1
|
Xu Z, Lei Z, Peng S, Fu X, Xu Y, Pan G. Dysregulation of deubiquitinases in gastric cancer progression. Front Oncol 2024; 14:1456710. [PMID: 39605891 PMCID: PMC11598704 DOI: 10.3389/fonc.2024.1456710] [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: 06/29/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024] Open
Abstract
Gastric cancer (GC), characterized by a high incidence rate, poses significant clinical challenges owing to its poor prognosis despite advancements in diagnostic and therapeutic approaches. Therefore, a comprehensive understanding of the molecular mechanisms driving GC progression is crucial for identifying predictive markers and defining treatment targets. Deubiquitinating enzymes (DUBs), also called deubiquitinases, function as reverse transcriptases within the ubiquitin-proteasome system to counteract protein degradation. Recent findings suggest that DUB dysregulation could be a crucial factor in GC pathogenesis. In this review, we examined recent research findings on DUBs in the context of GC, elucidating their molecular characteristics, categorizations, and roles while also exploring the potential mechanisms underlying their dysregulation in GC. Furthermore, we assessed the therapeutic efficacy of DUB inhibitors in treating malignancies and evaluated the prevalence of aberrant DUB expression in GC.
Collapse
Affiliation(s)
| | | | | | | | | | - Guoqing Pan
- First Affiliated Hospital of Kunming Medical University, Department of Pathology, Kunming, China
| |
Collapse
|
2
|
Lusci Gemignani A, Papotti R, Bomben R, Gattei V, Pozzi S, Donati V, Bettelli S, Forti E, Mansueto G, Di Napoli A, Cox MC, Flenghi L, Rossi P, Volpe G, Dardanis D, Bono C, Guerrini F, Morganti R, Sacchi S, Galimberti S. A new digital droplet PCR method for looking at epigenetics in diffuse large B-cell lymphomas: The role of BMI1, EZH2, and USP22 genes. Int J Lab Hematol 2024. [PMID: 39255961 DOI: 10.1111/ijlh.14363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 07/17/2024] [Indexed: 09/12/2024]
Abstract
INTRODUCTION Epigenetics has been shown to be relevant in oncology: BMI1 overexpression has been reported in leukemias, EZH2 mutations have been found in follicular lymphoma, and USP22 seems to stabilize BMI1 protein. In this study, we measured the expression of BMI1, EZH2, and USP22 in lymph nodes from 56 diffuse large B-cell lymphoma (DLBCL) patients. METHODS A new multiplex digital droplet PCR (ddPCR) has been set up to measure the expression of 4 genes (BMI1, EZH2, USP22, and GAPDH) in the same reaction on RNA extracted from paraffin-embedded tissues. RESULTS The specificity of ddPCR was confirmed by a 100% alignment on the BLAST platform and its repeatability demonstrated by duplicates. A strict correlation between expression of BMI1 and EZH2 and BMI1 and USP22 has been found, and high expression of these genes was correlated with extra-nodal lymphomas. Progression-free survival (PFS) and overall survival (OS) were conditioned by IPI, bone marrow infiltration, and the complete response achievement. High levels of BMI1 and USP22 did not condition the response to therapy, but impaired the PFS, especially for patients defined at "high risk" based on the cell of origin (no germinal center [GCB]), high BCL2 expression, and IPI 3-5. In this subgroup, the probability of relapse/progression was twice higher than that of patients carrying low BMI1 and USP22 levels. CONCLUSION High expression of BMI1 and of USP22 might be a poor prognostic factor in DLBCL, and might represent the target for novel inhibitors.
Collapse
Affiliation(s)
| | - Robel Papotti
- International PhD School in Clinical and Experimental Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Riccardo Bomben
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Valter Gattei
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Samantha Pozzi
- Dipartimento di Scienze Mediche e Chirurgiche Materno-Infantili e dell'Adulto, Università di Modena e Reggio Emilia, Modena, Italy
| | - Valentina Donati
- Pathology II, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Stefania Bettelli
- Patologia Molecolare e Medicina Predittiva, AOU Modena, Dipartimento di Scienze Mediche e Chirurgiche Materno-Infantili e dell'Adulto, Università di Modena e Reggio Emilia, Modena, Italy
| | - Elisa Forti
- Patologia Molecolare e Medicina Predittiva, AOU Modena, Dipartimento di Scienze Mediche e Chirurgiche Materno-Infantili e dell'Adulto, Università di Modena e Reggio Emilia, Modena, Italy
| | - Giovanna Mansueto
- IRCCS-CROB, Referral Cancer Center of Basilicata, Rionero in Vulture, Italy
| | - Arianna Di Napoli
- Department of Clinical and Molecular Medicine, Sapienza University, Sant'Andrea University Hospital, Rome, Italy
| | - Maria Christina Cox
- Haematology Department, King's College Hospital NHS Trust and UOC Ematologia, AOU Sant'Andrea, Roma, Italy
| | - Leonardo Flenghi
- Department of Emergency and Organ Transplantation, Azienda Ospedaliera di Perugia, Italy
| | - Pietro Rossi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Guido Volpe
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Dimitri Dardanis
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Clara Bono
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Francesca Guerrini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Riccardo Morganti
- SOD supporto statistico agli studi clinici, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | - Stefano Sacchi
- Dipartimento di Scienze Mediche e Chirurgiche Materno-Infantili e dell'Adulto, Università di Modena e Reggio Emilia, Modena, Italy
| | - Sara Galimberti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
3
|
Li KQ, Bai X, Ke AT, Ding SQ, Zhang CD, Dai DQ. Ubiquitin-specific proteases: From biological functions to potential therapeutic applications in gastric cancer. Biomed Pharmacother 2024; 173:116323. [PMID: 38401523 DOI: 10.1016/j.biopha.2024.116323] [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: 12/04/2023] [Revised: 02/17/2024] [Accepted: 02/19/2024] [Indexed: 02/26/2024] Open
Abstract
Deubiquitination, a post-translational modification regulated by deubiquitinases, is essential for cancer initiation and progression. Ubiquitin-specific proteases (USPs) are essential elements of the deubiquitinase family, and are overexpressed in gastric cancer (GC). Through the regulation of several signaling pathways, such as Wnt/β-Catenin and nuclear factor-κB signaling, and the promotion of the expression of deubiquitination- and stabilization-associated proteins, USPs promote the proliferation, metastasis, invasion, and epithelial-mesenchymal transition of GC. In addition, the expression of USPs is closely related to clinicopathological features, patient prognosis, and chemotherapy resistance. USPs therefore could be used as prognostic biomarkers. USP targeting small molecule inhibitors have demonstrated strong anticancer activity. However, they have not yet been tested in the clinic. This article provides an overview of the latest fundamental research on USPs in GC, aiming to enhance the understanding of how USPs contribute to GC progression, and identifying possible targets for GC treatment to improve patient survival.
Collapse
Affiliation(s)
- Kai-Qiang Li
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province 110032, China
| | - Xiao Bai
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province 110032, China
| | - Ang-Ting Ke
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province 110032, China
| | - Si-Qi Ding
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province 110032, China
| | - Chun-Dong Zhang
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province 110032, China
| | - Dong-Qiu Dai
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province 110032, China; Cancer Center, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province 110032, China.
| |
Collapse
|
4
|
Bolhuis DL, Emanuele MJ, Brown NG. Friend or foe? Reciprocal regulation between E3 ubiquitin ligases and deubiquitinases. Biochem Soc Trans 2024; 52:241-267. [PMID: 38414432 PMCID: PMC11349938 DOI: 10.1042/bst20230454] [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: 12/19/2023] [Revised: 01/31/2024] [Accepted: 02/06/2024] [Indexed: 02/29/2024]
Abstract
Protein ubiquitination is a post-translational modification that entails the covalent attachment of the small protein ubiquitin (Ub), which acts as a signal to direct protein stability, localization, or interactions. The Ub code is written by a family of enzymes called E3 Ub ligases (∼600 members in humans), which can catalyze the transfer of either a single ubiquitin or the formation of a diverse array of polyubiquitin chains. This code can be edited or erased by a different set of enzymes termed deubiquitinases (DUBs; ∼100 members in humans). While enzymes from these distinct families have seemingly opposing activities, certain E3-DUB pairings can also synergize to regulate vital cellular processes like gene expression, autophagy, innate immunity, and cell proliferation. In this review, we highlight recent studies describing Ub ligase-DUB interactions and focus on their relationships.
