1
|
Gao Q, Li N, Pan Y, Chu P, Zhou Y, Jia H, Cheng Y, Xue G, Song J, Zhang Y, Zhu H, Sun J, Zhang B, Sun Z, Fang D. Hepatocyte growth factor promotes melanoma metastasis through ubiquitin-specific peptidase 22-mediated integrins upregulation. Cancer Lett 2024; 604:217196. [PMID: 39222676 PMCID: PMC11542356 DOI: 10.1016/j.canlet.2024.217196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 07/15/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Hepatocyte growth factor (HGF) plays a critical role in promoting tumor migration, invasion, and metastasis, partly by upregulating integrins. The molecular mechanisms behind how HGF facilitates integrin-mediated tumorigenesis are not fully understood. In this study, we demonstrate that the ubiquitin-specific peptidase 22 (USP22) is essential for HGF-induced melanoma metastasis. HGF treatment dramatically increased the expression of both USP22 and multiple integrin family members in particular ITGAV, ITGB3, and ITGA1. An unbiased analysis of the TCGA database reveals integrins as common downstream targets of both USP22 and HGF across multiple human cancer types. Notably, CRISPR-mediated deletion of USP22 completely eliminates HGF-induced integrin expression in melanoma cells. At the molecular level, USP22 acts as a bona fide deubiquitinase for Sp1, a transcription factor for the ITGAV, ITGB3, and ITGA1 genes. USP22 interacts with and inhibits Sp1 ubiquitination, protecting against Sp1 proteasomal degradation. Supporting this, immunohistology analysis detects a positive correlation among USP22, Sp1, and integrin αv in human melanoma tissues. This study identifies the death from the signature gene USP22 as a critical positive regulator for HGF-induced integrin expression by deubiquitinating the Sp1 transcription factor during melanoma metastasis.
Collapse
Affiliation(s)
- Qiong Gao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China; Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Na Li
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China
| | - Yujie Pan
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China
| | - Peng Chu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China
| | - Yuanzhang Zhou
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China
| | - Huijun Jia
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China
| | - Yang Cheng
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Guoqing Xue
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China
| | - Jiankun Song
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China
| | - Yue Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China
| | - Houyu Zhu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China
| | - Jia Sun
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China
| | - Bin Zhang
- Department of Medicine, Hematology/Oncology Division, Robert H. Lurie Comprehensive Cancer Center, USA
| | - Zhaolin Sun
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Dalian Medical University, Dalian, 116044, China.
| | - Deyu Fang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Department of Medicine, Hematology/Oncology Division, Robert H. Lurie Comprehensive Cancer Center, USA.
| |
Collapse
|
2
|
Dolapchiev LI, Gonzales KA, Cruz LR, Gagea M, Stevenson HL, Kwan SY, Beretta L. Gut Microbiome and Hepatic Transcriptomic Determinants of HCC Development in Mice with Metabolic Dysfunction-Associated Steatohepatitis. J Hepatocell Carcinoma 2024; 11:1891-1905. [PMID: 39372712 PMCID: PMC11456366 DOI: 10.2147/jhc.s485532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 09/18/2024] [Indexed: 10/08/2024] Open
Abstract
Purpose Hepatocellular carcinoma (HCC) related to metabolic dysfunction-associated steatotic liver disease (MASLD) is often diagnosed at a late stage, and its incidence is increasing. Predictive biomarkers are therefore needed to identify individuals at high risk of HCC. We aimed to characterize the gut microbiome and hepatic transcriptome associated with HCC development in female mice with hepatocyte-deletion of Pten (HepPten -). These mice present with large variations in HCC development, making them a powerful model for biomarker discovery. Methods & Results Sequencing of stool 16S and hepatic RNA was performed on a first set of mice. Among all liver histology parameters measured, the strongest association with microbiome composition changes was with the number of tumors detected at necropsy, followed by inflammation. The gut microbiome of mice with more than 2 tumors was enriched with Lachnospiraceae UCG and depleted of Palleniella intestinalis and Odoribacter. In contrast, hepatic transcriptomic changes were most strongly associated with tumor burden, followed by liver fibrosis. The 840 differentially expressed genes correlating with tumor burden were enriched in leukocyte extravasation and interleukin 10 receptor A (IL10RA) pathways. In addition, the abundance of Spp1-high epithelial cells is correlated with tumor burden. Association between tumor number and depletion of Palleniella intestinalis, and between tumor burden and circulating levels of C-X-C motif chemokine ligand 13 (CXCL13) and stem cell factor (SCF), was further validated in an independent set of mice. Conclusion We identified microbiome components contributing to liver carcinogenesis by inducing inflammation, and changes in hepatic gene expression and hepatic cells distribution that contribute to tumor growth. Such information can be highly valuable for the development of new prevention strategies as well as of new biomarkers for risk modeling in HCC.
Collapse
Affiliation(s)
- Lillian I Dolapchiev
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kristyn A Gonzales
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Lorenzo R Cruz
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mihai Gagea
- Department of Veterinary Medicine & Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Heather L Stevenson
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA
| | - Suet-Ying Kwan
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Laura Beretta
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
3
|
Liu Y, Li D, Zhang X, Xia S, Qu Y, Ling X, Li Y, Kong X, Zhang L, Cui CP, Li D. A protein sequence-based deep transfer learning framework for identifying human proteome-wide deubiquitinase-substrate interactions. Nat Commun 2024; 15:4519. [PMID: 38806474 PMCID: PMC11133436 DOI: 10.1038/s41467-024-48446-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 04/26/2024] [Indexed: 05/30/2024] Open
Abstract
Protein ubiquitination regulates a wide range of cellular processes. The degree of protein ubiquitination is determined by the delicate balance between ubiquitin ligase (E3)-mediated ubiquitination and deubiquitinase (DUB)-mediated deubiquitination. In comparison to the E3-substrate interactions, the DUB-substrate interactions (DSIs) remain insufficiently investigated. To address this challenge, we introduce a protein sequence-based ab initio method, TransDSI, which transfers proteome-scale evolutionary information to predict unknown DSIs despite inadequate training datasets. An explainable module is integrated to suggest the critical protein regions for DSIs while predicting DSIs. TransDSI outperforms multiple machine learning strategies against both cross-validation and independent test. Two predicted DUBs (USP11 and USP20) for FOXP3 are validated by "wet lab" experiments, along with two predicted substrates (AR and p53) for USP22. TransDSI provides new functional perspective on proteins by identifying regulatory DSIs, and offers clues for potential tumor drug target discovery and precision drug application.
Collapse
Affiliation(s)
- Yuan Liu
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Dianke Li
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
- State Key Laboratory of Farm Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Xin Zhang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Simin Xia
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Yingjie Qu
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Xinping Ling
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
- College of Life Sciences, Hebei University, Baoding, 071002, China
| | - Yang Li
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Xiangren Kong
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Lingqiang Zhang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Chun-Ping Cui
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China.
| | - Dong Li
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China.
| |
Collapse
|
4
|
Rayginia TP, Keerthana CK, Shifana SC, Pellissery MJ, Abhishek A, Anto RJ. Phytochemicals as Potential Lead Molecules against Hepatocellular Carcinoma. Curr Med Chem 2024; 31:5199-5221. [PMID: 38213177 DOI: 10.2174/0109298673275501231213063902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 10/31/2023] [Accepted: 11/16/2023] [Indexed: 01/13/2024]
Abstract
Hepatocellular carcinoma (HCC) is the most prevalent form of liver cancer, accounting for 85-90% of liver cancer cases and is a leading cause of cancer-related mortality worldwide. The major risk factors for HCC include hepatitis C and B viral infections, along with chronic liver diseases, such as cirrhosis, fibrosis, and non-alcoholic steatohepatitis associated with metabolic syndrome. Despite the advancements in modern medicine, there is a continuous rise in the annual global incidence rate of HCC, and it is estimated to reach >1 million cases by 2025. Emerging research in phytomedicine and chemotherapy has established the anti-cancer potential of phytochemicals, owing to their diverse biological activities. In this review, we report the major phytochemicals that have been explored in combating hepatocellular carcinoma and possess great potential to be used as an alternative or in conjunction with the existing HCC treatment modalities. An overview of the pre-clinical observations, mechanism of action and molecular targets of some of these phytochemicals is also incorporated.
Collapse
Affiliation(s)
- Tennyson Prakash Rayginia
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
- Department of Biotechnology, University of Kerala, Thiruvananthapuram, Kerala, 695011, India
| | - Chenicheri Kizhakkeveettil Keerthana
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
- Department of Biotechnology, University of Kerala, Thiruvananthapuram, Kerala, 695011, India
| | | | - Maria Joy Pellissery
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
| | - Ajmani Abhishek
- Molecular Bioassay Laboratory, Institute of Advanced Virology, Thiruvananthapuram, Kerala, 695317, India
| | - Ruby John Anto
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
- Molecular Bioassay Laboratory, Institute of Advanced Virology, Thiruvananthapuram, Kerala, 695317, India
| |
Collapse
|
5
|
Friebus-Kardash J, Christ TC, Dietlein N, Elwy A, Abdelrahman H, Holnsteiner L, Hu Z, Rodewald HR, Lang KS. Usp22 Deficiency Leads to Downregulation of PD-L1 and Pathological Activation of CD8 + T Cells and Causes Immunopathology in Response to Acute LCMV Infection. Vaccines (Basel) 2023; 11:1563. [PMID: 37896966 PMCID: PMC10610587 DOI: 10.3390/vaccines11101563] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 09/07/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
Ubiquitin-specific peptidase 22 (Usp22) cleaves ubiquitin moieties from numerous proteins, including histone H2B and transcription factors. Recently, it was reported that Usp22 acts as a negative regulator of interferon-dependent responses. In the current study, we investigated the role of Usp22 deficiency in acute viral infection with lymphocytic choriomeningitis virus (LCMV). We found that the lack of Usp22 on bone marrow-derived cells (Usp22fl/fl Vav1-Cre mice) reduced the induction of type I and II interferons. A limited type I interferon response did not influence virus replication. However, restricted expression of PD-L1 led to increased frequencies of functional virus-specific CD8+ T cells and rapid death of Usp22-deficient mice. CD8+ T cell depletion experiments revealed that accelerated CD8+ T cells were responsible for enhanced lethality in Usp22 deficient mice. In conclusion, we found that the lack of Usp22 generated a pathological CD8+ T cell response, which gave rise to severe disease in mice.
Collapse
Affiliation(s)
- Justa Friebus-Kardash
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, 45147 Essen, Germany; (T.C.C.); (A.E.); (H.A.); (L.H.); (Z.H.); (K.S.L.)
- Department of Nephrology, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
| | - Theresa Charlotte Christ
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, 45147 Essen, Germany; (T.C.C.); (A.E.); (H.A.); (L.H.); (Z.H.); (K.S.L.)
| | - Nikolaus Dietlein
- Division of Cellular Immunology, German Cancer Research Center, 69120 Heidelberg, Germany; (N.D.)
| | - Abdelrahman Elwy
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, 45147 Essen, Germany; (T.C.C.); (A.E.); (H.A.); (L.H.); (Z.H.); (K.S.L.)
| | - Hossam Abdelrahman
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, 45147 Essen, Germany; (T.C.C.); (A.E.); (H.A.); (L.H.); (Z.H.); (K.S.L.)
| | - Lisa Holnsteiner
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, 45147 Essen, Germany; (T.C.C.); (A.E.); (H.A.); (L.H.); (Z.H.); (K.S.L.)
| | - Zhongwen Hu
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, 45147 Essen, Germany; (T.C.C.); (A.E.); (H.A.); (L.H.); (Z.H.); (K.S.L.)
| | - Hans-Reimer Rodewald
- Division of Cellular Immunology, German Cancer Research Center, 69120 Heidelberg, Germany; (N.D.)
| | - Karl Sebastian Lang
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, 45147 Essen, Germany; (T.C.C.); (A.E.); (H.A.); (L.H.); (Z.H.); (K.S.L.)
| |
Collapse
|
6
|
Di Q, Zhao X, Tang H, Li X, Xiao Y, Wu H, Wu Z, Quan J, Chen W. USP22 suppresses the NLRP3 inflammasome by degrading NLRP3 via ATG5-dependent autophagy. Autophagy 2023; 19:873-885. [PMID: 35900990 PMCID: PMC9980574 DOI: 10.1080/15548627.2022.2107314] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 07/24/2022] [Accepted: 07/25/2022] [Indexed: 11/02/2022] Open
Abstract
The NLRP3 inflammasome is involved in a diverse range of inflammatory diseases. The activation of inflammasomes must be tightly regulated to prevent excessive inflammation, and the protein ubiquitination system is reported to be one of the ways in which inflammasome activation is regulated. However, the deubiquitination regulatory mechanisms of inflammasome activation remain elusive. Here, we demonstrated that USP22 (ubiquitin specific peptidase 22) promotes NLRP3 degradation and inhibits NLRP3 inflammasome activation. USP22 deficiency or in vivo silencing significantly increases alum-induced peritonitis and lipopolysaccharide-induced systemic inflammation. Mechanistically, USP22 inhibits NLRP3 inflammasome activation via the promotion of ATG5-mediated macroautophagy/autophagy. USP22 stabilizes ATG5 via decreasing K27- and K48-linked ubiquitination of ATG5 at the Lys118 site. Taken together, these findings reveal the role USP22 plays in the regulation of NLRP3 inflammasome activation and suggest a potential therapeutic target to treat NLRP3 inflammasome-related diseases.Abbreviations: ATG5: autophagy related 5; ATP: adenosine triphosphate; CASP1: caspase 1; IL18: interleukin 18; IL1B/IL-1β: interleukin 1 beta; LPS: lipopolysaccharide; NLRC4: NLR family, CARD domain containing 4; NLRP3: NLR family, pyrin domain containing 3; PYCARD/ASC: PYD and CARD domain containing; TNF/TNF-α: tumor necrosis factor; USP22: ubiquitin specific peptidase 22.
