1
|
Ali FEM, Badran KSA, El-Maksoud MSA, Ibrahim IM, Althagafy HS, Hassanein EHM. The role of Wnt/β-catenin signaling in lung cancer progression and therapy: a comprehensive review. Med Oncol 2025; 42:183. [PMID: 40289194 DOI: 10.1007/s12032-025-02709-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/30/2025] [Indexed: 04/30/2025]
Abstract
Most instances of lung cancer (LC), which is the primary cause of cancer-related death worldwide, are non-small-cell lung cancer (NSCLC). Genetic predispositions, environmental exposures, and smoking are risk factors that lead to the development of LC, and the ineffectiveness of existing treatments emphasizes the need for innovative approaches to therapy. Through its regulation of cell proliferation, apoptosis, epithelial-to-mesenchymal transition (EMT), and cancer stem cell maintenance, the Wnt/β-catenin signaling system is essential to advancing LC. This study offers a thorough examination of Wnt/β-catenin signaling in LC, emphasizing how miRNAs, long non-coding RNAs (lncRNAs), circular RNAs (circRNAs), protein-coding genes, enzymes, and both natural and synthetic drugs affect this signaling. Recent research supports the dual function of Wnt/β-catenin signaling in tumor development and repression, which we describe. We also emphasize the therapeutic potential of Wnt/β-catenin inhibitors despite issues including off-target effects and bioavailability. This study highlights the potential of focusing on Wnt/β-catenin signaling to enhance LC patient outcomes by combining computational studies with molecular insights. It also lays the groundwork for further research and treatment development.
Collapse
Affiliation(s)
- Fares E M Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut, 71524, Egypt.
- Faculty of Pharmacy, Michael Sayegh, Aqaba University of Technology, Aqaba, 77110, Jordan.
| | - Khalid S A Badran
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut, 71524, Egypt
| | - Mostafa S Abd El-Maksoud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef, 62521, Egypt
| | - Islam M Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef, 62521, Egypt
| | - Hanan S Althagafy
- Department of Biochemistry, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut, 71524, Egypt
| |
Collapse
|
2
|
Shao N, Xi L, Lv Y, Idris M, Zhang L, Cao Y, Xiang J, Xu X, Ong BX, Zhang Q, Peng X, Yue X, Xu F, Liu C. USP5 stabilizes YTHDF1 to control cancer immune surveillance through mTORC1-mediated phosphorylation. Nat Commun 2025; 16:1313. [PMID: 39900921 PMCID: PMC11791202 DOI: 10.1038/s41467-025-56564-9] [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: 05/24/2024] [Accepted: 01/17/2025] [Indexed: 02/05/2025] Open
Abstract
The N6-methyladenosine binding protein YTHDF1, often upregulated in cancer, promotes tumor growth and hinders immune checkpoint blockade treatment. A comprehensive understanding of the molecular mechanisms governing YTHDF1 protein stability is pivotal for enhancing clinical response rates and the effectiveness of immune checkpoint blockade in cancer patients. Here, we report that USP5 interacts with YTHDF1, stabilizing it by removing K11-linked polyubiquitination. Insulin activates mTORC1, phosphorylating USP5 and promoting its dimerization, which binds to and protects YTHDF1 from degradation. Conversely, the CUL7-FBXW8 E3 ligase promotes YTHDF1 degradation. Deficiency in YTHDF1 or USP5 increases PD-L1 expression and suppresses immune-related gene expression, facilitating immune evasion. Combining USP5 inhibition with anti-PD-L1 therapy enhances anti-tumor immunity, suggesting USP5 as a potential biomarker for patient stratification. This study reveals a ubiquitination-dependent regulation of YTHDF1, proposing USP5 inhibition alongside PD-(L)1 blockade as a promising cancer treatment strategy.
Collapse
Affiliation(s)
- Na Shao
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), The Second Affiliated Hospital, Chongqing Medical University, Chongqing, PR China
| | - Lei Xi
- College of Biological and Food Engineering, Hubei Minzu University, Enshi, PR China
| | - Yangfan Lv
- Department of Pathology, Xinqiao Hospital, Third Military Medical University, Chongqing, PR China
| | - Muhammad Idris
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Lin Zhang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), The Second Affiliated Hospital, Chongqing Medical University, Chongqing, PR China
| | - Ya Cao
- Department of Pathology, Xinqiao Hospital, Third Military Medical University, Chongqing, PR China
| | - Jingyi Xiang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), The Second Affiliated Hospital, Chongqing Medical University, Chongqing, PR China
| | - Xi Xu
- Department of Pathology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Belinda X Ong
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Qiongyi Zhang
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Xu Peng
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Xiaoyan Yue
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Feng Xu
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore.
| | - Chungang Liu
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), The Second Affiliated Hospital, Chongqing Medical University, Chongqing, PR China.
| |
Collapse
|
3
|
Song J, Liu L, Wang F, Bao D. Targeting Ubiquitin-specific Protease 5 Overcomes Chemoresistance via Negatively Regulating p53 in Gastric Cancer. Curr Mol Med 2025; 25:211-221. [PMID: 38347776 DOI: 10.2174/0115665240278762240202103722] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 01/04/2024] [Accepted: 01/04/2024] [Indexed: 04/11/2025]
Abstract
BACKGROUND Resistance to chemotherapy is a major obstacle in the clinical management of gastric cancer, and the mechanisms underlying chemoresistance remain largely unknown. AIMS This study aimed to investigate the involvement of ubiquitin-specific protease 5 (USP5), a deubiquitinating enzyme, in gastric cancer chemoresistance. METHODS USP5 expression was analyzed in fifty paired gastric cancer and adjacent normal tissues, chemo-sensitive and chemo-resistant gastric cancer lines using quantitative ELISA. The role of USP5 was determined using loss-of-function and gainof- function methods. USP5-mediated downstream effectors were analyzed using biochemical methods focusing on p53. RESULTS USP5 expression was comparable in tumors and normal in the majority of the cohort. Following chemotherapy treatment, USP5 expression significantly increased in gastric cancer cells, while p53 levels remained unaltered. Overexpression of USP5 amplified growth and migration while decreasing apoptosis induced by serum withdrawal across multiple gastric cancer cell lines. Conversely, USP5 knockdown effectively heightened gastric cancer sensitivity to paclitaxel and 5-FU treatments, particularly targeting chemo-resistant gastric cancer cells by inhibiting proliferation and migration and inducing apoptosis. Additionally, USP5 knockdown increased levels of p53 but not MDM2, increased p53 activity and increased transcription of p53 target genes. In contrast, USP5 overexpression decreased the level and activity of p53 and inhibited transcription of p53 target genes. The anti-proliferative, anti-migratory, and pro-apoptotic effects of USP5 were significantly diminished upon p53 depletion, highlighting the interplay between p53 and USP5 in regulating gastric cancer cell activities. Additionally, USP5 inhibition suppressed chemo-resistant gastric cancer cell migration via suppressing epithelial-mesenchymal transition (EMT) and RhoA activity. CONCLUSION Targeting USP5 inhibition has emerged as a promising alternative therapeutic approach to overcoming chemoresistance in gastric cancer. Additionally, our study sheds light on the novel role of USP5 as a regulator of p53 in gastric cancer.
Collapse
Affiliation(s)
- Jing Song
- Department of Oncology, Xiangyang Central Hospital, Hubei University of Arts and Science, Xiangyang, 441000, People's Republic of China
| | - Lei Liu
- Department of Oncology, Xiangyang Central Hospital, Hubei University of Arts and Science, Xiangyang, 441000, People's Republic of China
| | - Fang Wang
- Department of Oncology, Xiangyang Central Hospital, Hubei University of Arts and Science, Xiangyang, 441000, People's Republic of China
| | - Di Bao
- Department of Oncology, Xiangyang Central Hospital, Hubei University of Arts and Science, Xiangyang, 441000, People's Republic of China
| |
Collapse
|
4
|
Wang J, Fang S, Jiang Y, Hua Q. Unraveling the Mechanism of Action of Ubiquitin-Specific Protease 5 and Its Inhibitors in Tumors. Clin Med Insights Oncol 2024; 18:11795549241281932. [PMID: 39391229 PMCID: PMC11465303 DOI: 10.1177/11795549241281932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 08/21/2024] [Indexed: 10/12/2024] Open
Abstract
Ubiquitin-specific protease 5 (USP5), a member of the ubiquitin-specific proteases (USPs) family, functions by specifically removing ubiquitin chains from target proteins for stabilization and degrading unbound polyubiquitin chains to maintain a steady-state monoubiquitin pool. Ubiquitin-specific protease 5 regulates various cellular activities, including DNA double-strand break repair, transmission of neuropathic and inflammatory pain signals, immune response, and tumor cell proliferation. Furthermore, USP5 is involved in the development of multiple tumors such as liver, lung, pancreatic, and breast cancers as well as melanoma. Downstream regulatory mechanisms associated with USP5 are complex and diverse. Ubiquitin-specific protease 5 has been revealed as an emerging target for tumor treatment. This study has introduced some molecules upstream to control the expression of USP5 at the levels of transcription, translation, and post-translation. Furthermore, the study incorporated inhibitors known to be associated with USP5, including partially selective deubiquitinase (DUB) inhibitors such as WP1130, EOAI3402143, vialinin A, and chalcone derivatives. It also included the ubiquitin-activating enzyme E1 inhibitor, PYR-41. These small molecule inhibitors impact the occurrence and development of various tumors. Therefore, this article comprehensively reviews the pivotal role of USP5 in different signaling pathways during tumor progression and resumes the progress made in developing USP5 inhibitors, providing a theoretical foundation for their clinical translation.
Collapse
Affiliation(s)
| | | | - Yang Jiang
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qingquan Hua
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
5
|
Koll R, Theilen J, Hauten E, Woodhouse JN, Thiel R, Möllmann C, Fabrizius A. Network-based integration of omics, physiological and environmental data in real-world Elbe estuarine Zander. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 942:173656. [PMID: 38830414 DOI: 10.1016/j.scitotenv.2024.173656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/05/2024]
Abstract
Coastal and estuarine environments are under endogenic and exogenic pressures jeopardizing survival and diversity of inhabiting biota. Information of possible synergistic effects of multiple (a)biotic stressors and holobiont interaction are largely missing in estuaries like the Elbe but are of importance to estimate unforeseen effects on animals' physiology. Here, we seek to leverage host-transcriptional RNA-seq and gill mucus microbial 16S rRNA metabarcoding data coupled with physiological and abiotic measurements in a network analysis approach to decipher the impact of multiple stressors on the health of juvenile Sander lucioperca along one of the largest European estuaries. We find mesohaline areas characterized by gill tissue specific transcriptional responses matching osmosensing and tissue remodeling. Liver transcriptomes instead emphasized that zander from highly turbid areas were undergoing starvation which was supported by compromised body condition. Potential pathogenic bacteria, including Shewanella, Acinetobacter, Aeromonas and Chryseobacterium, dominated the gill microbiome along the freshwater transition and oxygen minimum zone. Their occurrence coincided with a strong adaptive and innate transcriptional immune response in host gill and enhanced energy demand in liver tissue supporting their potential pathogenicity. Taken together, we show physiological responses of a fish species and its microbiome to abiotic factors whose impact is expected to increase with consequences of climate change. We further present a method for the close-meshed detection of the main stressors and bacterial species with disease potential in a highly productive ecosystem.
Collapse
Affiliation(s)
- Raphael Koll
- University of Hamburg, Institute of Cell- and Systems Biology of Animals, Molecular Animal Physiology, Germany.
| | - Jesse Theilen
- University of Hamburg, Department of Biology, Biodiversity Research, Germany
| | - Elena Hauten
- University of Hamburg, Institute of Marine Ecosystem and Fishery Science, Marine ecosystem dynamics, Germany
| | - Jason Nicholas Woodhouse
- University of Hamburg, Institute of Cell- and Systems Biology of Animals, Molecular Animal Physiology, Germany; Leibniz-Institute of Freshwater Ecology and Inland Fisheries (IGB), Microbial and phytoplankton Ecology, Germany
| | - Ralf Thiel
- Leibniz Institute for the Analysis of Biodiversity Change (LIB) - Hamburg site, Centre for Taxonomy & Morphology, Zoological Museum, Germany; University of Hamburg, Department of Biology, Biodiversity Research, Germany
| | - Christian Möllmann
- University of Hamburg, Institute of Marine Ecosystem and Fishery Science, Marine ecosystem dynamics, Germany
| | - Andrej Fabrizius
- University of Hamburg, Institute of Cell- and Systems Biology of Animals, Molecular Animal Physiology, Germany
| |
Collapse
|
6
|
Gu J, Chen C, He P, Du Y, Zhu B. Unraveling the Immune Regulatory Functions of USP5: Implications for Disease Therapy. Biomolecules 2024; 14:683. [PMID: 38927085 PMCID: PMC11201890 DOI: 10.3390/biom14060683] [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: 05/03/2024] [Revised: 05/30/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
Ubiquitin-specific protease 5 (USP5) belongs to the ubiquitin-specific protease (USP) family, which uniquely recognizes unanchored polyubiquitin chains to maintain the homeostasis of monoubiquitin chains. USP5 participates in a wide range of cellular processes by specifically cleaving isopeptide bonds between ubiquitin and substrate proteins or ubiquitin itself. In the process of immune regulation, USP5 affects important cellular signaling pathways, such as NF-κB, Wnt/β-catenin, and IFN, by regulating ubiquitin-dependent protein degradation. These pathways play important roles in immune regulation and inflammatory responses. In addition, USP5 regulates the activity and function of immunomodulatory signaling pathways via the deubiquitination of key proteins, thereby affecting the activity of immune cells and the regulation of immune responses. In the present review, the structure and function of USP5, its role in immune regulation, and the mechanism by which USP5 affects the development of diseases by regulating immune signaling pathways are comprehensively overviewed. In addition, we also introduce the latest research progress of targeting USP5 in the treatment of related diseases, calling for an interdisciplinary approach to explore the therapeutic potential of targeting USP5 in immune regulation.