Collapse
Affiliation(s)
- Derek L Bolhuis
- Department of Biochemistry and Biophysics, UNC Chapel Hill School of Medicine, Chapel Hill, NC, 27599
| | - Michael J Emanuele
- Department of Pharmacology and Lineberger Comprehensive Care Center, UNC Chapel Hill School of Medicine, Chapel Hill, NC, 27599
| | - Nicholas G Brown
- Department of Pharmacology and Lineberger Comprehensive Care Center, UNC Chapel Hill School of Medicine, Chapel Hill, NC, 27599
| |
Collapse
|
5
|
Zhang K, Sun T, Li W, Guo Y, Li A, Hsieh M, Wang J, Wu J, Arvanitis L, Raz DJ. Inhibition of USP7 upregulates USP22 and activates its downstream cancer-related signaling pathways in human cancer cells. Cell Commun Signal 2023; 21:319. [PMID: 37946202 PMCID: PMC10634000 DOI: 10.1186/s12964-023-01320-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 09/14/2023] [Indexed: 11/12/2023] Open
Abstract
Deubiquitinases (DUBs) play important roles in various human cancers and targeting DUBs is considered as a novel anticancer therapeutic strategy. Overexpression of ubiquitin specific protease 7 and 22 (USP7 and USP22) are associated with malignancy, therapy resistance, and poor prognosis in many cancers. Although both DUBs are involved in the regulation of similar genes and signaling pathways, such as histone H2B monoubiquitination (H2Bub1), c-Myc, FOXP3, and p53, the interdependence of USP22 and USP7 expression has never been described. In the study, we found that targeting USP7 via either siRNA-mediated knockdown or pharmaceutical inhibitors dramatically upregulates USP22 in cancer cells. Mechanistically, the elevated USP22 occurs through a transcriptional pathway, possibly due to desuppression of the transcriptional activity of SP1 via promoting its degradation upon USP7 inhibition. Importantly, increased USP22 expression leads to significant activation of downstream signal pathways including H2Bub1 and c-Myc, which may potentially enhance cancer malignancy and counteract the anticancer efficacy of USP7 inhibition. Importantly, targeting USP7 further suppresses the in vitro proliferation of USP22-knockout (USP22-Ko) A549 and H1299 lung cancer cells and induces a stronger activation of p53 tumor suppressor signaling pathway. In addition, USP22-Ko cancer cells are more sensitive to a combination of cisplatin and USP7 inhibitor. USP7 inhibitor treatment further suppresses in vivo angiogenesis and tumor growth and induced more apoptosis in USP22-Ko cancer xenografts. Taken together, our findings demonstrate that USP7 inhibition can dramatically upregulate USP22 in cancer cells; and targeting USP7 and USP22 may represent a more effective approach for targeted cancer therapy, which warrants further study. Video Abstract.
Collapse
Affiliation(s)
- Keqiang Zhang
- Division of Thoracic Surgery, City of Hope National Medical Center, Duarte, CA, USA.
| | - Ting Sun
- Division of Thoracic Surgery, City of Hope National Medical Center, Duarte, CA, USA
- Faculty of Health Science, University of Macau, Macau, China
| | - Wendong Li
- Division of Thoracic Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | - Yuming Guo
- Division of Comparative Medicine, City of Hope National Medical Center, Duarte, CA, USA
| | - Aimin Li
- Pathology Core of Shared Resources, City of Hope National Medical Center, Duarte, CA, USA
| | - Marcus Hsieh
- Division of Thoracic Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | - Jinghan Wang
- Department of Hepatobiliary and Pancreatic Surgery, East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jun Wu
- Division of Comparative Medicine, City of Hope National Medical Center, Duarte, CA, USA
| | - Leonidas Arvanitis
- Department of Pathology, City of Hope National Medical Center, Duarte, CA, USA
| | - Dan J Raz
- Division of Thoracic Surgery, City of Hope National Medical Center, Duarte, CA, USA.
| |
Collapse
|
6
|
Ma N, Zhao S, Yang W, Wang Y. B-cell-specific Moloney murine leukemia virus integration site 1 knockdown impairs adriamycin resistance of gastric cancer cells. Arab J Gastroenterol 2023; 24:168-174. [PMID: 36878814 DOI: 10.1016/j.ajg.2023.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 01/13/2023] [Accepted: 02/23/2023] [Indexed: 03/07/2023]
Abstract
BACKGROUND AND STUDY AIMS The B-cell-specific Moloney murine leukemia virus integration site 1 (BMI-1) is associated with the progression of gastric cancer (GC). However, its role in drug resistance of gastric cancer stem cell (GCSC) remains unclear. This study aimed to explore the biological function of BMI-1 in GC cells and its role in drug resistance of GCSCs. PATIENTS AND METHODS We assessed BMI-1 expression in the GEPIA database and in our collected samples from patients with GC. We silenced BMI-1 using siRNA to study the cell proliferation and migration of GC cells. We also used Hoechst 33342 staining to verify the effect of adriamycin (ADR) on side population (SP) cells, and measured the effects of BMI-1 on the expression of N-cadherin, E-cadherin, and drug-resistance-related proteins (multidrug resistance mutation 1 and lung resistance-related protein). Finally, we analyzed BMI-1-related proteins uing the STRING and GEPIA databases. RESULTS BMI-1 mRNA was upregulated in GC tissues and cell lines, especially in MKN-45 and HGC-27 cells. Silencing BMI-1 reduced the proliferation and migration of GC cells. Knocking down BMI-1 significantly decreased epithelial-mesenchymal transition progression, expression levels of drug-resistant proteins, and the number of SP cells in ADR-treated GC cells. Bioinformatics analysis showed that EZH2, CBX8, CBX4, and SUZ12 were positively correlated with BMI-1 in GC tissues. CONCLUSION Our study demonstrates that BMI-1 affects the cellular activity, proliferation, migration, and invasion of GC cells. Silencing the BMI-1 gene significantly reduces the number of SP cells and the expression of drug-resistant proteins in ADR-treated GC cells. We speculate that inhibition of BMI-1 increases the drug resistance of GC cells by affecting GCSCs, and that EZH2, CBX8, CBX4, and SUZ12 may participate in BMI-1-induced enhancement of GCSC-like phenotype and viability.
Collapse
Affiliation(s)
- Ning Ma
- Department of General Surgery Ⅱ, the First People's Hospital of Yunnan Province, the Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan Province, China.
| | - Sihui Zhao
- Department of General Surgery Ⅱ, the First People's Hospital of Yunnan Province, the Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan Province, China
| | - Wei Yang
- Department of General Surgery Ⅱ, the First People's Hospital of Yunnan Province, the Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan Province, China
| | - Yongfang Wang
- Department of General Surgery Ⅱ, the First People's Hospital of Yunnan Province, the Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan Province, China
| |
Collapse
|
7
|
Deng T, Zhong P, Lou R, Yang X. RNF220 promotes gastric cancer growth and stemness via modulating the USP22/wnt/β-catenin pathway. Tissue Cell 2023; 83:102123. [PMID: 37295272 DOI: 10.1016/j.tice.2023.102123] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 05/15/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023]
Abstract
Gastric cancer (GC) is a prevalent malignancy that seriously threatens the health and life of patients. Although Ring finger 220 (RNF220) has been demonstrated to participate in the development of various cancers, its role and mechanism in GC remain undiscovered. The expression of RNF220 was determined by The Cancer Genome Atlas (TCGA) database and Western blot. Additionally, the overall survival (OS) and post-progression survival (PPS) were analyzed based on the levels of RNF220 in the TCGA database. The role and mechanism of RNF220 in growth and stemness were investigated using cell counting kit-8, colony formation, sphere-formation, co-immunoprecipitation, and Western blot experiments. Furthermore, the role of RNF220 was investigated in a xenografted mouse model. The expression of RNF220 was found to be upregulated in GC, which predicted unfavorable OS and PPS in patients with GC. Knockdown of RNF220 reduced cell viability, colony numbers, numbers of spheres formation, and the relative protein levels of Nanog, Sox2, and Oct4 in both AGS and MKN-45 cells. Moreover, overexpression of RNF220 increased cell viability and the numbers of spheres formation in MKN-45 cells. Mechanistically, RNF220 bound to USP22, and interference of RNF220 downregulated the Wnt/β-catenin axis via USP22, which was confirmed by the overexpression of USP22 in both cell lines. Furthermore, silencing of RNF220 significantly decreased tumor volume and weight, the level of Ki-67, and the relative protein levels of USP22, β-catenin, c-myc, Nanog, Sox2, and Oct4. Taken together, downregulation of RNF220 suppressed GC cell growth and stemness by downregulating the USP22/Wnt/β-catenin axis.