Collapse
Affiliation(s)
- Qianqian Di
- Guangdong Provincial Key Laboratory for Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| | - Xibao Zhao
- Guangdong Provincial Key Laboratory for Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| | - Haimei Tang
- Guangdong Provincial Key Laboratory for Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| | - Xunwei Li
- Guangdong Provincial Key Laboratory for Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| | - Yue Xiao
- Guangdong Provincial Key Laboratory for Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| | - Han Wu
- Guangdong Provincial Key Laboratory for Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| | - Zherui Wu
- Guangdong Provincial Key Laboratory for Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| | - Jiazheng Quan
- Guangdong Provincial Key Laboratory for Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| | - Weilin Chen
- Guangdong Provincial Key Laboratory for Regional Immunity and Diseases, Department of Immunology, Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| |
Collapse
|
7
|
Gutiérrez-Cuevas J, Lucano-Landeros S, López-Cifuentes D, Santos A, Armendariz-Borunda J. Epidemiologic, Genetic, Pathogenic, Metabolic, Epigenetic Aspects Involved in NASH-HCC: Current Therapeutic Strategies. Cancers (Basel) 2022; 15:23. [PMID: 36612019 PMCID: PMC9818030 DOI: 10.3390/cancers15010023] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver cancer and is the sixth most frequent cancer in the world, being the third cause of cancer-related deaths. Nonalcoholic steatohepatitis (NASH) is characterized by fatty infiltration, oxidative stress and necroinflammation of the liver, with or without fibrosis, which can progress to advanced liver fibrosis, cirrhosis and HCC. Obesity, metabolic syndrome, insulin resistance, and diabetes exacerbates the course of NASH, which elevate the risk of HCC. The growing prevalence of obesity are related with increasing incidence of NASH, which may play a growing role in HCC epidemiology worldwide. In addition, HCC initiation and progression is driven by reprogramming of metabolism, which indicates growing appreciation of metabolism in the pathogenesis of this disease. Although no specific preventive pharmacological treatments have recommended for NASH, dietary restriction and exercise are recommended. This review focuses on the molecular connections between HCC and NASH, including genetic and risk factors, highlighting the metabolic reprogramming and aberrant epigenetic alterations in the development of HCC in NASH. Current therapeutic aspects of NASH/HCC are also reviewed.
Collapse
Affiliation(s)
- Jorge Gutiérrez-Cuevas
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, University of Guadalajara, CUCS, Guadalajara 44340, Jalisco, Mexico
| | - Silvia Lucano-Landeros
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, University of Guadalajara, CUCS, Guadalajara 44340, Jalisco, Mexico
| | - Daniel López-Cifuentes
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, University of Guadalajara, CUCS, Guadalajara 44340, Jalisco, Mexico
| | - Arturo Santos
- Tecnologico de Monterrey, EMCS, Campus Guadalajara, Zapopan 45201, Jalisco, Mexico
| | - Juan Armendariz-Borunda
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, University of Guadalajara, CUCS, Guadalajara 44340, Jalisco, Mexico
- Tecnologico de Monterrey, EMCS, Campus Guadalajara, Zapopan 45201, Jalisco, Mexico
| |
Collapse
|
8
|
Guo J, Zhao J, Fu W, Xu Q, Huang D. Immune Evasion and Drug Resistance Mediated by USP22 in Cancer: Novel Targets and Mechanisms. Front Immunol 2022; 13:918314. [PMID: 35935969 PMCID: PMC9347222 DOI: 10.3389/fimmu.2022.918314] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
Regulation of ubiquitination is involved in various processes in cancer occurrence and development, including cell cycle arrest, cell proliferation, apoptosis, invasion, metastasis, and immunity. Ubiquitination plays an important role not only at the transcriptional and post-translational levels but also at the protein level. When ubiquitination is in a pathological state, abnormally activated biological processes will not only induce cancer progression but also induce immune evasion. The main function of deubiquitinases (DUBs) is to remove ubiquitin chains from substrates, changing the biological activity of the substrates. It has great potential to improve the prognosis of cancer by targeting DUB to regulate proteome. Ubiquitin-specific peptidase 22 (USP22) belongs to the ubiquitin-specific protease (USP) family of DUBs and has been reported to be related to various physiological and pathological processes. USP22 is abnormally expressed in various malignant tumors such as prostate cancer, lung cancer, liver cancer, and colorectal cancer, which suggests that USP22 may play an important role in tumors. USP22 may stabilize programmed death ligand 1 (PD-L1) by deubiquitination while also regulating T-cell infiltration into tumors. Regulatory T cells (Tregs) are a unique class of immunosuppressive CD4+ T cells that primarily suppress the immune system by expressing the master transcription factor forkhead box protein 3 (FOXP3). USP22 was found to be a positive regulator of stable FOXP3 expression. Treg-specific ablation of USP22 leads to reduced tumor volume in multiple cancer models. This suggests that USP22 may regulate tumor resistance to immunotherapy. In this article, we review and summarize the biological functions of USP22 in multiple signal transduction pathways during tumorigenesis, immune evasion, and drug resistance. Furthermore, we propose a new possibility of combining USP22 with chemotherapeutic, targeted, and immunosuppressive drugs in the treatment of cancer.
Collapse
Affiliation(s)
- Jinhui Guo
- Qingdao Medical College, Qingdao University, Qingdao, China
- Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Jie Zhao
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Wen Fu
- Qingdao Medical College, Qingdao University, Qingdao, China
| | - Qiuran Xu
- Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Dongsheng Huang
- Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
9
|
Braghini MR, Lo Re O, Romito I, Fernandez-Barrena MG, Barbaro B, Pomella S, Rota R, Vinciguerra M, Avila MA, Alisi A. Epigenetic remodelling in human hepatocellular carcinoma. J Exp Clin Cancer Res 2022; 41:107. [PMID: 35331312 PMCID: PMC8943959 DOI: 10.1186/s13046-022-02297-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 02/19/2022] [Indexed: 04/13/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most frequent primary liver cancer, being the sixth most commonly diagnosed cancer and the fourth leading cause of cancer-related death. As other heterogeneous solid tumours, HCC results from a unique synergistic combination of genetic alterations mixed with epigenetic modifications.In HCC the patterns and frequencies of somatic variations change depending on the nearby chromatin. On the other hand, epigenetic alterations often induce genomic instability prone to mutations. Epigenetics refers to heritable states of gene expression without alteration to the DNA sequence itself and, unlike genetic changes, the epigenetic modifications are reversible and affect gene expression more extensively than genetic changes. Thus, studies of epigenetic regulation and the involved molecular machinery are greatly contributing to the understanding of the mechanisms that underline HCC onset and heterogeneity. Moreover, this knowledge may help to identify biomarkers for HCC diagnosis and prognosis, as well as future new targets for more efficacious therapeutic approaches.In this comprehensive review we will discuss the state-of-the-art knowledge about the epigenetic landscape in hepatocarcinogenesis, including evidence on the diagnostic and prognostic role of non-coding RNAs, modifications occurring at the chromatin level, and their role in the era of precision medicine.Apart from other better-known risk factors that predispose to the development of HCC, characterization of the epigenetic remodelling that occurs during hepatocarcinogenesis could open the way to the identification of personalized biomarkers. It may also enable a more accurate diagnosis and stratification of patients, and the discovery of new targets for more efficient therapeutic approaches.
Collapse
Affiliation(s)
- Maria Rita Braghini
- Unit of Molecular Genetics of Complex Phenotypes, Bambino Gesù Children's Hospital, IRCCS, Viale S. Paolo, 15, 00146, Rome, Italy
| | - Oriana Lo Re
- Department of Translational Stem Cell Biology, Research Institute of the Medical University of Varna, Varna, Bulgaria
| | - Ilaria Romito
- Unit of Molecular Genetics of Complex Phenotypes, Bambino Gesù Children's Hospital, IRCCS, Viale S. Paolo, 15, 00146, Rome, Italy
| | - Maite G Fernandez-Barrena
- Hepatology Program, CIMA, University of Navarra, Pamplona, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Barbara Barbaro
- Unit of Molecular Genetics of Complex Phenotypes, Bambino Gesù Children's Hospital, IRCCS, Viale S. Paolo, 15, 00146, Rome, Italy
| | - Silvia Pomella
- Department of Paediatric Haematology/Oncology and Cellular and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Rossella Rota
- Department of Paediatric Haematology/Oncology and Cellular and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Manlio Vinciguerra
- Department of Translational Stem Cell Biology, Research Institute of the Medical University of Varna, Varna, Bulgaria
| | - Matias A Avila
- Hepatology Program, CIMA, University of Navarra, Pamplona, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Anna Alisi
- Unit of Molecular Genetics of Complex Phenotypes, Bambino Gesù Children's Hospital, IRCCS, Viale S. Paolo, 15, 00146, Rome, Italy.
| |
Collapse
|
10
|
Chang YS, Su CW, Chen SC, Chen YY, Liang YJ, Wu JC. Upregulation of USP22 and ABCC1 during Sorafenib Treatment of Hepatocellular Carcinoma Contribute to Development of Resistance. Cells 2022; 11:cells11040634. [PMID: 35203285 PMCID: PMC8870465 DOI: 10.3390/cells11040634] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/28/2022] [Accepted: 02/10/2022] [Indexed: 12/10/2022] Open
Abstract
Sorafenib is a small molecule that blocks tumor proliferation by targeting the activity of multi-kinases for the treatment of advanced hepatocellular carcinoma (HCC). Increasing sorafenib resistance following long-term treatment is frequently encountered. Mechanisms underlying sorafenib resistance remain not completely clear. To further understand the mechanism of sorafenib resistance in HCC, we established sorafenib-resistant cell lines by slowly increasing sorafenib concentration in cell culture medium. Upregulation of USP22 and ABCC1 were found in Sorafenib-resistant cells. Sorafenib-resistant cells treated with USP22 siRNA showed significant reduction in endogenous mRNA and protein levels of ABCC1. During sorafenib treatment, upregulation of USP22 increases ABCC1 expression and subsequently contributes to sorafenib resistance in HCC cells. Immunohistochemical analysis revealed a positive correlation between USP22 and ABCC1 expression in tissue samples from sorafenib-resistant patients (Pearson’s correlation = 0.59, p = 0.03). Our findings indicate that upregulation of USP22 and ABCC1 expression during treatment contribute to sorafenib resistance in HCC cells and that USP22 has strong potential as a therapeutic target for overcoming sorafenib resistance in HCC patients.
Collapse
Affiliation(s)
- Yung-Sheng Chang
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; (Y.-S.C.); (S.-C.C.)
| | - Chien-Wei Su
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan;
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - San-Chi Chen
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; (Y.-S.C.); (S.-C.C.)
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan;
- Department of Oncology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Yen-Ying Chen
- Department of Pathology, Taipei Veterans General Hospital, Taipei 11217, Taiwan;
| | - Yuh-Jin Liang
- Medical Research Department, Taipei Veterans General Hospital, Taipei 11217, Taiwan;
| | - Jaw-Ching Wu
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; (Y.-S.C.); (S.-C.C.)
- Medical Research Department, Taipei Veterans General Hospital, Taipei 11217, Taiwan;
- Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
- Correspondence: ; Tel.: +886-2-28712121 (ext. 3218)
| |
Collapse
|
11
|
Tian Y, Tang B, Wang C, Wang Y, Mao J, Yao Y, Gao Z, Liang R, Ye M, Cai S, Wang L. Operative ubiquitin-specific protease 22 deubiquitination confers a more invasive phenotype to cholangiocarcinoma. Cell Death Dis 2021; 12:678. [PMID: 34226501 PMCID: PMC8257691 DOI: 10.1038/s41419-021-03940-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 06/14/2021] [Accepted: 06/15/2021] [Indexed: 11/22/2022]
Abstract
Oncogenic ubiquitin-specific protease 22 (USP22) is implicated in a variety of tumours; however, evidence of its role and underlying molecular mechanisms in cholangiocarcinoma (CCA) development remains unknown. We collected paired tumour and adjacent non-tumour tissues from 57 intrahepatic CCA (iCCA) patients and evaluated levels of the USP22 gene and protein by qPCR and immunohistochemistry. Both the mRNA and protein were significantly upregulated, correlated with the malignant invasion and worse OS of iCCA. In cell cultures, USP22 overexpression increased CCA cell proliferation and mobility, and induced epithelial-to-mesenchymal transition (EMT). Upon an interaction, USP22 deubiquitinated and stabilized sirtuin-1 (SIRT1), in conjunction with Akt/ERK activation. In implantation xenografts, USP22 overexpression stimulated tumour growth and metastasis to the lungs of mice. Conversely, the knockdown by USP22 shRNA attenuated the tumour growth and invasiveness in vitro and in vivo. Furthermore, SIRT1 overexpression reversed the USP22 functional deficiency, while the knockdown acetylated TGF-β-activated kinase 1 (TAK1) and Akt. Our present study defines USP22 as a poor prognostic predictor in iCCA that cooperates with SIRT1 and facilitates tumour development.
