201
|
Lin Z, Tan C, Qiu Q, Kong S, Yang H, Zhao F, Liu Z, Li J, Kong Q, Gao B, Barrett T, Yang GY, Zhang J, Fang D. Ubiquitin-specific protease 22 is a deubiquitinase of CCNB1. Cell Discov 2015; 1. [PMID: 27030811 PMCID: PMC4809424 DOI: 10.1038/celldisc.2015.28] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The elevated level of CCNB1 indicates more aggressive cancer and poor prognosis. However, the factors that cause CCNB1 upregulation remain enigmatic. Herein, we identify USP22 as a CCNB1 interactor and discover that both USP22 and CCNB1 are dramatically elevated with a strong positive correlation in colon cancer tissues. USP22 stabilizes CCNB1 by antagonizing proteasome-mediated degradation in a cell cycle-specific manner. Phosphorylation of USP22 by CDK1 enhances its activity in deubiquitinating CCNB1. The ubiquitin ligase anaphase-promoting complex (APC/C) targets USP22 for degradation by using the substrate adapter CDC20 during cell exit from M phase, presumably allowing CCNB1 degradation. Finally, we discover that USP22 knockdown leads to slower cell growth and reduced tumor size. Our study demonstrates that USP22 is a CCNB1 deubiquitinase, suggesting that targeting USP22 might be an effective approach to treat cancers with elevated CCNB1 expression.
Collapse
Affiliation(s)
- Zhenghong Lin
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Can Tan
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Quan Qiu
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Sinyi Kong
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Heeyoung Yang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Fang Zhao
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Zhaojian Liu
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jinping Li
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Qingfei Kong
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Beixue Gao
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Terry Barrett
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Guang-Yu Yang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jianing Zhang
- School of Life Science and Medicine, Dalian University of Technology, Panjin, China
| | - Deyu Fang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; School of Life Science and Medicine, Dalian University of Technology, Panjin, China
| |
Collapse
|
202
|
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: 5.0] [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
|
203
|
Yang H, Liu S, He WT, Zhao J, Jiang LL, Hu HY. Aggregation of Polyglutamine-expanded Ataxin 7 Protein Specifically Sequesters Ubiquitin-specific Protease 22 and Deteriorates Its Deubiquitinating Function in the Spt-Ada-Gcn5-Acetyltransferase (SAGA) Complex. J Biol Chem 2015. [PMID: 26195632 DOI: 10.1074/jbc.m114.631663] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Human ataxin 7 (Atx7) is a component of the deubiquitination module (DUBm) in the Spt-Ada-Gcn5-acetyltransferase (SAGA) complex for transcriptional regulation, and expansion of its polyglutamine (polyQ) tract leads to spinocerebellar ataxia type 7. However, how polyQ expansion of Atx7 affects DUBm function remains elusive. We investigated the effects of polyQ-expanded Atx7 on ubiquitin-specific protease (USP22), an interacting partner of Atx7 functioning in deubiquitination of histone H2B. The results showed that the inclusions or aggregates formed by polyQ-expanded Atx7 specifically sequester USP22 through their interactions mediated by the N-terminal zinc finger domain of Atx7. The mutation of the zinc finger domain in Atx7 that disrupts its interaction with USP22 dramatically abolishes sequestration of USP22. Moreover, polyQ expansion of Atx7 decreases the deubiquitinating activity of USP22 and, consequently, increases the level of monoubiquitinated H2B. Therefore, we propose that polyQ-expanded Atx7 forms insoluble aggregates that sequester USP22 into a catalytically inactive state, and then the impaired DUBm loses the function to deubiquitinate monoubiquitinated histone H2B or H2A. This may result in dysfunction of the SAGA complex and transcriptional dysregulation in spinocerebellar ataxia type 7 disease.
Collapse
Affiliation(s)
- Hui Yang
- From the State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences. 320 Yue-Yang Road, Shanghai 200031, China
| | - Shuai Liu
- From the State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences. 320 Yue-Yang Road, Shanghai 200031, China
| | - Wen-Tian He
- From the State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences. 320 Yue-Yang Road, Shanghai 200031, China
| | - Jian Zhao
- From the State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences. 320 Yue-Yang Road, Shanghai 200031, China
| | - Lei-Lei Jiang
- From the State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences. 320 Yue-Yang Road, Shanghai 200031, China
| | - Hong-Yu Hu
- From the State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences. 320 Yue-Yang Road, Shanghai 200031, China
| |
Collapse
|
204
|
Liu YL, Zheng J, Tang LJ, Han W, Wang JM, Liu DW, Tian QB. The deubiquitinating enzyme activity of USP22 is necessary for regulating HeLa cell growth. Gene 2015; 572:49-56. [PMID: 26143114 DOI: 10.1016/j.gene.2015.06.075] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 06/10/2015] [Accepted: 06/28/2015] [Indexed: 01/07/2023]
Abstract
Ubiquitin-specific protease 22 (USP22) can regulate the cell cycle and apoptosis in many cancer cell types, while it is still unclear whether the deubiquitinating enzyme activity of USP22 is necessary for these processes. As little is known about the impact of USP22 on the growth of HeLa cell, we observed whether USP22 can effectively regulate HeLa cell growth as well as the necessity of deubiquitinating enzyme activity for these processes in HeLa cell. In this study, we demonstrate that USP22 can regulate cell cycle but not apoptosis in HeLa cell. The deubiquitinating enzyme activity of USP22 is necessary for this process as confirmed by an activity-deleted mutant (C185S) and an activity-decreased mutant (Y513C). In addition, the deubiquitinating enzyme activity of USP22 is related to the levels of BMI-1, c-Myc, cyclin D2 and p53. Our findings indicate that the deubiquitinating enzyme activity of USP22 is necessary for regulating HeLa cell growth, and it promotes cell proliferation via the c-Myc/cyclin D2, BMI-1 and p53 pathways in HeLa cell.
Collapse
Affiliation(s)
- Ying-Li Liu
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang 050017, China
| | - Jie Zheng
- Department of Functional Neurosurgery, Hebei General Hospital, Shijiazhuang, China
| | - Li-Juan Tang
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang 050017, China
| | - Wei Han
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang 050017, China
| | - Jian-Min Wang
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang 050017, China
| | - Dian-Wu Liu
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang 050017, China
| | - Qing-Bao Tian
- Department of Epidemiology and Statistics, School of Public Health, Hebei Medical University, 361 East Zhongshan Road, Shijiazhuang 050017, China.
| |
Collapse
|
205
|
Qiu G, Li X, Che X, Wei C, He S, Lu J, Jia Z, Pang K, Fan L. SIRT1 is a regulator of autophagy: Implications in gastric cancer progression and treatment. FEBS Lett 2015; 589:2034-42. [PMID: 26049033 DOI: 10.1016/j.febslet.2015.05.042] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 05/22/2015] [Accepted: 05/27/2015] [Indexed: 12/20/2022]
Abstract
Silent mating type information regulation 1 (SIRT1) is implicated in tumorigenesis through its effect on autophagy. In gastric cancer (GC), SIRT1 is a marker for prognosis and is involved in cell invasion, proliferation, epithelial-mesenchymal transition (EMT) and drug resistance. Autophagy can function as a cell-survival mechanism or lead to cell death during the genesis and treatment of GC. This functionality is determined by factors including the stage of the tumor, cellular context and stress levels. Interestingly, SIRT1 can regulate autophagy through the deacetylation of autophagy-related genes (ATGs) and mediators of autophagy. Taken together, these findings support the need for continued research efforts to understand the mechanisms mediating the development of gastric cancer and unveil new strategies to eradicate this disease.
Collapse
Affiliation(s)
- Guanglin Qiu
- Department of General Surgery, The First Affiliated Hospital Medical College of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China
| | - Xuqi Li
- Department of General Surgery, The First Affiliated Hospital Medical College of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China
| | - Xiangming Che
- Department of General Surgery, The First Affiliated Hospital Medical College of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China
| | - Chao Wei
- Xi'an Health School, Xi'an 710054, Shaanxi Province, China
| | - Shicai He
- Department of General Surgery, The First Affiliated Hospital Medical College of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China
| | - Jing Lu
- Department of General Surgery, The First Affiliated Hospital Medical College of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China
| | - Zongliang Jia
- Department of General Surgery, The First Affiliated Hospital Medical College of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China
| | - Ke Pang
- Shaanxi Friendship Hospital, Xi'an 710068, Shaanxi Province, China
| | - Lin Fan
- Department of General Surgery, The First Affiliated Hospital Medical College of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China.
| |
Collapse
|
206
|
Song CL, Tang H, Ran LK, Ko BCB, Zhang ZZ, Chen X, Ren JH, Tao NN, Li WY, Huang AL, Chen J. Sirtuin 3 inhibits hepatocellular carcinoma growth through the glycogen synthase kinase-3β/BCL2-associated X protein-dependent apoptotic pathway. Oncogene 2015; 35:631-41. [PMID: 25915842 DOI: 10.1038/onc.2015.121] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 02/10/2015] [Accepted: 03/09/2015] [Indexed: 01/03/2023]
Abstract
SIRT3 is a class III histone deacetylase that has been implicated in a variety of cancers. The role of SIRT3 in hepatocellular carcinoma (HCC) remains elusive. In this study, we found that SIRT3 expression was frequently repressed in HCC and its downregulation was closely associated with tumor grade and size. Ectopic expression of SIRT3 inhibited cell growth and induced apoptosis in HCC cells, whereas depletion of SIRT3 in immortalized hepatocyte promoted cell growth and decreased epirubicin-induced apoptosis. Mechanistic studies revealed that SIRT3 deacetylated and activated glycogen synthase kinase-3β (GSK-3β), which subsequently induced expression and mitochondrial translocation of the pro-apoptotic protein BCL2-associated X protein (Bax) to promote apoptosis. GSK-3β inhibitor or gene silencing of BAX reversed SIRT3-induced growth inhibition and apoptosis. Furthermore, SIRT3 overexpression also suppressed tumor growth in vivo. Together, this study reveals a role of SIRT3/GSK-3β/Bax signaling pathway in the suppression of HCC growth, and also suggests that targeting this pathway may represent a potential therapeutic approach for HCC treatment.