Collapse
Affiliation(s)
- Taozhi Deng
- Department of Gastroenterology, Hainan Cancer Hospital, Haikou, Hainan 570000, China
| | - Ping Zhong
- Department of Gastroenterology, Hainan Cancer Hospital, Haikou, Hainan 570000, China
| | - Runlong Lou
- Department of Gastroenterology, Hainan Cancer Hospital, Haikou, Hainan 570000, China
| | - Xiaojun Yang
- Department of Gastroenterology, Suzhou BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Suzhou, Jiangsu 215010, China.
| |
Collapse
|
8
|
Tu X, Li C, Sun W, Tian X, Li Q, Wang S, Ding X, Huang Z. Suppression of Cancer Cell Stemness and Drug Resistance via MYC Destabilization by Deubiquitinase USP45 Inhibition with a Natural Small Molecule. Cancers (Basel) 2023; 15:cancers15030930. [PMID: 36765885 PMCID: PMC9913288 DOI: 10.3390/cancers15030930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/23/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
Cancer stem cells (CSCs) play significant roles in cancer development, drug resistance and cancer recurrence. In cancer treatments based on the CSC characteristics and inducing factors, MYC is a promising target for therapeutic molecules. Although it has been regarded as an undrugable target, its stability tightly regulated by the ubiquitin-proteasome system offers a new direction for molecule targeting and cancer treatment. Herein we report our discoveries in this research area, and we have found that deubiquitinase USP45 can directly bind with MYC, resulting in its deubiquitination and stabilization. Further, USP45 overexpressing can upregulate MYC, and this overexpressing can significantly enhance cancer development, cancer cell stemness and drug resistance. Interestingly, without enhancing cancer development, MYC silencing with shRNA can only suppress USP45-induced stemness and drug resistance. Moreover, we have identified that USP45 can be specifically bound and inhibited by a natural small molecule (α-mangostin), in turn significantly suppressing USP45-induced stemness and drug resistance. Since USP45 is significantly expressed in cervical tumors, we have discovered that the combination of α-mangostin and doxorubicin can significantly inhibit USP45-induced cervical tumorigenesis in an animal model. In general, on the basis of our USP45 discoveries on its MYC deubiquitination and α-mangostin inhibition, suppressing USP45 has opened a new window for suppressing cancer development, stemness and drug resistance.
Collapse
Affiliation(s)
- Xiao Tu
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu 610000, China
| | - Chuncheng Li
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu 610000, China
| | - Wen Sun
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu 610000, China
| | - Xi Tian
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu 610000, China
| | - Qiufu Li
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu 610000, China
| | - Shaoxin Wang
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu 610000, China
| | - Xiaoling Ding
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu 610000, China
| | - Zhen Huang
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Science, Sichuan University, Chengdu 610000, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 610000, China
- SeNA Research Institute and Szostak-CDHT Large Nucleic Acids Institute, Chengdu 610000, China
- Correspondence: ; Fax: +86-028-8550-2629
| |
Collapse
|
9
|
An T, Lu Y, Gong Z, Wang Y, Su C, Tang G, Hou J. Research Progress for Targeting Deubiquitinases in Gastric Cancers. Cancers (Basel) 2022; 14:cancers14235831. [PMID: 36497313 PMCID: PMC9735992 DOI: 10.3390/cancers14235831] [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: 10/24/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Gastric cancers (GCs) are malignant tumors with a high incidence that threaten global public health. Despite advances in GC diagnosis and treatment, the prognosis remains poor. Therefore, the mechanisms underlying GC progression need to be identified to develop prognostic biomarkers and therapeutic targets. Ubiquitination, a post-translational modification that regulates the stability, activity, localization, and interactions of target proteins, can be reversed by deubiquitinases (DUBs), which can remove ubiquitin monomers or polymers from modified proteins. The dysfunction of DUBs has been closely linked to tumorigenesis in various cancer types, and targeting certain DUBs may provide a potential option for cancer therapy. Multiple DUBs have been demonstrated to function as oncogenes or tumor suppressors in GC. In this review, we summarize the DUBs involved in GC and their associated upstream regulation and downstream mechanisms and present the benefits of targeting DUBs for GC treatment, which could provide new insights for GC diagnosis and therapy.
Collapse
Affiliation(s)
- Tao An
- School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
| | - Yanting Lu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250353, China
| | - Zhaoqi Gong
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
| | - Yongtao Wang
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
| | - Chen Su
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China
- Institute of Gastrointestinal Oncology, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Guimei Tang
- School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China
- Correspondence: (G.T.); (J.H.)
| | - Jingjing Hou
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China
- Institute of Gastrointestinal Oncology, School of Medicine, Xiamen University, Xiamen 361005, China
- Correspondence: (G.T.); (J.H.)
| |
Collapse
|
10
|
Guo J, Zhao J, Fu W, Xu Q, Huang D. Immune Evasion and Drug Resistance Mediated by USP22 in Cancer: Novel Targets and Mechanisms. Front Immunol 2022; 13:918314. [PMID: 35935969 PMCID: PMC9347222 DOI: 10.3389/fimmu.2022.918314] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
Regulation of ubiquitination is involved in various processes in cancer occurrence and development, including cell cycle arrest, cell proliferation, apoptosis, invasion, metastasis, and immunity. Ubiquitination plays an important role not only at the transcriptional and post-translational levels but also at the protein level. When ubiquitination is in a pathological state, abnormally activated biological processes will not only induce cancer progression but also induce immune evasion. The main function of deubiquitinases (DUBs) is to remove ubiquitin chains from substrates, changing the biological activity of the substrates. It has great potential to improve the prognosis of cancer by targeting DUB to regulate proteome. Ubiquitin-specific peptidase 22 (USP22) belongs to the ubiquitin-specific protease (USP) family of DUBs and has been reported to be related to various physiological and pathological processes. USP22 is abnormally expressed in various malignant tumors such as prostate cancer, lung cancer, liver cancer, and colorectal cancer, which suggests that USP22 may play an important role in tumors. USP22 may stabilize programmed death ligand 1 (PD-L1) by deubiquitination while also regulating T-cell infiltration into tumors. Regulatory T cells (Tregs) are a unique class of immunosuppressive CD4+ T cells that primarily suppress the immune system by expressing the master transcription factor forkhead box protein 3 (FOXP3). USP22 was found to be a positive regulator of stable FOXP3 expression. Treg-specific ablation of USP22 leads to reduced tumor volume in multiple cancer models. This suggests that USP22 may regulate tumor resistance to immunotherapy. In this article, we review and summarize the biological functions of USP22 in multiple signal transduction pathways during tumorigenesis, immune evasion, and drug resistance. Furthermore, we propose a new possibility of combining USP22 with chemotherapeutic, targeted, and immunosuppressive drugs in the treatment of cancer.
Collapse
Affiliation(s)
- Jinhui Guo
- Qingdao Medical College, Qingdao University, Qingdao, China
- Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Jie Zhao
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, China
| | - Wen Fu
- Qingdao Medical College, Qingdao University, Qingdao, China
| | - Qiuran Xu
- Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Dongsheng Huang
- Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
11
|
Wang Y, Jia Z, Gao J, Zhou T, Zhang X, Zu G. Clinicopathological and Prognostic Value of USP22 Expression in Gastric Cancer: A Systematic Review and Meta-Analysis and Database Validation. Front Surg 2022; 9:920595. [PMID: 35784926 PMCID: PMC9243499 DOI: 10.3389/fsurg.2022.920595] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/26/2022] [Indexed: 01/02/2023] Open
Abstract
Background It has been reported that there is a correlation between the level of ubiquitin-specific protease 22 (USP22) and the clinicopathological parameters and prognosis of gastric cancer (GC) patients, but the conclusions are inconsistent. Hence, a meta-analysis must be conducted to clarify the relationship between USP22 expression and clinicopathological and prognostic value of GC patients to provide more accurate evidence. Methods According to the predetermined selection criteria, systematic file retrieval was performed. The hazard ratio (HR) or odds ratio (OR) and its 95% confidence interval (CI) were used to evaluate the relationship between USP22 expression and clinicopathological and prognostic value of GC patients. Results In a total of 802 patients, those with GC were finally included in 6 studies. The pooled results demonstrated that the expression of USP22 was significantly increased in GC tissues compared with control tissues (OR = 9.947, 95% CI, 6.074–16.291, P = 0.000), and USP22 expression was related to lymph node metastasis (OR = 2.415, 95% CI, 1.082, P = 0.031), distant metastasis (OR = 3.956, 95% CI, 1.365–11.464, P = 0.011) and TNM stage (OR = 2.973, 95% CI, 1.153–7.666, P = 0.024). Nevertheless, the expression of USP22 was not correlated with gender (OR = 1.202, 95% CI, 0.877–1.648, P = 0.253), age (OR = 1.090, 95% CI, 0.811–1.466, P = 0.568), tumor size (OR = 0.693,95% CI, 0.348–1.380, P = 0.297), tumor differentiation (OR = 1.830, 95%CI, 0.948–3.531, P = 0.072) and depth of invasion (OR = 2.320, 95% CI, 0.684–7.871, P = 0.177). Moreover, a high expression of USP22 predicted a poor overall survival (OS) in GC patients (HR = 2.012, 95% CI, 1.522–2.658, P = 0.000). The database of Kaplan–Meier plotter confirmed that a high expression of USP22 was correlated with poor prognostics in GC patients (HR = 1.41, 95% CI, 1.18–1.68, P < 0.01). Conclusion USP22 overexpression in GC tissues is positively related to lymph node metastasis, distant metastasis and TNM stage and indicates a poor clinical outcome of GC patients, but it is not associated with age, gender, depth of invasion, tumor differentiation and tumor size of GC patients. Systematic Review Registration: https://www.crd.york.ac.uk/prospero/, identifier: 338361.