Collapse
Affiliation(s)
- Yu Tian
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Second Hospital of Dalian Medical University, Dalian, Liaoning, PR China
- Division of Vascular Surgery, Department of Surgery, The Second Hospital of Dalian Medical University, Dalian, Liaoning, PR China
- National Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, PR China
| | - Bo Tang
- Department of Health Sciences, Hiroshima Shudo University, Hiroshima, Japan.
| | - Chengye Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Second Hospital of Dalian Medical University, Dalian, Liaoning, PR China
- National Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, PR China
| | - Yan Wang
- National Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, PR China
| | - Jiakai Mao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Second Hospital of Dalian Medical University, Dalian, Liaoning, PR China
| | - Yifan Yao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Second Hospital of Dalian Medical University, Dalian, Liaoning, PR China
| | - Zhenming Gao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Second Hospital of Dalian Medical University, Dalian, Liaoning, PR China
| | - Rui Liang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Second Hospital of Dalian Medical University, Dalian, Liaoning, PR China
| | - Mingliang Ye
- National Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, PR China
| | - Shijie Cai
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK.
| | - Liming Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Second Hospital of Dalian Medical University, Dalian, Liaoning, PR China.
| |
Collapse
|
12
|
Li M, Xu Y, Liang J, Lin H, Qi X, Li F, Han P, Gao Y, Yang X. USP22 deficiency in melanoma mediates resistance to T cells through IFNγ-JAK1-STAT1 signal axis. Mol Ther 2021; 29:2108-2120. [PMID: 33601053 DOI: 10.1016/j.ymthe.2021.02.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 10/12/2020] [Accepted: 02/11/2021] [Indexed: 12/15/2022] Open
Abstract
Genome-wide clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated 9 (Cas9)-mediated loss-of-function screens are powerful tools for identifying genes responsible for diverse phenotypes. Here, we perturbed genes in melanoma cells to screen for genes involved in tumor escape from T cell-mediated killing. Multiple interferon gamma (IFNγ) signaling-related genes were enriched in melanoma cells resistant to T cell killing. In addition, deletion of the deubiquitinating protease ubiquitin specific peptidase 22 (USP22) in mouse melanoma (B16-OVA) cells decreased the efficacy of T cell-mediated killing, both in vitro and in vivo, while overexpression enhanced tumor-cell sensitivity to T (OT-I) cell-mediated killing. USP22 deficiency in both mouse and human melanoma cells showed impaired sensitivity to interferon pathway and USP22 was positively correlated with key molecules of interferon pathway in clinical melanoma samples. Mechanistically, USP22 may directly interact with signal transducer and activator of transcription 1 (STAT1), deubiquitinate it, and improve its stability in both human and mouse melanoma cells. Our findings identified a previously unknown function of USP22 and linked the loss of genes in tumor cells that are essential for escaping the effector function of CD8+ T cells during immunotherapy.
Collapse
Affiliation(s)
- Min Li
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, People's Republic of China; Joint International Research Laboratory of Metabolic & Developmental Sciences, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Yanqin Xu
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, People's Republic of China; Joint International Research Laboratory of Metabolic & Developmental Sciences, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Jie Liang
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Hao Lin
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Xinyue Qi
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Fanlin Li
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Ping Han
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Yanfeng Gao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, People's Republic of China
| | - Xuanming Yang
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, People's Republic of China; Joint International Research Laboratory of Metabolic & Developmental Sciences, Shanghai Jiao Tong University, Shanghai, People's Republic of China.
| |
Collapse
|
13
|
Roedig J, Kowald L, Juretschke T, Karlowitz R, Ahangarian Abhari B, Roedig H, Fulda S, Beli P, van Wijk SJL. USP22 controls necroptosis by regulating receptor-interacting protein kinase 3 ubiquitination. EMBO Rep 2021; 22:e50163. [PMID: 33369872 PMCID: PMC7857539 DOI: 10.15252/embr.202050163] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 10/21/2020] [Accepted: 11/03/2020] [Indexed: 12/27/2022] Open
Abstract
Dynamic control of ubiquitination by deubiquitinating enzymes is essential for almost all biological processes. Ubiquitin-specific peptidase 22 (USP22) is part of the SAGA complex and catalyzes the removal of mono-ubiquitination from histones H2A and H2B, thereby regulating gene transcription. However, novel roles for USP22 have emerged recently, such as tumor development and cell death. Apart from apoptosis, the relevance of USP22 in other programmed cell death pathways still remains unclear. Here, we describe a novel role for USP22 in controlling necroptotic cell death in human tumor cell lines. Loss of USP22 expression significantly delays TNFα/Smac mimetic/zVAD.fmk (TBZ)-induced necroptosis, without affecting TNFα-mediated NF-κB activation or extrinsic apoptosis. Ubiquitin remnant profiling identified receptor-interacting protein kinase 3 (RIPK3) lysines 42, 351, and 518 as novel, USP22-regulated ubiquitination sites during necroptosis. Importantly, mutation of RIPK3 K518 reduced necroptosis-associated RIPK3 ubiquitination and amplified necrosome formation and necroptotic cell death. In conclusion, we identify a novel role of USP22 in necroptosis and further elucidate the relevance of RIPK3 ubiquitination as crucial regulator of necroptotic cell death.
Collapse
Affiliation(s)
- Jens Roedig
- Institute for Experimental Cancer Research in PediatricsGoethe‐UniversityFrankfurt am MainGermany
| | - Lisa Kowald
- Institute for Experimental Cancer Research in PediatricsGoethe‐UniversityFrankfurt am MainGermany
| | | | - Rebekka Karlowitz
- Institute for Experimental Cancer Research in PediatricsGoethe‐UniversityFrankfurt am MainGermany
| | - Behnaz Ahangarian Abhari
- Lighthouse Core FacilityZentrum für Translationale ZellforschungUniversitaetsklinikum FreiburgKlinik für Innere Medizin IFreiburgGermany
| | - Heiko Roedig
- Pharmazentrum FrankfurtInstitut für Allgemeine Pharmakologie und ToxikologieGoethe‐UniversityFrankfurt am MainGermany
| | - Simone Fulda
- Institute for Experimental Cancer Research in PediatricsGoethe‐UniversityFrankfurt am MainGermany
| | - Petra Beli
- Institute of Molecular Biology (IMB)MainzGermany
| | - Sjoerd JL van Wijk
- Institute for Experimental Cancer Research in PediatricsGoethe‐UniversityFrankfurt am MainGermany
| |
Collapse
|
14
|
Wang S, Zhong X, Wang C, Luo H, Lin L, Sun H, Sun G, Zeng K, Zou R, Liu W, Sun N, Song H, Liu W, Zhang Q, Liao Z, Teng X, Zhou T, Sun X, Zhao Y. USP22 positively modulates ERα action via its deubiquitinase activity in breast cancer. Cell Death Differ 2020; 27:3131-3145. [PMID: 32494025 DOI: 10.1038/s41418-020-0568-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 12/19/2022] Open
Abstract
Estrogen receptor α (ERα) is the crucial factor in ERα-positive breast cancer progression. Endocrine therapies targeting ERα signaling is one of the widely used therapeutic strategies for breast cancer. However, a large number of the patients become refractory to therapy. Abnormal expression of ERα co-regulator facilitates breast cancer development and tendency of endocrine resistance. Thus, it is necessary to discover the novel co-regulators modulating ERα action. Here, we demonstrate that histone deubiquitinase USP22 is highly expressed in breast cancer samples compared with that in the benign tissue, and high expression of USP22 was significantly associated with poorer overall survival in BCa samples. Moreover, USP22 associates with ERα to be involved in maintenance of ERα stability. USP22 enhances ERα-induced transactivation. We further provide the evidence that USP22 is recruited together with ERα to cis-regulatory elements of ERα target gene. USP22 promotes cell growth even under hypoxia condition and with the treatment of ERα antagonist in breast cancer cells. Importantly, the deubiquitination activity of USP22 is required for its functions on maintenance of ERα stability, thereby enhancing ERα action and conferring endocrine resistance in breast cancer.
Collapse
Affiliation(s)
- Shengli Wang
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Xinping Zhong
- Department of General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, 110001, Liaoning, China
| | - Chunyu Wang
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Hao Luo
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Lin Lin
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Hongmiao Sun
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Ge Sun
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Kai Zeng
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Renlong Zou
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Wei Liu
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Ning Sun
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Huijuan Song
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Wensu Liu
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Qiang Zhang
- Department of mammary gland, LiaoNing Tumor Hospital & Institute, Shenyang, 110042, Liaoning, China
| | - Zhixuan Liao
- Department of mammary gland, LiaoNing Tumor Hospital & Institute, Shenyang, 110042, Liaoning, China
| | - Xiaochun Teng
- Department of Endocrinology and Metabolism, Institute of Endocrinology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, Liaoning, China
| | - Tingting Zhou
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Xun Sun
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, 110122, Liaoning, China
| | - Yue Zhao
- Department of Cell Biology, Key laboratory of Cell Biology, Ministry of Public Health, and Key laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang, 110122, Liaoning, China. .,Department of Endocrinology and Metabolism, Institute of Endocrinology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, Liaoning, China.
| |
Collapse
|
15
|
Higurashi M, Maruyama T, Nogami Y, Ishikawa F, Yoshida Y, Mori K, Fujita KI, Shibanuma M. High expression of FOXM1 critical for sustaining cell proliferation in mitochondrial DNA-less liver cancer cells. Exp Cell Res 2020; 389:111889. [PMID: 32032602 DOI: 10.1016/j.yexcr.2020.111889] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 01/06/2020] [Accepted: 02/03/2020] [Indexed: 12/16/2022]
Abstract
The copy number of mitochondrial DNA (mtDNA) is decreased in most cancer types, including hepatocellular carcinoma (HCC), compared to normal counterparts. However, a decrease in mtDNA usually leads to defects in cell proliferation, which contradicts the robustness of cancer cell proliferation. In this study, we found that four out of seven HCC cell lines were of the mtDNA-less type. Interestingly, FOXM1, a member of the FOX transcription factor family, was highly expressed in a subset of them with proliferative potential maintained. B-MYB, a partner of FOXM1, was also expressed in the same cell lines. RNAi-mediated experiments demonstrated that when FOXM1/B-MYB was silenced in the cell lines, cell cycle-related genes were downregulated, while p21Cip1 was induced with senescence-associated β-galactosidase, resulting in G1/S cell cycle arrest. These results suggest that high expression of FOXM1/B-MYB is critical for sustaining cell proliferation in mtDNA-less cells. In addition, we found that high expression of FOXM1 was mediated by the deubiquitinating enzyme, OTUB1, in one cell line. Thus, interference with FOXM1/B-MYB expression, such as through OTUB1 inhibition, may induce a dormant state of senescence-like proliferation arrest in mtDNA-less cancer cells. This finding may be utilized for the development of precision medicine for relevant cancers.
Collapse
Affiliation(s)
- Masato Higurashi
- Division of Cancer Cell Biology, Department of Pharmaceutical Sciences, Showa University School of Pharmacy, Tokyo, 142-8555, Japan
| | - Tsuyoshi Maruyama
- Division of Cancer Cell Biology, Department of Pharmaceutical Sciences, Showa University School of Pharmacy, Tokyo, 142-8555, Japan
| | - Yusuke Nogami
- Division of Cancer Cell Biology, Department of Pharmaceutical Sciences, Showa University School of Pharmacy, Tokyo, 142-8555, Japan
| | - Fumihiro Ishikawa
- Division of Cancer Cell Biology, Department of Pharmaceutical Sciences, Showa University School of Pharmacy, Tokyo, 142-8555, Japan
| | - Yukiko Yoshida
- Department of Advanced Science for Biomolecules, Tokyo Metropolitan Institute of Medical Science, Tokyo, 156-8506, Japan
| | - Kazunori Mori
- Division of Cancer Cell Biology, Department of Pharmaceutical Sciences, Showa University School of Pharmacy, Tokyo, 142-8555, Japan
| | - Ken-Ichi Fujita
- Division of Cancer Cell Biology, Department of Pharmaceutical Sciences, Showa University School of Pharmacy, Tokyo, 142-8555, Japan
| | - Motoko Shibanuma
- Division of Cancer Cell Biology, Department of Pharmaceutical Sciences, Showa University School of Pharmacy, Tokyo, 142-8555, Japan.
| |
Collapse
|
16
|
McCann JJ, Vasilevskaya IA, Poudel Neupane N, Shafi AA, McNair C, Dylgjeri E, Mandigo AC, Schiewer MJ, Schrecengost RS, Gallagher P, Stanek TJ, McMahon SB, Berman-Booty LD, Ostrander WF, Knudsen KE. USP22 Functions as an Oncogenic Driver in Prostate Cancer by Regulating Cell Proliferation and DNA Repair. Cancer Res 2020; 80:430-443. [PMID: 31740444 PMCID: PMC7814394 DOI: 10.1158/0008-5472.can-19-1033] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 10/02/2019] [Accepted: 11/13/2019] [Indexed: 02/06/2023]
Abstract
Emerging evidence indicates the deubiquitinase USP22 regulates transcriptional activation and modification of target substrates to promote pro-oncogenic phenotypes. Here, in vivo characterization of tumor-associated USP22 upregulation and unbiased interrogation of USP22-regulated functions in vitro demonstrated critical roles for USP22 in prostate cancer. Specifically, clinical datasets validated that USP22 expression is elevated in prostate cancer, and a novel murine model demonstrated a hyperproliferative phenotype with prostate-specific USP22 overexpression. Accordingly, upon overexpression or depletion of USP22, enrichment of cell-cycle and DNA repair pathways was observed in the USP22-sensitive transcriptome and ubiquitylome using prostate cancer models of clinical relevance. Depletion of USP22 sensitized cells to genotoxic insult, and the role of USP22 in response to genotoxic insult was further confirmed using mouse adult fibroblasts from the novel murine model of USP22 expression. As it was hypothesized that USP22 deubiquitylates target substrates to promote protumorigenic phenotypes, analysis of the USP22-sensitive ubiquitylome identified the nucleotide excision repair protein, XPC, as a critical mediator of the USP22-mediated response to genotoxic insult. Thus, XPC undergoes deubiquitylation as a result of USP22 function and promotes USP22-mediated survival to DNA damage. Combined, these findings reveal unexpected functions of USP22 as a driver of protumorigenic phenotypes and have significant implications for the role of USP22 in therapeutic outcomes. SIGNIFICANCE: The studies herein present a novel mouse model of tumor-associated USP22 overexpression and implicate USP22 in modulation of cellular survival and DNA repair, in part through regulation of XPC.