Collapse
Affiliation(s)
- C-L Song
- The Second Affiliated Hospital and the Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - H Tang
- The Second Affiliated Hospital and the Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - L-K Ran
- The Second Affiliated Hospital and the Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - B C B Ko
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China.,State Key Laboratory of Chirosciences, The Hong Kong Polytechnic University, Hong Kong, China
| | - Z-Z Zhang
- Department of Infectious Diseases, The Children's Hospital of Chongqing Medical University, Chongqing, China
| | - X Chen
- The Second Affiliated Hospital and the Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - J-H Ren
- The Second Affiliated Hospital and the Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - N-N Tao
- The Second Affiliated Hospital and the Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - W-Y Li
- The Second Affiliated Hospital and the Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - A-L Huang
- The Second Affiliated Hospital and the Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China.,The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Zhejiang, China
| | - J Chen
- The Second Affiliated Hospital and the Key Laboratory of Molecular Biology of Infectious Diseases Designated by the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| |
Collapse
|
207
|
Lin YH, Yuan J, Pei H, Liu T, Ann DK, Lou Z. KAP1 Deacetylation by SIRT1 Promotes Non-Homologous End-Joining Repair. PLoS One 2015; 10:e0123935. [PMID: 25905708 PMCID: PMC4408008 DOI: 10.1371/journal.pone.0123935] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 03/09/2015] [Indexed: 01/13/2023] Open
Abstract
Homologous recombination and non-homologous end joining are two major DNA double-strand-break repair pathways. While HR-mediated repair requires a homologous sequence as the guiding template to restore the damage site precisely, NHEJ-mediated repair ligates the DNA lesion directly and increases the risk of losing nucleotides. Therefore, how a cell regulates the balance between HR and NHEJ has become an important issue for maintaining genomic integrity over time. Here we report that SIRT1-dependent KAP1 deacetylation positively regulates NHEJ. We show that up-regulation of KAP1 attenuates HR efficiency while promoting NHEJ repair. Moreover, SIRT1-mediated KAP1 deacetylation further enhances the effect of NHEJ by stabilizing its interaction with 53BP1, which leads to increased 53BP1 focus formation in response to DNA damage. Taken together, our study suggests a SIRT1-KAP1 regulatory mechanism for HR-NHEJ repair pathway choice.
Collapse
Affiliation(s)
- Yi-Hui Lin
- Department of Biochemistry and Molecular Biology, Mayo Graduate School, Rochester, Minnesota, United States of America
| | - Jian Yuan
- Research Center for Translational Medicine, Tongji University School of Medicine, Shanghai, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Huadong Pei
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Tongzheng Liu
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - David K. Ann
- Department of Molecular Pharmacology and Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute, City of Hope, Duarte, California, United States of America
| | - Zhenkun Lou
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
208
|
Kobayashi T, Iwamoto Y, Takashima K, Isomura A, Kosodo Y, Kawakami K, Nishioka T, Kaibuchi K, Kageyama R. Deubiquitinating enzymes regulate Hes1 stability and neuronal differentiation. FEBS J 2015; 282:2411-23. [PMID: 25846153 DOI: 10.1111/febs.13290] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 03/03/2015] [Accepted: 03/30/2015] [Indexed: 11/30/2022]
Abstract
Hairy and enhancer of split 1 (Hes1), a basic helix-loop-helix transcriptional repressor protein, regulates the maintenance of neural stem/progenitor cells by repressing proneural gene expression via Notch signaling. Previous studies showed that Hes1 expression oscillates in both mouse embryonic stem cells and neural stem cells, and that the oscillation contributes to their potency and differentiation fates. This oscillatory expression depends on the stability of Hes1, which is rapidly degraded by the ubiquitin/proteasome pathway. However, the detailed molecular mechanisms governing Hes1 stability remain unknown. We analyzed Hes1-interacting deubiquitinases purified from mouse embryonic stem cells using an Hes1-specific antibody, and identified the ubiquitin-specific protease 27x (Usp27x) as a new regulator of Hes1. We found that Hes1 was deubiquitinated and stabilized by Usp27x and its homologs ubiquitin-specific protease 22 (Usp22) and ubiquitin-specific protease 51 (Usp51). Knockdown of Usp22 shortened the half-life of Hes1, delayed its oscillation, and enhanced neuronal differentiation in mouse developing brain, whereas mis-expression of Usp27x reduced neuronal differentiation. These results suggest that these deubiquitinases modulate Hes1 protein dynamics by removing ubiquitin molecules, and thereby regulate neuronal differentiation of stem cells.
Collapse
Affiliation(s)
- Taeko Kobayashi
- Institute for Virus Research, Kyoto University, Japan.,Japan Science and Technology Agency, Core Research for Evolutional Science and Technology (CREST), Kawaguchi, Japan.,Graduate School of Medicine, Kyoto University, Japan.,Graduate School of Biostudies, Kyoto University, Japan
| | | | | | - Akihiro Isomura
- Institute for Virus Research, Kyoto University, Japan.,Japan Science and Technology Agency, Core Research for Evolutional Science and Technology (CREST), Kawaguchi, Japan
| | - Yoichi Kosodo
- Department of Anatomy, Kawasaki Medical School, Kurashiki, Japan
| | - Koichi Kawakami
- Division of Molecular and Developmental Biology, National Institute of Genetics, Shizuoka, Japan
| | - Tomoki Nishioka
- Department of Cell Pharmacology, Graduate School of Medicine, Nagoya University, Japan
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Graduate School of Medicine, Nagoya University, Japan
| | - Ryoichiro Kageyama
- Institute for Virus Research, Kyoto University, Japan.,Japan Science and Technology Agency, Core Research for Evolutional Science and Technology (CREST), Kawaguchi, Japan.,Graduate School of Medicine, Kyoto University, Japan.,Graduate School of Biostudies, Kyoto University, Japan.,World Premier International Research Initiative/Institute for Integrated Cell and Material Sciences, Kyoto University, Japan
| |
Collapse
|
209
|
Ao N, Liu Y, Bian X, Feng H, Liu Y. Ubiquitin-specific peptidase 22 inhibits colon cancer cell invasion by suppressing the signal transducer and activator of transcription 3/matrix metalloproteinase 9 pathway. Mol Med Rep 2015; 12:2107-13. [PMID: 25902005 DOI: 10.3892/mmr.2015.3661] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 03/10/2015] [Indexed: 11/06/2022] Open
Abstract
Colon cancer is associated with increased cell migration and invasion. In the present study, the role of ubiquitin-specific peptidase 22 (USP22) in signal transducer and activator of transcription 3 (STAT3)-mediated colon cancer cell invasion was investigated. The messenger RNA levels of STAT3 target genes were measured by reverse transcription-quantitative polymerase chain reaction, following USP22 knockdown by RNA interference in SW480 colon cancer cells. The matrix metalloproteinase 9 (MMP9) proteolytic activity and invasion potential of SW480 cells were measured by zymography and Transwell assay, respectively, following combined USP22 and STAT3 short interfering (si)RNA treatment or STAT3 siRNA treatment alone. Similarly, a cell counting kit-8 assay was used to detect the proliferation potential of SW480 cells. The protein expression levels of USP22, STAT3 and MMP9 were detected by immunohistochemistry in colon cancer tissue microarrays (TMAs) and the correlation between USP22, STAT3 and MMP9 was analyzed. USP22/STAT3 co-depletion partly rescued the MMP9 proteolytic activity and invasion of SW480 cells, compared with that of STAT3 depletion alone. However, the proliferation of USP22/STAT3si-SW480 cells was decreased compared with that of STAT3si-SW480 cells. USP22 expression was positively correlated with STAT3 and MMP9 expression in colon cancer TMAs. In conclusion, USP22 attenuated the invasion capacity of colon cancer cells by inhibiting the STAT3/MMP9 signaling pathway.
Collapse
Affiliation(s)
- Ning Ao
- Department of Pathology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Tsinghua University, Beijing 100005, P.R. China
| | - Yanyan Liu
- Department of Pathology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Tsinghua University, Beijing 100005, P.R. China
| | - Xiaocui Bian
- Department of Pathology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Tsinghua University, Beijing 100005, P.R. China
| | - Hailiang Feng
- Department of Pathology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Tsinghua University, Beijing 100005, P.R. China
| | - Yuqin Liu
- Department of Pathology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Tsinghua University, Beijing 100005, P.R. China
| |
Collapse
|
210
|
Xiong J, Gong Z, Zhou X, Liu J, Jiang HE, Wu P, Li W. p38 mitogen-activated protein kinase inhibits USP22 transcription in HeLa cells. Biomed Rep 2015; 3:461-467. [PMID: 26171149 DOI: 10.3892/br.2015.450] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 03/12/2015] [Indexed: 12/24/2022] Open
Abstract
Elevated expression of ubiquitin-specific processing enzyme 22 (USP22) was identified in multiple types of human cancers, and was correlated with tumorigenesis and progression. Despite an increase in the numbers of studies in the physiological function of USP22, little is known regarding the regulation of its expression. The 5' flanking sequence of the USP22 gene was recently characterized. In the present study, USP22 transcription was regulated by p38 mitogen-activated protein kinase (MAPK). Treatment of human cervical carcinoma (HeLa) cells with SB203580, an inhibitor of p38 MAPK, enhanced basal USP22 promoter activity and mRNA abundance. Transfection of MAPK kinase 6 (MKK6), an upstream activator of p38 MAPK, resulted in a 40% decrease in USP22 mRNA, while the dominant negative MKK6 increased the transcription level of the USP22, similar to SB203580. Dual luciferase report assays showed that mutations of the Sp1 binding site ahead of the transcription start site abolished the promoting effect of the USP22 promoter by SB203580. Cisplatin, the activator of p38 MAPK, also suppressed USP22 expression. This suppression was blocked by SB203580. In conclusion, p38 MAPK acts as an upstream negative regulator of USP22 transcription in HeLa cells.
Collapse
Affiliation(s)
- Jianjun Xiong
- Key Laboratory of Jiangxi Province for the Systems Bio-Medicine, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China ; College of Basic Medical Science, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Zhen Gong
- College of Basic Medical Science, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Xiaou Zhou
- Key Laboratory of Jiangxi Province for the Systems Bio-Medicine, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China ; College of Basic Medical Science, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Jianyun Liu
- Key Laboratory of Jiangxi Province for the Systems Bio-Medicine, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - H E Jiang
- Key Laboratory of Jiangxi Province for the Systems Bio-Medicine, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Ping Wu
- Key Laboratory of Jiangxi Province for the Systems Bio-Medicine, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Weidong Li
- Key Laboratory of Jiangxi Province for the Systems Bio-Medicine, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| |
Collapse
|
211
|
Abstract
During the development of the central nervous system (CNS), neurons and glia are derived from multipotent neural stem cells (NSCs) undergoing self-renewal. NSC commitment and differentiation are tightly controlled by intrinsic and external regulatory mechanisms in space- and time-related fashions. SIRT1, a silent information regulator 2 (Sir2) ortholog, is expressed in several areas of the brain and has been reported to be involved in the self-renewal, multipotency, and fate determination of NSCs. Recent studies have highlighted the role of the deacetylase activity of SIRT1 in the determination of the final fate of NSCs. This review summarizes the roles of SIRT1 in the expansion and differentiation of NSCs, specification of neuronal subtypes and glial cells, and reprogramming of functional neurons from embryonic stem cells and fibroblasts. This review also discusses potential signaling pathways through which SIRT1 can exhibit versatile functions in NSCs to regulate the cell fate decisions of neurons and glia.
Collapse
|
212
|
Abstract
Precise regulation of gene expression programs during embryo development requires cooperation between transcriptional factors and histone-modifying enzymes, such as the Gcn5 histone acetyltransferase. Gcn5 functions within a multi-subunit complex, called SAGA, that is recruited to specific genes through interactions with sequence-specific DNA-binding proteins to aid in gene activation. Although the transcriptional programs regulated by SAGA in embryos are not well defined, deletion of either Gcn5 or USP22, the catalytic subunit of a deubiquitinase module in SAGA, leads to early embryonic lethality. Here, we review the known functions of Gcn5, USP22 and associated proteins during development and discuss how these functions might be related to human disease states, including cancer and neurodegenerative diseases.