Collapse
Affiliation(s)
- Yuhang Wang
- Department of General Surgery, The Dalian Municipal Central Hospital Affiliated of Dalian Medical University, Dalian, China
- Department of Graduate School, Dalian Medical University, Dalian, China
| | - Zirui Jia
- Department of General Surgery, The Dalian Municipal Central Hospital Affiliated of Dalian Medical University, Dalian, China
- Department of Graduate School, Dalian Medical University, Dalian, China
| | - Jiacheng Gao
- Department of General Surgery, The Dalian Municipal Central Hospital Affiliated of Dalian Medical University, Dalian, China
- Department of Graduate School, Dalian Medical University, Dalian, China
| | - Tingting Zhou
- Department of Neurology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiangwen Zhang
- Department of General Surgery, The Dalian Municipal Central Hospital Affiliated of Dalian Medical University, Dalian, China
- Correspondence: Guo Zu Xiangwen Zhang
| | - Guo Zu
- Department of General Surgery, The Dalian Municipal Central Hospital Affiliated of Dalian Medical University, Dalian, China
- Correspondence: Guo Zu Xiangwen Zhang
| |
Collapse
|
12
|
Yan J, Tan M, Yu L, Jin X, Li Y. Ring finger 220 promotes the stemness and progression of colon cancer cells via Ubiquitin specific peptidase 22-BMI1 axis. Bioengineered 2021; 12:12060-12069. [PMID: 34753387 PMCID: PMC8809949 DOI: 10.1080/21655979.2021.2003664] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 12/03/2022] Open
Abstract
Colorectal cancer (CRC) is ranked as the third most common malignancy worldwide. Therefore, it is urgent to screen novel and effective molecular drug targets for colorectal cancer therapeutics. In this study, the specific role and related mechanism underlying Ring finger (RNF) 220 in colon cancer were investigated. Firstly, RT-PCR assay was used to compare differences between expression levels of RNF220 in colorectal tumor and normal tissues. Western blot and RT-PCR assays were applied to examine the protein levels of RNF220 in normal colonic mucosa and colorectal cancer cells. We found that RNF220 was upregulated in colorectal cancer in patients and cell models. RNF220 promoted the proliferation and migration, invasion of colorectal cancer cells through BrdU incorporation, clone formation, transwell and wound healing assays. Spheroid formation and western blot assays illustrated that RNF220 promoted the stemness of colorectal cancer cells. Moreover, we found that RNF220 regulated BMI1 expression through USP22 by western blot. Finally, we discovered that RNF220 facilitated tumor growth in vivo through establishment of subcutaneous xenograft tumor mice model. In conclusion, RNF220 promoted the stemness and progression of colon cancer cells via the USP22-BMI1 axis.
Collapse
Affiliation(s)
- Jianwen Yan
- Department of Surgery 1, Guilin Tcm Hospital of China, Guilin City, Guangxi Zhuang Autonomous Region, China
| | - Min Tan
- Department of Surgery 1, Guilin Tcm Hospital of China, Guilin City, Guangxi Zhuang Autonomous Region, China
| | - Lin Yu
- Department of Surgery 1, Guilin Tcm Hospital of China, Guilin City, Guangxi Zhuang Autonomous Region, China
| | - Xichao Jin
- Department of Surgery 1, Guilin Tcm Hospital of China, Guilin City, Guangxi Zhuang Autonomous Region, China
| | - Yangcheng Li
- Department of General Surgery, Nantong Tumor Hospital, Nantong City, Jiangsu Province, China
| |
Collapse
|
13
|
Seifert C, Balz E, Herzog S, Korolev A, Gaßmann S, Paland H, Fink MA, Grube M, Marx S, Jedlitschky G, Tzvetkov MV, Rauch BH, Schroeder HWS, Bien-Möller S. PIM1 Inhibition Affects Glioblastoma Stem Cell Behavior and Kills Glioblastoma Stem-like Cells. Int J Mol Sci 2021; 22:ijms222011126. [PMID: 34681783 PMCID: PMC8541331 DOI: 10.3390/ijms222011126] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/05/2021] [Accepted: 10/09/2021] [Indexed: 12/15/2022] Open
Abstract
Despite comprehensive therapy and extensive research, glioblastoma (GBM) still represents the most aggressive brain tumor in adults. Glioma stem cells (GSCs) are thought to play a major role in tumor progression and resistance of GBM cells to radiochemotherapy. The PIM1 kinase has become a focus in cancer research. We have previously demonstrated that PIM1 is involved in survival of GBM cells and in GBM growth in a mouse model. However, little is known about the importance of PIM1 in cancer stem cells. Here, we report on the role of PIM1 in GBM stem cell behavior and killing. PIM1 inhibition negatively regulates the protein expression of the stem cell markers CD133 and Nestin in GBM cells (LN-18, U-87 MG). In contrast, CD44 and the astrocytic differentiation marker GFAP were up-regulated. Furthermore, PIM1 expression was increased in neurospheres as a model of GBM stem-like cells. Treatment of neurospheres with PIM1 inhibitors (TCS PIM1-1, Quercetagetin, and LY294002) diminished the cell viability associated with reduced DNA synthesis rate, increased caspase 3 activity, decreased PCNA protein expression, and reduced neurosphere formation. Our results indicate that PIM1 affects the glioblastoma stem cell behavior, and its inhibition kills glioblastoma stem-like cells, pointing to PIM1 targeting as a potential anti-glioblastoma therapy.
Collapse
Affiliation(s)
- Carolin Seifert
- Department of Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany; (C.S.); (E.B.); (S.H.); (A.K.); (S.G.); (H.P.); (M.A.F.); (M.G.); (G.J.); (M.V.T.); (B.H.R.)
- Department of Neurosurgery, University Medicine Greifswald, 17489 Greifswald, Germany; (S.M.); (H.W.S.S.)
| | - Ellen Balz
- Department of Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany; (C.S.); (E.B.); (S.H.); (A.K.); (S.G.); (H.P.); (M.A.F.); (M.G.); (G.J.); (M.V.T.); (B.H.R.)
- Department of Neurosurgery, University Medicine Greifswald, 17489 Greifswald, Germany; (S.M.); (H.W.S.S.)
| | - Susann Herzog
- Department of Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany; (C.S.); (E.B.); (S.H.); (A.K.); (S.G.); (H.P.); (M.A.F.); (M.G.); (G.J.); (M.V.T.); (B.H.R.)
| | - Anna Korolev
- Department of Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany; (C.S.); (E.B.); (S.H.); (A.K.); (S.G.); (H.P.); (M.A.F.); (M.G.); (G.J.); (M.V.T.); (B.H.R.)
| | - Sebastian Gaßmann
- Department of Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany; (C.S.); (E.B.); (S.H.); (A.K.); (S.G.); (H.P.); (M.A.F.); (M.G.); (G.J.); (M.V.T.); (B.H.R.)
| | - Heiko Paland
- Department of Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany; (C.S.); (E.B.); (S.H.); (A.K.); (S.G.); (H.P.); (M.A.F.); (M.G.); (G.J.); (M.V.T.); (B.H.R.)
- Department of Neurosurgery, University Medicine Greifswald, 17489 Greifswald, Germany; (S.M.); (H.W.S.S.)
| | - Matthias A. Fink
- Department of Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany; (C.S.); (E.B.); (S.H.); (A.K.); (S.G.); (H.P.); (M.A.F.); (M.G.); (G.J.); (M.V.T.); (B.H.R.)