Collapse
Affiliation(s)
- Jinyi Gu
- Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730030, China; (J.G.); (P.H.); (Y.D.)
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation, Lanzhou 730030, China
- Clinical Laboratory, Affiliated Hospital of Yunnan University, Kunming 650032, China
| | - Changshun Chen
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China;
- Department of Orthopedics and Trauma Surgery, Affiliated Hospital of Yunnan University, Kunming 650032, China
| | - Pu He
- Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730030, China; (J.G.); (P.H.); (Y.D.)
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation, Lanzhou 730030, China
| | - Yunjie Du
- Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730030, China; (J.G.); (P.H.); (Y.D.)
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation, Lanzhou 730030, China
| | - Bingdong Zhu
- Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730030, China; (J.G.); (P.H.); (Y.D.)
- Gansu Provincial Key Laboratory of Evidence Based Medicine and Clinical Translation, Lanzhou 730030, China
| |
Collapse
|
7
|
Foster BM, Wang Z, Schmidt CK. DoUBLing up: ubiquitin and ubiquitin-like proteases in genome stability. Biochem J 2024; 481:515-545. [PMID: 38572758 PMCID: PMC11088880 DOI: 10.1042/bcj20230284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/05/2024] [Accepted: 03/18/2024] [Indexed: 04/05/2024]
Abstract
Maintaining stability of the genome requires dedicated DNA repair and signalling processes that are essential for the faithful duplication and propagation of chromosomes. These DNA damage response (DDR) mechanisms counteract the potentially mutagenic impact of daily genotoxic stresses from both exogenous and endogenous sources. Inherent to these DNA repair pathways is the activity of protein factors that instigate repair processes in response to DNA lesions. The regulation, coordination, and orchestration of these DDR factors is carried out, in a large part, by post-translational modifications, such as phosphorylation, ubiquitylation, and modification with ubiquitin-like proteins (UBLs). The importance of ubiquitylation and UBLylation with SUMO in DNA repair is well established, with the modified targets and downstream signalling consequences relatively well characterised. However, the role of dedicated erasers for ubiquitin and UBLs, known as deubiquitylases (DUBs) and ubiquitin-like proteases (ULPs) respectively, in genome stability is less well established, particularly for emerging UBLs such as ISG15 and UFM1. In this review, we provide an overview of the known regulatory roles and mechanisms of DUBs and ULPs involved in genome stability pathways. Expanding our understanding of the molecular agents and mechanisms underlying the removal of ubiquitin and UBL modifications will be fundamental for progressing our knowledge of the DDR and likely provide new therapeutic avenues for relevant human diseases, such as cancer.
Collapse
Affiliation(s)
- Benjamin M. Foster
- Manchester Cancer Research Centre (MCRC), Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, 555 Wilmslow Road, Manchester M20 4GJ, U.K
| | - Zijuan Wang
- Manchester Cancer Research Centre (MCRC), Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, 555 Wilmslow Road, Manchester M20 4GJ, U.K
| | - Christine K. Schmidt
- Manchester Cancer Research Centre (MCRC), Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, 555 Wilmslow Road, Manchester M20 4GJ, U.K
| |
Collapse
|
8
|
Xu Y, Xing Z, Abdalla Ibrahim Suliman R, Liu Z, Tang F. Ferroptosis in liver cancer: a key role of post-translational modifications. Front Immunol 2024; 15:1375589. [PMID: 38650929 PMCID: PMC11033738 DOI: 10.3389/fimmu.2024.1375589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/26/2024] [Indexed: 04/25/2024] Open
Abstract
Ferroptosis is an emerging form of regulated cell death in an oxidative stress- and iron-dependent manner, primarily induced by the over-production of reactive oxygen species (ROS). Manipulation of ferroptosis has been considered a promising therapeutic approach to inhibit liver tumor growth. Nevertheless, the development of resistance to ferroptosis in liver cancer poses a significant challenge in cancer treatment. Post-translational modifications (PTMs) are crucial enzymatic catalytic reactions that covalently regulate protein conformation, stability and cellular activities. Additionally, PTMs play pivotal roles in various biological processes and divergent programmed cell death, including ferroptosis. Importantly, key PTMs regulators involved in ferroptosis have been identified as potential targets for cancer therapy. PTMs function of two proteins, SLC7A11, GPX4 involved in ferroptosis resistance have been extensively investigated in recent years. This review will summarize the roles of PTMs in ferroptosis-related proteins in hepatocellular carcinoma (HCC) treatment.
Collapse
Affiliation(s)
- Ying Xu
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Zhiyao Xing
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | | | - Zichuan Liu
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, China
| | - Fengyuan Tang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
- Thinking Biomed (Beijing) Co., Ltd, Beijing Economic and Technological Development Zone, Beijing, China
| |
Collapse
|
9
|
Gao ST, Xin X, Wang ZY, Hu YY, Feng Q. USP5: Comprehensive insights into structure, function, biological and disease-related implications, and emerging therapeutic opportunities. Mol Cell Probes 2024; 73:101944. [PMID: 38049041 DOI: 10.1016/j.mcp.2023.101944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 12/06/2023]
Abstract
Ubiquitin specific protease 5 (USP5) is a vital deubiquitinating enzyme that regulates various physiological functions by removing ubiquitin chains from target proteins. This review provides an overview of the structural and functional characteristics of USP5. Additionally, we discuss the role of USP5 in regulating diverse cellular processes, including cell proliferation, apoptosis, DNA double-strand damage, methylation, heat stress, and protein quality control, by targeting different substrates. Furthermore, we describe the involvement of USP5 in several pathological conditions such as tumors, pathological pain, developmental abnormalities, inflammatory diseases, and virus infection. Finally, we introduce newly developed inhibitors of USP5. In conclusion, investigating the novel functions and substrates of USP5, elucidating the underlying mechanisms of USP5-substrate interactions, intensifying the development of inhibitors, and exploring the upstream regulatory mechanisms of USP5 in detail can provide a new theoretical basis for the treatment of various diseases, including cancer, which is a promising research direction with considerable potential. Overall, USP5 plays a critical role in regulating various physiological and pathological processes, and investigating its novel functions and regulatory mechanisms may have significant implications for the development of therapeutic strategies for cancer and other diseases.
Collapse
Affiliation(s)
- Si-Ting Gao
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xin Xin
- Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Ministry of Education, Shanghai, China
| | - Zhuo-Yuan Wang
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yi-Yang Hu
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China; Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Ministry of Education, Shanghai, China; Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China.
| | - Qin Feng
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China; Key Laboratory of Liver and Kidney Diseases, Shanghai University of Traditional Chinese Medicine, Ministry of Education, Shanghai, China; Central Laboratory, ShuGuang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China; Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China.
| |
Collapse
|
10
|
Yan B, Guo J, Wang Z, Ning J, Wang H, Shu L, Hu K, Chen L, Shi Y, Zhang L, Liu S, Tao Y, Xiao D. The ubiquitin-specific protease 5 mediated deubiquitination of LSH links metabolic regulation of ferroptosis to hepatocellular carcinoma progression. MedComm (Beijing) 2023; 4:e337. [PMID: 37492786 PMCID: PMC10363799 DOI: 10.1002/mco2.337] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 06/20/2023] [Accepted: 06/26/2023] [Indexed: 07/27/2023] Open
Abstract
Epigenetic regulators and posttranslational modifications of proteins play important roles in various kinds of cancer cell death, including ferroptosis, a non-apoptotic form of cell death. However, the interplay of chromatin modifiers and deubiquitinase (DUB) in ferroptosis remains unclear. Here, we found that ubiquitin-specific protease 5 (USP5) is regarded as a bona fide DUB of lymphoid-specific helicase (LSH), a DNA methylation repressor, in hepatocellular carcinoma (HCC). Functional studies reveal that USP5 interacts with LSH and stabilizes LSH by a deubiquitylation activity-dependent process. Furthermore, the USP5-mediated deubiquitination of LSH facilitates the tumorigenesis of HCC by upregulating solute carrier family 7 member 11 (SLC7A11) to suppress ferroptosis of liver cancer cells. Moreover, the USP5 inhibitor degrasyn inhibits DUB activities of USP5 to LSH to suppress the progression of HCC. Additionally, USP5 and LSH are positively correlated and both are overexpressed and linked to poor prognosis in HCC patients. Together, our findings show that USP5 interacts with LSH directly and enhances LSH protein stability through deubiquitination, which, in turn, promotes the development of HCC by suppressing ferroptosis of liver cancer cells, suggesting that USP5 may be a potential therapeutic target for HCC.
Collapse
Affiliation(s)
- Bokang Yan
- Department of PathologyZhuzhou Hospital Affiliated to Xiangya School of MedicineCentral South UniversityZhuzhouHunanChina
- Department of PathologyXiangya HospitalCentral South UniversityChangshaHunanChina
- NHC Key Laboratory of Carcinogenesis (Central South University), Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education)Cancer Research Institute and School of Basic MedicineCentral South UniversityChangshaHunanChina
| | - Jiaxing Guo
- Department of PathologyXiangya HospitalCentral South UniversityChangshaHunanChina
- NHC Key Laboratory of Carcinogenesis (Central South University), Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education)Cancer Research Institute and School of Basic MedicineCentral South UniversityChangshaHunanChina
| | - Zuli Wang
- Department of PathologyXiangya HospitalCentral South UniversityChangshaHunanChina
- NHC Key Laboratory of Carcinogenesis (Central South University), Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education)Cancer Research Institute and School of Basic MedicineCentral South UniversityChangshaHunanChina
| | - Jieling Ning
- NHC Key Laboratory of Carcinogenesis (Central South University), Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education)Cancer Research Institute and School of Basic MedicineCentral South UniversityChangshaHunanChina
| | - Haiyan Wang
- Department of PathologyXiangya HospitalCentral South UniversityChangshaHunanChina
- NHC Key Laboratory of Carcinogenesis (Central South University), Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education)Cancer Research Institute and School of Basic MedicineCentral South UniversityChangshaHunanChina
| | - Long Shu
- NHC Key Laboratory of Carcinogenesis (Central South University), Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education)Cancer Research Institute and School of Basic MedicineCentral South UniversityChangshaHunanChina
| | - Kuan Hu
- NHC Key Laboratory of Carcinogenesis (Central South University), Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education)Cancer Research Institute and School of Basic MedicineCentral South UniversityChangshaHunanChina
- Department of Hepatobiliary SurgeryXiangya HospitalCentral South UniversityChangshaHunanChina
| | - Ling Chen
- Department of PathologyXiangya HospitalCentral South UniversityChangshaHunanChina
- NHC Key Laboratory of Carcinogenesis (Central South University), Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education)Cancer Research Institute and School of Basic MedicineCentral South UniversityChangshaHunanChina
| | - Ying Shi
- Department of PathologyXiangya HospitalCentral South UniversityChangshaHunanChina
- NHC Key Laboratory of Carcinogenesis (Central South University), Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education)Cancer Research Institute and School of Basic MedicineCentral South UniversityChangshaHunanChina
| | - Lingqiang Zhang
- State Key Laboratory of ProteomicsBeijing Proteome Research CenterBeijing Institute of Radiation MedicineCollaborative Innovation Center for Cancer MedicineBeijingChina
| | - Shuang Liu
- Department of OncologyInstitute of Medical SciencesNational Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunanChina
| | - Yongguang Tao
- Department of PathologyXiangya HospitalCentral South UniversityChangshaHunanChina
- NHC Key Laboratory of Carcinogenesis (Central South University), Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education)Cancer Research Institute and School of Basic MedicineCentral South UniversityChangshaHunanChina
- Department of Thoracic SurgeryHunan Key Laboratory of Early Diagnosis and Precision Therapy in Lung CancerSecond Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Desheng Xiao
- Department of PathologyXiangya HospitalCentral South UniversityChangshaHunanChina
- Department of PathologySchool of Basic MedicineCentral South UniversityChangshaHunanChina
| |
Collapse
|
11
|
Yan B, Guo J, Deng S, Chen D, Huang M. A pan-cancer analysis of the role of USP5 in human cancers. Sci Rep 2023; 13:8972. [PMID: 37268697 DOI: 10.1038/s41598-023-35793-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 05/24/2023] [Indexed: 06/04/2023] Open
Abstract
Posttranslational modifications (PTM) such as acetylation, deubiquitination, and phosphorylation of proteins, play important roles in various kinds of cancer progression. Ubiquitin-specific proteinase 5 (USP5), a unique member of deubiquitinating enzymes (DUBs) which recognizes unanchored polyubiquitin specifically, could regulate the stability of many tumorigenesis-associated proteins to influence cancer initiation and progression. However, the diverse biological significance of USP5 in pan-cancer has not been systematically and comprehensively studied. Here, we explored the role of USP5 in pan-cancer using The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) database, and we also acquired and analyzed data via various software and web platforms such as R, GEPIA2.0, HPA, TISIDB, cBioPortal, UALCAN, TIMER 2.0, CancerSEA and BioGRID. USP5 expression was high in most cancers and differed significantly in different molecular and immune subtypes of cancers. In addition, USP5 had certain diagnostic value in multiple cancers, and high expression of USP5 generally predicted poor prognosis for cancer patients. We also found that the most frequent genetic alterations type of USP5 was mutation, and the DNA methylation level of USP5 decreased in various cancers. Furthermore, USP5 expression correlated with cancer-associated fibroblasts (CAFs), endothelial cells (EC) and genetic markers of immunodulators in cancers. Moreover, the result from single cell sequencing showed that USP5 could regulate several tumor biological behaviors such as apoptosis, DNA damage and metastasis. Gene enrichment analysis indicated "spliceosome" and "RNA splicing" may be the critical mechanism for USP5 to involve in cancer. Taken together, our study elucidates the biological significance of USP5 in the diagnosis, prognosis and immune in human pan-cancer.