Collapse
Affiliation(s)
- Li Wang
- Program in Molecular Carcinogenesis, Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Science Park, Smithville, TX 78957, USA
| | | |
Collapse
|
213
|
Hu J, Yang D, Zhang H, Liu W, Zhao Y, Lu H, Meng Q, Pang H, Chen X, Liu Y, Cai L. USP22 promotes tumor progression and induces epithelial-mesenchymal transition in lung adenocarcinoma. Lung Cancer 2015; 88:239-45. [PMID: 25907317 DOI: 10.1016/j.lungcan.2015.02.019] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 02/26/2015] [Accepted: 02/27/2015] [Indexed: 01/30/2023]
Abstract
OBJECTIVES Our previous study showed that USP22 as an oncogene may mediate cancer development and progression in NSCLC, but the underlying molecular mechanism remains uncharacterized. Epithelial-mesenchymal transition (EMT) has been reported to play an important role in migration and invasion of the tumor cells. Thus, this study aims to determine the clinical significance and the possible roles of USP22 in EMT and progression of lung adenocarcinoma. METHODS Immunohistochemistry was used to determine the expression of USP22 in clinical samples. The clinical correlations and prognostic significance of the aberrantly expressed proteins were evaluated by statistical analysis. Moreover, we evaluated whether USP22 could induce EMT in cultured lung cancer cells. RESULTS The USP22 expression was positive in 76.03% of specimens and was correlated with advanced clinicopathologic classifications (differentiation, T and AJCC stages) and TGF-β1 expression (p=0.008). Multivariate Cox regression analysis revealed that USP22 expression level was an independent prognostic factor for both overall survival and disease-free survival (HR, 2.060; p=0.013 and HR, 1.993; p=0.016). In vitro study revealed that USP22 can regulate proliferation and invasive properties, and induce EMT of lung adenocarcinoma cells. Moreover, USP22 may up-regulate TGF-β1 expression. CONCLUSIONS Our data indicated that USP22 may promote lung adenocarcinoma cell invasion by the induction of EMT.
Collapse
Affiliation(s)
- Jing Hu
- The 4th Department of Internal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China.
| | - Dongdong Yang
- Department of Oncological Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Huijuan Zhang
- The 4th Department of Internal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Wei Liu
- The 4th Department of Internal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Yanbin Zhao
- The 4th Department of Internal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Hailing Lu
- The 4th Department of Internal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Qingwei Meng
- The 4th Department of Internal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Hui Pang
- The 4th Department of Internal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Xuesong Chen
- The 4th Department of Internal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Yanlong Liu
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China.
| | - Li Cai
- The 4th Department of Internal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China.
| |
Collapse
|
214
|
Deubiquitinase inhibition as a cancer therapeutic strategy. Pharmacol Ther 2015; 147:32-54. [DOI: 10.1016/j.pharmthera.2014.11.002] [Citation(s) in RCA: 200] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 09/16/2014] [Indexed: 12/27/2022]
|
215
|
Peng L, Yuan Z, Li Y, Ling H, Izumi V, Fang B, Fukasawa K, Koomen J, Chen J, Seto E. Ubiquitinated sirtuin 1 (SIRT1) function is modulated during DNA damage-induced cell death and survival. J Biol Chem 2015; 290:8904-12. [PMID: 25670865 DOI: 10.1074/jbc.m114.612796] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Indexed: 11/06/2022] Open
Abstract
Downstream signaling of physiological and pathological cell responses depends on post-translational modification such as ubiquitination. The mechanisms regulating downstream DNA damage response (DDR) signaling are not completely elucidated. Sirtuin 1 (SIRT1), the founding member of Class III histone deacetylases, regulates multiple steps in DDR and is closely associated with many physiological and pathological processes. However, the role of post-translational modification or ubiquitination of SIRT1 during DDR is unclear. We show that SIRT1 is dynamically and distinctly ubiquitinated in response to DNA damage. SIRT1 was ubiquitinated by the MDM2 E3 ligase in vitro and in vivo. SIRT1 ubiquitination under normal conditions had no effect on its enzymatic activity or rate of degradation; hypo-ubiquitination, however, reduced SIRT1 nuclear localization. Ubiquitination of SIRT1 affected its function in cell death and survival in response to DNA damage. Our results suggest that ubiquitination is required for SIRT1 function during DDR.
Collapse
Affiliation(s)
- Lirong Peng
- From the Department of Molecular Oncology, Moffitt Cancer Center and Research Institute, Tampa, Florida 33612
| | - Zhigang Yuan
- From the Department of Molecular Oncology, Moffitt Cancer Center and Research Institute, Tampa, Florida 33612
| | - Yixuan Li
- From the Department of Molecular Oncology, Moffitt Cancer Center and Research Institute, Tampa, Florida 33612
| | - Hongbo Ling
- From the Department of Molecular Oncology, Moffitt Cancer Center and Research Institute, Tampa, Florida 33612
| | - Victoria Izumi
- From the Department of Molecular Oncology, Moffitt Cancer Center and Research Institute, Tampa, Florida 33612
| | - Bin Fang
- From the Department of Molecular Oncology, Moffitt Cancer Center and Research Institute, Tampa, Florida 33612
| | - Kenji Fukasawa
- From the Department of Molecular Oncology, Moffitt Cancer Center and Research Institute, Tampa, Florida 33612
| | - John Koomen
- From the Department of Molecular Oncology, Moffitt Cancer Center and Research Institute, Tampa, Florida 33612
| | - Jiandong Chen
- From the Department of Molecular Oncology, Moffitt Cancer Center and Research Institute, Tampa, Florida 33612
| | - Edward Seto
- From the Department of Molecular Oncology, Moffitt Cancer Center and Research Institute, Tampa, Florida 33612
| |
Collapse
|
216
|
Abstract
Deubiquitinases (DUBs) play important roles and therefore are potential drug targets in various diseases including cancer and neurodegeneration. In this review, we recapitulate structure-function studies of the most studied DUBs including USP7, USP22, CYLD, UCHL1, BAP1, A20, as well as ataxin 3 and connect them to regulatory mechanisms and their growing protein interaction networks. We then describe DUBs that have been associated with endocrine carcinogenesis with a focus on prostate, ovarian, and thyroid cancer, pheochromocytoma, and adrenocortical carcinoma. The goal is enhancing our understanding of the connection between dysregulated DUBs and cancer to permit the design of therapeutics and to establish biomarkers that could be used in diagnosis and prognosis.
Collapse
Affiliation(s)
- Roland Pfoh
- Department of BiologyYork University, 4700 Keele Street, Toronto, Ontario, Canada, M3J1P3
| | - Ira Kay Lacdao
- Department of BiologyYork University, 4700 Keele Street, Toronto, Ontario, Canada, M3J1P3
| | - Vivian Saridakis
- Department of BiologyYork University, 4700 Keele Street, Toronto, Ontario, Canada, M3J1P3
| |
Collapse
|
217
|
Abstract
Ubiquitination has traditionally been viewed in the context of polyubiquitination that is essential for marking proteins for degradation via the proteasome. Recent discoveries have shed light on key cellular roles for monoubiquitination, including as a post-translational modification (PTM) of histones such as histone H2B. Monoubiquitination plays a significant role as one of the largest histone PTMs, alongside smaller, better-studied modifications such as methylation, acetylation and phosphorylation. Monoubiquitination of histone H2B at lysine 120 (H2Bub1) has been shown to have key roles in transcription, the DNA damage response and stem cell differentiation. The H2Bub1 enzymatic cascade involves E3 RING finger ubiquitin ligases, with the main E3 generally accepted to be the RNF20-RNF40 complex, and deubiquitinases including ubiquitin-specific protease 7 (USP7), USP22 and USP44. H2Bub1 has been shown to physically disrupt chromatin strands, fostering a more open chromatin structure accessible to transcription factors and DNA repair proteins. It also acts as a recruiting signal, actively attracting proteins with roles in transcription and DNA damage. H2Bub1 also appears to play central roles in histone cross-talk, influencing methylation events on histone H3, including H3K4 and H3K79. Most significantly, global levels of H2Bub1 are low to absent in advanced cancers including breast, colorectal, lung and parathyroid, marking H2Bub1 and the enzymes that regulate it as key molecules of interest as possible new therapeutic targets for the treatment of cancer. This review offers an overview of current knowledge regarding H2Bub1 and highlights links between dysregulation of H2Bub1-associated enzymes, stem cells and malignancy.
Collapse
Affiliation(s)
- Alexander J Cole
- Hormones and Cancer GroupKolling Institute of Medical Research, Royal North Shore Hospital, University of Sydney, Sydney, New South Wales 2065, Australia
| | - Roderick Clifton-Bligh
- Hormones and Cancer GroupKolling Institute of Medical Research, Royal North Shore Hospital, University of Sydney, Sydney, New South Wales 2065, Australia
| | - Deborah J Marsh
- Hormones and Cancer GroupKolling Institute of Medical Research, Royal North Shore Hospital, University of Sydney, Sydney, New South Wales 2065, Australia
| |
Collapse
|
218
|
XIONG JIANJUN, ZHOU XIAOOU, GONG ZHEN, WANG TING, ZHANG CHAO, XU XIAOYUAN, LIU JIANYUN, LI WEIDONG. PKA/CREB regulates the constitutive promoter activity of the USP22 gene. Oncol Rep 2015; 33:1505-11. [DOI: 10.3892/or.2015.3740] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 11/14/2014] [Indexed: 11/06/2022] Open
|
219
|
Sasaki T. Age-Associated Weight Gain, Leptin, and SIRT1: A Possible Role for Hypothalamic SIRT1 in the Prevention of Weight Gain and Aging through Modulation of Leptin Sensitivity. Front Endocrinol (Lausanne) 2015; 6:109. [PMID: 26236282 PMCID: PMC4504171 DOI: 10.3389/fendo.2015.00109] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Accepted: 07/01/2015] [Indexed: 12/14/2022] Open
Abstract
The hypothalamus is the principal regulator of body weight and energy balance. It modulates both energy intake and energy expenditure by sensing the energy status of the body through neural inputs from the periphery as well as direct humoral inputs. Leptin, an adipokine, is one of the humoral factors responsible for alerting the hypothalamus that enough energy is stored in the periphery. Plasma leptin levels are positively linked to adiposity; leptin suppress energy intake and stimulates energy expenditure. However, prolonged increases in plasma leptin levels due to obesity cause leptin resistance, affecting both leptin access to hypothalamic neurons and leptin signal transduction within hypothalamic neurons. Decreased sensing of peripheral energy status through leptin may lead to a positive energy balance and gradual gains in weight and adiposity, further worsening leptin resistance. Leptin resistance, increased adiposity, and weight gain are all associated with aging in both humans and animals. Central insulin resistance is associated with similar observations. Therefore, improving the action of humoral factors in the hypothalamus may prevent gradual weight gain, especially during middle age. SIRT1 is a NAD(+)-dependent protein deacetylase with numerous substrates, including histones, transcription factors, co-factors, and various enzymes. SIRT1 improves both leptin sensitivity and insulin sensitivity by decreasing the levels of several molecules that impair leptin and insulin signal transduction. SIRT1 and NAD(+) levels decrease with age in the hypothalamus; increased hypothalamic SIRT1 levels prevent age-associated weight gain and improve leptin sensitivity in mice. Therefore, preventing the age-dependent loss of SIRT1 function in the hypothalamus could improve the action of humoral factors in the hypothalamus as well as central regulation of energy balance.