- Department of Neurosurgery, University Medicine Greifswald, 17489 Greifswald, Germany; (S.M.); (H.W.S.S.)
| | - Markus Grube
- Department of Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany; (C.S.); (E.B.); (S.H.); (A.K.); (S.G.); (H.P.); (M.A.F.); (M.G.); (G.J.); (M.V.T.); (B.H.R.)
| | - Sascha Marx
- Department of Neurosurgery, University Medicine Greifswald, 17489 Greifswald, Germany; (S.M.); (H.W.S.S.)
| | - Gabriele Jedlitschky
- Department of Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany; (C.S.); (E.B.); (S.H.); (A.K.); (S.G.); (H.P.); (M.A.F.); (M.G.); (G.J.); (M.V.T.); (B.H.R.)
| | - Mladen V. Tzvetkov
- Department of Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany; (C.S.); (E.B.); (S.H.); (A.K.); (S.G.); (H.P.); (M.A.F.); (M.G.); (G.J.); (M.V.T.); (B.H.R.)
| | - Bernhard H. Rauch
- Department of Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany; (C.S.); (E.B.); (S.H.); (A.K.); (S.G.); (H.P.); (M.A.F.); (M.G.); (G.J.); (M.V.T.); (B.H.R.)
- Department of Pharmacology and Toxicology, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
| | - Henry W. S. Schroeder
- Department of Neurosurgery, University Medicine Greifswald, 17489 Greifswald, Germany; (S.M.); (H.W.S.S.)
| | - Sandra Bien-Möller
- Department of Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany; (C.S.); (E.B.); (S.H.); (A.K.); (S.G.); (H.P.); (M.A.F.); (M.G.); (G.J.); (M.V.T.); (B.H.R.)
- Department of Neurosurgery, University Medicine Greifswald, 17489 Greifswald, Germany; (S.M.); (H.W.S.S.)
- Correspondence: ; Tel.: +49-03834-865646
| |
Collapse
|
14
|
Jing T, Wang B, Yang Z, Liu Y, Xu G, Xu X, Jiao K, Chen Z, Xiang L, Zhang L, Liu Y. Deubiquitination of the repressor E2F6 by USP22 facilitates AKT activation and tumor growth in hepatocellular carcinoma. Cancer Lett 2021; 518:266-277. [PMID: 34339800 DOI: 10.1016/j.canlet.2021.07.044] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 07/16/2021] [Accepted: 07/28/2021] [Indexed: 01/20/2023]
Abstract
Dysregulated ubiquitination of tumor-related proteins plays a critical role in tumor development and progression. The deubiquitinase USP22 is aberrantly expressed in certain types of cancer and contributes to aggressive tumor progression. However, the precise mechanism underlying the pro-tumorigenic function of USP22 in hepatocellular carcinoma (HCC) remains unclear. Here, we report that E2F6, a pocket protein-independent transcription repressor, is essential for HCC cell growth, and that its activities are controlled by USP22-mediated deubiquitination. USP22 interacts with and stabilizes E2F6, resulting in the transcriptional repression of phosphatase DUSP1. Moreover, the process involving DUSP1 repression by E2F6 strengthens AKT activation in HCC cells. Therefore, these findings provide mechanistic insights into the USP22-mediated control of oncogenic AKT signaling, emphasizing the importance of USP22-E2F6 regulation in HCC development.
Collapse
Affiliation(s)
- Tiantian Jing
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, China
| | - Boshi Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, China
| | - Zhaojuan Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, China
| | - Yun Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, China
| | - Guiqin Xu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, China
| | - Xiaoli Xu
- Shanghai Jiao Tong University School of Biomedical Engineering, Shanghai, 200030, China
| | - Kun Jiao
- Shanghai Jiao Tong University School of Biomedical Engineering, Shanghai, 200030, China
| | - Zehong Chen
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, China
| | - Lvzhu Xiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, China
| | - Li Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, China.
| | - Yongzhong Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, China.
| |
Collapse
|
15
|
Guo JN, Xia BR, Deng SH, Yang C, Pi YN, Cui BB, Jin WL. Deubiquitinating Enzymes Orchestrate the Cancer Stem Cell-Immunosuppressive Niche Dialogue: New Perspectives and Therapeutic Potential. Front Cell Dev Biol 2021; 9:680100. [PMID: 34179009 PMCID: PMC8220152 DOI: 10.3389/fcell.2021.680100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 05/17/2021] [Indexed: 11/13/2022] Open
Abstract
Cancer stem cells (CSCs) are sparks for igniting tumor recurrence and the instigators of low response to immunotherapy and drug resistance. As one of the important components of tumor microenvironment, the tumor associated immune microenvironment (TAIM) is driving force for the heterogeneity, plasticity and evolution of CSCs. CSCs create the inhibitory TAIM (ITAIM) mainly through four stemness-related signals (SRSs), including Notch-nuclear factor-κB axis, Hedgehog, Wnt and signal transducer and activator of transcription. Ubiquitination and deubiquitination in proteins related to the specific stemness of the CSCs have a profound impact on the regulation of ITAIM. In regulating the balance between ubiquitination and deubiquitination, it is crucial for deubiquitinating enzymes (DUBs) to cleave ubiquitin chains from substrates. Ubiquitin-specific peptidases (USPs) comprise the largest family of DUBs. Growing evidence suggests that they play novel functions in contribution of ITAIM, including regulating tumor immunogenicity, activating stem cell factors, upregulating the SRSs, stabilizing anti-inflammatory receptors, and regulating anti-inflammatory cytokines. These overactive or abnormal signaling may dampen antitumor immune responses. The inhibition of USPs could play a regulatory role in SRSs and reversing ITAIM, and also have great potential in improving immune killing ability against tumor cells, including CSCs. In this review, we focus on the USPs involved in CSCs signaling pathways and regulating ITAIM, which are promising therapeutic targets in antitumor therapy.
Collapse
Affiliation(s)
- Jun-Nan Guo
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Bai-Rong Xia
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, Anhui Provincial Cancer Hospital, University of Science and Technology of China, Hefei, China
| | - Shen-Hui Deng
- Department of Anesthesiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chang Yang
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ya-Nan Pi
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Bin-Bin Cui
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Wei-Lin Jin
- Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, Institute of Cancer Neuroscience, The First Clinical Medical College of Lanzhou University, Lanzhou, China
| |
Collapse
|
16
|
Xiong B, Huang J, Liu Y, Zou M, Zhao Z, Gong J, Wu X, Qiu C. Ubiquitin-specific protease 2a promotes hepatocellular carcinoma progression via deubiquitination and stabilization of RAB1A. Cell Oncol (Dordr) 2021; 44:329-343. [PMID: 33074477 DOI: 10.1007/s13402-020-00568-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2020] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Deubiquitination, the inverse process of ubiquitination, is catalyzed by deubiquitinases (DUBs) that remove ubiquitin from target proteins and subsequently prevent their degradation by proteasomes. Previously, deubiquitination has been found to be involved in hepatocellular carcinoma (HCC) progression. As yet, however, little is known about the exact role of deubiquitination in the development and/or progression of this type of cancer. METHODS HCC tissues and tissue microarrays were used to detect expression of the DUB ubiquitin-specific protease 2a (USP2a). The critical role of USP2a in HCC development and progression was assessed in both in vitro cell and in vivo animal models. LC-MS/MS analyses were performed to identify potential targets of USP2a in HCC cells, after which regulation of target protein stability and ubiquitin status by USP2a were investigated. RESULTS We found that USP2a was significantly upregulated in HCC tissues, and that a high expression was positively associated with a poor prognosis. Subsequently, we found that USP2a silencing resulted in inhibition of HCC cell proliferation, migration and invasion, whereas exogenous USP2a overexpression resulted in the opposite effects, both in vitro and in vivo. Mechanistically, LC-MS/MS analysis revealed that RAB1A, a key regulator of the ER and Golgi vesicular transport system, serves as a potential target of USP2a in HCC cells. In addition, we found that USP2a can deubiquitinate and stabilize RAB1A and prevent its degradation, and that this process is required for inducing HCC progression by USP2a. CONCLUSIONS Our data indicate that USP2a can promote HCC progression via deubiquitination and stabilization of RAB1A. This observation indicates that DUB targeting may serve as a novel approach to improve the treatment of HCC.
Collapse
Affiliation(s)
- Bin Xiong
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Junwei Huang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Yan Liu
- Department of Gastroenterology, The Fifth people's Hospital of Chengdu, Chengdu, Sichuan, 611130, People's Republic of China
| | - Min Zou
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Zhibo Zhao
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Jianping Gong
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Xiaoling Wu
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Chan Qiu
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China.
| |
Collapse
|
17
|
Jin D, Wei W, Song C, Han P, Leng X. RETRACTED: Knockdown EZH2 attenuates cerebral ischemia-reperfusion injury via regulating microRNA-30d-3p methylation and USP22. Brain Res Bull 2021; 169:25-34. [PMID: 33388376 DOI: 10.1016/j.brainresbull.2020.12.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 11/30/2020] [Accepted: 12/28/2020] [Indexed: 02/07/2023]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief as there are concerns about the reliability of the results. Concerns have been raised about a portion of Figure 5B, ‘DMSO’ group appears to contain image similarities with Figure 4e, ‘Inhibitor NC’ group, published in Yang et al., 2021 doi: 10.1080/15384101.2020.1856498. A portion of Figure 5B, ‘DZNeP+miR-30d-3p antagomir’ group appears to contain image similarities with Figure 4e, ‘Inhibitor NC’ group, published in Yang et al., 2021. Figure 7/G western blot bands have the same eyebrow shaped phenotype as many other publications as detailed here (https://pubpeer.com/publications/B26AE47AC0E71E0EF339B40893B2C2).