Collapse
Affiliation(s)
- Bokang Yan
- Department of Pathology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412007, Hunan, China
| | - Jiaxing Guo
- Department of Hematology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412007, Hunan, China
| | - Shuang Deng
- Department of Pathology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412007, Hunan, China
| | - Dongliang Chen
- Department of Pathology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412007, Hunan, China.
| | - Meiyuan Huang
- Department of Pathology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412007, Hunan, China.
| |
Collapse
|
12
|
Xiao X, Shi J, He C, Bu X, Sun Y, Gao M, Xiang B, Xiong W, Dai P, Mao Q, Xing X, Yao Y, Yu H, Xu G, Li S, Ren Y, Chen B, Jiang C, Meng G, Lee YR, Wei W, Freeman GJ, Xie C, Zhang J. ERK and USP5 govern PD-1 homeostasis via deubiquitination to modulate tumor immunotherapy. Nat Commun 2023; 14:2859. [PMID: 37208329 PMCID: PMC10199079 DOI: 10.1038/s41467-023-38605-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 05/10/2023] [Indexed: 05/21/2023] Open
Abstract
The programmed cell death protein 1 (PD-1) is an inhibitory receptor on T cells and plays an important role in promoting cancer immune evasion. While ubiquitin E3 ligases regulating PD-1 stability have been reported, deubiquitinases governing PD-1 homeostasis to modulate tumor immunotherapy remain unknown. Here, we identify the ubiquitin-specific protease 5 (USP5) as a bona fide deubiquitinase for PD-1. Mechanistically, USP5 interacts with PD-1, leading to deubiquitination and stabilization of PD-1. Moreover, extracellular signal-regulated kinase (ERK) phosphorylates PD-1 at Thr234 and promotes PD-1 interaction with USP5. Conditional knockout of Usp5 in T cells increases the production of effector cytokines and retards tumor growth in mice. USP5 inhibition in combination with Trametinib or anti-CTLA-4 has an additive effect on suppressing tumor growth in mice. Together, this study describes a molecular mechanism of ERK/USP5-mediated regulation of PD-1 and identifies potential combinatorial therapeutic strategies for enhancing anti-tumor efficacy.
Collapse
Affiliation(s)
- Xiangling Xiao
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University; Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan, 430071, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Jie Shi
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University; Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan, 430071, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Chuan He
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University; Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan, 430071, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xia Bu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Yishuang Sun
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University; Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan, 430071, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Minling Gao
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University; Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan, 430071, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Bolin Xiang
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University; Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan, 430071, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Wenjun Xiong
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University; Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan, 430071, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Panpan Dai
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University; Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan, 430071, China
| | - Qi Mao
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University; Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan, 430071, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xixin Xing
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University; Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan, 430071, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Yingmeng Yao
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University; Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan, 430071, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Haisheng Yu
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University; Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan, 430071, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Gaoshan Xu
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University; Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan, 430071, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Siqi Li
- Department of Biology, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Yan Ren
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Baoxiang Chen
- Department of Colorectal and Anal Surgery, Low Rectal Cancer Diagnosis and Treatment Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Congqing Jiang
- Department of Colorectal and Anal Surgery, Low Rectal Cancer Diagnosis and Treatment Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Geng Meng
- College of Veterinary Medicine, China Agricultural University, Beijing, 100094, China
| | - Yu-Ru Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115201, Taiwan
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Conghua Xie
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University; Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan, 430071, China.
| | - Jinfang Zhang
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University; Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan, 430071, China.
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
13
|
Ali MY, Gadotti VM, Huang S, Garcia-Caballero A, Antunes FTT, Jung HA, Choi JS, Zamponi GW. Icariside II, a Prenyl-Flavonol, Alleviates Inflammatory and Neuropathic Pain by Inhibiting T-Type Calcium Channels and USP5-Cav3.2 Interactions. ACS Chem Neurosci 2023; 14:1859-1869. [PMID: 37116219 DOI: 10.1021/acschemneuro.3c00083] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2023] Open
Abstract
Cav3.2 channels play an important role in the afferent nociceptive pathway, which is responsible for both physiological and pathological pain transmission. Cav3.2 channels are upregulated during neuropathic pain or peripheral inflammation in part due to an increased association with the deubiquitinase USP5. In this study, we investigated nine naturally occurring flavonoid derivatives which we tested for their abilities to inhibit transiently expressed Cav3.2 channels and their interactions with USP5. Icariside II (ICA-II), one of the flavonols studied, inhibited the biochemical interactions between USP5 and Cav3.2 and concomitantly and effectively blocked Cav3.2 channels. Molecular docking analysis predicts that ICA-II binds to the cUBP domain and the Cav3.2 interaction region. In addition, ICA-II was predicted to interact with residues in close proximity to the Cav3.2 channel's fenestrations, thus accounting for the observed blocking activity. In mice with inflammatory and neuropathic pain, ICA-II inhibited both phases of the formalin-induced nocifensive responses and abolished thermal hyperalgesia induced by injection of complete Freund's adjuvant (CFA) into the hind paw. Furthermore, ICA-II produced significant and long-lasting thermal anti-hyperalgesia in female mice, whereas Cav3.2 null mice were resistant to the action of ICA-II. Altogether, our data show that ICA-II has analgesic activity via an action on Cav3.2 channels.
Collapse
Affiliation(s)
- Md Yousof Ali
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB T2N4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N4N1, Canada
- Zymedyne Therapeutics, Calgary, AB T2N4G4, Canada
| | - Vinicius M Gadotti
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB T2N4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N4N1, Canada
- Zymedyne Therapeutics, Calgary, AB T2N4G4, Canada
| | - Sun Huang
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB T2N4N1, Canada
| | - Agustin Garcia-Caballero
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB T2N4N1, Canada
- Zymedyne Therapeutics, Calgary, AB T2N4G4, Canada
| | - Flavia T T Antunes
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB T2N4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N4N1, Canada
| | - Hyun Ah Jung
- Department of Food Science and Human Nutrition, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Jae Sue Choi
- Department of Food and Life Science, Pukyong National University, Busan 48513, Republic of Korea
| | - Gerald W Zamponi
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB T2N4N1, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N4N1, Canada
| |
Collapse
|
14
|
Ubiquitin-Specific Proteases as Potential Therapeutic Targets in Bladder Cancer—In Vitro Evaluation of Degrasyn and PR-619 Activity Using Human and Canine Models. Biomedicines 2023; 11:biomedicines11030759. [PMID: 36979739 PMCID: PMC10045593 DOI: 10.3390/biomedicines11030759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/24/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Background: The inhibition of ubiquitin-specific proteases (USPs) is a novel and promising direction in the development of molecularly targeted therapies in oncology. The aim of the present study was to examine whether Degrasyn could be a potential therapeutic agent against bladder cancer (BC). Also, we aimed to determine whether Degrasyn is more effective in terms of anti-cancer activity compared to the non-selective DUB inhibitor PR-619. To facilitate the translational value of the obtained results, our experiments were performed using both human and canine in vitro models of BC. Methods: Human T24 (urothelial grade III BC) and SV-HUC-1 (non-tumorigenic urothelial cell line), as well as canine K9TCC-PU-NK and RDSVS-TCC1 (both derived from invasive grade III urothelial bladder tumors) cell lines, were used in the present study. Cell proliferation was determined using the MTT assay and Ki-67 proliferation assay, and the level of apoptosis induced by Degrasyn and PR-619 was evaluated by Annexin V-FITC staining and caspase 3/7 activation assay. Western blot was used to assess DNA damage and key proteins involved in apoptosis. Results: Degrasyn inhibited the proliferation of all BC cell lines in a concentration- and time-dependent manner. Lower concentrations of Degrasyn were more potent against human and canine BC cell lines compared to PR-619. Degrasyn induced caspase-dependent apoptosis and triggered DNA damage. PR-619 did not show a significant pro-apoptotic effect. Conclusions: Our results demonstrate that Degrasyn significantly impairs the growth of in vitro models of human and canine BC. Selective USP inhibition with Degrasyn seems to be more effective in reducing BC cell proliferation and inducing apoptosis and DNA damage than non-selective USP inhibition with PR-619.
Collapse
|
15
|
Zheng Y, Wang L, Niu X, Guo Y, Zhao J, Li L, Zhao J. EOAI, a ubiquitin-specific peptidase 5 inhibitor, prevents non-small cell lung cancer progression by inducing DNA damage. BMC Cancer 2023; 23:28. [PMID: 36611139 PMCID: PMC9826599 DOI: 10.1186/s12885-023-10506-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 01/02/2023] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE Targeting deubiquitinases (DUBs) has emerged as a promising avenue for anticancer drug development. However, the effect and mechanism of pan-DUB inhibitor EOAI on non-small cell lung cancer (NSCLC) remains to be studied. MATERIALS AND METHODS The expression of ubiquitin-specific peptidase 5 (USP5) in NSCLC was evaluated by immunohistochemistry. The effect of the USP5 inhibitor, EOAI, on NSCLC cell growth and cell cycle was evaluated by CCK-8 and PI staining. Apoptosis was detected by Annexin V-FITC/PI double staining. Autophagy was examined by LC3 immunofluorescence. Comet assay and γ-H2AX immunofluorescence staining were used to detect DNA damage, and Western blotting was used to detect the expression of apoptosis, cycle, autophagy and DNA damage-related proteins. In vivo experiments demonstrated the effect of EOAI on NSCLC. RESULTS We also found that USP5 was significantly upregulated in NSCLC tissues in this study. In addition, we show that EOAI can cause DNA damage in NSCLC cells while modulating the transcriptional activity of P53, thereby inducing cell cycle arrest in NSCLC cells, autophagy and apoptosis. In vivo experiments have shown that EOAI can inhibit tumors and synergistically enhance the anti-tumor effect of cisplatin. CONCLUSION USP5-mediated epigenetic regulation of oncogenes promotes the occurrence of NSCLC, which provides ideas for developing potential targeted therapy.
Collapse
Affiliation(s)
- Yuanyuan Zheng
- grid.412633.10000 0004 1799 0733Internet Medical and System Applications of National Engineering Laboratory, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Longhao Wang
- grid.412633.10000 0004 1799 0733Internet Medical and System Applications of National Engineering Laboratory, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Xiaoyu Niu
- grid.414008.90000 0004 1799 4638Department of Anesthesiology, the Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, 450008 Henan China
| | - Yongjun Guo
- grid.414008.90000 0004 1799 4638Department of Molecular Pathology, the Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, 450008 Henan China ,Henan Key Laboratory of Molecular Pathology, Zhengzhou, 450008 Henan China
| | - Jiuzhou Zhao
- grid.414008.90000 0004 1799 4638Department of Molecular Pathology, the Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, 450008 Henan China ,Henan Key Laboratory of Molecular Pathology, Zhengzhou, 450008 Henan China
| | - Lifeng Li
- grid.412633.10000 0004 1799 0733Internet Medical and System Applications of National Engineering Laboratory, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Jie Zhao
- grid.412633.10000 0004 1799 0733Internet Medical and System Applications of National Engineering Laboratory, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China ,grid.412633.10000 0004 1799 0733Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| |
Collapse
|
16
|
Sparks A, Kelly CJ, Saville MK. Ubiquitin receptors play redundant roles in the proteasomal degradation of the p53 repressor MDM2. FEBS Lett 2022; 596:2746-2767. [PMID: 35735670 PMCID: PMC9796813 DOI: 10.1002/1873-3468.14436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/01/2022] [Accepted: 06/05/2022] [Indexed: 01/07/2023]
Abstract
Much remains to be determined about the participation of ubiquitin receptors in proteasomal degradation and their potential as therapeutic targets. Suppression of the ubiquitin receptor S5A/PSMD4/hRpn10 alone stabilises p53/TP53 but not the key p53 repressor MDM2. Here, we observed S5A and the ubiquitin receptors ADRM1/PSMD16/hRpn13 and RAD23A and B functionally overlap in MDM2 degradation. We provide further evidence that degradation of only a subset of ubiquitinated proteins is sensitive to S5A knockdown because ubiquitin receptor redundancy is commonplace. p53 can be upregulated by S5A modulation while degradation of substrates with redundant receptors is maintained. Our observations and analysis of Cancer Dependency Map (DepMap) screens show S5A depletion/loss substantially reduces cancer cell line viability. This and selective S5A dependency of proteasomal substrates make S5A a target of interest for cancer therapy.