Collapse
Affiliation(s)
- Tsutomu Sasaki
- Laboratory for Metabolic Signaling, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
- *Correspondence: Tsutomu Sasaki, Laboratory for Metabolic Signaling, Institute for Molecular and Cellular Regulation, Gunma University, 3-39-15 Showa-machi, Maebashi, Gunma 371-8512, Japan,
| |
Collapse
|
220
|
Yin YW, Jin HJ, Zhao W, Gao B, Fang J, Wei J, Zhang DD, Zhang J, Fang D. The Histone Acetyltransferase GCN5 Expression Is Elevated and Regulated by c-Myc and E2F1 Transcription Factors in Human Colon Cancer. Gene Expr 2015; 16:187-96. [PMID: 26637399 PMCID: PMC5584536 DOI: 10.3727/105221615x14399878166230] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The histone acetyltransferase GCN5 has been suggested to be involved in promoting cancer cell growth. But its role in human colon cancer development remains unknown. Herein we discovered that GCN5 expression is significantly upregulated in human colon adenocarcinoma tissues. We further demonstrate that GCN5 is upregulated in human colon cancer at the mRNA level. Surprisingly, two transcription factors, the oncogenic c-Myc and the proapoptotic E2F1, are responsible for GCN5 mRNA transcription. Knockdown of c-Myc inhibited colon cancer cell proliferation largely through downregulating GCN5 transcription, which can be fully rescued by the ectopic GCN5 expression. In contrast, E2F1 expression induced human colon cancer cell death, and suppression of GCN5 expression in cells with E2F1 overexpression further facilitated cell apoptosis, suggesting that GCN5 expression is induced by E2F1 as a possible negative feedback in suppressing E2F1-mediated cell apoptosis. In addition, suppression of GCN5 with its specific inhibitor CPTH2 inhibited human colon cancer cell growth. Our studies reveal that GCN5 plays a positive role in human colon cancer development, and its suppression holds a great therapeutic potential in antitumor therapy.
Collapse
Affiliation(s)
- Yan-Wei Yin
- *Department of Oncology, Linyi People’s Hospital, and Linyi Tumor Hospital, Linyi, P.R. China
| | - Hong-Jian Jin
- †Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Wenjing Zhao
- *Department of Oncology, Linyi People’s Hospital, and Linyi Tumor Hospital, Linyi, P.R. China
| | - Beixue Gao
- ‡Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jiangao Fang
- ‡Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Junmin Wei
- §Department of Chemotherapy, Cancer Center, Qilu Hospital, Shandong University, Jinan, P.R. China
| | - Donna D. Zhang
- ¶Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA
| | - Jianing Zhang
- #School of Life Science and Medicine, Dalian University of Technology, Panjin, P.R. China
| | - Deyu Fang
- ‡Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- #School of Life Science and Medicine, Dalian University of Technology, Panjin, P.R. China
| |
Collapse
|
221
|
USP22 promotes NSCLC tumorigenesis via MDMX up-regulation and subsequent p53 inhibition. Int J Mol Sci 2014; 16:307-20. [PMID: 25547493 PMCID: PMC4307248 DOI: 10.3390/ijms16010307] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 12/15/2014] [Indexed: 12/15/2022] Open
Abstract
Increasing evidence suggests that ubiquitin-specific protease 22 (USP22) has great clinicopathologic significance in oncology. In this study, we investigated the role of USP22 in human NSCLC tumorigenesis along with the underlying mechanisms of action. First, we determined the expression of USP22 in human NSCLC, as well as normal tissues and cell lines. We then studied the effects of USP22 silencing by shRNA on NSCLC cell growth in vitro and tumorigenesis in vivo, along with the effect on the p53 pathway. We found that USP22 is overexpressed in human NSCLC tissues and cell lines. USP22 silencing by shRNA inhibits proliferation, induces apoptosis and arrests cells at the G0/G1 phases in NSCLC cells and curbs human NSCLC tumor growth in a mouse xenograft model. Additionally, USP22 silencing downregulates MDMX protein expression and activates the p53 pathway. Our co-immunoprecipitation analysis shows that USP22 interacts with MDMX in NSCLC cells. Furthermore, MDMX silencing leads to growth arrest and apoptosis in NSCLC cells, and over-expression of MDMX reverses the USP22 silencing-induced effects. Taken together, our results suggest that USP22 promotes NSCLC tumorigenesis in vitro and in vivo through MDMX upregulation and subsequent p53 inhibition. USP22 may represent a novel target for NSCLC treatment.
Collapse
|
222
|
Kozako T, Suzuki T, Yoshimitsu M, Arima N, Honda SI, Soeda S. Anticancer agents targeted to sirtuins. Molecules 2014; 19:20295-313. [PMID: 25486244 PMCID: PMC6270850 DOI: 10.3390/molecules191220295] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Revised: 11/28/2014] [Accepted: 12/01/2014] [Indexed: 12/16/2022] Open
Abstract
Sirtuins are nicotinamide adenine dinucleotide+-dependent deacetylases of which there are seven isoforms (SIRT1–7). Sirtuin activity is linked to gene expression, lifespan extension, neurodegeneration, and age-related disorders. Numerous studies have suggested that sirtuins could be of great significance with regard to both antiaging and tumorigenesis, depending on its targets in specific signaling pathways or in specific cancers. Recent studies have identified small chemical compounds that modulate sirtuins, and these modulators have enabled a greater understanding of the biological function and molecular mechanisms of sirtuins. This review highlights the possibility of sirtuins, especially SIRT1 and SIRT2, for cancer therapy targets, and focuses on the therapeutic potential of sirtuin modulators both in cancer prevention and treatment.
Collapse
Affiliation(s)
- Tomohiro Kozako
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Takayoshi Suzuki
- Faculty of Medicine, Kyoto Prefectural University of Medicine, 1-5 Shimogamohangi-Cho, Sakyo-Ku, Kyoto 606-0823, Japan.
| | - Makoto Yoshimitsu
- Department of Hematology and Immunology, Kagoshima University Hospital, Kagoshima, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan.
| | - Naomichi Arima
- Department of Hematology and Immunology, Kagoshima University Hospital, Kagoshima, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan.
| | - Shin-ichiro Honda
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Shinji Soeda
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| |
Collapse
|
223
|
Liu T, Lin YH, Leng W, Jung SY, Zhang H, Deng M, Evans D, Li Y, Luo K, Qin B, Qin J, Yuan J, Lou Z. A divergent role of the SIRT1-TopBP1 axis in regulating metabolic checkpoint and DNA damage checkpoint. Mol Cell 2014; 56:681-95. [PMID: 25454945 DOI: 10.1016/j.molcel.2014.10.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 07/02/2014] [Accepted: 10/06/2014] [Indexed: 11/16/2022]
Abstract
DNA replication is executed only when cells have sufficient metabolic resources and undamaged DNA. Nutrient limitation and DNA damage cause a metabolic checkpoint and DNA damage checkpoint, respectively. Although SIRT1 activity is regulated by metabolic stress and DNA damage, its function in these stress-mediated checkpoints remains elusive. Here we report that the SIRT1-TopBP1 axis functions as a switch for both checkpoints. With glucose deprivation, SIRT1 is activated and deacetylates TopBP1, resulting in TopBP1-Treslin disassociation and DNA replication inhibition. Conversely, SIRT1 activity is inhibited under genotoxic stress, resulting in increased TopBP1 acetylation that is important for the TopBP1-Rad9 interaction and activation of the ATR-Chk1 pathway. Mechanistically, we showed that acetylation of TopBP1 changes the conformation of TopBP1, thereby facilitating its interaction with distinct partners in DNA replication and checkpoint activation. Taken together, our studies identify the SIRT1-TopBP1 axis as a key signaling mode in the regulation of the metabolic checkpoint and the DNA damage checkpoint.
Collapse
Affiliation(s)
- Tongzheng Liu
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Yi-Hui Lin
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Wenchuan Leng
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sung Yun Jung
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Haoxing Zhang
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Min Deng
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Debra Evans
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Yunhui Li
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Kuntian Luo
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Bo Qin
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Jun Qin
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jian Yuan
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China.
| | - Zhenkun Lou
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
224
|
Han L, Zhao G, Wang H, Tong T, Chen J. Calorie restriction upregulated sirtuin 1 by attenuating its ubiquitin degradation in cancer cells. Clin Exp Pharmacol Physiol 2014; 41:165-8. [PMID: 24471483 DOI: 10.1111/1440-1681.12199] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 12/19/2013] [Accepted: 12/19/2013] [Indexed: 11/29/2022]
Abstract
Sirtuin (SIRT) 1 is a key protein in mediating the benefits of calorie restriction (CR) in mammals. However, the molecular mechanisms underlying CR-induced SIRT1 upregulation in mammals remain unclear. Herein we show that the elevated SIRT1 levels are not due to increased SIRT1 mRNA expression. but rather to enhanced SIRT1 protein stability as a result of reduced ubiquitin-proteasome degradation of SIRT1 under limited nutrient conditions. Our observations have important implications for improving healthy aging in humans.
Collapse
Affiliation(s)
- Limin Han
- Peking University Research Center on Ageing, Peking University Health Science Center, Beijing, China; Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | | | | | | | | |
Collapse
|
225
|
Ji M, Shi H, Xie Y, Zhao Z, Li S, Chang C, Cheng X, Li Y. Ubiquitin specific protease 22 promotes cell proliferation and tumor growth of epithelial ovarian cancer through synergy with transforming growth factor β1. Oncol Rep 2014; 33:133-40. [PMID: 25369910 DOI: 10.3892/or.2014.3580] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 09/22/2014] [Indexed: 11/06/2022] Open
Abstract
Ubiquitin specific protease 22 (USP22) is an oncogene that is upregulated in many cancer types, and aberrant expression of USP22 correlates with clinical outcome. However, its potential functional impact in epithelial ovarian cancer (EOC) has not been determined. Here, we report that USP22 was upregulated in EOC specimens and EOC cell lines with important functional consequences. A high level of USP22 in EOC tissues was associated with advanced clinical FIGO stage, lymph node metastasis and worse prognosis. Patients with higher USP22 expression had shorter relapse-free and overall survival. Depletion of USP22 suppressed cell proliferation in vitro and tumor growth in vivo. We found that inhibition of USP22 suppressed cell proliferation by inducing G1 phase cell cycle arrest through synergy with oncogenic transforming growth factor-β1 (TGFB1). Our results indicate that USP22 functions as an oncogene in EOC, and thus USP22 may serve as a potential therapeutic target for individualized EOC treatment.
