polyubiquitination of Dvl. Instead of targeting Dvl for degradation, ubiquitylation of the DIX domain of Dvl by Huwe1 inhibited Dvl multimerization, which is necessary for its function. Our findings indicate that Huwe1 is part of an evolutionarily conserved negative feedback loop in the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Reinoud E A de Groot
- 1Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Uppsalalaan 8, 3584CT Utrecht, the Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Gao C, Xiao G, Hu J. Regulation of Wnt/β-catenin signaling by posttranslational modifications. Cell Biosci 2014; 4:13. [PMID: 24594309 PMCID: PMC3977945 DOI: 10.1186/2045-3701-4-13] [Citation(s) in RCA: 170] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 01/07/2014] [Indexed: 02/07/2023] Open
Abstract
The canonical Wnt signaling pathway (or Wnt/β-catenin pathway) plays a pivotal role in embryonic development and adult homeostasis; deregulation of the Wnt pathway contributes to the initiation and progression of human diseases including cancer. Despite its importance in human biology and disease, how regulation of the Wnt/β-catenin pathway is achieved remains largely undefined. Increasing evidence suggests that post-translational modifications (PTMs) of Wnt pathway components are essential for the activation of the Wnt/β-catenin pathway. PTMs create a highly dynamic relay system that responds to Wnt stimulation without requiring de novo protein synthesis and offer a platform for non-Wnt pathway components to be involved in the regulation of Wnt signaling, hence providing alternative opportunities for targeting the Wnt pathway. This review highlights the current status of PTM-mediated regulation of the Wnt/β-catenin pathway with a focus on factors involved in Wnt-mediated stabilization of β-catenin.
Collapse
Affiliation(s)
| | | | - Jing Hu
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| |
Collapse
|