Collapse
Affiliation(s)
| | - Christopher J. Kelly
- School of MedicineUniversity of DundeeUK,Institute of Infection, Immunity and InflammationUniversity of GlasgowUK
| | - Mark K. Saville
- School of MedicineUniversity of DundeeUK,Silver River EditingDundeeUK
| |
Collapse
|
17
|
Usp5, Usp34, and Otu1 deubiquitylases mediate DNA repair in Drosophila melanogaster. Sci Rep 2022; 12:5870. [PMID: 35393473 PMCID: PMC8990000 DOI: 10.1038/s41598-022-09703-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 03/21/2022] [Indexed: 11/29/2022] Open
Abstract
Ubiquitylation is critical for preventing aberrant DNA repair and for efficient maintenance of genome stability. As deubiquitylases (DUBs) counteract ubiquitylation, they must have a great influence on many biological processes, including DNA damage response. To elucidate the role of DUBs in DNA repair in Drosophila melanogaster, systematic siRNA screening was applied to identify DUBs with a reduced survival rate following exposure to ultraviolet and X-ray radiations. As a secondary validation, we applied the direct repeat (DR)-white reporter system with which we induced site-specific DSBs and affirmed the importance of the DUBs Ovarian tumor domain-containing deubiquitinating enzyme 1 (Otu1), Ubiquitin carboxyl-terminal hydrolase 5 (Usp5), and Ubiquitin carboxyl-terminal hydrolase 34 (Usp34) in DSB repair pathways using Drosophila. Our results indicate that the loss of Otu1 and Usp5 induces strong position effect variegation in Drosophila eye following I-SceI-induced DSB deployment. Otu1 and Usp5 are essential in DNA damage-induced cellular response, and both DUBs are required for the fine-tuned regulation of the non-homologous end joining pathway. Furthermore, the Drosophila DR-white assay demonstrated that homologous recombination does not occur in the absence of Usp34, indicating an indispensable role of Usp34 in this process.
Collapse
|
18
|
Huang W, Liu X, Zhang Y, Deng M, Li G, Chen G, Yu L, Jin L, Liu T, Wang Y, Chen Y. USP5 promotes breast cancer cell proliferation and metastasis by stabilizing HIF2α. J Cell Physiol 2022; 237:2211-2219. [PMID: 35102545 DOI: 10.1002/jcp.30686] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 01/04/2022] [Accepted: 01/10/2022] [Indexed: 12/19/2022]
Abstract
Hypoxia-inducible factor 2α (HIF2α) plays a pivotal role in breast tumor growth and metastasis. However, the regulatory mechanisms of HIF2α protein stability remain poorly understood. The precise role of the deubiquitinase (DUB) ubiquitin-specific peptidase 5 (USP5) in breast cancer and the underlying mechanism remains largely unknown. Here, we identified USP5 as a novel DUB for HIF2α. Physically, USP5 interacts with HIF2α and protects HIF2α from ubiquitin-proteasome degradation, thereby promoting the transcription of HIF2α target genes, such as SLC2A1, PLOD2, P4HA1, and VEGFA. USP5 ablation impairs breast cancer cells proliferation, colony formation, migration, and invasion. Moreover, USP5 is highly expressed in breast cancer, and the protein levels of USP5 are positively correlated with HIF2α protein levels in human breast cancer tissues. Importantly, high levels of USP5 leads to poor clinical outcome in patients with breast cancer. Collectively, USP5 stabilizes HIF2α through its DUB activity and provides a potential therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Weixiao Huang
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, Guangdong, China.,Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Xiong Liu
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Yao Zhang
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Mingxia Deng
- Biomedical Translational Research Institute, Jinan University, Guangzhou, Guangdong, China
| | - Guangqiang Li
- Biomedical Translational Research Institute, Jinan University, Guangzhou, Guangdong, China
| | - Guo Chen
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Li Yu
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Lai Jin
- Department of Pharmacy, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Tongzheng Liu
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, China
| | - Yijie Wang
- Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Yan Chen
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, Guangdong, China.,Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| |
Collapse
|
19
|
Li G, Yang T, Chen Y, Bao J, Wu D, Hu X, Feng C, Xu L, Li M, Li G, Jin M, Xu Y, Zhang R, Qian G, Pan J. USP5 Sustains the Proliferation of Glioblastoma Through Stabilization of CyclinD1. Front Pharmacol 2021; 12:720307. [PMID: 34483932 PMCID: PMC8415357 DOI: 10.3389/fphar.2021.720307] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/04/2021] [Indexed: 12/21/2022] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most malignant primary tumors in humans. Despite standard therapeutic strategy with tumor resection combined with radiochemotherapy, the prognosis remains disappointed. Recently, deubiquitinating enzymes (DUBs) has been reported as potential cancer therapy targets due to their multifunctions involved in the regulation of tumorigenesis, cell cycle, apoptosis, and autophagy. In this study, we found that knockdown of ubiquitin specific protease (USP5), a family member of DUB, could significantly suppress GBM cell line U251 and DBTRG-05MG proliferation and colony formation by inducing cell cycle G1/S arrest, which was correlated with downregulation of CyclinD1 protein level. CyclinD1 had been reported to play a critical role in the tumorigenesis and development of GBM via regulating cell cycle transition. Overexpression of USP5 could significantly extend the half-life of CyclinD1, while knockdown of USP5 decreased the protein level of CyclinD1, which could be restored by proteasome inhibitor MG-132. Indeed, USP5 was found to directly interact with CyclinD1, and decrease its K48-linked polyubiquitination level. Furthermore, knockdown of USP5 in U251 cells remarkably inhibited tumor growth in vivo. Taken together, these findings demonstrate that USP5 plays a critical role in tumorigenesis and progression of GBM by stabilizing CyclinD1 protein. Targeting USP5 could be a potential therapeutic strategy for GBM.
Collapse
Affiliation(s)
- Gen Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China.,Laboratory of Molecular Neuropathology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Tianquan Yang
- Department of Neurosurgery, Children's Hospital of Soochow University, Suzhou, China
| | - Yanling Chen
- School of Basic Medicine and Biological Sciences, Soochow University, Suzhou, China
| | - Jianping Bao
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, China
| | - Di Wu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Xiaohan Hu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Chenxi Feng
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Lixiao Xu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Mei Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Gang Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Meifang Jin
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Yunyun Xu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Rui Zhang
- Clinical Pediatrics School, Soochow University, Suzhou, China
| | - Guanghui Qian
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Jian Pan
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| |
Collapse
|
20
|
Cai B, Zhao J, Zhang Y, Liu Y, Ma C, Yi F, Zheng Y, Zhang L, Chen T, Liu H, Liu B, Gao C. USP5 attenuates NLRP3 inflammasome activation by promoting autophagic degradation of NLRP3. Autophagy 2021; 18:990-1004. [PMID: 34486483 DOI: 10.1080/15548627.2021.1965426] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
ABBREVIATIONS 3-MA: 3-methyladenine; AIM2: absent in melanoma 2; ATG5: autophagy related 5; BafA1: bafilomycin A1; CASP1: caspase 1; CHX: cycloheximide; Co-IP: co-immunoprecipitation; CQ: chloroquine; DUBs: deubiquitinases; IL1B/IL-1β: interleukin 1 beta; LAMP1: lysosomal associated membrane protein 1; LPS: lipopolysaccharide; MARCHF7/MARCH7: membrane associated RING-CH-type finger 7; NFKB/NF-κB: nuclear factor kappa B; Nig.: nigericin; NLRC4: NLR family CARD domain containing 4; NLRP3: NLR family pyrin domain containing 3; PECs: peritoneal exudate cells; PMN: polymorphonuclear; PMs: peritoneal macrophages; PYCARD/ASC: PYD and CARD domain containing; TLRs: toll like receptors; TNF/TNF-α: tumor necrosis factor; Ub: ubiquitin; USP5: ubiquitin specific peptidase 5; WT: wild type.
Collapse
Affiliation(s)
- Baoshan Cai
- Key Laboratory of Infection and Immunity of Shandong Province & Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, China
| | - Jian Zhao
- Key Laboratory of Infection and Immunity of Shandong Province & Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, China
| | - Yuling Zhang
- Key Laboratory of Infection and Immunity of Shandong Province & Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, China
| | - Yaxing Liu
- Key Laboratory of Infection and Immunity of Shandong Province & Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, China
| | - Chunhong Ma
- Key Laboratory of Infection and Immunity of Shandong Province & Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, China
| | - Fan Yi
- Department of Pharmacology, School of Biomedical Sciences, Shandong University, Jinan, China
| | - Yi Zheng
- Key Laboratory of Infection and Immunity of Shandong Province & Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, China
| | - Lei Zhang
- Key Laboratory of Infection and Immunity of Shandong Province & Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, China
| | - Tian Chen
- Department of Pathogenic Biology, School of Biomedical Sciences, Shandong University, Jinan, China
| | - Huiqing Liu
- Department of Pharmacology, School of Biomedical Sciences, Shandong University, Jinan, China
| | - Bingyu Liu
- Key Laboratory of Infection and Immunity of Shandong Province & Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, China
| | - Chengjiang Gao
- Key Laboratory of Infection and Immunity of Shandong Province & Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, China
| |
Collapse
|
21
|
Snyder NA, Silva GM. Deubiquitinating enzymes (DUBs): Regulation, homeostasis, and oxidative stress response. J Biol Chem 2021; 297:101077. [PMID: 34391779 PMCID: PMC8424594 DOI: 10.1016/j.jbc.2021.101077] [Citation(s) in RCA: 160] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 12/17/2022] Open
Abstract
Ubiquitin signaling is a conserved, widespread, and dynamic process in which protein substrates are rapidly modified by ubiquitin to impact protein activity, localization, or stability. To regulate this process, deubiquitinating enzymes (DUBs) counter the signal induced by ubiquitin conjugases and ligases by removing ubiquitin from these substrates. Many DUBs selectively regulate physiological pathways employing conserved mechanisms of ubiquitin bond cleavage. DUB activity is highly regulated in dynamic environments through protein-protein interaction, posttranslational modification, and relocalization. The largest family of DUBs, cysteine proteases, are also sensitive to regulation by oxidative stress, as reactive oxygen species (ROS) directly modify the catalytic cysteine required for their enzymatic activity. Current research has implicated DUB activity in human diseases, including various cancers and neurodegenerative disorders. Due to their selectivity and functional roles, DUBs have become important targets for therapeutic development to treat these conditions. This review will discuss the main classes of DUBs and their regulatory mechanisms with a particular focus on DUB redox regulation and its physiological impact during oxidative stress.
Collapse
Affiliation(s)
- Nathan A Snyder
- Department of Biology, Duke University, Durham, North Carolina, USA
| | - Gustavo M Silva
- Department of Biology, Duke University, Durham, North Carolina, USA.
| |
Collapse
|
22
|
Qu Y, Xu Y, Jiang Y, Yu D, Jiang X, Zhao L. Macrophage-derived extracellular vesicles regulates USP5-mediated HDAC2/NRF2 axis to ameliorate inflammatory pain. FASEB J 2021; 35:e21332. [PMID: 34423867 DOI: 10.1096/fj.202001185rr] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 11/11/2022]
Abstract
Emerging research has highlighted the capacity of microRNA-23a-3p (miR-23a-3p) to alleviate inflammatory pain. However, the molecular mechanism by which miR-23a-3p attenuates inflammatory pain is yet to be fully understood. Hence, the current study aimed to elucidate the mechanism by which miR-23a-3p influences inflammatory pain. Bioinformatics was initially performed to predict the inflammatory pain related downstream targets of miR-23a-3p in macrophage-derived extracellular vesicles (EVs). An animal inflammatory pain model was established using Complete Freund's Adjuvant (CFA). The miR-23a-3p expression was downregulated in the microglia of CFA-induced mice, after which the inflammatory factors were determined by ELISA. FISH and immunofluorescence were performed to analyze the co-localization of miR-23a-3p and microglia. Interestingly, miR-23a-3p was transported to the microglia via M2 macrophage-EVs, which elevated the mechanical allodynia and the thermal hyperalgesia thresholds in mice model. The miR-23a-3p downstream target, USP5, was found to stabilize HDAC2 via deubiquitination to promote its expression while inhibiting the expression of NRF2. Taken together, the key findings of the current study demonstrate that macrophage-derived EVs containing miR-23a-3p regulates the HDAC2/NRF2 axis by decreasing USP5 expression to alleviate inflammatory pain, which may provide novel therapeutic targets for the treatment of inflammatory pain.