Collapse
Affiliation(s)
- Mei Ji
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Huirong Shi
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Ya Xie
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Zhao Zhao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Shunshuang Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Cheng Chang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Xinghan Cheng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yue Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
226
|
Xiong J, Xu X, Zhou X, Liu J, Gong Z, Wu P, Li W. USP22 transcriptional activity is negatively regulated by the histone deacetylase inhibitor trichostatin A. Mol Med Rep 2014; 10:3343-7. [PMID: 25323692 DOI: 10.3892/mmr.2014.2666] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 08/29/2014] [Indexed: 11/05/2022] Open
Abstract
The ubiquitin‑specific protease 22 (USP22) gene is overexpressed in the majority of types of cancer cells, and has been implicated in tumorigenesis. However, the mechanisms that regulate its expression remain unclear. The results of the present study demonstrated that the expression of USP22 is negatively regulated by trichostatin A (TSA), a classical histone deacetylase inhibitor. Furthermore, TSA was revealed to interfere with the binding of RNA polymerase II to the USP22 promoter, directly suppressing its transcription. In addition, the overexpression of USP22 was observed to attenuate TSA‑induced apoptosis in HeLa cells. To the best of our knowledge, these results provide the first insight into the regulation of the USP22 gene by antitumor drugs and into the mechanisms underlying the anticancer activity of TSA.
Collapse
Affiliation(s)
- Jianjun Xiong
- Jiangxi Province Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Xiaoyuan Xu
- Jiangxi Province Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Xiaou Zhou
- Jiangxi Province Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Jianyun Liu
- Jiangxi Province Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Zhen Gong
- College of Basic Medical Science, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Ping Wu
- Jiangxi Province Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Weidong Li
- Jiangxi Province Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| |
Collapse
|
227
|
Pan J, Deng Q, Jiang C, Wang X, Niu T, Li H, Chen T, Jin J, Pan W, Cai X, Yang X, Lu M, Xiao J, Wang P. USP37 directly deubiquitinates and stabilizes c-Myc in lung cancer. Oncogene 2014; 34:3957-67. [DOI: 10.1038/onc.2014.327] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 08/03/2014] [Accepted: 08/19/2014] [Indexed: 01/07/2023]
|
228
|
Li L, Osdal T, Ho Y, Chun S, McDonald T, Agarwal P, Lin A, Chu S, Qi J, Li L, Hsieh YT, Dos Santos C, Yuan H, Ha TQ, Popa M, Hovland R, Bruserud Ø, Gjertsen BT, Kuo YH, Chen W, Lain S, McCormack E, Bhatia R. SIRT1 activation by a c-MYC oncogenic network promotes the maintenance and drug resistance of human FLT3-ITD acute myeloid leukemia stem cells. Cell Stem Cell 2014; 15:431-446. [PMID: 25280219 PMCID: PMC4305398 DOI: 10.1016/j.stem.2014.08.001] [Citation(s) in RCA: 172] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 07/11/2014] [Accepted: 08/08/2014] [Indexed: 12/12/2022]
Abstract
The FLT3-ITD mutation is frequently observed in acute myeloid leukemia (AML) and is associated with poor prognosis. In such patients, FLT3 tyrosine kinase inhibitors (TKIs) are only partially effective and do not eliminate the leukemia stem cells (LSCs) that are assumed to be the source of treatment failure. Here, we show that the NAD-dependent SIRT1 deacetylase is selectively overexpressed in primary human FLT3-ITD AML LSCs. This SIRT1 overexpression is related to enhanced expression of the USP22 deubiquitinase induced by c-MYC, leading to reduced SIRT1 ubiquitination and enhanced stability. Inhibition of SIRT1 expression or activity reduced the growth of FLT3-ITD AML LSCs and significantly enhanced TKI-mediated killing of the cells. Therefore, these results identify a c-MYC-related network that enhances SIRT1 protein expression in human FLT3-ITD AML LSCs and contributes to their maintenance. Inhibition of this oncogenic network could be an attractive approach for targeting FLT3-ITD AML LSCs to improve treatment outcomes.
Collapse
MESH Headings
- Animals
- Antigens, CD34/metabolism
- Benzothiazoles/pharmacology
- Cell Proliferation/drug effects
- Cell Survival/drug effects
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Gene Duplication/drug effects
- Gene Knockdown Techniques
- Gene Regulatory Networks
- Humans
- Leukemia, Myeloid, Acute/enzymology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Mice, SCID
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/enzymology
- Neoplastic Stem Cells/pathology
- Phenylurea Compounds/pharmacology
- Protein Binding/drug effects
- Proto-Oncogene Proteins c-myc/genetics
- Proto-Oncogene Proteins c-myc/metabolism
- Sirtuin 1/antagonists & inhibitors
- Sirtuin 1/metabolism
- Thiolester Hydrolases/metabolism
- Ubiquitin Thiolesterase
- fms-Like Tyrosine Kinase 3/metabolism
Collapse
Affiliation(s)
- Ling Li
- Division of Hematopoietic Stem Cell and Leukemia Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Tereza Osdal
- Department of Clinical Science, Hematology Section, University of Bergen, Bergen 5021, Norway
| | - Yinwei Ho
- Division of Hematopoietic Stem Cell and Leukemia Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Sookhee Chun
- Division of Hematopoietic Stem Cell and Leukemia Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Tinisha McDonald
- Division of Hematopoietic Stem Cell and Leukemia Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Puneet Agarwal
- Division of Hematopoietic Stem Cell and Leukemia Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Allen Lin
- Division of Hematopoietic Stem Cell and Leukemia Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Su Chu
- Division of Hematopoietic Stem Cell and Leukemia Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Jing Qi
- Division of Hematopoietic Stem Cell and Leukemia Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Liang Li
- Division of Hematopoietic Stem Cell and Leukemia Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Yao-Te Hsieh
- Division of Hematopoietic Stem Cell and Leukemia Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Cedric Dos Santos
- Division of Hematopoietic Stem Cell and Leukemia Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Hongfeng Yuan
- Department of Cancer Biology, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Trung-Quang Ha
- Department of Clinical Science, Hematology Section, University of Bergen, Bergen 5021, Norway
| | | | - Randi Hovland
- Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen 5021, Norway
| | - Øystein Bruserud
- Department of Clinical Science, Hematology Section, University of Bergen, Bergen 5021, Norway; Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Bergen 5021, Norway
| | - Bjørn Tore Gjertsen
- Department of Clinical Science, Hematology Section, University of Bergen, Bergen 5021, Norway; Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Bergen 5021, Norway
| | - Ya-Huei Kuo
- Division of Hematopoietic Stem Cell and Leukemia Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Wenyong Chen
- Department of Cancer Biology, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Sonia Lain
- Karolinska Institutet, Stockholm 17177, Sweden
| | - Emmet McCormack
- Department of Clinical Science, Hematology Section, University of Bergen, Bergen 5021, Norway; Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Bergen 5021, Norway.
| | - Ravi Bhatia
- Division of Hematopoietic Stem Cell and Leukemia Research, City of Hope National Medical Center, Duarte, CA 91010, USA.
| |
Collapse
|
229
|
Herbert KJ, Holloway A, Cook AL, Chin SP, Snow ET. Arsenic exposure disrupts epigenetic regulation of SIRT1 in human keratinocytes. Toxicol Appl Pharmacol 2014; 281:136-45. [PMID: 25281835 DOI: 10.1016/j.taap.2014.09.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 09/09/2014] [Accepted: 09/22/2014] [Indexed: 02/08/2023]
Abstract
Arsenic is an environmental toxin which increases skin cancer risk for exposed populations worldwide; however the underlying biomolecular mechanism for arsenic-induced carcinogenesis is complex and poorly defined. Recent investigations show that histone deacetylase and DNA methyltransferase activity is impaired, and epigenetic patterns of gene regulation are consistently altered in cancers associated with arsenic exposure. Expression of the histone deacetylase SIRT1 is altered in solid tumours and haematological malignancies; however its role in arsenic-induced pathology is unknown. In this study we investigated the effect of arsenic on epigenetic regulation of SIRT1 and its targeting microRNA, miR-34a in primary human keratinocytes. Acetylation of histone H4 at lysine 16 (H4K16) increased in keratinocytes exposed to 0.5μM arsenite [As(III)]; and this was associated with chromatin remodelling at the miR-34a promoter. Moreover, although SIRT1 protein initially increased in these As(III)-exposed cells, after 24days expression was not significantly different from untreated controls. Extended exposure to low-dose As(III) (0.5μM; >5weeks) compromised the pattern of CpG methylation at SIRT1 and miR-34a gene promoters, and this was associated with altered expression for both genes. We have found that arsenic alters epigenetic regulation of SIRT1 expression via structural reorganisation of chromatin at the miR-34a gene promoter in the initial 24h of exposure; and over time, through shifts in miR-34a and SIRT1 gene methylation. Taken together, this investigation demonstrates that arsenic produces cumulative disruptions to epigenetic regulation of miR-34a expression, and this is associated with impaired coordination of SIRT1 functional activity.
Collapse
Affiliation(s)
- Katharine J Herbert
- School of Health Sciences, University of Tasmania, Launceston, TAS 7250, Australia
| | - Adele Holloway
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, TAS 7000, Australia
| | - Anthony L Cook
- School of Health Sciences, University of Tasmania, Launceston, TAS 7250, Australia
| | - Suyin P Chin
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, TAS 7000, Australia
| | - Elizabeth T Snow
- School of Health Sciences, University of Tasmania, Launceston, TAS 7250, Australia.
| |
Collapse
|
230
|
Ning Z, Wang A, Liang J, Xie Y, Liu J, Yan Q, Wang Z. USP22 promotes epithelial-mesenchymal transition via the FAK pathway in pancreatic cancer cells. Oncol Rep 2014; 32:1451-8. [PMID: 25070659 DOI: 10.3892/or.2014.3354] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2014] [Accepted: 06/27/2014] [Indexed: 11/06/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) contributes to the occurrence and development of tumors, particularly to the promotion of tumor invasion and metastasis. As a newly discovered ubiquitin hydrolase family member, USP22 plays a key role in the malignant transformation of tumors and the regulation of the cell cycle. However, recent studies on USP22 have primarily focused on its role in cell cycle regulation, and the potential mechanism underlying the promotion of tumor invasion and metastasis by abnormal USP22 expression has not been reported. Our studies revealed that the overexpression of USP22 in PANC-1 cells promoted Ezrin redistribution and phosphorylation and cytoskeletal remodeling, upregulated expression of the transcription factors Snail and ZEB1 to promote EMT, and increased cellular invasion and migration. In contrast, blockade of USP22 expression resulted in the opposite effects. In addition, the focal adhesion kinase (FAK) signaling pathway was shown to play a key role in the process of EMT induction in PANC-1 cells by USP22. Thus, the present study suggests that USP22 acts as a regulatory protein for EMT in pancreatic cancer, which may provide a new approach for the targeted therapy of pancreatic cancer.