Collapse
Affiliation(s)
- Jennifer J McCann
- Department of Cancer Biology, Sidney Kimmel Medical College, Philadelphia, Pennsylvania
| | - Irina A Vasilevskaya
- Department of Cancer Biology, Sidney Kimmel Medical College, Philadelphia, Pennsylvania
| | | | - Ayesha A Shafi
- Department of Cancer Biology, Sidney Kimmel Medical College, Philadelphia, Pennsylvania
| | - Christopher McNair
- Department of Cancer Biology, Sidney Kimmel Medical College, Philadelphia, Pennsylvania
| | - Emanuela Dylgjeri
- Department of Cancer Biology, Sidney Kimmel Medical College, Philadelphia, Pennsylvania
| | - Amy C Mandigo
- Department of Cancer Biology, Sidney Kimmel Medical College, Philadelphia, Pennsylvania
| | - Matthew J Schiewer
- Department of Cancer Biology, Sidney Kimmel Medical College, Philadelphia, Pennsylvania
| | - Randy S Schrecengost
- Department of Cancer Biology, Sidney Kimmel Medical College, Philadelphia, Pennsylvania
| | - Peter Gallagher
- Department of Cancer Biology, Sidney Kimmel Medical College, Philadelphia, Pennsylvania
| | - Timothy J Stanek
- Department of Biochemistry & Molecular Biology, Sidney Kimmel Medical College, Philadelphia, Pennsylvania
| | - Steven B McMahon
- Department of Biochemistry & Molecular Biology, Sidney Kimmel Medical College, Philadelphia, Pennsylvania
| | - Lisa D Berman-Booty
- Department of Cancer Biology, Sidney Kimmel Medical College, Philadelphia, Pennsylvania
| | - William F Ostrander
- Department of Cancer Biology, Sidney Kimmel Medical College, Philadelphia, Pennsylvania
| | - Karen E Knudsen
- Department of Cancer Biology, Sidney Kimmel Medical College, Philadelphia, Pennsylvania.
| |
Collapse
|
17
|
Abstract
Prostate cancer (PCa) is the leading cause of cancer death in men. With more therapeutic modalities available, the overall survival in PCa has increased significantly in recent years. Patients with relapses after advanced secondgeneration anti-androgen therapy however, often show poor disease prognosis. This group of patients often die from cancer-related complicacies. Multiple approaches have been taken to understand disease recurrence and to correlate the gene expression profile. In one such study, an 11-gene signature was identified to be associated with PCa recurrence and poor survival. Amongst them, a specific deubiquitinase called ubiquitin-specific peptidase 22 (USP22) was selectively and progressively overexpressed with PCa progression. Subsequently, it was shown to regulate androgen receptors and Myc, the two most important regulators of PCa progression. Furthermore, USP22 has been shown to be associated with the development of therapy resistant PCa. Inhibiting USP22 was also found to be therapeutically advantageous, especially in clinically challenging and advanced PCa. This review provides an update of USP22 related functions and challenges associated with PCa research and explains why targeting this axis is beneficial for PCa relapse cases.
Collapse
Affiliation(s)
- Nivedita Nag
- Department of Microbiology, Sister Nibedita Government General Degree College for Girls, Kolkata 700027, India
| | - Samikshan Dutta
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| |
Collapse
|
18
|
Wen X, Ling S, Wu W, Shan Q, Liu P, Wang C, Wei X, Ding W, Teng X, Xu X. Ubiquitin-Specific Protease 22/Silent Information Regulator 1 Axis Plays a Pivotal Role in the Prognosis and 5-Fluorouracil Resistance in Hepatocellular Carcinoma. Dig Dis Sci 2020; 65:1064-1073. [PMID: 31587155 PMCID: PMC7069902 DOI: 10.1007/s10620-019-05844-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 09/11/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Ubiquitin-specific protease 22 (USP22) is described as a key subunit of the Spt-Ada-Gcn5 acetyl transferase complex, which plays an important role in the prognosis and resistance to chemotherapy drugs in hepatocellular carcinoma (HCC). Silent information regulator 1 (SIRT1) is a member of the sirtuin family that is deubiquitinated by USP22. However, it is still unknown whether USP22 and SIRT1 co-expression is associated with disease progression and 5-Fluorouracil (5-FU) resistance in HCC. METHODS 141 patients who received hepatectomy at our hospital from January 2010 to December 2014 were enrolled in this study. The expression of USP22 and SIRT1 was detected by immunohistochemical staining. Clinicopathological features, including age, gender, tumor number, tumor size, tumor differentiation, tumor stage, alpha-fetoprotein and microscopic vascular invasion, were assessed. Further experiments confirmed the role of SIRT1 in 5-FU drug resistance in vivo. RESULTS Immunohistochemical staining showed that the high expression of USP22 and SIRT1 was frequently observed in HCC tissues relative to normal liver tissues. Overexpression of USP22 is associated with microscopic vascular invasion (MVI). Further analysis showed that the co-expression of USP22 and SIRT1 was more effective in predicting the prognosis of HCC. The SIRT1 inhibitor EX-527 dramatically inhibited the expression of Cyclin B1 and resistance-associated protein 3 (MRP3) to reduce 5-FU drug resistance in vivo. CONCLUSION These findings suggest that the co-expression of USP22 and SIRT1 is significantly associated with unfavorable HCC progression. The inhibition of SIRT1 in vivo could be valuable in improving 5-FU drug sensitivity and inhibiting tumor cell proliferation and inducing apoptosis.
Collapse
Affiliation(s)
- Xue Wen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003 China ,Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of Organ Transplantation, Ministry of Public Health, Hangzhou, China ,Key Laboratory of Organ Transplantation, Hangzhou, Zhejiang China ,Department of Pathology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Sunbin Ling
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003 China ,Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of Organ Transplantation, Ministry of Public Health, Hangzhou, China ,Key Laboratory of Organ Transplantation, Hangzhou, Zhejiang China
| | - Weiqiang Wu
- Department of Pathology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qiaonan Shan
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003 China ,Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of Organ Transplantation, Ministry of Public Health, Hangzhou, China ,Key Laboratory of Organ Transplantation, Hangzhou, Zhejiang China
| | - Peng Liu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003 China ,Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of Organ Transplantation, Ministry of Public Health, Hangzhou, China ,Key Laboratory of Organ Transplantation, Hangzhou, Zhejiang China
| | - Chao Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003 China ,Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of Organ Transplantation, Ministry of Public Health, Hangzhou, China ,Key Laboratory of Organ Transplantation, Hangzhou, Zhejiang China
| | - Xuyong Wei
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003 China ,Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of Organ Transplantation, Ministry of Public Health, Hangzhou, China ,Key Laboratory of Organ Transplantation, Hangzhou, Zhejiang China
| | - Wei Ding
- Department of Pathology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaodong Teng
- Department of Pathology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiao Xu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003 China ,Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of Organ Transplantation, Ministry of Public Health, Hangzhou, China ,Key Laboratory of Organ Transplantation, Hangzhou, Zhejiang China
| |
Collapse
|
19
|
Functional analysis of deubiquitylating enzymes in tumorigenesis and development. Biochim Biophys Acta Rev Cancer 2019; 1872:188312. [DOI: 10.1016/j.bbcan.2019.188312] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/16/2019] [Accepted: 08/16/2019] [Indexed: 02/06/2023]
|
20
|
Liu H, Liu N, Zhao Y, Zhu X, Wang C, Liu Q, Gao C, Zhao X, Li J. Oncogenic USP22 supports gastric cancer growth and metastasis by activating c-Myc/NAMPT/SIRT1-dependent FOXO1 and YAP signaling. Aging (Albany NY) 2019; 11:9643-9660. [PMID: 31689236 PMCID: PMC6874452 DOI: 10.18632/aging.102410] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 10/26/2019] [Indexed: 12/28/2022]
Abstract
In this study, we investigated the role of ubiquitin-specific protease 22 (USP22) in the growth and progression of gastric cancer (GC). USP22 mRNA and protein levels were significantly higher in GC tissue samples and GC cell lines than in adjacent noncancerous tissue samples and a normal gastric mucosal epithelial cell line (GES1), respectively. USP22 knockdown significantly decreased in vitro survival, proliferation, migration, and invasiveness of GC cells compared with the controls. Western blot analysis of control and USP22-silenced GC cells showed that USP22 modulates the c-Myc/NAMPT/SIRT1-dependent FOXO1 and YAP signaling pathways. Subcutanenous injection of USP22-silenced GC cells into SCID mice generated significantly smaller xenograft tumors than did control cells. Moreover, USP22-silenced GC cells showed less lung metastasis than the controls following tail vein injection in SCID mice. In addition, high USP22 expression correlated positively with tumor size, advanced stage and metastasis, and correlated negatively with tumor differentiation and prognosis in GC patients. These results show that USP22 regulates growth and progression of GC via the c-Myc/NAMPT/SIRT1-dependent FOXO1 and YAP signaling pathways.
Collapse
Affiliation(s)
- Hongxia Liu
- Jujube Scientific Research and Applied Center, Life Science College, Luoyang Normal University, Luoyang, Henan 471934, China
| | - Ningning Liu
- Centre of Inflammation and Cancer Research, 150th Central Hospital of PLA, Luoyang, Henan 471031, China
| | - Yali Zhao
- Centre of Inflammation and Cancer Research, 150th Central Hospital of PLA, Luoyang, Henan 471031, China
| | - Xiaoshan Zhu
- Centre of Inflammation and Cancer Research, 150th Central Hospital of PLA, Luoyang, Henan 471031, China
| | - Changsong Wang
- Department of Pathology, 150th Central Hospital of PLA, Luoyang, Henan 471031, China
| | - Qinqin Liu
- Centre of Inflammation and Cancer Research, 150th Central Hospital of PLA, Luoyang, Henan 471031, China
| | - Chunfang Gao
- Centre of Inflammation and Cancer Research, 150th Central Hospital of PLA, Luoyang, Henan 471031, China
| | - Xusheng Zhao
- Jujube Scientific Research and Applied Center, Life Science College, Luoyang Normal University, Luoyang, Henan 471934, China
| | - Juntang Li
- Centre of Inflammation and Cancer Research, 150th Central Hospital of PLA, Luoyang, Henan 471031, China.,Department of Pathology, 150th Central Hospital of PLA, Luoyang, Henan 471031, China.,State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China.,State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| |
Collapse
|
21
|
Yuan X, Wang H, Xu A, Zhu X, Zhan Y, Wang W. Ubiquitin-specific peptidase 22 promotes proliferation and metastasis in human colon cancer. Oncol Lett 2019; 18:5567-5576. [PMID: 31612065 PMCID: PMC6781658 DOI: 10.3892/ol.2019.10872] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 08/13/2019] [Indexed: 01/08/2023] Open
Abstract
Colon cancer is one of the most common malignant tumors in the world; however, the mechanism underlying the progression of colon cancer remains unclear. In the present study, the expression of ubiquitin-specific peptidase 22 (USP22) in paraffin sections of human colon cancer tissues and normal colon tissues were examined using immunohistochemistry. The human colon cancer cell lines HCT116 and HT29 were used for USP22 knockdown experiments, and functional assays were performed. The results demonstrated that compared with normal colon tissues, human colon cancer tissues exhibited upregulated expression of USP22 and this was associated with tumor lymph node metastasis and tumor stage in colon cancer tissues. In addition, upregulated expression of USP22 was significantly correlated with both lower relapse-free survival and lower overall survival rates in patients with colon cancer. When USP22 was silenced in colon cancer cell lines, this resulted in a decrease in cell proliferation and metastatic behaviors. Furthermore, Bmi-1 and Cyclin D2 were found to be positively regulated by USP22, which may have mediated the tumorigenic effects of USP22 in human colon cancer. The results of the present study may have significant implications for examining the underlying mechanisms of cancer development and the potential development of cancer therapeutics.
Collapse
Affiliation(s)
- Xiao Yuan
- Department of General Surgery, The Fourth Affiliated Hospital of Anhui Medical University, Anhui, Hefei 230022, P.R. China
| | - Hao Wang
- Department of Pathology, Anhui Medical University, Anhui, Hefei 230032, P.R. China
| | - Aman Xu
- Department of General Surgery, The Fourth Affiliated Hospital of Anhui Medical University, Anhui, Hefei 230022, P.R. China
| | - Xingyang Zhu
- Department of General Surgery, The Fourth Affiliated Hospital of Anhui Medical University, Anhui, Hefei 230022, P.R. China
| | - Yanqing Zhan
- Department of General Surgery, The Fourth Affiliated Hospital of Anhui Medical University, Anhui, Hefei 230022, P.R. China
| | - Wenbin Wang
- Department of General Surgery, The Fourth Affiliated Hospital of Anhui Medical University, Anhui, Hefei 230022, P.R. China
| |
Collapse
|
22
|
Classification of early and late stage liver hepatocellular carcinoma patients from their genomics and epigenomics profiles. PLoS One 2019; 14:e0221476. [PMID: 31490960 PMCID: PMC6730898 DOI: 10.1371/journal.pone.0221476] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 08/07/2019] [Indexed: 02/07/2023] Open
Abstract
Background Liver Hepatocellular Carcinoma (LIHC) is one of the major cancers worldwide, responsible for millions of premature deaths every year. Prediction of clinical staging is vital to implement optimal therapeutic strategy and prognostic prediction in cancer patients. However, to date, no method has been developed for predicting the stage of LIHC from the genomic profile of samples. Methods The Cancer Genome Atlas (TCGA) dataset of 173 early stage (stage-I), 177 late stage (stage-II, Stage-III and stage-IV) and 50 adjacent normal tissue samples for 60,483 RNA transcripts and 485,577 methylation CpG sites, was extensively analyzed to identify the key transcriptomic expression and methylation-based features using different feature selection techniques. Further, different classification models were developed based on selected key features to categorize different classes of samples implementing different machine learning algorithms. Results In the current study, in silico models have been developed for classifying LIHC patients in the early vs. late stage and cancerous vs. normal samples using RNA expression and DNA methylation data. TCGA datasets were extensively analyzed to identify differentially expressed RNA transcripts and methylated CpG sites that can discriminate early vs. late stages and cancer vs. normal samples of LIHC with high precision. Naive Bayes model developed using 51 features that combine 21 CpG methylation sites and 30 RNA transcripts achieved maximum MCC (Matthew’s correlation coefficient) 0.58 with an accuracy of 78.87% on the validation dataset in discrimination of early and late stage. Additionally, the prediction models developed based on 5 RNA transcripts and 5 CpG sites classify LIHC and normal samples with an accuracy of 96–98% and AUC (Area Under the Receiver Operating Characteristic curve) 0.99. Besides, multiclass models also developed for classifying samples in the normal, early and late stage of cancer and achieved an accuracy of 76.54% and AUC of 0.86. Conclusion Our study reveals stage prediction of LIHC samples with high accuracy based on the genomics and epigenomics profiling is a challenging task in comparison to the classification of cancerous and normal samples. Comprehensive analysis, differentially expressed RNA transcripts, methylated CpG sites in LIHC samples and prediction models are available from CancerLSP (http://webs.iiitd.edu.in/raghava/cancerlsp/).