Collapse
Affiliation(s)
- Dianshi Jin
- The Affliated Dalian Central Hospital of Dalian Medical University, Dalian, 116033 Liaoning, China.
| | - Wei Wei
- The Affliated Dalian Central Hospital of Dalian Medical University, Dalian, 116033 Liaoning, China
| | - Chong Song
- The Affliated Dalian Central Hospital of Dalian Medical University, Dalian, 116033 Liaoning, China
| | - Peng Han
- The Affliated Dalian Central Hospital of Dalian Medical University, Dalian, 116033 Liaoning, China
| | - Xiaolei Leng
- The Affliated Dalian Central Hospital of Dalian Medical University, Dalian, 116033 Liaoning, China
| |
Collapse
|
18
|
Baek D, Park KH, Lee KM, Jung S, Joung S, Kim J, Lee JW. Ubiquitin-specific protease 53 promotes osteogenic differentiation of human bone marrow-derived mesenchymal stem cells. Cell Death Dis 2021; 12:238. [PMID: 33664230 PMCID: PMC7933275 DOI: 10.1038/s41419-021-03517-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 01/29/2021] [Accepted: 02/04/2021] [Indexed: 12/11/2022]
Abstract
The ubiquitin protease pathway plays important role in human bone marrow-derived mesenchymal stem cell (hBMSC) differentiation, including osteogenesis. However, the function of deubiquitinating enzymes in osteogenic differentiation of hBMSCs remains poorly understood. In this study, we aimed to investigate the role of ubiquitin-specific protease 53 (USP53) in the osteogenic differentiation of hBMSCs. Based on re-analysis of the Gene Expression Omnibus database, USP53 was selected as a positive regulator of osteogenic differentiation in hBMSCs. Overexpression of USP53 by lentivirus enhanced osteogenesis in hBMSCs, whereas knockdown of USP53 by lentivirus inhibited osteogenesis in hBMSCs. In addition, USP53 overexpression increased the level of active β-catenin and enhanced the osteogenic differentiation of hBMSCs. This effect was reversed by the Wnt/β-catenin inhibitor DKK1. Mass spectrometry showed that USP53 interacted with F-box only protein 31 (FBXO31) to promote proteasomal degradation of β-catenin. Inhibition of the osteogenic differentiation of hBMSCs by FBXO31 was partially rescued by USP53 overexpression. Animal studies showed that hBMSCs with USP53 overexpression significantly promoted bone regeneration in mice with calvarial defects. These results suggested that USP53 may be a target for gene therapy for bone regeneration.
Collapse
Affiliation(s)
- Dawoon Baek
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
- Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Kwang Hwan Park
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Kyoung-Mi Lee
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Sujin Jung
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
- Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Soyeong Joung
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
- Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Jihyun Kim
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
- Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Jin Woo Lee
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea.
- Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea.
- Severance Biomedical Science Institute, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea.
| |
Collapse
|
19
|
Molecular Mechanisms of DUBs Regulation in Signaling and Disease. Int J Mol Sci 2021; 22:ijms22030986. [PMID: 33498168 PMCID: PMC7863924 DOI: 10.3390/ijms22030986] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/15/2021] [Accepted: 01/18/2021] [Indexed: 02/07/2023] Open
Abstract
The large family of deubiquitinating enzymes (DUBs) are involved in the regulation of a plethora of processes carried out inside the cell by protein ubiquitination. Ubiquitination is a basic pathway responsible for the correct protein homeostasis in the cell, which could regulate the fate of proteins through the ubiquitin–proteasome system (UPS). In this review we will focus on recent advances on the molecular mechanisms and specificities found for some types of DUBs enzymes, highlighting illustrative examples in which the regulatory mechanism for DUBs has been understood in depth at the molecular level by structural biology. DUB proteases are responsible for cleavage and regulation of the multiple types of ubiquitin linkages that can be synthesized inside the cell, known as the ubiquitin-code, which are tightly connected to specific substrate functions. We will display some strategies carried out by members of different DUB families to provide specificity on the cleavage of particular ubiquitin linkages. Finally, we will also discuss recent progress made for the development of drug compounds targeting DUB proteases, which are usually correlated to the progress of many pathologies such as cancer and neurodegenerative diseases.
Collapse
|
20
|
Exosome-transferred LINC01559 promotes the progression of gastric cancer via PI3K/AKT signaling pathway. Cell Death Dis 2020; 11:723. [PMID: 32895368 PMCID: PMC7477231 DOI: 10.1038/s41419-020-02810-5] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 06/24/2020] [Accepted: 06/29/2020] [Indexed: 12/29/2022]
Abstract
Increasing evidence indicates that long non-coding RNAs (lncRNAs) are associated with the progression of human cancers. However, the expression level and function of LINC01559 (long intergenic non-protein coding RNA 1559) in gastric cancer (GC) are rarely reported. Here we found that LINC01559 was upregulated in GC tissues based on GEPIA (Gene Expression Profiling Interactive Analysis) and TCGA (The Cancer Genome Atlas) databases. Also, LINC01559 was expressed at a lower level in GC cells than in mesenchymal stem cells (MSCs). In vitro experiments revealed that silencing LINC01559 remarkably hindered GC cell proliferation, migration and stemness. Then, we identified that LINC01559 was transmitted form MSCs to GC cells via the exosomes. Immunofluorescence staining and electron microscope validated the existence of exosomes in GC cells. Mechanistically, LINC01559 sponged miR-1343-3p to upregulate PGK1 (phosphoglycerate kinase 1), therefore activating PI3K/AKT pathway. Moreover, LINC01559 recruited EZH2 (enhancer of zeste 2 polycomb repressive complex 2 subunit) to PTEN (phosphatase and tensin homolog) promoter, inducing the methylation of PTEN promoter and finally resulting in PTEN repression. Of note, LINC01559 targeted both PGK1 and PTEN to promote GC progression by activating PI3K/AKT pathway. Taken together, our study demonstrated that LINC01559 accelerated GC progression via upregulating PGK1 and downregulating PTEN to trigger phosphatidylinositol 3-kinase/AKT serine/threonine kinase (PI3K/AKT) pathway, indicating LINC01559 as a potential biomarker for GC treatment.
Collapse
|
21
|
Zhao M, Hou Y, Du YE, Yang L, Qin Y, Peng M, Liu S, Wan X, Qiao Y, Zeng H, Cui X, Teng Y, Liu M. Drosha-independent miR-6778-5p strengthens gastric cancer stem cell stemness via regulation of cytosolic one-carbon folate metabolism. Cancer Lett 2020; 478:8-21. [PMID: 32142918 DOI: 10.1016/j.canlet.2020.02.040] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 02/26/2020] [Accepted: 02/28/2020] [Indexed: 02/07/2023]
Abstract
Drosha-dependent canonical microRNAs (miRNAs) play a crucial role in the biological functions and development of cancer. However, the effects of Drosha-independent non-canonical miRNAs remain poorly understood. In our previous work, we found a set of aberrant miRNAs, including some upregulated miRNAs, called Drosha-independent noncanonical miRNAs, in Drosha-knockdown gastric cancer (GC) cells. Surprisingly, Drosha-silenced GC cells still retained strong malignant properties (e.g., proliferation ability and cancer stem cell (CSC) characteristics), indicating that aberrantly upregulated non-canonical miRNAs may play an important role in the maintenance of the malignant properties in GC cells that express low Drosha levels. Here, we report that miR-6778-5p, a noncanonical miRNA, acts as a crucial regulator for maintenance of CSC stemness in Drosha-silenced GC cells. MiR-6778-5p belongs to the 5'-tail mirtron type of non-canonical miRNAs and is transcript splice-derived from intron 5 of SHMT1 (coding cytoplasmic serine hydroxymethyltransferase). It positively regulates expression of its host gene, SHMT1, via targeting YWHAE in Drosha-knockdown GC cells. Similar to its family member SHMT2, SHMT1 plays a crucial role in folate-dependent serine/glycine inter-conversion in one-carbon metabolism. In Drosha wild type GC cells, SHMT2 mediates a mitochondrial-carbon metabolic pathway, which is a major pathway of one-carbon metabolism in normal cells and most cancer cells. However, in Drosha-silenced or Drosha low-expressing GC cells, miR-6778-5p positively regulates SHMT1, instead of SHMT2, thus mediating a compensatory activation of cytoplasmic carbon metabolism that plays an essential role in the maintenance of CSCs in gastric cancer (GCSCs). Drosha wild type GCSCs with SHMT2 are sensitive to 5-fluorouracil; however, Drosha low-expressing GCSCs with SHMT1 are 5-FU-resistant. The loss of miR-6778-5p or SHMT1 notably mitigates GCSC sphere formation and increases sensitivity to 5-fluorouracil in Drosha-knockdown gastric cancer cells. Thus, our study reveals a novel function of Drosha-independent noncanonical miRNAs in maintaining the stemness of GCSCs.