Collapse
Affiliation(s)
- Yao Qu
- Department of Pain Management, The First Hospital of Jilin University, Changchun, P.R. China
| | - Yunhe Xu
- Department of Stomatology, The First Hospital of Jilin University, Changchun, P.R. China
| | - Yuncheng Jiang
- Department of Anesthesiology, Dehui People's Hospital, Dehui, P.R. China
| | - Dehai Yu
- The Laboratory of Cancer Precision Medicine, The First Hospital of Jilin University, Changchun, P.R. China
| | - Xi Jiang
- Health Promotion Center, The First Hospital of Jilin University, Changchun, P.R. China
| | - Ling Zhao
- Department of Rheumatology, The First Hospital of Jilin University, Changchun, P.R. China
| |
Collapse
|
23
|
Ubiquitin-Specific Peptidase 5 is Involved in the Proliferation of Trophoblast Cells by Regulating Wnt/β-Catenin Signaling. Mol Biotechnol 2021; 63:686-693. [PMID: 33977498 DOI: 10.1007/s12033-021-00330-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 04/23/2021] [Indexed: 12/19/2022]
Abstract
Preeclampsia (PE) is a pathologic condition in pregnant women which accounts for the inhibition of proliferation, migration and invasion of trophoblast cells. This study aimed to investigate the regulation of ubiquitin-specific peptidase 5 (USP5) on the trophoblast cells in PE. Expressions of USP5 in the placentas of PE patients and healthy donors were examined by qRT-PCR and Western blot. Hypoxia/reoxygenation (H/R) model in trophoblast cells was further established. Cell viability was examined using CCK-8 assay. Finally, the effect of overexpression and silence of USP5 using lentivirus transduction was studied. Our results showed that USP5 was lowly expressed in the placentas of PE patients as well as in H/R-induced trophoblast cells. In the experiments of overexpression, USP5 promoted the proliferation of trophoblast cells, and up-regulated the expressions of β-catenin and the downstream signals c-Myc and Cyclin D1 in trophoblast cells. On the other hand, silence of USP5 elicited the opposite results. The overexpression of USP5 in the H/R model greatly released the H/R-induced inhibition in the trophoblast cells, and moderated the down-regulation of β-catenin and c-Myc induced by H/R. We concluded that USP5 promoted the proliferation of trophoblast cells via the up-regulation of the Wnt/β-catenin signaling pathway.
Collapse
|
24
|
Zhang H, Zheng H, Zhu J, Dong Q, Wang J, Fan H, Chen Y, Zhang X, Han X, Li Q, Lu J, Tong Y, Chen Z. Ubiquitin-Modified Proteome of SARS-CoV-2-Infected Host Cells Reveals Insights into Virus-Host Interaction and Pathogenesis. J Proteome Res 2021; 20:2224-2239. [PMID: 33666082 PMCID: PMC7945586 DOI: 10.1021/acs.jproteome.0c00758] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Indexed: 12/12/2022]
Abstract
The outbreak of coronavirus disease 2019 (COVID-19), which is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has posed a serious threat to global public health. The mechanism of pathogenesis and the host immune response to SARS-CoV-2 infection are largely unknown. In the present study, we applied a quantitative proteomic technology to identify and quantify the ubiquitination changes that occur in both the virus and the Vero E6 cells during SARS-CoV-2 infection. By applying label-free, quantitative liquid chromatography with tandem mass spectrometry proteomics, 8943 lysine ubiquitination sites on 3086 proteins were identified, of which 138 sites on 104 proteins were quantified as significantly upregulated, while 828 sites on 447 proteins were downregulated at 72 h post-infection. Bioinformatics analysis suggested that SARS-CoV-2 infection might modulate host immune responses through the ubiquitination of important proteins, including USP5, IQGAP1, TRIM28, and Hsp90. Ubiquitination modification was also observed on 11 SAR-CoV-2 proteins, including proteins involved in virus replication and inhibition of the host innate immune response. Our study provides new insights into the interaction between SARS-CoV-2 and the host as well as potential targets for the prevention and treatment of COVID-19.
Collapse
Affiliation(s)
- Huan Zhang
- Key Laboratory of Zoonotic of Liaoning Province,
College of Animal Science and Veterinary Medicine, Shenyang Agricultural
University, Shenyang 110866, Liaoning Province, P. R.
China
| | - Huanying Zheng
- Guangdong Provincial Center for Disease
Control and Prevention, Guangzhou 511430, P. R.
China
| | - Jinying Zhu
- Key Laboratory of Zoonotic of Liaoning Province,
College of Animal Science and Veterinary Medicine, Shenyang Agricultural
University, Shenyang 110866, Liaoning Province, P. R.
China
| | - Qiao Dong
- Key Laboratory of Zoonotic of Liaoning Province,
College of Animal Science and Veterinary Medicine, Shenyang Agricultural
University, Shenyang 110866, Liaoning Province, P. R.
China
| | - Jin Wang
- School of Public Health, Sun Yat-sen
University, Guangzhou 510080, P. R. China
| | - Huahao Fan
- Beijing Advanced Innovation Center for Soft Matter
Science and Engineering, Beijing University of Chemical
Technology, Beijing 100029, P. R. China
| | - Yangzhen Chen
- Beijing Advanced Innovation Center for Soft Matter
Science and Engineering, Beijing University of Chemical
Technology, Beijing 100029, P. R. China
| | - Xi Zhang
- Key Laboratory of Zoonotic of Liaoning Province,
College of Animal Science and Veterinary Medicine, Shenyang Agricultural
University, Shenyang 110866, Liaoning Province, P. R.
China
| | - Xiaohu Han
- Key Laboratory of Zoonotic of Liaoning Province,
College of Animal Science and Veterinary Medicine, Shenyang Agricultural
University, Shenyang 110866, Liaoning Province, P. R.
China
| | - Qianlin Li
- School of Public Health, Sun Yat-sen
University, Guangzhou 510080, P. R. China
| | - Jiahai Lu
- School of Public Health, Sun Yat-sen
University, Guangzhou 510080, P. R. China
| | - Yigang Tong
- Beijing Advanced Innovation Center for Soft Matter
Science and Engineering, Beijing University of Chemical
Technology, Beijing 100029, P. R. China
| | - Zeliang Chen
- Key Laboratory of Zoonotic of Liaoning Province,
College of Animal Science and Veterinary Medicine, Shenyang Agricultural
University, Shenyang 110866, Liaoning Province, P. R.
China
- School of Public Health, Sun Yat-sen
University, Guangzhou 510080, P. R. China
- Beijing Advanced Innovation Center for Soft Matter
Science and Engineering, Beijing University of Chemical
Technology, Beijing 100029, P. R. China
| |
Collapse
|
25
|
DNA double-strand break repair: Putting zinc fingers on the sore spot. Semin Cell Dev Biol 2021; 113:65-74. [DOI: 10.1016/j.semcdb.2020.09.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/22/2020] [Accepted: 09/07/2020] [Indexed: 12/15/2022]
|
26
|
Lian J, Liu C, Guan X, Wang B, Yao Y, Su D, Ma Y, Fang L, Zhang Y. Ubiquitin specific peptidase 5 enhances STAT3 signaling and promotes migration and invasion in Pancreatic Cancer. J Cancer 2020; 11:6802-6811. [PMID: 33123271 PMCID: PMC7592018 DOI: 10.7150/jca.48536] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/14/2020] [Indexed: 12/29/2022] Open
Abstract
Purpose: Ubiquitin specific peptidase 5 (USP5) has been reported to promote the progression of several malignant tumors. It may affect cancer development via modulating cell cycle and colony formation. In pancreatic cancer, the biological function of USP5, especially in migration and invasion remains unclear. Methods: USP5 protein expression levels in primary pancreatic cancer and lymph node metastasis tissues were detected using immunohistochemistry (IHC). χ2 test, Kaplan-Meier analysis, univariate and multivariate analyses were used to evaluate the relationship between USP5 expression and clinicopathological feature. RT-qPCR were carried out to quantitate the mRNA expression levels of USP5 in pancreatic cancer cell lines. CCK8 and Colony formation assay were performed to prove how USP5 works in proliferation. Evaluation of tumor metastasis was made by Transwell and wound healing assay. EMT and STAT3 signaling related markers were detected by western blot. Results: (1) USP5 protein expression levels were related to tumor differentiation, CEA and CA19-9 level. (2) Univariate and multivariate analyses showed that high USP5 expression is an unfavorable prognostic factor for pancreatic cancer. Kaplan-Meier analysis directly indicated that patients with high USP5 expression had shorter overall survival. (3) Increased USP5 expression is related to pancreatic cancer in both proliferation and metastasis. (4) USP5 was proved to mediate STAT3 signaling in pancreatic cancer cells. Conclusions: The results suggest that USP5 is highly expressed and might have clinical significance for pancreatic cancer patients. High USP5 expression promotes both progression and metastasis by activating STAT3 signaling. Thus, USP5 might be a potential target in pancreatic cancer treatment.
Collapse
Affiliation(s)
- Jie Lian
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China.,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang Province, China
| | - Chao Liu
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China.,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang Province, China
| | - Xin Guan
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China.,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang Province, China
| | - Bojun Wang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China.,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang Province, China
| | - Yuanfei Yao
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China.,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang Province, China
| | - Dan Su
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China
| | - Yue Ma
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China
| | - Lin Fang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China.,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang Province, China
| | - Yanqiao Zhang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China.,Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, Heilongjiang Province, China
| |
Collapse
|
27
|
Yao J, Li C, Shi L, Lu Y, Liu X. Zebrafish ubiquitin-specific peptidase 5 (USP5) activates interferon resistance to the virus by increase the expression of RIG-I. Gene 2020; 751:144761. [DOI: 10.1016/j.gene.2020.144761] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/02/2020] [Accepted: 05/08/2020] [Indexed: 12/15/2022]
|
28
|
Jia P, Zhang W, Xiang Y, Lu X, Liu W, Jia K, Yi M. Ubiquitin-specific protease 5 was involved in the interferon response to RGNNV in sea perch (Lateolabrax japonicus). FISH & SHELLFISH IMMUNOLOGY 2020; 103:239-247. [PMID: 32437860 DOI: 10.1016/j.fsi.2020.04.065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 03/25/2020] [Accepted: 04/29/2020] [Indexed: 06/11/2023]
Abstract
Deubiquitinases are widely involved in the regulation of the virus-triggered type I interferon (IFN) signaling. Here, we found sea perch (Lateolabrax japonicus) ubiquitin-specific protease 5 (LjUSP5) was a negative regulatory factor of the red-spotted grouper nervous necrosis virus (RGNNV)-triggered IFN response. LjUSP5 encoded a polypeptide of 830 amino acids, containing a zinc finger UBP domain (residues 197-270 aa), two ubiquitin-associated domains (residues 593-607 aa; 628-665 aa), and one UBP domain (residues 782-807 aa), and shared the closest genetic relationship with the USP5 of Larimichthys crocea. Quantitative RT-PCR analysis showed that LjUSP5 was ubiquitously expressed and up-regulated significantly in all inspected tissues post RGNNV infection, and its transcripts significantly increased in brain, liver and kidney tissues post RGNNV infection. LjUSP5 was up-regulated in cultured LJB cells after poly I:C and RGNNV treatments. In addition, overexpression of LjUSP5 significantly inhibited the activation of zebrafish IFN 1 promoter and promoted RGNNV replication in vitro. Furthermore, LjUSP5 inhibited the activation of zebrafish IFN 1 promoter induced by key genes of retinoic acid-inducible gene I-like receptors signaling pathway. Our findings provides useful information for further elucidating the mechanism underlying NNV infection.
Collapse
Affiliation(s)
- Peng Jia
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, Guangdong, China; Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangdong, China.
| | - Wanwan Zhang
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, Guangdong, China; Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangdong, China.
| | - Yangxi Xiang
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, Guangdong, China; Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangdong, China.
| | - Xiaobing Lu
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, Guangdong, China; Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangdong, China.
| | - Wei Liu
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, Guangdong, China; Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangdong, China.
| | - Kuntong Jia
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, Guangdong, China; Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangdong, China.
| | - Meisheng Yi
- School of Marine Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, Guangdong, China; Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangdong, China.
| |
Collapse
|
29
|
The role of deubiquitinating enzymes in cancer drug resistance. Cancer Chemother Pharmacol 2020; 85:627-639. [PMID: 32146496 DOI: 10.1007/s00280-020-04046-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 02/19/2020] [Indexed: 12/18/2022]
Abstract
Drug resistance is a well-known phenomenon leading to a reduction in the effectiveness of pharmaceutical treatments. Resistance to chemotherapeutic agents can involve various intrinsic cellular processes including drug efflux, increased resistance to apoptosis, increased DNA damage repair capabilities in response to platinum salts or other DNA-damaging drugs, drug inactivation, drug target alteration, epithelial-mesenchymal transition (EMT), inherent cell heterogeneity, epigenetic effects, or any combination of these mechanisms. Deubiquitinating enzymes (DUBs) reverse ubiquitination of target proteins, maintaining a balance between ubiquitination and deubiquitination of proteins to maintain cell homeostasis. Increasing evidence supports an association of altered DUB activity with development of several cancers. Thus, DUBs are promising candidates for targeted drug development. In this review, we outline the involvement of DUBs, particularly ubiquitin-specific proteases, and their roles in drug resistance in different types of cancer. We also review potential small molecule DUB inhibitors that can be used as drugs for cancer treatment.
Collapse
|
30
|
Wu L, Zhang C, Chu M, Fan Y, Wei L, Li Z, Yao Y, Zhuang W. miR-125a suppresses malignancy of multiple myeloma by reducing the deubiquitinase USP5. J Cell Biochem 2020; 121:642-650. [PMID: 31452281 DOI: 10.1002/jcb.29309] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 07/15/2019] [Indexed: 12/14/2022]
Abstract
miR-125a is a microRNA that is frequently diminished in various human malignancies. However, the mechanism by which impaired miR-125a promotes cancer growth remains undefined. In this study, we investigated the role of miR-125a in the proliferation and apoptosis of multiple myeloma (MM). To do this, we used MM tissue samples (from 40 anonymous patients), normal matched control samples, and five MM-derived cell lines. We also established a mouse model of MM xenograft to explore the effect of overexpression of miR-125a on the MM growth in vivo. Quantitative real-time polymerase chain reaction revealed that the miR-125a expression was broadly reduced in MM tissues and cell lines. The impairment of miR-125a in MM tissues was functionally relevant because the overexpression of miR-125a remarkably decreased the cell viability and colony-forming activity, at least in part, by promoting apoptosis in two miR-125a-deficient MM cell lines: NCI-H929 and U266. Interestingly, we also discovered that the human gene encoding the ubiquitin-specific peptidase 5 (USP5), which is known to promote cellular deubiquitination and ubiquitin/proteasome-dependent proteolysis, was a direct transcriptional target for miR-125a to repress. More importantly, the heterologous expression of USP5 significantly reversed the growth-inhibitory effects of miR-125a on MM cells in vitro. In the mouse xenograft model, overexpressed miR-125a prominently inhibited the growth of MM tumors and concomitantly reduced the expression of USP5 in tumor tissues. These results suggest that miR-125a inhibits the expression of USP5, thereby mitigating the proliferation and survival of malignant MM cells. We propose that USP5 acts as an oncoprotein in miR-125a-missing cancers.