Collapse
|
23
|
Huang X, Zhang Q, Lou Y, Wang J, Zhao X, Wang L, Zhang X, Li S, Zhao Y, Chen Q, Liang T, Bai X. USP22 Deubiquitinates CD274 to Suppress Anticancer Immunity. Cancer Immunol Res 2019; 7:1580-1590. [PMID: 31399419 DOI: 10.1158/2326-6066.cir-18-0910] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 04/08/2019] [Accepted: 08/05/2019] [Indexed: 11/16/2022]
Abstract
PD-1 (CD279)-PD-L1 (CD274) inhibitory signaling is critical for cancer immune evasion, and thus has become one of the major targets in anticancer immunotherapy. There are several studies that demonstrate the potent effects of posttranslational modifications of CD274 on immune inactivation and suppression, such as ubiquitination, phosphorylation, glycosylation, and palmitoylation. However, the regulatory mechanisms for CD274 deubiquitination are still largely unclear. Here, we identified ubiquitin-specific protease 22 (USP22) as a novel deubiquitinase of CD274. USP22 directly interacted with the C terminus of CD274, inducing its deubiquitination and stabilization. Across multiple cancer types, USP22 was highly expressed and frequently altered in liver cancer, closely correlating with poor prognosis of these patients. Genetic depletion of USP22 inhibited liver cancer growth in an immune system-dependent manner, increased tumor immunogenicity and tumor-infiltrating lymphocytes, and improved therapeutic efficacy of CD274-targeted immunotherapy and CDDP-based chemotherapy in mice. We demonstrate that targeting USP22 is a promising strategy to potentiate anticancer immunity for CD274-amplified cancer.
Collapse
Affiliation(s)
- Xing Huang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China. .,The Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing, Jiangsu, China
| | - Qi Zhang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yu Lou
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, Hangzhou, Zhejiang, China
| | - Junli Wang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, Hangzhou, Zhejiang, China
| | - Xinyu Zhao
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, Hangzhou, Zhejiang, China
| | - Lin Wang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaozhen Zhang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, Hangzhou, Zhejiang, China
| | - Shanshan Li
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yulan Zhao
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qi Chen
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, Hangzhou, Zhejiang, China
| | - Tingbo Liang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China. .,Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, Hangzhou, Zhejiang, China
| | - Xueli Bai
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China. .,Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, Hangzhou, Zhejiang, China
| |
Collapse
|
24
|
Poondla N, Chandrasekaran AP, Kim KS, Ramakrishna S. Deubiquitinating enzymes as cancer biomarkers: new therapeutic opportunities? BMB Rep 2019. [PMID: 30760385 PMCID: PMC6476481 DOI: 10.5483/bmbrep.2019.52.3.048] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cancer remains a life-threatening disease and accounts for the major mortality rates worldwide. The practice of using biomarkers for early detection, staging, and customized therapy may increase cancer patients’ survival. Deubiquitinating enzymes (DUBs) are a family of proteases that remove ubiquitin tags from proteins of interest undergoing proteasomal degradation. DUBs play several functional roles other than deubiquitination. One of the important roles of DUBs is regulation of tumor progression. Several reports have suggested that the DUB family members were highly-elevated in various cancer cells and tissues in different stages of cancer. These findings suggest that the DUBs could be used as drug targets in cancer therapeutics. In this review, we recapitulate the role of the DUB family members, including ubiquitin-specific protease, otubain protease, and important candidates from other family members. Our aim was to better understand the connection between DUB expression profiles and cancers to allow researchers to design inhibitors or gene therapies to improve diagnosis and prognosis of cancers.
Collapse
Affiliation(s)
- Naresh Poondla
- Graduate School of Biomedical Science and Engineering, Department of Biomedical Science, Hanyang University, Seoul 04763, Korea
| | - Arun Pandian Chandrasekaran
- Graduate School of Biomedical Science and Engineering, Department of Biomedical Science, Hanyang University, Seoul 04763, Korea
| | - Kye-Seong Kim
- Graduate School of Biomedical Science and Engineering, Department of Biomedical Science, Hanyang University, Seoul 04763; College of Medicine, Hanyang University, Seoul 04763, Korea
| | - Suresh Ramakrishna
- Graduate School of Biomedical Science and Engineering, Department of Biomedical Science, Hanyang University, Seoul 04763; College of Medicine, Hanyang University, Seoul 04763, Korea
| |
Collapse
|
25
|
Liao Y, Liang X, Liang W, Li Z, Wang Y, Wang L, Zhen S, Tang B, Wang Z. High expression of ubiquitin carboxyl-terminal hydrolase 22 is associated with poor prognosis in hepatitis B virus-associated liver cancer. Oncol Lett 2019; 17:5159-5168. [PMID: 31186731 DOI: 10.3892/ol.2019.10154] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 12/03/2018] [Indexed: 12/16/2022] Open
Abstract
Deubiquitinating enzymes regulate protein activity and cell homeostasis by removing ubiquitin moieties from various substrates. Ubiquitin carboxyl-terminal hydrolase 22 (USP22) is a member of the deubiquitinating protease family and is associated with the development of several tumor types. A previous study demonstrated that USP22 is highly expressed in liver cancer, and its high expression is associated with resistance to chemotherapy. However, the role of USP22 in hepatitis B virus (HBV)-associated liver cancer has not yet been elucidated. The current study demonstrated that USP22 was highly expressed in the tissues of patients with HBV-associated liver cancer, and its high expression was associated with clinicopathological characteristics, including tumor size, clinical stage and prognosis. Further results indicated that USP22 may regulate the proliferative and apoptotic abilities of HepG2.2.15 cells. Additionally, investigation into the underlying mechanism, using small interfering RNA, revealed that the downregulation of USP22 inhibited proliferation and promoted apoptosis though the phosphoinositide 3-kinase/protein kinase B signaling pathway. Therefore, USP22 has the potential to be used as an independent predictor of patient prognosis, as well as a therapeutic target for the treatment of HBV-associated liver cancer.
Collapse
Affiliation(s)
- Yong Liao
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541000, P.R. China
| | - Xingsi Liang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541000, P.R. China
| | - Wenjin Liang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541000, P.R. China
| | - Zeming Li
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541000, P.R. China
| | - Yan Wang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541000, P.R. China
| | - Lin Wang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541000, P.R. China
| | - Siqi Zhen
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541000, P.R. China
| | - Bo Tang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541000, P.R. China
| | - Zhenran Wang
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541000, P.R. China
| |
Collapse
|
26
|
Zhou X, Gan L, Liu J, Xie X, Wang T, Xiong J. Pirarubicin reduces USP22 expression by inhibiting CREB-1 phosphorylation in HeLa cells. Exp Ther Med 2019; 17:4230-4236. [PMID: 31007754 DOI: 10.3892/etm.2019.7447] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 11/02/2018] [Indexed: 12/21/2022] Open
Abstract
The expression of ubiquitin specific peptidase 22 (USP22) is upregulated in several types of cancer, and has been implicated in tumorigenesis. Pirarubicin (THP), an anthracycline antineoplastic drug, can induce apoptosis of several types of cancer cells. However, the molecular mechanisms underlying the action of THP remain to be elucidated. In the current study, treatment with THP induced HeLa cell apoptosis and decreased USP22 expression in a dose- and time-dependent manner. THP reduced the USP22 promoter-regulated luciferase activity, regardless of the mutation of transcriptional activator MYB or E3 ubiquitin-protein ligase SP1 binding sequences; however, this effect was abrogated by the mutation of cyclic AMP-responsive element-binding protein (CREB) binding sequence in HeLa cells. Furthermore, the inhibition on the USP22 promoter activity by THP was not affected by overexpression of CREB-1 in HeLa cells. Additionally, treatment with THP significantly decreased the phosphorylation of CREB-1 at ser133 in HeLa cells. Quantitative chromatin immunoprecipitation assay revealed that THP significantly inhibited the binding of CREB-1 to the USP22 promoter in HeLa cells. The present study demonstrated that THP decreased USP22 expression and promoted HeLa cell apoptosis partially by inhibiting the phosphorylation of CREB-1. The current results may provide novel insights into the molecular mechanisms underlying the pharmacological effect of THP on cancer cell apoptosis.
Collapse
Affiliation(s)
- Xiaoou Zhou
- Department of Pharmacology, College of Basic Medical Science, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Lijun Gan
- Department of Clinical Nursing, College of Nursing, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Jianyun Liu
- Key Laboratory of Jiangxi Province for The Systems Biomedicine, Jiujiang, Jiangxi 332000, P.R. China
| | - Xin Xie
- Department of Pharmacology, College of Basic Medical Science, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Tao Wang
- Department of Pharmacology, College of Basic Medical Science, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Jianjun Xiong
- Department of Pharmacology, College of Basic Medical Science, Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China.,Key Laboratory of Jiangxi Province for The Systems Biomedicine, Jiujiang, Jiangxi 332000, P.R. China
| |
Collapse
|
27
|
Yang F, Zheng Z, Xue X, Zheng L, Qin J, Li H, Zhou Y, Fang G. Targeted eradication of gastric cancer stem cells by CD44 targeting USP22 small interfering RNA-loaded nanoliposomes. Future Oncol 2018; 15:281-295. [PMID: 30543303 DOI: 10.2217/fon-2018-0295] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
AIM USP22, a member of ubiquitin-specific proteases (USPs), is a well-defined protein that promotes poor prognosis, invasion and metastasis, and also participates in the maintenance of cancer stem cells. USP22 siRNA-loaded nanoliposomes conjugated with CD44 antibodies (USP22-NLs-CD44) were constructed to enhance the therapeutic effect of USP22 siRNA against gastric cancer stem cells. MATERIALS & METHODS The targeting and therapeutic efficacies of USP22-NLs-CD44 against gastric cancer stem cells were evaluated. RESULTS & CONCLUSION USP22-NLs-CD44 was demonstrated to be able to effectively deliver USP22 siRNA to CD44+ gastric cancer stem cells, achieving superior therapeutic effects against CD44+ gastric cancer stem cells than nontargeted nanoliposomes. USP22-NLs-CD44 may provide a novel approach to eradicate gastric cancer stem cells in the near future.
Collapse
Affiliation(s)
- Feng Yang
- Department of General Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 700 Moyubei Road, Shanghai 201805, PR China
| | - Zhi Zheng
- Department of General Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 700 Moyubei Road, Shanghai 201805, PR China
| | - Xuchao Xue
- Department of General Surgery, Changhai Hospital, Second Military Medical University, 168 Changhai Road, Shanghai 200433, PR China
| | - Luming Zheng
- Department of General Surgery, General Hospital of Jinan Military Area, 25 Shifan Road, Jinan 250031, PR China
| | - Jianmin Qin
- Department of General Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 700 Moyubei Road, Shanghai 201805, PR China
| | - Haijia Li
- Department of General Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 700 Moyubei Road, Shanghai 201805, PR China
| | - Yuhong Zhou
- Department of General Surgery, Changhai Hospital, Second Military Medical University, 168 Changhai Road, Shanghai 200433, PR China
| | - Guoen Fang
- Department of General Surgery, Changhai Hospital, Second Military Medical University, 168 Changhai Road, Shanghai 200433, PR China
| |
Collapse
|
28
|
Stoppacciaro A, Di Vito S, Filetici P. Epigenetic Factors and Mitochondrial Biology in Yeast: A New Paradigm for the Study of Cancer Metabolism? Front Pharmacol 2018; 9:1349. [PMID: 30524288 PMCID: PMC6258771 DOI: 10.3389/fphar.2018.01349] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 11/02/2018] [Indexed: 12/14/2022] Open
Abstract
Bidirectional cross-talk between nuclear and mitochondrial DNA is fundamental for cell homeostasis. Epigenetic mechanisms regulate the inter-organelle communication between nucleus and mitochondria. Recent research highlights not only the retrograde activation of nuclear gene transcription in case of mitochondria dysfunction, but also the role of post-translational modifications of mitochondrial proteins in respiratory metabolism. Here we discuss some aspects and novel findings in Saccharomyces cerevisiae. In yeast, KAT-Gcn5 and DUB-Ubp8 have a role in respiration and are localized, as single proteins, into mitochondria. These findings, beside the canonical and widely known nuclear activity of SAGA complex in chromatin regulation, provide novel clues on promising aspects linking evolutionary conserved epigenetic factors to the re-programmed metabolism of cancer cells.