Collapse
Affiliation(s)
- Zhen Ning
- Department of General Surgery, The First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, P.R. China
| | - Aman Wang
- Department of Oncology, The First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, P.R. China
| | - Jinxiao Liang
- Department of General Surgery, The First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, P.R. China
| | - Yunpeng Xie
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Liaoning Provincial Core Laboratory of Glycobiology and Glycoengineering, Dalian, Liaoning, P.R. China
| | - Jiwei Liu
- Department of Oncology, The First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, P.R. China
| | - Qiu Yan
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Liaoning Provincial Core Laboratory of Glycobiology and Glycoengineering, Dalian, Liaoning, P.R. China
| | - Zhongyu Wang
- Department of General Surgery, The First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, P.R. China
| |
Collapse
|
231
|
McCormick MA, Mason AG, Guyenet SJ, Dang W, Garza RM, Ting MK, Moller RM, Berger SL, Kaeberlein M, Pillus L, La Spada AR, Kennedy BK. The SAGA histone deubiquitinase module controls yeast replicative lifespan via Sir2 interaction. Cell Rep 2014; 8:477-86. [PMID: 25043177 DOI: 10.1016/j.celrep.2014.06.037] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 05/20/2014] [Accepted: 06/19/2014] [Indexed: 11/19/2022] Open
Abstract
We have analyzed the yeast replicative lifespan of a large number of open reading frame (ORF) deletions. Here, we report that strains lacking genes SGF73, SGF11, and UBP8 encoding SAGA/SLIK complex histone deubiquitinase module (DUBm) components are exceptionally long lived. Strains lacking other SAGA/SALSA components, including the acetyltransferase encoded by GCN5, are not long lived; however, these genes are required for the lifespan extension observed in DUBm deletions. Moreover, the SIR2-encoded histone deacetylase is required, and we document both a genetic and physical interaction between DUBm and Sir2. A series of studies assessing Sir2-dependent functions lead us to propose that DUBm strains are exceptionally long lived because they promote multiple prolongevity events, including reduced rDNA recombination and altered silencing of telomere-proximal genes. Given that ataxin-7, the human Sgf73 ortholog, causes the neurodegenerative disease spinocerebellar ataxia type 7, our findings indicate that the genetic and epigenetic interactions between DUBm and SIR2 will be relevant to neurodegeneration and aging.
Collapse
Affiliation(s)
- Mark A McCormick
- Buck Institute for Research on Aging, Novato, CA 94945, USA; Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Amanda G Mason
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA; Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Stephan J Guyenet
- Department of Medical Genetics, University of Washington, Seattle, WA 98195, USA
| | - Weiwei Dang
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Huffington Center on Aging, Baylor College of Medicine, Houston, TX 76798, USA
| | - Renee M Garza
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA; University of California, San Diego, Moores Cancer Center, La Jolla, CA 92093, USA
| | - Marc K Ting
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Rick M Moller
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Shelley L Berger
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Matt Kaeberlein
- Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Lorraine Pillus
- University of California, San Diego, Moores Cancer Center, La Jolla, CA 92093, USA; Department of Pathology, University of Washington, Seattle, WA 98195, USA
| | - Albert R La Spada
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA; Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Department of Medical Genetics, University of Washington, Seattle, WA 98195, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Department of Neuroscience, University of California, San Diego, La Jolla, CA 92093, USA; Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Brian K Kennedy
- Buck Institute for Research on Aging, Novato, CA 94945, USA; Department of Biochemistry, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
232
|
Ning Z, Wang A, Liang J, Xie Y, Liu J, Feng L, Yan Q, Wang Z. USP22 promotes the G1/S phase transition by upregulating FoxM1 expression via β-catenin nuclear localization and is associated with poor prognosis in stage II pancreatic ductal adenocarcinoma. Int J Oncol 2014; 45:1594-608. [PMID: 24993031 DOI: 10.3892/ijo.2014.2531] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 06/02/2014] [Indexed: 11/05/2022] Open
Abstract
Ubiquitin-specific protease 22 (USP22), a newly discovered member of ubiquitin hydrolase family, exhibits a critical function in cell cycle progression and tumorigenesis. The forkhead box M1 (FoxM1) transcription factor plays a crucial role in cell proliferation, differentiation and transformation. However, the expression and functions of USP22 in pancreatic ductal adenocarcinoma (PDA) and whether FoxM1 is involved in USP22-mediated cell cycle regulation have not been studied. We examined the expression of USP22 and FoxM1 in 136 stage II PDA tissues by immunohistochemistry. Clinical significance was analyzed by multivariate Cox regression analysis, Kaplan-Meier curves and log-rank test. RT-PCR, western blot analysis, luciferase and immunofluorescence assays were used to investigate the molecular function of USP22 and FoxM1 in PDA fresh tissues and cell lines. USP22 and FoxM1 were significantly upregulated in PDA tissues compared with the paired normal carcinoma-adjacent tissues. A statistical correlation was observed between USP22 and FoxM1 expression. The expression of USP/FoxM1 and co-expression of both factors correlated with tumor size, lymph node metastasis and overall survival. Multivariate Cox regression analysis revealed that the expression of USP22/FoxM1, especially the co-expression of both factors, is an independent, unfavorable prognostic factor. USP22 overexpression is accompanied by an increase in FoxM1 expression and USP22 increases FoxM1 expression to promote G1/S transition and cell proliferation through promoting β-catenin nuclear translocation in PDA cell lines. USP22 promotes the G1/S phase transition by upregulating FoxM1 expression via promoting β-catenin nuclear localization. USP22 and FoxM1 may act as prognostic markers and potential targets for PDA.
Collapse
Affiliation(s)
- Zhen Ning
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, P.R. China
| | - Aman Wang
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, P.R. China
| | - Jinxiao Liang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, P.R. China
| | - Yunpeng Xie
- Department of Pathology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, P.R. China
| | - Jiwei Liu
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, P.R. China
| | - Lu Feng
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian 116044, P.R. China
| | - Qiu Yan
- Department of Pathology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, P.R. China
| | - Zhongyu Wang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, P.R. China
| |
Collapse
|
233
|
Xiong J, Wang Y, Gong Z, Liu J, Li W. Identification of a functional nuclear localization signal within the human USP22 protein. Biochem Biophys Res Commun 2014; 449:14-8. [PMID: 24802393 DOI: 10.1016/j.bbrc.2014.04.133] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 04/25/2014] [Indexed: 01/30/2023]
Abstract
Ubiquitin-specific processing enzyme 22 (USP22), a member of the deubiquitinase family, is over-expressed in most human cancers and has been implicated in tumorigenesis. Because it is an enzymatic subunit of the human SAGA transcriptional cofactor, USP22 deubiquitylates histone H2A and H2B in the nucleus, thus participating in gene regulation and cell-cycle progression. However, the mechanisms regulating its nuclear translocation have not yet been elucidated. It was here demonstrated that USP22 is imported into the nucleus through a mechanism mediated by nuclear localization signal (NLS). The bipartite NLS sequence KRELELLKHNPKRRKIT (aa152-168), was identified as the functional NLS for its nuclear localization. Furthermore, a short cluster of basic amino acid residues KRRK within this bipartite NLS plays the primary role in nuclear localization and is evolutionarily conserved in USP22 homologues. In the present study, a functional NLS and the minimal sequences required for the active targeting of USP22 to the nucleus were identified. These findings may provide a molecular basis for the mechanism underlying USP22 nuclear trafficking and function.
Collapse
Affiliation(s)
- Jianjun Xiong
- Key Laboratory of Jiangxi Province for the Systems Bio-Medicine, Jiujiang, Jiangxi Province 332000, China; College of Basic Medical Science, Jiujiang University, Jiujiang, Jiangxi Province 332000, China
| | - Yaqin Wang
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China
| | - Zhen Gong
- Key Laboratory of Jiangxi Province for the Systems Bio-Medicine, Jiujiang, Jiangxi Province 332000, China
| | - Jianyun Liu
- College of Basic Medical Science, Jiujiang University, Jiujiang, Jiangxi Province 332000, China
| | - Weidong Li
- College of Basic Medical Science, Jiujiang University, Jiujiang, Jiangxi Province 332000, China.
| |
Collapse
|
234
|
Zhang J, Zhang X, Xie F, Zhang Z, van Dam H, Zhang L, Zhou F. The regulation of TGF-β/SMAD signaling by protein deubiquitination. Protein Cell 2014; 5:503-17. [PMID: 24756567 PMCID: PMC4085288 DOI: 10.1007/s13238-014-0058-8] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 03/28/2014] [Indexed: 01/17/2023] Open
Abstract
Transforming growth factor-β (TGF-β) members are key cytokines that control embryogenesis and tissue homeostasis via transmembrane TGF-β type II (TβR II) and type I (TβRI) and serine/threonine kinases receptors. Aberrant activation of TGF-β signaling leads to diseases, including cancer. In advanced cancer, the TGF-β/SMAD pathway can act as an oncogenic factor driving tumor cell invasion and metastasis, and thus is considered to be a therapeutic target. The activity of TGF-β/SMAD pathway is known to be regulated by ubiquitination at multiple levels. As ubiquitination is reversible, emerging studies have uncovered key roles for ubiquitin-removals on TGF-β signaling components by deubiquitinating enzymes (DUBs). In this paper, we summarize the latest findings on the DUBs that control the activity of the TGF-β signaling pathway. The regulatory roles of these DUBs as a driving force for cancer progression as well as their underlying working mechanisms are also discussed.
Collapse
Affiliation(s)
- Juan Zhang
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058 China
- Department of Molecular Cell Biology, Cancer Genomics Centre Netherlands and Centre of Biomedical Genetics, Leiden University Medical Center, Postbus 9600, 2300 RC Leiden, The Netherlands
| | - Xiaofei Zhang
- Department of Molecular Cell Biology, Cancer Genomics Centre Netherlands and Centre of Biomedical Genetics, Leiden University Medical Center, Postbus 9600, 2300 RC Leiden, The Netherlands
| | - Feng Xie
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058 China
| | - Zhengkui Zhang
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058 China
| | - Hans van Dam
- Department of Molecular Cell Biology, Cancer Genomics Centre Netherlands and Centre of Biomedical Genetics, Leiden University Medical Center, Postbus 9600, 2300 RC Leiden, The Netherlands
| | - Long Zhang
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058 China
- Department of Molecular Cell Biology, Cancer Genomics Centre Netherlands and Centre of Biomedical Genetics, Leiden University Medical Center, Postbus 9600, 2300 RC Leiden, The Netherlands
| | - Fangfang Zhou
- Department of Molecular Cell Biology, Cancer Genomics Centre Netherlands and Centre of Biomedical Genetics, Leiden University Medical Center, Postbus 9600, 2300 RC Leiden, The Netherlands
| |
Collapse
|
235
|
Belle JI, Nijnik A. H2A-DUBbing the mammalian epigenome: expanding frontiers for histone H2A deubiquitinating enzymes in cell biology and physiology. Int J Biochem Cell Biol 2014; 50:161-74. [PMID: 24647359 DOI: 10.1016/j.biocel.2014.03.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 03/05/2014] [Accepted: 03/07/2014] [Indexed: 12/16/2022]
Abstract
Posttranslational modifications of histone H2A through the attachment of ubiquitin or poly-ubiquitin conjugates are common in mammalian genomes and play an important role in the regulation of chromatin structure, gene expression, and DNA repair. Histone H2A deubiquitinases (H2A-DUBs) are a group of structurally diverse enzymes that catalyze the removal ubiquitin from histone H2A. In this review we provide a concise summary of the mechanisms that mediate histone H2A ubiquitination in mammalian cells, and review our current knowledge of mammalian H2A-DUBs, their biochemical activities, and recent developments in our understanding of their functions in mammalian physiology.