Collapse
Affiliation(s)
- Liting Wu
- Medical Laboratory, Shanghai Shidong Hospital, Shanghai, Yangpu, China
| | - Cui Zhang
- Medical Laboratory, Shanghai Shidong Hospital, Shanghai, Yangpu, China
| | - Min Chu
- Medical Laboratory, Shanghai Shidong Hospital, Shanghai, Yangpu, China
| | - Yingchao Fan
- Medical Laboratory, Shanghai Shidong Hospital, Shanghai, Yangpu, China
| | - Lu Wei
- Medical Laboratory, Shanghai Shidong Hospital, Shanghai, Yangpu, China
| | - Zhumeng Li
- Medical Laboratory, Shanghai Shidong Hospital, Shanghai, Yangpu, China
| | - Yonghua Yao
- Medical Laboratory, Shanghai Shidong Hospital, Shanghai, Yangpu, China
| | - Wenfang Zhuang
- Medical Laboratory, Shanghai Shidong Hospital, Shanghai, Yangpu, China
| |
Collapse
|
31
|
Pavey S, Pinder A, Fernando W, D'Arcy N, Matigian N, Skalamera D, Lê Cao KA, Loo-Oey D, Hill MM, Stark M, Kimlin M, Burgess A, Cloonan N, Sturm RA, Gabrielli B. Multiple interaction nodes define the postreplication repair response to UV-induced DNA damage that is defective in melanomas and correlated with UV signature mutation load. Mol Oncol 2019; 14:22-41. [PMID: 31733171 PMCID: PMC6944116 DOI: 10.1002/1878-0261.12601] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/06/2019] [Accepted: 11/14/2019] [Indexed: 01/25/2023] Open
Abstract
Ultraviolet radiation‐induced DNA mutations are a primary environmental driver of melanoma. The reason for this very high level of unrepaired DNA lesions leading to these mutations is still poorly understood. The primary DNA repair mechanism for UV‐induced lesions, that is, the nucleotide excision repair pathway, appears intact in most melanomas. We have previously reported a postreplication repair mechanism that is commonly defective in melanoma cell lines. Here we have used a genome‐wide approach to identify the components of this postreplication repair mechanism. We have used differential transcript polysome loading to identify transcripts that are associated with UV response, and then functionally assessed these to identify novel components of this repair and cell cycle checkpoint network. We have identified multiple interaction nodes, including global genomic nucleotide excision repair and homologous recombination repair, and previously unexpected MASTL pathway, as components of the response. Finally, we have used bioinformatics to assess the contribution of dysregulated expression of these pathways to the UV signature mutation load of a large melanoma cohort. We show that dysregulation of the pathway, especially the DNA damage repair components, are significant contributors to UV mutation load, and that dysregulation of the MASTL pathway appears to be a significant contributor to high UV signature mutation load.
Collapse
Affiliation(s)
- Sandra Pavey
- Diamantina Institute, TRI, The University of Queensland, Woolloongabba, QLD, Australia
| | - Alex Pinder
- Diamantina Institute, TRI, The University of Queensland, Woolloongabba, QLD, Australia
| | - Winnie Fernando
- Mater Research, TRI, The University of Queensland, Woolloongabba, QLD, Australia
| | - Nicholas D'Arcy
- Mater Research, TRI, The University of Queensland, Woolloongabba, QLD, Australia
| | - Nicholas Matigian
- Diamantina Institute, TRI, The University of Queensland, Woolloongabba, QLD, Australia.,QFAB Bioinformatics, The University of Queensland, Brisbane, QLD, Australia
| | - Dubravka Skalamera
- Diamantina Institute, TRI, The University of Queensland, Woolloongabba, QLD, Australia.,Mater Research, TRI, The University of Queensland, Woolloongabba, QLD, Australia
| | - Kim-Anh Lê Cao
- Diamantina Institute, TRI, The University of Queensland, Woolloongabba, QLD, Australia
| | - Dorothy Loo-Oey
- Diamantina Institute, TRI, The University of Queensland, Woolloongabba, QLD, Australia
| | - Michelle M Hill
- Diamantina Institute, TRI, The University of Queensland, Woolloongabba, QLD, Australia.,QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Mitchell Stark
- Diamantina Institute, TRI, The University of Queensland, Woolloongabba, QLD, Australia
| | - Michael Kimlin
- University of the Sunshine Coast, Sippy Downs, QLD, Australia
| | | | - Nicole Cloonan
- QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Richard A Sturm
- Diamantina Institute, TRI, The University of Queensland, Woolloongabba, QLD, Australia
| | - Brian Gabrielli
- Diamantina Institute, TRI, The University of Queensland, Woolloongabba, QLD, Australia.,Mater Research, TRI, The University of Queensland, Woolloongabba, QLD, Australia
| |
Collapse
|
32
|
Mann MK, Franzoni I, de Freitas RF, Tempel W, Houliston S, Smith L, Vedadi M, Arrowsmith CH, Harding RJ, Schapira M. Discovery of Small Molecule Antagonists of the USP5 Zinc Finger Ubiquitin-Binding Domain. J Med Chem 2019; 62:10144-10155. [PMID: 31663737 DOI: 10.1021/acs.jmedchem.9b00988] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
USP5 disassembles unanchored polyubiquitin chains to recycle free monoubiquitin, and is one of the 12 ubiquitin specific proteases featuring a zinc finger ubiquitin-binding domain (ZnF-UBD). This distinct structural module has been associated with substrate positioning or allosteric modulation of catalytic activity, but its cellular function remains unclear. We screened a chemical library focused on the ZnF-UBD of USP5, crystallized hits in complex with the protein, and generated a preliminary structure-activity relationship, which enables the development of more potent and selective compounds. This work serves as a framework for the discovery of a chemical probe to delineate the function of USP5 ZnF-UBD in proteasomal degradation and other ubiquitin signaling pathways in health and disease.
Collapse
Affiliation(s)
- Mandeep K Mann
- Structural Genomics Consortium, University of Toronto, MaRS Centre , South Tower, 101 College St., Suite 700 , Toronto , Ontario M5G 1L7 , Canada.,Department of Pharmacology and Toxicology , University of Toronto , 1 King's College Circle , Toronto , Ontario M5S 1A8 , Canada
| | - Ivan Franzoni
- Department of Chemistry , University of Toronto , 80 St. George St. , Toronto , Ontario M5S 3H6 , Canada
| | - Renato Ferreira de Freitas
- Structural Genomics Consortium, University of Toronto, MaRS Centre , South Tower, 101 College St., Suite 700 , Toronto , Ontario M5G 1L7 , Canada
| | - Wolfram Tempel
- Structural Genomics Consortium, University of Toronto, MaRS Centre , South Tower, 101 College St., Suite 700 , Toronto , Ontario M5G 1L7 , Canada
| | - Scott Houliston
- University Health Network , 661 University Avenue , Toronto , Ontario M5G 2C4 , Canada
| | - Leanna Smith
- Structural Genomics Consortium, University of Toronto, MaRS Centre , South Tower, 101 College St., Suite 700 , Toronto , Ontario M5G 1L7 , Canada
| | - Masoud Vedadi
- Structural Genomics Consortium, University of Toronto, MaRS Centre , South Tower, 101 College St., Suite 700 , Toronto , Ontario M5G 1L7 , Canada.,Department of Pharmacology and Toxicology , University of Toronto , 1 King's College Circle , Toronto , Ontario M5S 1A8 , Canada
| | - Cheryl H Arrowsmith
- Structural Genomics Consortium, University of Toronto, MaRS Centre , South Tower, 101 College St., Suite 700 , Toronto , Ontario M5G 1L7 , Canada.,University Health Network , 661 University Avenue , Toronto , Ontario M5G 2C4 , Canada
| | - Rachel J Harding
- Structural Genomics Consortium, University of Toronto, MaRS Centre , South Tower, 101 College St., Suite 700 , Toronto , Ontario M5G 1L7 , Canada
| | - Matthieu Schapira
- Structural Genomics Consortium, University of Toronto, MaRS Centre , South Tower, 101 College St., Suite 700 , Toronto , Ontario M5G 1L7 , Canada.,Department of Pharmacology and Toxicology , University of Toronto , 1 King's College Circle , Toronto , Ontario M5S 1A8 , Canada
| |
Collapse
|
33
|
Ning F, Xin H, Liu J, Lv C, Xu X, Wang M, Wang Y, Zhang W, Zhang X. Structure and function of USP5: Insight into physiological and pathophysiological roles. Pharmacol Res 2019; 157:104557. [PMID: 31756387 DOI: 10.1016/j.phrs.2019.104557] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 11/18/2019] [Accepted: 11/18/2019] [Indexed: 11/16/2022]
Abstract
Deubiquitinase (DUB)-mediated cleavage of ubiquitin chains from substrate proteins plays a crucial role in various cellular processes, such as DNA repair and protein stabilization and localization. DUBs can be classified into five families based on their sequence and structural homology, and the majority belong to the ubiquitin-specific proteinase (USP) family. As one of the USPs, ubiquitin-specific proteinase 5 (USP5) is unique in that it can specifically recognize unanchored (not conjugated to target proteins) polyubiquitin and is essential for maintaining homeostasis of the monoubiquitin pool. USP5 has also been implicated in a wide variety of cellular events. In the present review, we focus on USP5 and provide a comprehensive overview of the current knowledge regarding its structure, physiological roles in multiple cellular events, and pathophysiological roles in relevant diseases, especially cancer. Signaling pathways and emerging pharmacological profiles of USP5 are also introduced, which fully embody the therapeutic potential of USP5 for human diseases ranging from cancer to neurological diseases.
Collapse
Affiliation(s)
- Fengling Ning
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Hong Xin
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Junqiu Liu
- Laboratory of Medicinal Plant Biotechnology, College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Chao Lv
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xin Xu
- Suzhou Institute of Systems Medicine, Center for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou 215123, China
| | - Mengling Wang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yinhang Wang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Weidong Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Xuemei Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China.
| |
Collapse
|
34
|
Du Y, Lin J, Zhang R, Yang W, Quan H, Zang L, Han Y, Li B, Sun H, Wu J. Ubiquitin specific peptidase 5 promotes ovarian cancer cell proliferation through deubiquitinating HDAC2. Aging (Albany NY) 2019; 11:9778-9793. [PMID: 31727867 PMCID: PMC6874447 DOI: 10.18632/aging.102425] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 10/28/2019] [Indexed: 12/14/2022]
Abstract
Globally, epithelial ovarian cancer (EOC) is the most common gynecological malignancy with poor prognosis. The expression and oncogenic roles of ubiquitin specific peptidase 5 (USP5) have been reported in several cancers except EOC. In the current study, USP5 amplification was highly prevalent in patients with EOC and associated with higher mRNA expression of USP5. USP5 amplification and overexpression was positively correlated with poor prognosis of patients of ovarian serous carcinomas. Disruption of USP5 profoundly repressed cell proliferation by inducing cell cycle G0/G1 phase arrest in ovarian cancer cells. Additionally, USP5 knockdown inhibited xenograft growth in nude mice. Knockdown of USP5 decreased histone deacetylase 2 (HDAC2) expression and increased p27 (an important cell cycle inhibitor) expression in vitro and in vivo. The promoting effects of USP5 overexpression on cell proliferation and cell cycle transition, as well as the inhibitory effects of USP5 overexpression on p27 expression were mediated by HDAC2. Moreover, USP5 interacted with HDAC2, and disruption of USP5 enhanced the ubiquitination of HDAC2. HDAC2 protein was positively correlated USP5 protein, and negatively correlated with p27 protein in ovarian serous carcinomas tissues. Collectively, our data suggest the oncogenic function of USP5 and the potential regulatory mechanisms in ovarian carcinogenesis.