Collapse
Affiliation(s)
- Antonella Stoppacciaro
- Surgical Pathology Units, Department of Clinical and Molecular Medicine, Ospedale Sant'Andrea, Sapienza University of Rome, Rome, Italy
| | - Serena Di Vito
- Institute of Molecular Biology and Pathology, CNR, Sapienza University of Rome, Rome, Italy
| | - Patrizia Filetici
- Institute of Molecular Biology and Pathology, CNR, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
29
|
Li C, Huang L, Lu H, Wang W, Chen G, Gu Y, Zhou Q, Peng Z, Feng Z. Expression and clinical significance of ubiquitin‑specific‑processing protease 34 in diffuse large B‑cell lymphoma. Mol Med Rep 2018; 18:4543-4554. [PMID: 30221700 DOI: 10.3892/mmr.2018.9447] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Accepted: 10/13/2017] [Indexed: 11/06/2022] Open
Affiliation(s)
- Chunyao Li
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Lanshan Huang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Huiping Lu
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Wei Wang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Yongyao Gu
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Qianping Zhou
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Zhigang Peng
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Zhenbo Feng
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| |
Collapse
|
30
|
Gong Z, Liu J, Xie X, Xu X, Wu P, Li H, Wang Y, Li W, Xiong J. Identification of potential target genes of USP22 via ChIP-seq and RNA-seq analysis in HeLa cells. Genet Mol Biol 2018; 41:488-495. [PMID: 30088609 PMCID: PMC6082230 DOI: 10.1590/1678-4685-gmb-2017-0164] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 10/14/2016] [Indexed: 12/18/2022] Open
Abstract
The ubiquitin-specific protease 22 (USP22) is an oncogene and its expression is
upregulated in many types of cancer. In the nucleus, USP22 functions as one
subunit of the SAGA to regulate gene transcription. However, the genome-wide
USP22 binding sites and its direct target genes are yet clear. In this study, we
characterized the potential genomic binding sites of UPS22 and GCN5 by ChIP-seq
using specific antibodies in HeLa cells. There were 408 overlapping putative
target genes bound by both USP22 and GCN5. Motif analysis showed that the
sequences bound by USP22 and GCN5 shared two common motifs. Gene ontology (GO)
and pathway analysis indicated that the genes targeted by USP22 and GCN5 were
involved in different physiological processes and pathways. Further RNA-seq, GO
and pathway analyses revealed that knockdown of UPS22 induced differential
expression of many genes that participated in diverse physiological processes,
such as metabolic process. Integration of ChIP-seq and RNA-seq data revealed
that UPS22 bound to the promoters of 56 genes. These findings may provide new
insights into the regulation of USP22 on gene expression during the development
of cervical cancer.
Collapse
Affiliation(s)
- Zhen Gong
- College of Basic Medical Science, Jiujiang University, Jiujiang, Jiangxi, China
| | - Jianyun Liu
- Key Laboratory of Jiangxi Province for the Systemic Bio-medicine, Jiujiang University, Jiujiang, Jiangxi, China
| | - Xin Xie
- Key Laboratory of Jiangxi Province for the Systemic Bio-medicine, Jiujiang University, Jiujiang, Jiangxi, China
| | - Xiaoyuan Xu
- Key Laboratory of Jiangxi Province for the Systemic Bio-medicine, Jiujiang University, Jiujiang, Jiangxi, China
| | - Ping Wu
- Key Laboratory of Jiangxi Province for the Systemic Bio-medicine, Jiujiang University, Jiujiang, Jiangxi, China
| | - Huimin Li
- College of Basic Medical Science, Jiujiang University, Jiujiang, Jiangxi, China
| | - Yaqin Wang
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Weidong Li
- Key Laboratory of Jiangxi Province for the Systemic Bio-medicine, Jiujiang University, Jiujiang, Jiangxi, China
| | - Jianjun Xiong
- College of Basic Medical Science, Jiujiang University, Jiujiang, Jiangxi, China.,Key Laboratory of Jiangxi Province for the Systemic Bio-medicine, Jiujiang University, Jiujiang, Jiangxi, China
| |
Collapse
|
31
|
Yang X, Zang H, Luo Y, Wu J, Fang Z, Zhu W, Li Y. High expression of USP22 predicts poor prognosis and advanced clinicopathological features in solid tumors: a meta-analysis. Onco Targets Ther 2018; 11:3035-3046. [PMID: 29872315 PMCID: PMC5973323 DOI: 10.2147/ott.s148662] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Introduction The expression of USP22 has been demonstrated to play a pivotal role in solid tumors. However, the prognostic value of USP22 still remains unknown. Materials and methods A systematic meta-analysis was performed to assess the prognostic value of USP22 in cancers. A literature collection was conducted from inception to June 8, 2017 by searching PubMed, Cochrane Library, Embase, Ovid and Web of Science databases. The pooled hazard ratio (HR) and odds ratio (OR) were used to correlate high expression of USP22 with overall survival (OS) and clinicopathological features. Results The results, pooled by 19 studies with 2,876 cases, indicated that high expression of USP22 predicted poor OS (HR=2.48, 95% CI: 2.11–2.84, p<0.001) and disease-free survival (DFS; HR=2.55, 95% CI: 2.05–3.05, p<0.001) of cancer patients. Furthermore, high expression of USP22 was also significantly associated with advanced clinicopathological parameters, including tumor stage, tumor differentiation, metastasis, nodal status and tumor size. Conclusion Our finding revealed that USP22 might be an indicator of poor prognosis and advanced clinicopathological features of solid tumors and could be served as a novel biomarker.
Collapse
Affiliation(s)
- Xiaohui Yang
- Department of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Haiyang Zang
- Department of Spleen and Stomach, Xinyi Municipal Hospital of Traditional Chinese Medicine, Xinyi, Jiangsu, China
| | - Yingbin Luo
- Department of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianchun Wu
- Department of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhihong Fang
- Department of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weikang Zhu
- Department of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Li
- Department of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
32
|
USP22 knockdown enhanced chemosensitivity of hepatocellular carcinoma cells to 5-Fu by up-regulation of Smad4 and suppression of Akt. Oncotarget 2018; 8:24728-24740. [PMID: 28445968 PMCID: PMC5421883 DOI: 10.18632/oncotarget.15798] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 02/13/2017] [Indexed: 12/31/2022] Open
Abstract
USP22, a member of the deubiquitinases (DUBs) family, is known to be a key subunit of the human Spt-Ada-Gcn5 acetyltransferase (hSAGA) transcriptional cofactor complex. Within hSAGA, USP22 removes ubiquitin from histone proteins, thus regulating the transcription and expression of downstream genes. USP22 plays important roles in many cancers; however, its effect and the mechanism underlying HCC chemoresistance remain unclear. In the present study, we found that USP22 was highly expressed in chemoresistant HCC tissues and cells and was correlated with the prognosis of HCC patients who received chemotherapy. Silencing USP22 in chemoresistant HCC Bel/Fu cells dramatically inhibited proliferation, migration, invasion and epithelial-mesenchymal transition in vitro; suppressed tumorigenic and metastatic capacities in vivo; and inhibited drug resistance-related proteins (MDR1, LRP, MRP1). Mechanistically, we found that USP22 knockdown exerts its function through down-regulating PI3K and activating Smad4, which inhibited phosphorylation of Akt. Silencing Smad4 blocked USP22 knockdown-induced Akt inhibition in Bel/Fu cells. Our results, for the first time, provide evidence that USP22 plays a critical role in the development of chemoresistant HCC cells and that high USP22 expression serves as a molecular marker for the prognosis of HCC patients who undergo chemotherapy.
Collapse
|
33
|
Li Y, Yang Y, Li J, Liu H, Chen F, Li B, Cui B, Liu Y. USP22 drives colorectal cancer invasion and metastasis via epithelial-mesenchymal transition by activating AP4. Oncotarget 2018; 8:32683-32695. [PMID: 28427243 PMCID: PMC5464819 DOI: 10.18632/oncotarget.15950] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 02/22/2017] [Indexed: 12/20/2022] Open
Abstract
Ubiquitin specific peptidase 22 (USP22), a putative cancer stem cell marker, is overexpressed in liver metastases of colorectal cancer (CRC). However, the mechanism by which USP22 promotes CRC metastasis remains largely unknown. Here, we report that USP22 and AP4 are simultaneously overexpressed during TGF-β1-induced CRC cell epithelial-mesenchymal transition (EMT). USP22 up-regulation enhances CRC cell migration and invasion and EMT-related marker and AP4 expression, but these effects are partly blocked by AP4 knockdown. In addition, USP22 binds to the promoter region of AP4 to activate its transcription. In vivo, elevated USP22 expression promotes CRC cell metastasis to the lungs in nude mice, as evidenced by the fact that CRC metastatic nodules stain deeply positive for USP22 and AP4. In human CRC tissues, the genes encoding USP22 and AP4 are overexpressed in metastatic liver lesions compared with primary cancer tissues, and their overexpression is significantly associated with poor CRC patient survival. These findings indicate that USP22 and AP4 may serve as prognostic markers for predicting the risk of developing distant metastases in CRC.
Collapse
Affiliation(s)
- Yongmin Li
- Department of Colorectal Surgery, The Affiliated Tumor Hospital of Harbin Medical University, Harbin 150081, China
| | - Yanmei Yang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, China
| | - Jingwen Li
- Department of Colorectal Surgery, The Affiliated Tumor Hospital of Harbin Medical University, Harbin 150081, China
| | - He Liu
- Department of Colorectal Surgery, The Affiliated Tumor Hospital of Harbin Medical University, Harbin 150081, China
| | - Fuxun Chen
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, China
| | - Bingyang Li
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, China
| | - Binbin Cui
- Department of Colorectal Surgery, The Affiliated Tumor Hospital of Harbin Medical University, Harbin 150081, China
| | - Yanlong Liu
- Department of Colorectal Surgery, The Affiliated Tumor Hospital of Harbin Medical University, Harbin 150081, China
| |
Collapse
|
34
|
Ma Y, Fu HL, Wang Z, Huang H, Ni J, Song J, Xia Y, Jin WL, Cui DX. USP22 maintains gastric cancer stem cell stemness and promotes gastric cancer progression by stabilizing BMI1 protein. Oncotarget 2018; 8:33329-33342. [PMID: 28415621 PMCID: PMC5464871 DOI: 10.18632/oncotarget.16445] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 03/09/2017] [Indexed: 12/11/2022] Open
Abstract
Increased ubiquitin-specific protease 22 (USP22) has been associated with poor prognosis in several cancers including gastric cancer. However, the role of USP22 in gastric tumorigenesis is still unclear. Gastric cancer stem cells have been identified and shown to correlate with gastric cancer initiation and metastasis. In this study, we found that silencing of USP22 inhibited proliferation of gastric cancer cells and suppressed the cancer stem cell spheroid formation in serum-free culture. Furthermore, cancer stem cell markers, such as CD133, SOX2, OCT4 and NANOG were down-regulated. Additionally, knockdown of USP22 inhibited gastric cancer xenografts growth. Our analysis of TCGA database indicated that BMI1 overexpression may predict gastric cancer patient survival, and TAT-BMI1 proteins reversed the USP22 knockdown-mediated decreased in cancer stem cell properties, and elevated the expression of stemness-associated genes. Furthermore, we found that overexpression of USP22 stabilized the BMI1 protein in gastric cancer cells. Taken together, our study demonstrates that USP22 is indispensable for gastric cancer stem cell self-renewal through stabilization of BMI1. These results may provide novel approaches to the theranostics of gastric cancer in the near future.
Collapse
Affiliation(s)
- Yue Ma
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.,School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hua-Lin Fu
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.,National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Zhen Wang
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Science and Technology, Xi'an Jiao Tong University, Xi'an 710049, China
| | - Hai Huang
- Department of Clinical Biochemistry, School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou 550005, China
| | - Jian Ni
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.,National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Jie Song
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.,National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Ying Xia
- Department of Clinical Biochemistry, School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, Guizhou 550005, China
| | - Wei-Lin Jin
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.,National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Da-Xiang Cui
- Institute of Nano Biomedicine and Engineering, Shanghai Engineering Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.,School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.,National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, Shanghai 200240, PR China
| |
Collapse
|
35
|
Lei X, Xu JF, Chang RM, Fang F, Zuo CH, Yang LY. JARID2 promotes invasion and metastasis of hepatocellular carcinoma by facilitating epithelial-mesenchymal transition through PTEN/AKT signaling. Oncotarget 2018; 7:40266-40284. [PMID: 27259236 PMCID: PMC5130007 DOI: 10.18632/oncotarget.9733] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 04/28/2016] [Indexed: 02/07/2023] Open
Abstract
JARID2 is crucial for maintenance of pluripotency and differentiation of embryonic stem cells. However, little is known about the role of JARID2 in metastasis of hepatocellular carcinoma (HCC). This study found that JARID2 expression was significantly higher in HCC tissues than that in adjacent non-tumor liver tissues (ANLTs), and its expression level correlated with HCC metastasis. High JARID2 expression was significantly correlated with multiple tumor nodules, high Edmondson-Steiner grade, microvascular invasion, advanced TNM stage and advanced BCLC stage (all P < 0.05) and indicated poor prognosis of HCC in training and validation cohorts (all P < 0.05) totaling 182 patients. High JARID2 expression was an independent and significant risk factor for disease-free survival (DFS; P = 0.017) and overall survival (OS; P = 0.041) after curative liver resection in training cohort, and also validated as an independent and significant risk factor for DFS (P = 0.033) and OS (P = 0.031) in validation cohort. Moreover, down-regulation of JARID2 dramatically inhibited HCC cell migration, invasion, proliferation in vitro and metastasis in vivo, whereas overexpression of JARID2 significantly increased migration, invasion, proliferation in vitro and metastasis in vivo. Mechanistically, the data showed that JARID2 exerted its function by repressing PTEN expression through increasing H3K27 trimethylation (H3K27me3) at PTEN promoter region, which subsequently resulted in activation of protein kinase B (AKT) and enhanced epithelial-mesenchymal transition (EMT). In conclusion, this study revealed that JARID2 promotes invasion and metastasis of HCC by facilitating EMT through PTEN/AKT signaling.