Collapse
Affiliation(s)
- Maojia Zhao
- Key Laboratory of Laboratory Medical Diagnostics Designated By Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Yixuan Hou
- Experimental Teaching Center of Basic Medicine Science, Chongqing Medical University, Chongqing, 400016, China
| | - Yan-E Du
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Liping Yang
- Key Laboratory of Laboratory Medical Diagnostics Designated By Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Yilu Qin
- Key Laboratory of Laboratory Medical Diagnostics Designated By Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Meixi Peng
- Key Laboratory of Laboratory Medical Diagnostics Designated By Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Shuiqing Liu
- Key Laboratory of Laboratory Medical Diagnostics Designated By Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Xueying Wan
- Key Laboratory of Laboratory Medical Diagnostics Designated By Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Yina Qiao
- Key Laboratory of Laboratory Medical Diagnostics Designated By Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Huan Zeng
- Key Laboratory of Laboratory Medical Diagnostics Designated By Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Xiaojiang Cui
- Department of Surgery, Department of Obstetrics and Gynecology, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center. Los Angeles, CA, 91006, USA
| | - Yong Teng
- Department of Oral Biology, Dental College of Georgia, Georgia Cancer Center, Augusta University, Augusta, GA, USA
| | - Manran Liu
- Key Laboratory of Laboratory Medical Diagnostics Designated By Chinese Ministry of Education, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
22
|
Lim C, Xu JC, Chen TY, Xu JX, Chen WF, Hu JW, Li QL, Zhang YQ. Ubiquitin-specific peptide 22 acts as an oncogene in gastric cancer in a son of sevenless 1-dependent manner. Cancer Cell Int 2020; 20:45. [PMID: 32063746 PMCID: PMC7011508 DOI: 10.1186/s12935-020-1137-y] [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: 12/16/2019] [Accepted: 02/04/2020] [Indexed: 02/08/2023] Open
Abstract
Background Aberrant expression of ubiquitin-specific peptide 22 (USP22) has been detected in various cancers. This study aimed to investigate the role of USP22 and the underlying mechanism in human gastric cancer. Methods The expression pattern of USP22 in human gastric cancer was detected in a tissue microarray containing 88 pairs of gastric cancer tissue and adjacent normal tissue samples from patients with primary gastric cancer using immunohistochemical staining. The correlation of USP22 expression with clinical characteristics of patients, as well as their prognostic values in the overall survival of patients, were evaluated. USP22-overexpressing SGC7901 and USP22-silencing AGS cells were used to explore the role of USP22 in gastric cancer cell behavior in vitro and in vivo. Chromatin immunoprecipitation was performed to identify differentially expressed genes induced by USP22 overexpression. Western blot analysis was conducted to detect the activation of RAS/ERK and PI3K/AKT signaling in USP22-overexpressing SGC7901 cells and xenograft tumor tissues. Knockdown of RAS activator son of sevenless 1 (SOS1) was performed to investigate the role of SOS1 in USP22-regulated gastric cancer cell behavior and RAS signaling both in vitro and in vivo. Results USP22 protein expression was significantly increased in human gastric cancer tissues, compared with adjacent normal tissues, and was positively correlated with local tumor stage. Gain- and loss-of-function assays showed that USP22 promoted gastric cancer cell growth and cell cycle transition while suppressing apoptosis in vitro. Consistent results were observed in a xenograft mouse model. Chromatin immunoprecipitation revealed that the overexpression of USP22 induced the upregulation of RAS activator son of sevenless 1 (SOS1) in SGC7901 cells. Western blot analysis showed that USP22 overexpression also induced activation of the RAS/ERK and PI3K/AKT pathways in SGC7901 cells and xenograft tumor tissues. Furthermore, SOS1 silencing could reverse the effects of USP22 on gastric cancer cell behavior and RAS signaling both in vitro and in vivo. Conclusions Our results suggest that USP22 acts as an oncogene in gastric cancer in a SOS1-dependent manner, identifying the USP22/SOS1/RAS axis as a potential therapeutic target in gastric cancer.
Collapse
Affiliation(s)
- ChitChoon Lim
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, No. 180 FengLin Road, Shanghai, 200032 China
| | - Jia-Cheng Xu
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, No. 180 FengLin Road, Shanghai, 200032 China
| | - Tian-Yin Chen
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, No. 180 FengLin Road, Shanghai, 200032 China
| | - Jia-Xin Xu
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, No. 180 FengLin Road, Shanghai, 200032 China
| | - Wei-Feng Chen
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, No. 180 FengLin Road, Shanghai, 200032 China
| | - Jian-Wei Hu
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, No. 180 FengLin Road, Shanghai, 200032 China
| | - Quan-Lin Li
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, No. 180 FengLin Road, Shanghai, 200032 China
| | - Yi-Qun Zhang
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, No. 180 FengLin Road, Shanghai, 200032 China
| |
Collapse
|
23
|
Abstract
Prostate cancer (PCa) is the leading cause of cancer death in men. With more therapeutic modalities available, the overall survival in PCa has increased significantly in recent years. Patients with relapses after advanced secondgeneration anti-androgen therapy however, often show poor disease prognosis. This group of patients often die from cancer-related complicacies. Multiple approaches have been taken to understand disease recurrence and to correlate the gene expression profile. In one such study, an 11-gene signature was identified to be associated with PCa recurrence and poor survival. Amongst them, a specific deubiquitinase called ubiquitin-specific peptidase 22 (USP22) was selectively and progressively overexpressed with PCa progression. Subsequently, it was shown to regulate androgen receptors and Myc, the two most important regulators of PCa progression. Furthermore, USP22 has been shown to be associated with the development of therapy resistant PCa. Inhibiting USP22 was also found to be therapeutically advantageous, especially in clinically challenging and advanced PCa. This review provides an update of USP22 related functions and challenges associated with PCa research and explains why targeting this axis is beneficial for PCa relapse cases.
Collapse
Affiliation(s)
- Nivedita Nag
- Department of Microbiology, Sister Nibedita Government General Degree College for Girls, Kolkata 700027, India
| | - Samikshan Dutta
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| |
Collapse
|
24
|
Sun J, Shi X, Mamun MAA, Gao Y. The role of deubiquitinating enzymes in gastric cancer. Oncol Lett 2019; 19:30-44. [PMID: 31897112 PMCID: PMC6924028 DOI: 10.3892/ol.2019.11062] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 10/22/2019] [Indexed: 12/24/2022] Open
Abstract
The epigenetic regulation of gene expression (via DNA methylation, histone modification and microRNA interference) contributes to a variety of diseases, particularly cancer. Protein deubiquitination serves a key role in the mechanism underlying histone modification, and consequently influences tumor development and progression. Improved characterization of the role of ubiquitinating enzymes has led to the identification of numerous deubiquitinating enzymes (DUBs) with various functions. Gastric cancer (GC) is a highly prevalent cancer type that exhibits a high mortality rate. Latest analysis about cancer patient revealed that GC is sixth deadliest cancer type, which frequently occur in male (7.2%) than female (4.1%). Complex associations between DUBs and GC progression have been revealed in multiple studies; however, the molecular mechanism underpinning the metastasis and recurrence of GC is yet to be elucidated. Generally, DUBs were upregulated in gastric cancer. The relation of DUBs and tumor size, classification and staging was observed in GC. Besides, 5-yar survival rate of patients with GC is effeccted by expression level of DUBs. Among the highly expressed DUBs, specifically six DUBs namely UCHs, USPs, OTUs, MJDs, JAMMs and MCPIPs effect on this survival rate. Consequently, the association between GC and DUBs has received increasing attention in recent years. Therefore, in the present review, literature investigating the association between DUBs and GC pathophysiology was analyzed and critically appraised.