Collapse
Affiliation(s)
- Yanhua Du
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, P. R. China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, P. R. China
| | - Jun Lin
- Pathology Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P. R. China
| | - Rulin Zhang
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, P. R. China
| | - Wanli Yang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
| | - Heng Quan
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, P. R. China
| | - Lijuan Zang
- Pathology Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P. R. China
| | - Yaqin Han
- Pathology Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, P. R. China
| | - Bing Li
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
| | - Hong Sun
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200011, P. R. China
| | - Jun Wu
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, P. R. China
| |
Collapse
|
35
|
Zachariah S, Gray DA. Deubiquitinating Enzymes in Model Systems and Therapy: Redundancy and Compensation Have Implications. Bioessays 2019; 41:e1900112. [DOI: 10.1002/bies.201900112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/06/2019] [Indexed: 01/09/2023]
Affiliation(s)
- Sarah Zachariah
- Centre for Cancer TherapeuticsOttawa Hospital Research Institute 501 Smyth Box 926 Ottawa ON K1H 8L6 Canada
- Department of Biochemistry, Microbiology and ImmunologyUniversity of Ottawa 451 Smyth Rd Ottawa ON K1H 8M5 Canada
| | - Douglas A. Gray
- Centre for Cancer TherapeuticsOttawa Hospital Research Institute 501 Smyth Box 926 Ottawa ON K1H 8L6 Canada
- Department of Biochemistry, Microbiology and ImmunologyUniversity of Ottawa 451 Smyth Rd Ottawa ON K1H 8M5 Canada
| |
Collapse
|
36
|
Young MJ, Hsu KC, Lin TE, Chang WC, Hung JJ. The role of ubiquitin-specific peptidases in cancer progression. J Biomed Sci 2019; 26:42. [PMID: 31133011 PMCID: PMC6537419 DOI: 10.1186/s12929-019-0522-0] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 04/16/2019] [Indexed: 12/13/2022] Open
Abstract
Protein ubiquitination is an important mechanism for regulating the activity and levels of proteins under physiological conditions. Loss of regulation by protein ubiquitination leads to various diseases, such as cancer. Two types of enzymes, namely, E1/E2/E3 ligases and deubiquitinases, are responsible for controlling protein ubiquitination. The ubiquitin-specific peptidases (USPs) are the main members of the deubiquitinase family. Many studies have addressed the roles of USPs in various diseases. An increasing number of studies have indicated that USPs are critical for cancer progression, and some USPs have been used as targets to develop inhibitors for cancer prevention. Herein we collect and organize most of the recent studies on the roles of USPs in cancer progression and discuss the development of USP inhibitors for cancer therapy in the future.
Collapse
Affiliation(s)
- Ming-Jer Young
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, 701, Taiwan
| | - Kai-Cheng Hsu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Biomedical Commercialization Center, Taipei Medical University, Taipei, Taiwan
| | - Tony Eight Lin
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Wen-Chang Chang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jan-Jong Hung
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, 701, Taiwan. .,The Ph.D. Program for Neural Regenerative Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
37
|
Wang Z, Zhang H, Liu C, Xing J, Chen XL. A Deubiquitinating Enzyme Ubp14 Is Required for Development, Stress Response, Nutrient Utilization, and Pathogenesis of Magnaporthe oryzae. Front Microbiol 2018; 9:769. [PMID: 29720973 PMCID: PMC5915541 DOI: 10.3389/fmicb.2018.00769] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 04/04/2018] [Indexed: 02/02/2023] Open
Abstract
Ubiquitination is an essential protein modification in eukaryotic cells, which is reversible. Deubiquitinating enzymes (DUBs) catalyze deubiquitination process to reverse ubiquitination, maintain ubiquitin homeostasis or promote protein degradation by recycling ubiquitins. In order to investigate effects of deubiquitination process in plant pathogenic fungus Magnaporthe oryzae, we generated deletion mutants of MoUBP14. Ortholog of MoUbp14 was reported to play general roles in ubiquitin-mediated protein degradation in Saccharomyces cerevisiae. The ΔMoubp14 mutant lost its pathogenicity and was severely reduced in mycelial growth, sporulation, carbon source utilization, and increased in sensitivity to distinct stresses. The mutant was blocked in penetration, which could due to defect in turgor generation. It is also blocked in invasive growth, which could due to reduction in stress tolerance and nutrient utilization. Deletion of UBP14 also led to accumulation of free polyubiquitin chains. Pulldown assay identified some proteins related to carbohydrate metabolism and stress response may putatively interact with MoUbp14, including two key rate-limiting enzymes of gluconeogenesis, MoFbp1 and MoPck1. These two proteins were degraded when the glucose was supplied to M. oryzae grown in low glucose media for a short period of time (∼12 h), and this process required MoUbp14. In summary, pleiotropic phenotypes of the deletion mutants indicated that MoUbp14 is required for different developments and pathogenicity of M. oryzae.
Collapse
Affiliation(s)
- Zhao Wang
- The Provincial Key Lab of Plant Pathology of Hubei Province, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Hong Zhang
- The Provincial Key Lab of Plant Pathology of Hubei Province, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Caiyun Liu
- The Provincial Key Lab of Plant Pathology of Hubei Province, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Junjie Xing
- State Key Laboratory of Hybrid Rice, Hunan Hybrid Rice Research Center, Changsha, China
| | - Xiao-Lin Chen
- The Provincial Key Lab of Plant Pathology of Hubei Province, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China.,State Key Laboratory of Hybrid Rice, Hunan Hybrid Rice Research Center, Changsha, China
| |
Collapse
|
38
|
Nishi R. Balancing act: To be, or not to be ubiquitylated. Mutat Res 2017; 803-805:43-50. [PMID: 28764946 DOI: 10.1016/j.mrfmmm.2017.07.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 06/30/2017] [Accepted: 07/17/2017] [Indexed: 01/13/2023]
Abstract
DNA double-strand breaks (DSBs) are one of the most deleterious DNA lesions. Appropriate repair of DSB either by homologous recombination or non-homologous end-joining is critical for maintaining genome stability and fitness. DSB repair cooperates with cellular signalling networks, namely DSB response (DDR), which plays pivotal roles in the choice of DSB repair pathway, orchestrating recruitment of DDR factors to site of damage, transcription suppression and cell cycle checkpoint activation. It has been revealed that these mechanisms are strictly regulated, in time and space, by complex and minute ubiquitylation-mediated reactions. Furthermore, balancing the ubiquitylation status of the DDR and DSB repair proteins by deubiquitylation, which is carried out by deubiquitylating enzymes (DUBs), is also found to be important. Recent findings have uncovered that DUBs are involved in various aspects of both DDR and DSB repair by counteracting non-proteolytic ubiquitylations in addition to protecting substrates from proteasomal degradation by removing proteolytic ubiquitylation. An advanced understanding of the detailed molecular mechanisms of the "balancing act" between ubiquitylation and deubiquitylation will provide novel therapeutic targets for diseases caused by dysfunction of DDR and DSB repair.
Collapse
Affiliation(s)
- Ryotaro Nishi
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Shiga, 525-8577, Japan.
| |
Collapse
|
39
|
USP5 promotes tumorigenesis and progression of pancreatic cancer by stabilizing FoxM1 protein. Biochem Biophys Res Commun 2017; 492:48-54. [PMID: 28807830 DOI: 10.1016/j.bbrc.2017.08.040] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 08/10/2017] [Indexed: 02/08/2023]
Abstract
Increased ubiquitin-specific protease 5 (USP5) has been associated with tumorigenesis of malignancy including glioblastoma, melanoma and hepatocellular carcinoma. However, the role of USP5 in tumorigenesis of pancreatic ductal adenocarcinoma (PDAC) has not been studied yet. In this study, we demonstrated that USP5 was significantly upregulated in a panel of PDAC cell lines and correlated with FoxM1 protein expression. USP5 knockdown inhibited proliferation of PANC-1 and SW1990, two PDAC cell lines. In the mouse xenografted pancreatic tumor model, suppression of USP5 significantly decreased tumor growth, correlated with down regulation of FoxM1. Additionally, we found that overexpression of USP5 stabilized the FoxM1 protein in PDAC cells. Overexpression of USP5 extended the half-life of FoxM1. Knockdown of USP5 in PANC-1 cells decreased FoxM1 protein level while the proteasome inhibitor MG-132 treatment restored FoxM1 expression. We also found that endogenous USP5 was coimmunoprecipitated with an endogenous FoxM1 from PANC-1 cells while FoxM1 was also coimmunoprecipitated with USP5. Furthermore, we also confirmed that USP5 regulated proliferation of PDAC via FoxM1 by rescuing the inhibitory effect of USP5 knockdown with ectopic expression of FoxM1 in USP5-depleted cells. Taken together, our study demonstrates that USP5 plays a critical role in tumorigenesis and progression of pancreatic cancer by stabilizing FoxM1 protein, and provides a rationale for USP5 being a potential therapeutic approach against PDAC.
Collapse
|
40
|
Kaistha BP, Krattenmacher A, Fredebohm J, Schmidt H, Behrens D, Widder M, Hackert T, Strobel O, Hoheisel JD, Gress TM, Buchholz M. The deubiquitinating enzyme USP5 promotes pancreatic cancer via modulating cell cycle regulators. Oncotarget 2017; 8:66215-66225. [PMID: 29029505 PMCID: PMC5630405 DOI: 10.18632/oncotarget.19882] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 07/11/2017] [Indexed: 11/25/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal solid tumors. With an overall five-year survival rate remaining below 6%, there is an explicit need to search for new molecular targets for therapeutic interventions. We undertook a barcode labelled short-hairpin (shRNA) library screen in pancreatic cancer cells in order to identify novel genes promoting cancer survival and progression. Among the candidate genes identified in this screen was the deubiquitinase USP5, which subsequent gene expression analyses demonstrated to be significantly upregulated in primary human pancreatic cancer tissues. Using different knockdown approaches, we show that expression of USP5 is essential for the proliferation and survival of pancreatic cancer cells, tested under different 2D and 3D cell culture conditions as well as in in vivo experiments. These growth inhibition effects upon knockdown of USP5 are mediated primarily by the attenuation of G1/S phase transition in the cells, which is accompanied by accumulation of DNA damage, upregulation of p27, and increased apoptosis rates. Since USP5 is overexpressed in cancer tissues, it can thus potentially serve as a new target for therapeutic interventions, especially given the fact that deubiquitinases are currently emerging as new class of attractive drug targets in cancer.
Collapse
Affiliation(s)
- Brajesh P Kaistha
- Department of Medicine, Division of Gastroenterology, Endocrinology and Metabolism, Philipps-University Marburg, Marburg, Germany
| | - Anja Krattenmacher
- Department of Medicine, Division of Gastroenterology, Endocrinology and Metabolism, Philipps-University Marburg, Marburg, Germany
| | - Johannes Fredebohm
- Division of Functional Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Harald Schmidt
- Department of Medicine, Division of Gastroenterology, Endocrinology and Metabolism, Philipps-University Marburg, Marburg, Germany
| | - Diana Behrens
- Experimantal Pharmacology and Oncology (EPO Berlin-Buch), Berlin, Germany
| | - Miriam Widder
- Institute for Bioprocessing and Analytical Measurement Techniques (IBA-Heiligenstadt), Heilbad Heiligenstadt, Germany
| | - Thilo Hackert
- Department of Surgery, University Clinic Heidelberg, Heidelberg, Germany
| | - Oliver Strobel
- Department of Surgery, University Clinic Heidelberg, Heidelberg, Germany
| | - Jörg D Hoheisel
- Division of Functional Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thomas M Gress
- Department of Medicine, Division of Gastroenterology, Endocrinology and Metabolism, Philipps-University Marburg, Marburg, Germany
| | - Malte Buchholz
- Department of Medicine, Division of Gastroenterology, Endocrinology and Metabolism, Philipps-University Marburg, Marburg, Germany
| |
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
|
Ng CKY, Piscuoglio S, Geyer FC, Burke KA, Pareja F, Eberle CA, Lim RS, Natrajan R, Riaz N, Mariani O, Norton L, Vincent-Salomon A, Wen YH, Weigelt B, Reis-Filho JS. The Landscape of Somatic Genetic Alterations in Metaplastic Breast Carcinomas. Clin Cancer Res 2017; 23:3859-3870. [PMID: 28153863 DOI: 10.1158/1078-0432.ccr-16-2857] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 01/04/2017] [Accepted: 01/23/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Metaplastic breast carcinoma (MBC) is a rare and aggressive histologic type of breast cancer, predominantly of triple-negative phenotype, and characterized by the presence of malignant cells showing squamous and/or mesenchymal differentiation. We sought to define the repertoire of somatic genetic alterations and the mutational signatures of MBCs.Experimental Design: Whole-exome sequencing was performed in 35 MBCs, with 16, 10, and 9 classified as harboring chondroid, spindle, and squamous metaplasia as the predominant metaplastic component. The genomic landscape of MBCs was compared with that of triple-negative invasive ductal carcinomas of no special type (IDC-NST) from The Cancer Genome Atlas. Wnt and PI3K/AKT/mTOR pathway activity was assessed using a qPCR assay.Results: MBCs harbored complex genomes with frequent TP53 (69%) mutations. In contrast to triple-negative IDC-NSTs, MBCs more frequently harbored mutations in PIK3CA (29%), PIK3R1 (11%), ARID1A (11%), FAT1 (11%), and PTEN (11%). PIK3CA mutations were not found in MBCs with chondroid metaplasia. Compared with triple-negative IDC-NSTs, MBCs significantly more frequently harbored mutations in PI3K/AKT/mTOR pathway-related (57% vs. 22%) and canonical Wnt pathway-related (51% vs. 28%) genes. MBCs with somatic mutations in PI3K/AKT/mTOR or Wnt pathway-related genes displayed increased activity of the respective pathway.Conclusions: MBCs are genetically complex and heterogeneous, and are driven by a repertoire of somatic mutations distinct from that of triple-negative IDC-NSTs. Our study highlights the genetic basis and the importance of PI3K/AKT/mTOR and Wnt pathway dysregulation in MBCs and provides a rationale for the metaplastic phenotype and the reported responses to PI3K/AKT/mTOR inhibitors in these tumors. Clin Cancer Res; 23(14); 3859-70. ©2017 AACR.