Collapse
Affiliation(s)
- Xiong Lei
- Liver Cancer Laboratory, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Jiang-Feng Xu
- Liver Cancer Laboratory, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Rui-Min Chang
- Liver Cancer Laboratory, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Feng Fang
- Liver Cancer Laboratory, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Chao-Hui Zuo
- Department of Abdominal Surgical Oncology, Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China
| | - Lian-Yue Yang
- Liver Cancer Laboratory, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.,Department of Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| |
Collapse
|
36
|
Liu Y, Wang WM, Zou LY, Li L, Feng L, Pan MZ, Lv MY, Cao Y, Wang H, Kung HF, Pang JX, Fu WM, Zhang JF. Ubiquitin specific peptidase 5 mediates Histidine-rich protein Hpn induced cell apoptosis in hepatocellular carcinoma through P14-P53 signaling. Proteomics 2017; 17. [PMID: 28523650 DOI: 10.1002/pmic.201600350] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 04/07/2017] [Accepted: 05/12/2017] [Indexed: 12/15/2022]
Abstract
Hpn is a small histidine-rich cytoplasmic protein from Helicobacter pylori and has been recognized as a high-risk factor for several cancers including gastric cancer, colorectal cancer, and MALT lymphoma. However, the relationship between Hpn and cancers remains elusive. In this study, we discovered that Hpn protein effectively suppressed cell growth and induced apoptosis in hepatocellular carcinoma (HCC). A two-dimensional gel electrophoresis and mass spectrometry-based comparative proteomics was performed to find the molecular targets of Hpn in HCC cells. It was identified that twelve proteins were differentially expressed, with USP5 being one of the most significantly downregulated protein. The P14ARF -P53 signaling was activated by USP5 knockdown in HCC cells. Furthermore, USP5 overexpression significantly rescued the suppressive effect of Hpn on the viability of HCC cells. In conclusion, our study suggests that Hpn plays apoptosis-inducing roles through suppressing USP5 expression and activating the P14ARF -P53 signaling. Therefore, Hpn may be a potential candidate for developing novel anti-HCC drugs.
Collapse
Affiliation(s)
- Yi Liu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, Guangdong, P. R. China
| | - Wei-Mao Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, P. R. China
| | - Li-Yi Zou
- Department of Pharmacology, Guangdong Medical University, Dongguan, Guangdong
| | - Li Li
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, Guangdong, P. R. China
| | - Lu Feng
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, P. R. China
| | - Ming-Zhu Pan
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, Guangdong, P. R. China
| | - Min-Yi Lv
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Ying Cao
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Hua Wang
- Stanley Ho Centre for Emerging Infectious Diseases, The Chinese University of Hong Kong, Hong Kong, P. R. China
| | - Hsiang-Fu Kung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, P. R. China.,Stanley Ho Centre for Emerging Infectious Diseases, The Chinese University of Hong Kong, Hong Kong, P. R. China
| | - Jian-Xin Pang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Wei-Ming Fu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jin-Fang Zhang
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, P. R. China.,School of medicine, South China Unversity of Technlogy, Guangzhou, P. R. China
| |
Collapse
|
37
|
lncRNA HULC promotes the growth of hepatocellular carcinoma cells via stabilizing COX-2 protein. Biochem Biophys Res Commun 2017. [PMID: 28634076 DOI: 10.1016/j.bbrc.2017.06.103] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Highly upregulated in liver cancer (HULC), a lncRNA overexpressed in hepatocellular carcinoma (HCC), has been demonstrated to be involved in the carcinogenesis and progression of HCC. However, the mechanisms of HULC promoting the abnormal growth of HCC cells are still not well elucidated. In the present study, we for the first time demonstrated that HULC promoted the growth of HCC cells through elevating COX-2 protein. Moreover, the study of the corresponding mechanism by which HULC upregulated COX-2 showed that HULC enhanced the level of ubiquitin-specific peptidase 22 (USP22), which decreased ubiquitin-mediated degradation of COX-2 protein by removing the conjugated polyubiquitin chains from COX-2 and finally stabilized COX2 protein. In addition, knockdown of USP22 or COX-2 attenuated HULC-mediated abnormal growth of HCC cells. In conclusion, our results demonstrated that "USP22/COX-2" axis played an important role in HULC promoting growth of HCC cells. The identification of this novel pathway may pave a road for developing new potential anti-HCC strategies.
Collapse
|
38
|
Spolverini A, Fuchs G, Bublik DR, Oren M. let-7b and let-7c microRNAs promote histone H2B ubiquitylation and inhibit cell migration by targeting multiple components of the H2B deubiquitylation machinery. Oncogene 2017; 36:5819-5828. [PMID: 28604753 PMCID: PMC5600258 DOI: 10.1038/onc.2017.187] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 04/18/2017] [Accepted: 05/10/2017] [Indexed: 02/07/2023]
Abstract
Monoubiquitylation of histone H2B (H2Bub1) is catalyzed mainly by the RNF20/RNF40 complex and erased by multiple deubiquitylating enzymes (DUBs). H2Bub1 influences many aspects of chromatin function, including transcription regulation and DNA repair. Cancer cells often display reduced levels of H2Bub1, and this reduction may contribute to cancer progression. The let-7 family of microRNAs comprises multiple members with reported tumor suppressive features, whose expression is frequently downregulated in cancer. We now report that let-7b and let-7c can positively regulate cellular H2Bub1 levels. Overexpression of let-7b and let-7c in a variety of non-transformed and cancer-derived cell lines results in H2Bub1 elevation. The positive effect of let-7b and let-7c on H2Bub1 levels is achieved through targeting of multiple mRNAs, coding for distinct components of the H2B deubiquitylation machinery. Specifically, let-7b and let-7c bind directly and inhibit the mRNAs encoding the DUBs USP42 and USP44, and also the mRNA encoding the adapter protein ATXN7L3, which is part of the DUB module of the SAGA complex. RNF20 knockdown strongly reduces H2Bub1 levels and increases the migration of non-transformed mammary epithelial cells and breast cancer-derived cells. Remarkably, overexpression of let-7b, which partly counteracts the effect of RNF20 knockdown on H2Bub1 levels, also reverses the pro-migratory effect of RNF20 knockdown. Likewise, ATXN7L3 knockdown also increases H2Bub1 levels and reduces cell migration, and this anti-migratory effect is abolished by simultaneous knockdown of RNF20. Together, our findings uncover a novel function of let-7 microRNAs as regulators of H2B ubiquitylation, suggesting an additional mechanism whereby these microRNAs can exert their tumor suppressive effects.
Collapse
Affiliation(s)
- A Spolverini
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - G Fuchs
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - D R Bublik
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - M Oren
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
39
|
Kim D, Hong A, Park HI, Shin WH, Yoo L, Jeon SJ, Chung KC. Deubiquitinating enzyme USP22 positively regulates c-Myc stability and tumorigenic activity in mammalian and breast cancer cells. J Cell Physiol 2017; 232:3664-3676. [PMID: 28160502 DOI: 10.1002/jcp.25841] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 02/03/2017] [Accepted: 02/03/2017] [Indexed: 12/22/2022]
Abstract
The proto-oncogene c-Myc has a pivotal function in growth control, differentiation, and apoptosis and is frequently affected in human cancer, including breast cancer. Ubiquitin-specific protease 22 (USP22), a member of the USP family of deubiquitinating enzymes (DUBs), mediates deubiquitination of target proteins, including histone H2B and H2A, telomeric repeat binding factor 1, and cyclin B1. USP22 is also a component of the mammalian SAGA transcriptional co-activating complex. In this study, we explored the functional role of USP22 in modulating c-Myc stability and its physiological relevance in breast cancer progression. We found that USP22 promotes deubiquitination of c-Myc in several breast cancer cell lines, resulting in increased levels of c-Myc. Consistent with this, USP22 knockdown reduces c-Myc levels. Furthermore, overexpression of USP22 stimulates breast cancer cell growth and colony formation, and increases c-Myc tumorigenic activity. In conclusion, the present study reveals that USP22 in breast cancer cell lines increases c-Myc stability through c-Myc deubiquitination, which is closely correlated with breast cancer progression.
Collapse
Affiliation(s)
- Dongyeon Kim
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Ahyoung Hong
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Hye In Park
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Woo Hyun Shin
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Lang Yoo
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Seo Jeong Jeon
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Kwang Chul Chung
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| |
Collapse
|
40
|
Lawson AP, Bak DW, Shannon DA, Long MJC, Vijaykumar T, Yu R, Oualid FE, Weerapana E, Hedstrom L. Identification of deubiquitinase targets of isothiocyanates using SILAC-assisted quantitative mass spectrometry. Oncotarget 2017; 8:51296-51316. [PMID: 28881649 PMCID: PMC5584250 DOI: 10.18632/oncotarget.17261] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 03/22/2017] [Indexed: 01/14/2023] Open
Abstract
Cruciferous vegetables such as broccoli and kale have well documented chemopreventative and anticancer effects that are attributed to the presence of isothiocyanates (ITCs). ITCs modulate the levels of many oncogenic proteins, but the molecular mechanisms of ITC action are not understood. We previously reported that phenethyl isothiocyanate (PEITC) inhibits two deubiquitinases (DUBs), USP9x and UCH37. DUBs regulate many cellular processes and DUB dysregulation is linked to the pathogenesis of human diseases including cancer, neurodegeneration, and inflammation. Using SILAC assisted quantitative mass spectrometry, here we identify 9 new PEITC-DUB targets: USP1, USP3, USP10, USP11, USP16, USP22, USP40, USP48 and VCPIP1. Seven of these PEITC-sensitive DUBs have well-recognized roles in DNA repair or chromatin remodeling. PEITC both inhibits USP1 and increases its ubiquitination and degradation, thus decreasing USP1 activity by two mechanisms. The loss of USP1 activity increases the level of mono-ubiquitinated DNA clamp PCNA, impairing DNA repair. Both the inhibition/degradation of USP1 and the increase in mono-ubiquitinated PCNA are new activities for PEITC that can explain the previously recognized ability of ITCs to enhance cancer cell sensitivity to cisplatin treatment. Our work also demonstrates that PEITC reduces the mono-ubiquityl histones H2A and H2B. Understanding the mechanism of action of ITCs should facilitate their use as therapeutic agents.
Collapse
Affiliation(s)
- Ann P Lawson
- Department of Biology, Brandeis University, Waltham, MA 02453-9110, USA
| | - Daniel W Bak
- Department of Chemistry, Merkert Center, Boston College, Chestnut Hill, MA 02467-3860, USA
| | - D Alexander Shannon
- Department of Chemistry, Merkert Center, Boston College, Chestnut Hill, MA 02467-3860, USA
| | - Marcus J C Long
- Graduate Program in Biochemistry and Biophysics, Brandeis University, Waltham, MA 02453-9110, USA.,Current address: Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Tushara Vijaykumar
- Graduate Program in Molecular and Cellular Biology, Brandeis University, Waltham, MA 02453-9110, USA.,Current address: Sanofi Genzyme, Framingham, MA 01701, USA
| | - Runhan Yu
- Department of Chemistry, Brandeis University, Waltham, MA 02453-9110, USA
| | | | - Eranthie Weerapana
- Department of Chemistry, Merkert Center, Boston College, Chestnut Hill, MA 02467-3860, USA
| | - Lizbeth Hedstrom
- Department of Biology, Brandeis University, Waltham, MA 02453-9110, USA.,Department of Chemistry, Brandeis University, Waltham, MA 02453-9110, USA
| |
Collapse
|
41
|
Usp5 functions as an oncogene for stimulating tumorigenesis in hepatocellular carcinoma. Oncotarget 2017; 8:50655-50664. [PMID: 28881591 PMCID: PMC5584183 DOI: 10.18632/oncotarget.16901] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 03/28/2017] [Indexed: 12/30/2022] Open
Abstract
As deubiquitinases, several ubiquitin specific protease members have been reported to mediate tumorigenesis. Although ubiquitin specific protease 5 (Usp5) was previously demonstrated to suppress p53 transcriptional activity and DNA repair, its role in carcinogenesis remains elusive. In this study, we sought to define a novel role of Usp5 in tumorigenesis. It was found that Usp5 was significantly upregulated in hepatocellular carcinoma (HCC) cells and most clinical specimens. Further functional investigation also showed that Usp5 knockdown suppressed cell proliferation, migration, drug resistance and induced apoptosis; on the other hand, Usp5 overexpression promoted colony formation, migration, drug resistance and tumorigenesis. Additionally, the inactivated p14ARF-p53 signaling was observed in Usp5 overexpressed HCC cells, while this signaling was activated by Usp5 knockdown. Therefore, our data demonstrated that Usp5 contributed to hepatocarcinogenesis by acting as an oncogene, which provides new insights into the pathogenesis of HCC and explores a promising molecular target for HCC diagnosis and therapy.
Collapse
|
42
|
LncRNA HULC triggers autophagy via stabilizing Sirt1 and attenuates the chemosensitivity of HCC cells. Oncogene 2017; 36:3528-3540. [PMID: 28166203 DOI: 10.1038/onc.2016.521] [Citation(s) in RCA: 285] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 11/21/2016] [Accepted: 12/27/2016] [Indexed: 02/07/2023]
Abstract
Considerable evidences have shown that autophagy has an important role in tumor chemoresistance. However, it is still unknown whether the lncRNA HULC (highly upregulated in liver cancer) is involved in autophagy and chemoresistance of hepatocellular carcinoma (HCC). In this study, we for the first time demonstrated that treatment with antitumor reagents such as oxaliplatin, 5-fluorouracil and pirarubicin (THP) dramatically induced HULC expression and protective autophagy. Silencing of HULC sensitized HCC cells to the three antitumor reagents via inhibiting protective autophagy. Ectopic expression of HULC elicited the autophagy of HCC cells through stabilizing silent information regulator 1 (Sirt1) protein. The investigation for the corresponding mechanism by which HULC stabilized Sirt1 revealed that HULC upregulated ubiquitin-specific peptidase 22 (USP22), leading to the decrease of ubiquitin-mediated degradation of Sirt1 protein by removing the conjugated polyubiquitin chains from Sirt1. Moreover, we found that miR-6825-5p, miR-6845-5p and miR-6886-3p could decrease the level of USP22 protein by binding to the 3'-untranlated region of USP22 mRNA. All the three microRNAs (miRNAs) were downregulated by HULC, which resulted in the elevation of USP22. In addition, we showed that the level of HULC was positively correlated with that of Sirt1 protein in human HCC tissues. Collectively, our data reveals that the pathway 'HULC/USP22/Sirt1/ protective autophagy' attenuates the sensitivity of HCC cells to chemotherapeutic agents, suggesting that this pathway may be a novel target for developing sensitizing strategy to HCC chemotherapy.