Collapse
Affiliation(s)
- Jad I Belle
- Department of Physiology, McGill University, Canada; Complex Traits Group, McGill University, Canada
| | - Anastasia Nijnik
- Department of Physiology, McGill University, Canada; Complex Traits Group, McGill University, Canada.
| |
Collapse
|
236
|
Gao Y, Lin F, Xu P, Nie J, Chen Z, Su J, Tang J, Wu Q, Li Y, Guo Z, Gao Z, Li D, Shen J, Ge S, Tsun A, Li B. USP22 is a positive regulator of NFATc2 on promoting IL2 expression. FEBS Lett 2014; 588:878-83. [PMID: 24561192 DOI: 10.1016/j.febslet.2014.02.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 02/02/2014] [Accepted: 02/06/2014] [Indexed: 11/25/2022]
Abstract
Nuclear factor of activated T cells (NFAT) is an important regulator of T cell activation. However, the molecular mechanism whereby NFATc2 regulates IL2 transcription is not fully understood. In this study, we showed that ubiquitin-specific protease 22 (USP22), known as a cancer stem cell marker, specifically interacted with and deubiquitinated NFATc2. USP22 stabilized NFATc2 protein levels, which required its deubiquitinase activity. Consistent with these observations, depletion of USP22 in T cells reduced the expression of IL2, which is a cytokine that signifies T effector cell activation. Our findings thus unveil a previously uncharacterized positive regulator of NFATc2, suggesting that targeting the deubiquitinase activity of USP22 could have therapeutic benefit to control IL2 expression and T cell function.
Collapse
Affiliation(s)
- Yayi Gao
- Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Fang Lin
- Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Peng Xu
- Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Jia Nie
- Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zuojia Chen
- Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jinsong Su
- Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Department of Surgery, Shanghai Public Health Center, Fudan University, Shanghai 201508, China
| | - Jiayou Tang
- Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Institute for Immunity and Transplantation, Shanghai East Hospital, Tongji University, Shanghai 200120, China
| | - Qingsi Wu
- Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Department of Immunology, Anhui Medical University, Hefei 230032, China
| | - Yangyang Li
- Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhixiang Guo
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Zhimei Gao
- Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Dan Li
- Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jijia Shen
- Department of Immunology, Anhui Medical University, Hefei 230032, China
| | - Shenglin Ge
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Andy Tsun
- Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Bin Li
- Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
237
|
Lin Z, Yang H, Tan C, Li J, Liu Z, Quan Q, Kong S, Ye J, Gao B, Fang D. USP10 antagonizes c-Myc transcriptional activation through SIRT6 stabilization to suppress tumor formation. Cell Rep 2013; 5:1639-49. [PMID: 24332849 DOI: 10.1016/j.celrep.2013.11.029] [Citation(s) in RCA: 156] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 09/13/2013] [Accepted: 11/14/2013] [Indexed: 11/17/2022] Open
Abstract
The reduced protein expression of SIRT6 tumor suppressor is involved in tumorigenesis. The molecular mechanisms underlying SIRT6 protein downregulation in human cancers remain unknown. Using a proteomic approach, we have identified the ubiquitin-specific peptidase USP10, another tumor suppressor, as one of the SIRT6-interacting proteins. USP10 suppresses SIRT6 ubiquitination to protect SIRT6 from proteasomal degradation. USP10 antagonizes the transcriptional activity of the c-Myc oncogene through SIRT6, as well as p53, to inhibit cell-cycle progression, cancer cell growth, and tumor formation. To support this conclusion, we detected significant reductions in both USP10 and SIRT6 protein expression in human colon cancers. Our study discovered crosstalk between two tumor-suppressive genes in regulating cell-cycle progression and proliferation and showed that dysregulated USP10 function promotes tumorigenesis through SIRT6 degradation.
Collapse
Affiliation(s)
- Zhenghong Lin
- Department of Pathology, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Heeyoung Yang
- Department of Pathology, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Can Tan
- Department of Pathology, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Jinping Li
- Department of Pathology, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Zhaojian Liu
- Department of Pathology, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Qiu Quan
- Department of Pathology, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Sinyi Kong
- Department of Pathology, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Junsheng Ye
- Department of Pathology, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Beixue Gao
- Department of Pathology, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA
| | - Deyu Fang
- Department of Pathology, Northwestern University Feinberg School of Medicine, 303 East Chicago Avenue, Chicago, IL 60611, USA.
| |
Collapse
|
238
|
Gurskiy DY, Kopytova DV, Georgieva SG, Nabirochkina EN. SAGA complex: Role in viability and development. Mol Biol 2013. [DOI: 10.1134/s0026893313060071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
239
|
Yang H, Lee SM, Gao B, Zhang J, Fang D. Histone deacetylase sirtuin 1 deacetylates IRF1 protein and programs dendritic cells to control Th17 protein differentiation during autoimmune inflammation. J Biol Chem 2013; 288:37256-66. [PMID: 24214980 DOI: 10.1074/jbc.m113.527531] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The type III histone deacetylase Sirt1 has recently emerged as a critical immune regulator by suppressing T cell immunity and macrophage activation during inflammation, but its role in dendritic cells (DCs) remains unknown. Here, we show that mice with genetic Sirt1 deletion specifically in DCs are resistant to MOG-induced experimental autoimmune encephalomyelitis. Loss of Sirt1 functions in DCs enhances their ability to produce IL-27 and interferon β (IFN-β). Co-cultivation of Sirt1-null DCs with CD4(+) T cells inhibited Th17 differentiation, which is reversed by anti-IL27 and anti-IFN-β neutralization antibodies. Sirt1 antagonizes acetylation of IRF1, a transcription factor that drives IL-27 production. Genetic deletion of IRF1 in Sirt1-null DCs abolishes IL-27 production and suppresses Th17 differentiation. Our results show that the histone deacetylase Sirt1 programs DCs to regulate Th17 differentiation during inflammation.
Collapse
Affiliation(s)
- Heeyoung Yang
- From the Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611 and
| | | | | | | | | |
Collapse
|
240
|
Schrecengost RS, Dean JL, Goodwin JF, Schiewer MJ, Urban MW, Stanek TJ, Sussman RT, Hicks JL, Birbe RC, Draganova-Tacheva RA, Visakorpi T, DeMarzo AM, McMahon SB, Knudsen KE. USP22 regulates oncogenic signaling pathways to drive lethal cancer progression. Cancer Res 2013; 74:272-86. [PMID: 24197134 DOI: 10.1158/0008-5472.can-13-1954] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Increasing evidence links deregulation of the ubiquitin-specific proteases 22 (USP22) deubitiquitylase to cancer development and progression in a select group of tumor types, but its specificity and underlying mechanisms of action are not well defined. Here we show that USP22 is a critical promoter of lethal tumor phenotypes that acts by modulating nuclear receptor and oncogenic signaling. In multiple xenograft models of human cancer, modeling of tumor-associated USP22 deregulation demonstrated that USP22 controls androgen receptor accumulation and signaling, and that it enhances expression of critical target genes coregulated by androgen receptor and MYC. USP22 not only reprogrammed androgen receptor function, but was sufficient to induce the transition to therapeutic resistance. Notably, in vivo depletion experiments revealed that USP22 is critical to maintain phenotypes associated with end-stage disease. This was a significant finding given clinical evidence that USP22 is highly deregulated in tumors, which have achieved therapeutic resistance. Taken together, our findings define USP22 as a critical effector of tumor progression, which drives lethal phenotypes, rationalizing this enzyme as an appealing therapeutic target to treat advanced disease.
Collapse
Affiliation(s)
- Randy S Schrecengost
- Authors' Affiliations: Departments of Cancer Biology, Urology, Radiation Oncology, Pathology, and Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania; Sidney Kimmel Comprehensive Cancer Center; Department of Pathology, Johns Hopkins University, Baltimore, Maryland; and Institute of Biomedical Technology, University of Tampere and Tampere University Hospital, Tampere, Finland
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
241
|
Clague MJ, Barsukov I, Coulson JM, Liu H, Rigden DJ, Urbé S. Deubiquitylases from genes to organism. Physiol Rev 2013; 93:1289-315. [PMID: 23899565 DOI: 10.1152/physrev.00002.2013] [Citation(s) in RCA: 354] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Ubiquitylation is a major posttranslational modification that controls most complex aspects of cell physiology. It is reversed through the action of a large family of deubiquitylating enzymes (DUBs) that are emerging as attractive therapeutic targets for a number of disease conditions. Here, we provide a comprehensive analysis of the complement of human DUBs, indicating structural motifs, typical cellular copy numbers, and tissue expression profiles. We discuss the means by which specificity is achieved and how DUB activity may be regulated. Generically DUB catalytic activity may be used to 1) maintain free ubiquitin levels, 2) rescue proteins from ubiquitin-mediated degradation, and 3) control the dynamics of ubiquitin-mediated signaling events. Functional roles of individual DUBs from each of five subfamilies in specific cellular processes are highlighted with an emphasis on those linked to pathological conditions where the association is supported by whole organism models. We then specifically consider the role of DUBs associated with protein degradative machineries and the influence of specific DUBs upon expression of receptors and channels at the plasma membrane.
Collapse
Affiliation(s)
- Michael J Clague
- Cellular and Molecular Physiology, Institute of Translational Medicine, and Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom.
| | | | | | | | | | | |
Collapse
|
242
|
Kemp KL, Lin Z, Zhao F, Gao B, Song J, Zhang K, Fang D. The serine-threonine kinase inositol-requiring enzyme 1α (IRE1α) promotes IL-4 production in T helper cells. J Biol Chem 2013; 288:33272-82. [PMID: 24100031 DOI: 10.1074/jbc.m113.493171] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The inositol-requiring enzyme 1α (IRE1α) is a serine-threonine kinase that plays crucial roles in activating the unfolded protein response. Studies suggest that IRE1α is activated during thymic T cell development and in effector CD8(+) T cells. However, its role in regulating T helper cell differentiation remains unknown. We find that IRE1α is up-regulated and activated upon CD4(+) T cell activation and plays an important role in promoting cytokine IL-4 production. CD4(+) T cells from IRE1α KO mice have reduced IL-4 protein expression, and this impaired IL-4 production is not due to the altered expression of Th2 lineage-specific transcription factors, such as GATA3. Instead, IL-4 mRNA stability is reduced in IRE1α KO T cells. Furthermore, treatment of T cells with an IRE1α-specific inhibitor, 4μ8C, leads to a block in IL-4, IL-5, and IL-13 production, confirming the role of IRE1α in the regulation of IL-4. This study identifies a regulatory function for IRE1α in the promotion of IL-4 in T cells.
Collapse
Affiliation(s)
- Kyeorda L Kemp
- From the Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611
| | | | | | | | | | | | | |
Collapse
|
243
|
Zou T, Yang Y, Xia F, Huang A, Gao X, Fang D, Xiong S, Zhang J. Resveratrol Inhibits CD4+ T cell activation by enhancing the expression and activity of Sirt1. PLoS One 2013; 8:e75139. [PMID: 24073240 PMCID: PMC3779207 DOI: 10.1371/journal.pone.0075139] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Accepted: 08/10/2013] [Indexed: 12/15/2022] Open
Abstract
Resveratrol, a natural polyphenol compound, has broad effects on critical events, including inflammation, oxidation, cancer and aging. However, the function and molecular mechanisms of resveratrol on T cell activation are controversial. In the present study, we found that resveratrol significantly inhibits the activation and cytokine production of T cells in vitro and in vivo. Sirt1 expression was up-regulated in resveratrol-treated T cells. Once Sirt1 was down-regulated in the T cells, the resveratrol-induced inhibition of T cell activation noticeably diminished. The acetylation of c-Jun decreased and its translocation was impeded in the resveratrol-treated T cells. The incidence and severity of collagen-induced arthritis in the resveratrol-treated mice were considerably reduced.