Collapse
Affiliation(s)
- Charlotte K Y Ng
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York.,Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Salvatore Piscuoglio
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York.,Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Felipe C Geyer
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Pathology, Hospital Israelita Albert Einstein, Instituto Israelita de Ensino e Pesquisa, São Paulo, Brazil
| | - Kathleen A Burke
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Fresia Pareja
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Carey A Eberle
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Raymond S Lim
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Rachael Natrajan
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Nadeem Riaz
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Larry Norton
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Y Hannah Wen
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York. .,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
43
|
Jin WL, Mao XY, Qiu GZ. Targeting Deubiquitinating Enzymes in Glioblastoma Multiforme: Expectations and Challenges. Med Res Rev 2016; 37:627-661. [PMID: 27775833 DOI: 10.1002/med.21421] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 09/06/2016] [Accepted: 09/25/2016] [Indexed: 12/16/2022]
Abstract
Glioblastoma (GBM) is regarded as the most common primary intracranial neoplasm. Despite standard treatment with tumor resection and radiochemotherapy, the outcome remains gloomy. It is evident that a combination of oncogenic gain of function and tumor-suppressive loss of function has been attributed to glioma initiation and progression. The ubiquitin-proteasome system is a well-orchestrated system that controls the fate of most proteins by striking a dynamic balance between ubiquitination and deubiquitination of substrates, having a profound influence on the modulation of oncoproteins, tumor suppressors, and cellular signaling pathways. In recent years, deubiquitinating enzymes (DUBs) have emerged as potential anti-cancer targets due to their targeting several key proteins involved in the regulation of tumorigenesis, apoptosis, senescence, and autophagy. This review attempts to summarize recent studies of GBM-associated DUBs, their roles in various cellular processes, and discuss the relation between DUBs deregulation and gliomagenesis, especially how DUBs regulate glioma stem cells pluripotency, microenvironment, and resistance of radiation and chemotherapy through core stem-cell transcriptional factors. We also review recent achievements and progress in the development of potent and selective reversible inhibitors of DUBs, and attempted to find a potential GBM treatment by DUBs intervention.
Collapse
Affiliation(s)
- Wei-Lin Jin
- Institute of Nano Biomedicine and Engineering, 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, P. R. China.,National Centers for Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Xiao-Yuan Mao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, P. R. China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, P. R. China
| | - Guan-Zhong Qiu
- Department of Neurosurgery, General Hospital of Jinan Military Command, Jinan, 250031, P. R. China
| |
Collapse
|
44
|
Qian G, Ren Y, Zuo Y, Yuan Y, Zhao P, Wang X, Cheng Q, Liu J, Zhang L, Guo T, Liu C, Zheng H. Smurf1 represses TNF-α production through ubiquitination and destabilization of USP5. Biochem Biophys Res Commun 2016; 474:491-496. [DOI: 10.1016/j.bbrc.2016.04.135] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 04/26/2016] [Indexed: 02/02/2023]
|
45
|
Calabretta S, Bielli P, Passacantilli I, Pilozzi E, Fendrich V, Capurso G, Delle Fave G, Sette C. Modulation of PKM alternative splicing by PTBP1 promotes gemcitabine resistance in pancreatic cancer cells. Oncogene 2016; 35:2031-2039. [PMID: 26234680 PMCID: PMC4650269 DOI: 10.1038/onc.2015.270] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 05/11/2015] [Accepted: 06/05/2015] [Indexed: 02/07/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive and incurable disease. Poor prognosis is due to multiple reasons, including acquisition of resistance to gemcitabine, the first-line chemotherapeutic approach. Thus, there is a strong need for novel therapies, targeting more directly the molecular aberrations of this disease. We found that chronic exposure of PDAC cells to gemcitabine selected a subpopulation of cells that are drug-resistant (DR-PDAC cells). Importantly, alternative splicing (AS) of the pyruvate kinase gene (PKM) was differentially modulated in DR-PDAC cells, resulting in promotion of the cancer-related PKM2 isoform, whose high expression also correlated with shorter recurrence-free survival in PDAC patients. Switching PKM splicing by antisense oligonucleotides to favor the alternative PKM1 variant rescued sensitivity of DR-PDAC cells to gemcitabine and cisplatin, suggesting that PKM2 expression is required to withstand drug-induced genotoxic stress. Mechanistically, upregulation of the polypyrimidine-tract binding protein (PTBP1), a key modulator of PKM splicing, correlated with PKM2 expression in DR-PDAC cell lines. PTBP1 was recruited more efficiently to PKM pre-mRNA in DR- than in parental PDAC cells. Accordingly, knockdown of PTBP1 in DR-PDAC cells reduced its recruitment to the PKM pre-mRNA, promoted splicing of the PKM1 variant and abolished drug resistance. Thus, chronic exposure to gemcitabine leads to upregulation of PTBP1 and modulation of PKM AS in PDAC cells, conferring resistance to the drug. These findings point to PKM2 and PTBP1 as new potential therapeutic targets to improve response of PDAC to chemotherapy.
Collapse
Affiliation(s)
- Sara Calabretta
- Department of biomedicine and prevention, University of Rome Tor Vergata, Rome, Italy
- Department of science medical/chirurgic and translational medicine, University of Rome La Sapienza, Rome, Italy
| | - Pamela Bielli
- Laboratory of neuroembriology, Fondazione Santa Lucia, Rome, Italy
| | - Ilaria Passacantilli
- Department of biomedicine and prevention, University of Rome Tor Vergata, Rome, Italy
- Department of science medical/chirurgic and translational medicine, University of Rome La Sapienza, Rome, Italy
| | - Emanuela Pilozzi
- Department of clinic and molecular medicine, University of Rome La Sapienza, Rome, Italy
| | - Volker Fendrich
- Department of surgery, Philipps-University Marburg, Marburg, Germany
| | - Gabriele Capurso
- Department of science medical/chirurgic and translational medicine, University of Rome La Sapienza, Rome, Italy
| | - Gianfranco Delle Fave
- Department of science medical/chirurgic and translational medicine, University of Rome La Sapienza, Rome, Italy
| | - Claudio Sette
- Department of biomedicine and prevention, University of Rome Tor Vergata, Rome, Italy
- Laboratory of neuroembriology, Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
46
|
Abstract
Deubiquitinases are deubiquitinating enzymes (DUBs), which remove ubiquitin from proteins, thus regulating their proteasomal degradation, localization and activity. Here, we discuss DUBs as anti-cancer drug targets.
Collapse
|
47
|
Franklin JL, Mirzaei M, Wearne TA, Homewood J, Goodchild AK, Haynes PA, Cornish JL. Quantitative Proteomic Analysis of the Orbital Frontal Cortex in Rats Following Extended Exposure to Caffeine Reveals Extensive Changes to Protein Expression: Implications for Neurological Disease. J Proteome Res 2016; 15:1455-71. [DOI: 10.1021/acs.jproteome.5b01043] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Jane L. Franklin
- Department of Psychology, ‡Department of Chemistry and Biomolecular Sciences, §Faculty of Human Sciences, and ⊥Department of
Biomedical Sciences, Macquarie University, North Ryde, New South Wales 2109, Australia
| | - Mehdi Mirzaei
- Department of Psychology, ‡Department of Chemistry and Biomolecular Sciences, §Faculty of Human Sciences, and ⊥Department of
Biomedical Sciences, Macquarie University, North Ryde, New South Wales 2109, Australia
| | - Travis A. Wearne
- Department of Psychology, ‡Department of Chemistry and Biomolecular Sciences, §Faculty of Human Sciences, and ⊥Department of
Biomedical Sciences, Macquarie University, North Ryde, New South Wales 2109, Australia
| | - Judi Homewood
- Department of Psychology, ‡Department of Chemistry and Biomolecular Sciences, §Faculty of Human Sciences, and ⊥Department of
Biomedical Sciences, Macquarie University, North Ryde, New South Wales 2109, Australia
| | - Ann K. Goodchild
- Department of Psychology, ‡Department of Chemistry and Biomolecular Sciences, §Faculty of Human Sciences, and ⊥Department of
Biomedical Sciences, Macquarie University, North Ryde, New South Wales 2109, Australia
| | - Paul A. Haynes
- Department of Psychology, ‡Department of Chemistry and Biomolecular Sciences, §Faculty of Human Sciences, and ⊥Department of
Biomedical Sciences, Macquarie University, North Ryde, New South Wales 2109, Australia
| | - Jennifer L. Cornish
- Department of Psychology, ‡Department of Chemistry and Biomolecular Sciences, §Faculty of Human Sciences, and ⊥Department of
Biomedical Sciences, Macquarie University, North Ryde, New South Wales 2109, Australia
| |
Collapse
|
48
|
Abstract
Both proteolytic and nonproteolytic functions of ubiquitination are essential regulatory mechanisms for promoting DNA repair and the DNA damage response in mammalian cells. Deubiquitinating enzymes (DUBs) have emerged as key players in the maintenance of genome stability. In this minireview, we discuss the recent findings on human DUBs that participate in genome maintenance, with a focus on the role of DUBs in the modulation of DNA repair and DNA damage signaling.
Collapse
|
49
|
Potu H, Peterson LF, Pal A, Verhaegen M, Cao J, Talpaz M, Donato NJ. Usp5 links suppression of p53 and FAS levels in melanoma to the BRAF pathway. Oncotarget 2015; 5:5559-69. [PMID: 24980819 PMCID: PMC4170643 DOI: 10.18632/oncotarget.2140] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Usp5 is a deubiquitinase (DUB) previously shown to regulate unanchored polyubiquitin (Ub) chains, p53 transcriptional activity and double-strand DNA repair. In BRAF mutant melanoma cells, Usp5 activity was suppressed by BRAF inhibitor (vemurafenib) in sensitive but not in acquired or intrinsically resistant cells. Usp5 knockdown overcame acquired vemurafenib resistance and sensitized BRAF and NRAS mutant melanoma cells to apoptosis initiated by MEK inhibitor, cytokines or DNA-damaging agents. Knockdown and overexpression studies demonstrated that Usp5 regulates p53 (and p73) levels and alters cell growth and cell cycle distribution associated with p21 induction. Usp5 also regulates the intrinsic apoptotic pathway by modulating p53-dependent FAS expression. A small molecule DUB inhibitor (EOAI3402143) phenocopied the FAS induction and apoptotic sensitization of Usp5 knockdown and fully blocked melanoma tumor growth in mice. Overall, our results demonstrate that BRAF activates Usp5 to suppress cell cycle checkpoint control and apoptosis by blocking p53 and FAS induction; all of which can be restored by small molecule-mediated Usp5 inhibition. These results suggest that Usp5 inhibition can provide an alternate approach in recovery of diminished p53 (or p73) function in melanoma and can add to the targeted therapies already used in the treatment of melanoma.
Collapse
Affiliation(s)
- Harish Potu
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| | - Luke F Peterson
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| | - Anupama Pal
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| | - Monique Verhaegen
- Department of Dermatology, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| | - Juxiang Cao
- Department of Dermatology, Boston University School of Medicine, Boston, Massachusetts
| | - Moshe Talpaz
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| | - Nicholas J Donato
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI
| |
Collapse
|
50
|
Kovács L, Nagy O, Pál M, Udvardy A, Popescu O, Deák P. Role of the deubiquitylating enzyme DmUsp5 in coupling ubiquitin equilibrium to development and apoptosis in Drosophila melanogaster. PLoS One 2015; 10:e0120875. [PMID: 25806519 PMCID: PMC4373725 DOI: 10.1371/journal.pone.0120875] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 01/27/2015] [Indexed: 01/07/2023] Open
Abstract
Protein ubiquitylation is a dynamic process that affects the function and stability of proteins and controls essential cellular processes ranging from cell proliferation to cell death. This process is regulated through the balanced action of E3 ubiquitin ligases and deubiquitylating enzymes (DUB) which conjugate ubiquitins to, and remove them from target proteins, respectively. Our genetic analysis has revealed that the deubiquitylating enzyme DmUsp5 is required for maintenance of the ubiquitin equilibrium, cell survival and normal development in Drosophila. Loss of the DmUsp5 function leads to late larval lethality accompanied by the induction of apoptosis. Detailed analyses at a cellular level demonstrated that DmUsp5 mutants carry multiple abnormalities, including a drop in the free monoubiquitin level, the excessive accumulation of free polyubiquitins, polyubiquitylated proteins and subunits of the 26S proteasome. A shortage in free ubiquitins results in the induction of a ubiquitin stress response previously described only in the unicellular budding yeast. It is characterized by the induction of the proteasome-associated deubiquitylase DmUsp14 and sensitivity to cycloheximide. Removal of DmUsp5 also activates the pro-apoptotic machinery thereby resulting in widespread apoptosis, indicative of an anti-apoptotic role of DmUsp5. Collectively, the pleiotropic effects of a loss of DmUsp5 function can be explained in terms of the existence of a limited pool of free monoubiquitins which makes the ubiquitin-dependent processes mutually interdependent.
Collapse
Affiliation(s)
- Levente Kovács
- Department of Genetics, University of Szeged, Szeged, Hungary
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Olga Nagy
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Margit Pál
- Department of Genetics, University of Szeged, Szeged, Hungary
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Andor Udvardy
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Octavian Popescu
- Molecular Biology Center, Interdisciplinary Research Institute on Bio-Nano-Sciences, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Péter Deák
- Department of Genetics, University of Szeged, Szeged, Hungary
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
- * E-mail:
| |
Collapse
|