Collapse
|
43
|
Dwane L, Gallagher WM, Ní Chonghaile T, O'Connor DP. The Emerging Role of Non-traditional Ubiquitination in Oncogenic Pathways. J Biol Chem 2017; 292:3543-3551. [PMID: 28154183 DOI: 10.1074/jbc.r116.755694] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The addition of ubiquitin to a target protein has long been implicated in the process of degradation and is the primary mediator of protein turnover in the cell. Recently, however, many non-proteolytic functions of ubiquitination have emerged as key regulators of cellular homeostasis. In this review, we will describe the various non-traditional functions of ubiquitination, with particular focus on how they can be used as signaling entities in cancer formation and progression. Elaboration of this topic can lead to a better understanding of oncogenic mechanisms, as well as the discovery of novel druggable proteins within the ubiquitin pathway.
Collapse
Affiliation(s)
- Lisa Dwane
- From Molecular and Cellular Therapeutics and
| | - William M Gallagher
- the Cancer Biology and Therapeutics Laboratory, UCD School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Tríona Ní Chonghaile
- the Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland and
| | | |
Collapse
|
44
|
Zhai R, Tang F, Gong J, Zhang J, Lei B, Li B, Wei Y, Liang X, Tang B, He S. The relationship between the expression of USP22, BMI1, and EZH2 in hepatocellular carcinoma and their impacts on prognosis. Onco Targets Ther 2016; 9:6987-6998. [PMID: 27920552 PMCID: PMC5125798 DOI: 10.2147/ott.s110985] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Recent studies have shown that deubiquitination plays a key role in tumor progression, metastasis, resistance to chemotherapy drugs, and prognosis. In this study, we investigated the effects of the deubiquitinating enzyme USP22 on the expression of the drug-resistance genes BMI1 and EZH2 in hepatocellular carcinoma (HCC) and on prognosis. Downregulation of USP22 expression with interference ribonucleic acid in resistant HCC cell lines with high USP22 expression resulted in decreased BMI1 expression, but had no effect on EZH2 expression. USP22, BMI1, and EZH2 were highly expressed in HCC tissue, and the expression levels were positively correlated with tumor grade and clinical stage. Correlation analysis showed that USP22 expression was positively correlated with that of BMI1. Kaplan–Meier analysis showed that high levels of USP22 and BMI1 expression were associated with poor overall survival and relapse-free survival in all of the cases and in patients treated with transcatheter arterial chemoembolization. These results suggested that high levels of USP22 expression played an important role in drug resistance to chemotherapeutic drugs in HCC patients by upregulating the expression of BMI1; therefore, coexpression of USP22 and BMI1 may become a new predictor for HCC prognosis and may help guide clinical treatment.
Collapse
Affiliation(s)
- Run Zhai
- Laboratory of Liver Injury and Repair Molecular Medicine, Guilin Medical University, Guilin, Guangxi, People's Republic of China; Department of Hepatobiliary Surgery, Affiliated Hospital, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Fang Tang
- Pathology Department, Affiliated Hospital, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Jianhua Gong
- Laboratory of Liver Injury and Repair Molecular Medicine, Guilin Medical University, Guilin, Guangxi, People's Republic of China; Department of Hepatobiliary Surgery, Affiliated Hospital, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Jing Zhang
- Laboratory of Liver Injury and Repair Molecular Medicine, Guilin Medical University, Guilin, Guangxi, People's Republic of China; Department of Hepatobiliary Surgery, Affiliated Hospital, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Biao Lei
- Laboratory of Liver Injury and Repair Molecular Medicine, Guilin Medical University, Guilin, Guangxi, People's Republic of China; Department of Hepatobiliary Surgery, Affiliated Hospital, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Bo Li
- Department of Hepatobiliary Surgery, Affiliated Hospital, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Yangchao Wei
- Laboratory of Liver Injury and Repair Molecular Medicine, Guilin Medical University, Guilin, Guangxi, People's Republic of China; Department of Hepatobiliary Surgery, Affiliated Hospital, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Xingsi Liang
- Laboratory of Liver Injury and Repair Molecular Medicine, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Bo Tang
- Laboratory of Liver Injury and Repair Molecular Medicine, Guilin Medical University, Guilin, Guangxi, People's Republic of China; Department of Hepatobiliary Surgery, Affiliated Hospital, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| | - Songqing He
- Laboratory of Liver Injury and Repair Molecular Medicine, Guilin Medical University, Guilin, Guangxi, People's Republic of China; Department of Hepatobiliary Surgery, Affiliated Hospital, Guilin Medical University, Guilin, Guangxi, People's Republic of China
| |
Collapse
|
45
|
EXPRESSION OF UBIQUITIN SPECIFIC PEPTIDASE GENES IN IRE1 KNOCKDOWN U87 GLIOMA CELLS UPON GLUCOSE DEPRIVATION. BIOTECHNOLOGIA ACTA 2016. [DOI: 10.15407/biotech9.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
46
|
Pinto-Fernandez A, Kessler BM. DUBbing Cancer: Deubiquitylating Enzymes Involved in Epigenetics, DNA Damage and the Cell Cycle As Therapeutic Targets. Front Genet 2016; 7:133. [PMID: 27516771 PMCID: PMC4963401 DOI: 10.3389/fgene.2016.00133] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 07/12/2016] [Indexed: 12/21/2022] Open
Abstract
Controlling cell proliferation is one of the hallmarks of cancer. A number of critical checkpoints ascertain progression through the different stages of the cell cycle, which can be aborted when perturbed, for instance by errors in DNA replication and repair. These molecular checkpoints are regulated by a number of proteins that need to be present at the right time and quantity. The ubiquitin system has emerged as a central player controlling the fate and function of such molecules such as cyclins, oncogenes and components of the DNA repair machinery. In particular, proteases that cleave ubiquitin chains, referred to as deubiquitylating enzymes (DUBs), have attracted recent attention due to their accessibility to modulation by small molecules. In this review, we describe recent evidence of the critical role of DUBs in aspects of cell cycle checkpoint control, associated DNA repair mechanisms and regulation of transcription, representing pathways altered in cancer. Therefore, DUBs involved in these processes emerge as potentially critical targets for the treatment of not only hematological, but potentially also solid tumors.
Collapse
Affiliation(s)
- Adan Pinto-Fernandez
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford Oxford, UK
| | - Benedikt M Kessler
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford Oxford, UK
| |
Collapse
|
47
|
Melo-Cardenas J, Zhang Y, Zhang DD, Fang D. Ubiquitin-specific peptidase 22 functions and its involvement in disease. Oncotarget 2016; 7:44848-44856. [PMID: 27057639 PMCID: PMC5190139 DOI: 10.18632/oncotarget.8602] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 03/10/2016] [Indexed: 12/24/2022] Open
Abstract
Deubiquitylases remove ubiquitin moieties from different substrates to regulate protein activity and cell homeostasis. Since this posttranslational modification plays a role in several different cellular functions, its deregulation has been associated with different pathologies. Aberrant expression of the Ubiquitin-Specific Peptidase 22 (USP22) has been associated with poor cancer prognosis and neurological disorders. However, little is known about USP22 role in these pathologies or in normal physiology. This review summarizes the current knowledge about USP22 function from yeast to human and provides an overview of the possible mechanisms by which USP22 is emerging as a potential oncogene.
Collapse
Affiliation(s)
- Johanna Melo-Cardenas
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yusi Zhang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Donna D. Zhang
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA
| | - Deyu Fang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
48
|
Marine JC, Jochemsen AG. MDMX (MDM4), a Promising Target for p53 Reactivation Therapy and Beyond. Cold Spring Harb Perspect Med 2016; 6:6/7/a026237. [PMID: 27371671 DOI: 10.1101/cshperspect.a026237] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The MDMX protein was identified as a p53-interacting protein with a strong similarity to MDM2. Like Mdm2, Mdmx expression is essential for curbing p53 activity during embryonic development, indicating nonredundant functions of Mdmx and Mdm2. There is now a large body of evidence indicating that cancers frequently up-regulate MDMX expression as a means to dampen p53 tumor-suppressor function. Importantly, MDMX also shows p53-independent oncogenic functions. These data make MDMX an attractive therapeutic target for cancer therapy. Here, we summarize the mechanisms used by cancer cells to increase MDMX expression and promising pharmacological strategies to target MDMX in cancer-in particular, the recent findings that antisense oligonucleotides (ASOs) can be used to efficiently modulate MDMX messenger RNA (mRNA) splicing.
Collapse
Affiliation(s)
- Jean-Christophe Marine
- Laboratory for Molecular Cancer Biology, Center for the Biology of Disease, VIB, 3000 Leuven, Belgium Laboratory for Molecular Cancer Biology, Center of Human Genetics, KULeuven, 3000 Leuven, Belgium
| | - Aart G Jochemsen
- Department of Molecular Cell Biology, Leiden University Medical Center, 2300 RA Leiden, The Netherlands
| |
Collapse
|
49
|
IRE-1α regulates expression of ubiquitin specific peptidases during hypoxic response in U87 glioma cells. ENDOPLASMIC RETICULUM STRESS IN DISEASES 2016. [DOI: 10.1515/ersc-2016-0003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
AbstractIRE-1α (inositol requiring enzyme-1α), the most evolutionarily conserved of the endoplasmic reticulum stress signaling pathways, is highly implicated in sustaining the proliferation of glioma cells and subsequent tumor growth, which is decreased by the inhibition of IRE-1α. To explore the IRE-1α mediated regulation of ubiquitin system in glioma cells, the expression of a subset of ubiquitin specific peptidases (USP) and of ubiquitin activating enzyme E1-like protein/autophagy related 7 (GSA7/ATG7) genes was studied, during hypoxic stress in wild type and U87 glioma cells with inhibited IRE-1α. Hypoxic treatment of wild type glioma cells leads to the up-regulation of USP25 and the concomitant downregulation of USP1, USP10, USP14, and GSA7 genes. USP4 and USP22 genes expression did not significantly change with hypoxic treatment. Inhibition of IRE-1α activity led to up-regulation of USP1, USP4, USP10, USP22, and USP25, while USP14 and GSA7 genes were down-regulated. Therefore, IRE-1α activity modifies substrate-targeting specificity to proteasome during hypoxic stress, which in turn can affect cell survival. Inhibition of IRE-1α correlates directly with deregulation of ubiquitin specific peptidases and GSA7 in a fashion that ultimately slows tumor growth.
Collapse
|
50
|
Tang B, Liang X, Tang F, Zhang J, Zeng S, Jin S, Zhou L, Kudo Y, Qi G. Expression of USP22 and Survivin is an indicator of malignant behavior in hepatocellular carcinoma. Int J Oncol 2015; 47:2208-16. [PMID: 26497847 DOI: 10.3892/ijo.2015.3214] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 09/16/2015] [Indexed: 01/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary malignant liver tumor type, ranking as the third leading cause of all cancer-related deaths in the world. The post-surgical 5-year survival rate is low, largely due to the high recurrence rate. Therefore, the identification of target molecules that control the biological characteristics of HCC is of great importance. Ubiquitin-specific protease 22 (USP22) is a newly discovered deubiquitinating enzyme and is a cancer stem cell marker that plays a role in tumorigenesis, therapy resistance and cell cycle progression. Survivin is a member of the inhibitor of apoptosis protein (IAP) family and is known to function either as an inhibitor for apoptosis or as a regulator of cell division. Levels of survivin are correlated with the aggressiveness of tumors and a poor prognosis in various cancers including HCC. In the present study, we examined the USP22 expression and its association with survivin expression and clinicopathological features in HCC. First, we examined the expression of USP22 and survivin in 151 HCC cases by immunohistochemistry. High expression of USP22 and survivin was frequently observed in HCC cases, in comparison with normal adjacent liver tissues. Expression of USP22 and survivin was well correlated with malignant behavior including tumor size, stage and differentiation in HCC cases. Importantly, HCC patients with high expression of USP22 and survivin showed poor prognosis. USP22 expression was well correlated with survivin expression in HCC cases. This correlation was confirmed in HCC cell lines and tissues by RT-PCR and western blot analysis. Next, to investigate the biological role of USP22 in HCC, we examined the effect of USP22 knockdown on the cell growth and the expression of cell cycle-related protein including survivin in HCC cells. USP22 siRNA suppressed cell growth. Moreover, USP22 siRNA decreased survivin expression together with upregulation of CDK inhibitor, p21 and downregulation of cyclin B. These findings suggest that USP22 may be involved in HCC progression in cooperation with survivin. We suggest that USP22 can be useful as a new prognostic marker and therapeutic target in HCC patients.
Collapse
Affiliation(s)
- Bo Tang
- Department of Hepatobiliary Surgery, Guilin Medical University, Affiliated Hospital, Guilin, Guangxi 541001, P.R. China
| | - Xingsi Liang
- Department of Hepatobiliary Surgery, Guilin Medical University, Affiliated Hospital, Guilin, Guangxi 541001, P.R. China
| | - Fang Tang
- Department of Pathology, the Affiliated Hospital, Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Jing Zhang
- Department of Hepatobiliary Surgery, Guilin Medical University, Affiliated Hospital, Guilin, Guangxi 541001, P.R. China
| | - Sien Zeng
- Department of Pathology and Physiopathology, Guilin Medical University, Guilin, Guangxi 541004, P.R. China
| | - Shengjian Jin
- Department of Pathology and Physiopathology, Guilin Medical University, Guilin, Guangxi 541004, P.R. China
| | - Lihua Zhou
- Department of Pathology and Physiopathology, Guilin Medical University, Guilin, Guangxi 541004, P.R. China
| | - Yasusei Kudo
- Department of Oral Molecular Pathology, Institute of Health Biosciences, University of Tokushima Graduate School, Tokushima 770-8504, Japan
| | - Guangying Qi
- Department of Pathology and Physiopathology, Guilin Medical University, Guilin, Guangxi 541004, P.R. China
| |
Collapse
|