Collapse
Affiliation(s)
- Ting Zou
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province, People’s Republic of China
| | - Yi Yang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province, People’s Republic of China
| | - Fei Xia
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province, People’s Republic of China
| | - Anfei Huang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province, People’s Republic of China
| | - Xiaoming Gao
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province, People’s Republic of China
| | - Deyu Fang
- Department of Pathology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Sidong Xiong
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province, People’s Republic of China
- * E-mail: (JPZ); (SDX)
| | - Jinping Zhang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province, People’s Republic of China
- * E-mail: (JPZ); (SDX)
| |
Collapse
|
244
|
Abstract
The sirtuin family has emerged as important regulators of diverse physiological and pathological events, including life-span extension, neurodegeneration, age-related disorders, obesity, heart disease, inflammation, and cancer. In mammals, there are 7 members (SIRT1-SIRT7) in the sirtuin family, with the function of SIRT1 being extensively studied in the past decade. SIRT1 can deacetylate histones and a number of nonhistone substrates, which are involved in multiple signaling pathways. Numerous studies have suggested that SIRT1 could act as either a tumor suppressor or tumor promoter depending on its targets in specific signaling pathways or in specific cancers. This review highlights the major pathways regulated by SIRT1 involved in tumorigenesis.
Collapse
Affiliation(s)
- Zhenghong Lin
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | |
Collapse
|
245
|
Abstract
The cellular NAD(+)/NADH level controls Sir2 (silent information regulator 2) deacetylase activity in regulating aging in lower species. Much work has been put forth to identify ways to activate SIRT1, the mammalian ortholog of Sir2. The identification of p53 as a bona fide substrate of SIRT1 deacetylation has linked SIRT1 to a role in tumorigenesis. Here, we review the various SIRT1 endogenous and small molecular activators and inhibitors that regulate p53 acetylation and subsequent activation of p53 tumor suppression activity.
Collapse
|
246
|
McBurney MW, Clark-Knowles KV, Caron AZ, Gray DA. SIRT1 is a Highly Networked Protein That Mediates the Adaptation to Chronic Physiological Stress. Genes Cancer 2013; 4:125-34. [PMID: 24020004 DOI: 10.1177/1947601912474893] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
SIRT1 is a NAD(+)-dependent protein deacetylase that has a very large number of established protein substrates and an equally impressive list of biological functions thought to be regulated by its activity. Perhaps as notable is the remarkable number of points of conflict concerning the role of SIRT1 in biological processes. For example, evidence exists suggesting that SIRT1 is a tumor suppressor, is an oncogene, or has no effect on oncogenesis. Similarly, SIRT1 is variably reported to induce, inhibit, or have no effect on autophagy. We believe that the resolution of many conflicting results is possible by considering recent reports indicating that SIRT1 is an important hub interacting with a complex network of proteins that collectively regulate a wide variety of biological processes including cancer and autophagy. A number of the interacting proteins are themselves hubs that, like SIRT1, utilize intrinsically disordered regions for their promiscuous interactions. Many studies investigating SIRT1 function have been carried out on cell lines carrying undetermined numbers of alterations to the proteins comprising the SIRT1 network or on inbred mouse strains carrying fixed mutations affecting some of these proteins. Thus, the effects of modulating SIRT1 amount and/or activity are importantly determined by the genetic background of the cell (or the inbred strain of mice), and the effects attributed to SIRT1 are synthetic with the background of mutations and epigenetic differences between cells and organisms. Work on mice carrying alterations to the Sirt1 gene suggests that the network in which SIRT1 functions plays an important role in mediating physiological adaptation to various sources of chronic stress such as calorie restriction and calorie overload. Whether the catalytic activity of SIRT1 and the nuclear concentration of the co-factor, NAD(+), are responsible for modulating this activity remains to be determined. However, the effect of modulating SIRT1 activity must be interpreted in the context of the cell or tissue under investigation. Indeed, for SIRT1, we argue that context is everything.
Collapse
Affiliation(s)
- Michael W McBurney
- Program in Cancer Therapeutics, Ottawa Hospital Research Institute ; Department of Medicine, University of Ottawa, Ottawa, ON, Canada
| | | | | | | |
Collapse
|
247
|
Kong S, Yeung P, Fang D. The class III histone deacetylase sirtuin 1 in immune suppression and its therapeutic potential in rheumatoid arthritis. J Genet Genomics 2013; 40:347-54. [PMID: 23876775 PMCID: PMC4007159 DOI: 10.1016/j.jgg.2013.04.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Revised: 02/18/2013] [Accepted: 04/07/2013] [Indexed: 11/27/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic debilitating disease of the joints. Both the innate and adaptive immune responses participate in the development and progression of RA. While several therapeutic reagents, such as TNF-α agonists, have been successfully developed for the clinical use in the treatment of RA, more than half of the patients do not respond to anti-TNF therapy. Therefore, new therapeutic reagents are needed. Recent studies have shown that sirtuin 1 (Sirt1), a nicotinamide adenine dinucleotide (NAD)-dependent histone deacetylase, is a critical negative regulator of both the innate and adaptive immune response in mice, and its altered functions are likely to be involved in autoimmune diseases. Small molecules that modulate Sirt1 functions are potential therapeutic reagents for autoimmune inflammatory diseases. This review highlights the role of Sirt1 in immune regulation and RA.
Collapse
Affiliation(s)
- Sinyi Kong
- Department of Pathology, Northwestern University, Feinberg School of Medicine, 303 E Chicago Ave, Chicago, IL 60612, USA
| | - Pricilla Yeung
- Department of Pathology, Northwestern University, Feinberg School of Medicine, 303 E Chicago Ave, Chicago, IL 60612, USA
| | - Deyu Fang
- Department of Pathology, Northwestern University, Feinberg School of Medicine, 303 E Chicago Ave, Chicago, IL 60612, USA
| |
Collapse
|
248
|
Sussman RT, Stanek TJ, Esteso P, Gearhart JD, Knudsen KE, McMahon SB. The epigenetic modifier ubiquitin-specific protease 22 (USP22) regulates embryonic stem cell differentiation via transcriptional repression of sex-determining region Y-box 2 (SOX2). J Biol Chem 2013; 288:24234-46. [PMID: 23760504 DOI: 10.1074/jbc.m113.469783] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Pluripotent embryonic stem cells (ESCs) undergo self-renewal until stimulated to differentiate along specific lineage pathways. Many of the transcriptional networks that drive reprogramming of a self-renewing ESC to a differentiating cell have been identified. However, fundamental questions remain unanswered about the epigenetic programs that control these changes in gene expression. Here we report that the histone ubiquitin hydrolase ubiquitin-specific protease 22 (USP22) is a critical epigenetic modifier that controls this transition from self-renewal to differentiation. USP22 is induced as ESCs differentiate and is necessary for differentiation into all three germ layers. We further report that USP22 is a transcriptional repressor of the locus encoding the core pluripotency factor sex-determining region Y-box 2 (SOX2) in ESCs, and this repression is required for efficient differentiation. USP22 occupies the Sox2 promoter and hydrolyzes monoubiquitin from ubiquitylated histone H2B and blocks transcription of the Sox2 locus. Our study reveals an epigenetic mechanism that represses the core pluripotency transcriptional network in ESCs, allowing ESCs to transition from a state of self-renewal into lineage-specific differentiation programs.
Collapse
Affiliation(s)
- Robyn T Sussman
- Department of Cancer Biology, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | | | | | | | | | |
Collapse
|
249
|
Mialki RK, Zhao J, Wei J, Mallampalli DF, Zhao Y. Overexpression of USP14 protease reduces I-κB protein levels and increases cytokine release in lung epithelial cells. J Biol Chem 2013; 288:15437-41. [PMID: 23615914 DOI: 10.1074/jbc.c112.446682] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The ubiquitin-proteasome system is the major pathway of non-lysosomal intracellular protein degradation, playing an important role in a variety of cellular responses including cell division, proliferation, and apoptosis. Ubiquitin-specific protease 14 (USP14) is a component of proteasome regulatory subunit 19 S that regulates deubiquitinated proteins entering inside the proteasome core 20 S. The role of USP14 in protein degradation is still controversial. Several studies suggest that USP14 plays an inhibitory role in protein degradation. Here, in contrast, overexpression of USP14 induced I-κB degradation, which increased cytokine release in lung epithelial cells. Overexpression of HA-tagged USP14 (HA-USP14) reduced I-κB protein levels by increasing the I-κB degradation rate in mouse lung epithelial cells (MLE12). I-κB polyubiquitination was reduced in HA-USP14-overexpressed MLE12 cells, suggesting that USP14 regulates I-κB degradation by removing its ubiquitin chain, thus promoting the deubiquitinated I-κB degradation within the proteasome. Interestingly, we found that USP14 was associated with RelA, a binding partner of I-κB, suggesting that RelA is the linker between USP14 and I-κB. Lipopolysaccharide (LPS) treatment induced serine phosphorylation of USP14 as well as further reducing I-κB levels in HA-USP14-overexpressed MLE12 cells as compared with empty vector transfected cells. Further, overexpression of HA-USP14 increased the LPS-, TNFα-, or Escherichia coli-induced IL-8 release in human lung epithelial cells. This study suggests that USP14 removes the ubiquitin chain of I-κB, therefore inducing I-κB degradation and increasing cytokine release in lung epithelial cells.
Collapse
Affiliation(s)
- Rachel K Mialki
- Department of Medicine and the Acute Lung Injury Center of Excellence, the University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA
| | | | | | | | | |
Collapse
|
250
|
Satija YK, Bhardwaj A, Das S. A portrayal of E3 ubiquitin ligases and deubiquitylases in cancer. Int J Cancer 2013; 133:2759-68. [PMID: 23436247 DOI: 10.1002/ijc.28129] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Accepted: 02/14/2013] [Indexed: 02/03/2023]
Abstract
E3 ubiquitin ligases and deubiquitylating enzymes (DUBs) are the key components of ubiquitin proteasome system which plays a critical role in cellular protein homeostasis. Any shortcoming in their biological roles can lead to various diseases including cancer. The dynamic interplay between ubiquitylation and deubiquitylation determines the level and activity of several proteins including p53, which is crucial for cellular stress response and tumor suppression pathways. In this review, we describe the different types of E3 ubiquitin ligases including those targeting tumor suppressor p53, SCF ligases and RING type ligases and accentuate on biological functions of few important E3 ligases in the cellular regulatory networks. Tumor suppressor p53 level is tightly regulated by multiple E3 ligases including Mdm2, COP1, Pirh2, etc. SCF ubiquitin ligase complexes are key regulators of cell cycle and signal transduction. BRCA1 and VHL RING type ligases function as tumor suppressors and play an important role in DNA repair and hypoxia response respectively. Further, we discuss the biological consequences of deregulation of the E3 ligases and the implications for cancer development. We also describe deubiquitylases which reverse the process of ubiquitylation and regulate diverse cellular pathways including metabolism, cell cycle control and chromatin remodelling. As the E3 ubiquitin ligases and DUBs work in a substrate specific manner, an improved understanding of them can lead to better therapeutics for cancer.
Collapse
Affiliation(s)
- Yatendra Kumar Satija
- Molecular Oncology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | | | | |
Collapse
|