Collapse
Affiliation(s)
- Jiangang Sun
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Xiaojing Shi
- Zhengzhou University School of Pharmaceutical Science, Zhengzhou, Henan 450001, P.R. China
| | - M A A Mamun
- Zhengzhou University School of Pharmaceutical Science, Zhengzhou, Henan 450001, P.R. China
| | - Yongshun Gao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
25
|
Yuan X, Wang H, Xu A, Zhu X, Zhan Y, Wang W. Ubiquitin-specific peptidase 22 promotes proliferation and metastasis in human colon cancer. Oncol Lett 2019; 18:5567-5576. [PMID: 31612065 PMCID: PMC6781658 DOI: 10.3892/ol.2019.10872] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 08/13/2019] [Indexed: 01/08/2023] Open
Abstract
Colon cancer is one of the most common malignant tumors in the world; however, the mechanism underlying the progression of colon cancer remains unclear. In the present study, the expression of ubiquitin-specific peptidase 22 (USP22) in paraffin sections of human colon cancer tissues and normal colon tissues were examined using immunohistochemistry. The human colon cancer cell lines HCT116 and HT29 were used for USP22 knockdown experiments, and functional assays were performed. The results demonstrated that compared with normal colon tissues, human colon cancer tissues exhibited upregulated expression of USP22 and this was associated with tumor lymph node metastasis and tumor stage in colon cancer tissues. In addition, upregulated expression of USP22 was significantly correlated with both lower relapse-free survival and lower overall survival rates in patients with colon cancer. When USP22 was silenced in colon cancer cell lines, this resulted in a decrease in cell proliferation and metastatic behaviors. Furthermore, Bmi-1 and Cyclin D2 were found to be positively regulated by USP22, which may have mediated the tumorigenic effects of USP22 in human colon cancer. The results of the present study may have significant implications for examining the underlying mechanisms of cancer development and the potential development of cancer therapeutics.
Collapse
Affiliation(s)
- Xiao Yuan
- Department of General Surgery, The Fourth Affiliated Hospital of Anhui Medical University, Anhui, Hefei 230022, P.R. China
| | - Hao Wang
- Department of Pathology, Anhui Medical University, Anhui, Hefei 230032, P.R. China
| | - Aman Xu
- Department of General Surgery, The Fourth Affiliated Hospital of Anhui Medical University, Anhui, Hefei 230022, P.R. China
| | - Xingyang Zhu
- Department of General Surgery, The Fourth Affiliated Hospital of Anhui Medical University, Anhui, Hefei 230022, P.R. China
| | - Yanqing Zhan
- Department of General Surgery, The Fourth Affiliated Hospital of Anhui Medical University, Anhui, Hefei 230022, P.R. China
| | - Wenbin Wang
- Department of General Surgery, The Fourth Affiliated Hospital of Anhui Medical University, Anhui, Hefei 230022, P.R. China
| |
Collapse
|
26
|
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: 99] [Impact Index Per Article: 16.5] [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
|
27
|
He J, Lee HJ, Saha S, Ruan D, Guo H, Chan CH. Inhibition of USP2 eliminates cancer stem cells and enhances TNBC responsiveness to chemotherapy. Cell Death Dis 2019; 10:285. [PMID: 30918246 PMCID: PMC6437220 DOI: 10.1038/s41419-019-1512-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 02/28/2019] [Accepted: 03/08/2019] [Indexed: 01/17/2023]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer that harbors enriched cancer stem cell (CSC) populations in tumors. Conventional chemotherapy is a standard treatment for TNBC, but it spares the CSC populations, which cause tumor recurrence and progression. Therefore, identification of the core molecular pathway that controls CSC activity and expansion is essential for developing effective therapeutics for TNBC. In this study, we identify that USP2 deubiquitinating enzyme is upregulated in CSCs and is a novel regulator of CSCs. Genetic and pharmacological targeting of USP2 substantially inhibits the self-renewal, expansion and chemoresistance of CSCs. We show that USP2 maintains the CSC population by activating self-renewing factor Bmi1 and epithelial-mesenchymal transition through Twist upregulation. Mechanistically, USP2 promotes Twist stabilization by removing β-TrCP-mediated ubiquitination of Twist. Animal studies indicate that pharmacological inhibition of USP2 suppresses tumor progression and sensitizes tumor responses to chemotherapy in TNBC. Furthermore, the histological analyses reveal a positive correlation between USP2 upregulation and lymph node metastasis. Our findings together demonstrate a previously unrecognized role of USP2 in mediating Twist activation and CSC enrichment, suggesting that targeting USP2 is a novel therapeutic strategy to tackle TNBC.
Collapse
Affiliation(s)
- Jiabei He
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Hong-Jen Lee
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, 11794, USA.,Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Suchandrima Saha
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, 11794, USA.,Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Diane Ruan
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Hua Guo
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Chia-Hsin Chan
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, 11794, USA. .,Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, 11794, USA.
| |
Collapse
|
28
|
Is co-expression of USP22 and HSP90 more effective in predicting prognosis of gastric cancer? Pathol Res Pract 2018; 215:653-659. [PMID: 30598339 DOI: 10.1016/j.prp.2018.12.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/01/2018] [Accepted: 12/24/2018] [Indexed: 12/25/2022]
Abstract
The ubiquitin-specific peptidase 22 (USP22) belongs to the largest subfamily of deubiquitylases and recent studies indicate that overexpression of USP22 may promote gastric cancer progression and predict prognosis. But little is known about the interaction network of USP22 in gastric cancer. In this study, we applied bioinformatics methods and found that USP22 was correlated with the heat shock protein 90 (HSP90) which is now considered to be a biomarker to predict the prognosis of gastric cancer. Then the siRNA transfection and western blotting were used to testify the correlation of USP22 and HSP90 in gastric cancer cells. The immunohistochemistry staining of the microarrays was applied to confirm the correlation of USP22 and HSP90 expression in gastric cancer tissue and further analysis showed that co-expression of USP22 and HSP90 was related to lymph node metastasis and more effective in predicting the prognosis of gastric cancer. In summary, our data demonstrate that correlation exists between USP22 and HSP90 expressions in gastric cancer and co-expression of USP22 and HSP90 may be more effective in predicting prognosis of gastric cancer.
Collapse
|
29
|
Yang F, Zheng Z, Xue X, Zheng L, Qin J, Li H, Zhou Y, Fang G. Targeted eradication of gastric cancer stem cells by CD44 targeting USP22 small interfering RNA-loaded nanoliposomes. Future Oncol 2018; 15:281-295. [PMID: 30543303 DOI: 10.2217/fon-2018-0295] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
AIM USP22, a member of ubiquitin-specific proteases (USPs), is a well-defined protein that promotes poor prognosis, invasion and metastasis, and also participates in the maintenance of cancer stem cells. USP22 siRNA-loaded nanoliposomes conjugated with CD44 antibodies (USP22-NLs-CD44) were constructed to enhance the therapeutic effect of USP22 siRNA against gastric cancer stem cells. MATERIALS & METHODS The targeting and therapeutic efficacies of USP22-NLs-CD44 against gastric cancer stem cells were evaluated. RESULTS & CONCLUSION USP22-NLs-CD44 was demonstrated to be able to effectively deliver USP22 siRNA to CD44+ gastric cancer stem cells, achieving superior therapeutic effects against CD44+ gastric cancer stem cells than nontargeted nanoliposomes. USP22-NLs-CD44 may provide a novel approach to eradicate gastric cancer stem cells in the near future.
Collapse
Affiliation(s)
- Feng Yang
- Department of General Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 700 Moyubei Road, Shanghai 201805, PR China
| | - Zhi Zheng
- Department of General Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 700 Moyubei Road, Shanghai 201805, PR China
| | - Xuchao Xue
- Department of General Surgery, Changhai Hospital, Second Military Medical University, 168 Changhai Road, Shanghai 200433, PR China
| | - Luming Zheng
- Department of General Surgery, General Hospital of Jinan Military Area, 25 Shifan Road, Jinan 250031, PR China
| | - Jianmin Qin
- Department of General Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 700 Moyubei Road, Shanghai 201805, PR China
| | - Haijia Li
- Department of General Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 700 Moyubei Road, Shanghai 201805, PR China
| | - Yuhong Zhou
- Department of General Surgery, Changhai Hospital, Second Military Medical University, 168 Changhai Road, Shanghai 200433, PR China
| | - Guoen Fang
- Department of General Surgery, Changhai Hospital, Second Military Medical University, 168 Changhai Road, Shanghai 200433, PR China
| |
Collapse
|
30
|
Fu Y, Du P, Zhao J, Hu C, Qin Y, Huang G. Gastric Cancer Stem Cells: Mechanisms and Therapeutic Approaches. Yonsei Med J 2018; 59:1150-1158. [PMID: 30450848 PMCID: PMC6240570 DOI: 10.3349/ymj.2018.59.10.1150] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 10/11/2018] [Accepted: 10/12/2018] [Indexed: 12/12/2022] Open
Abstract
Gastric cancer (GC) is the third leading cause of cancer-related deaths worldwide. GC stem-like cells (GCSCs), with unlimited self-renewal, differentiation, and tumor-regenerating capacities, contribute significantly to the refractory features of GC and have gained increasing attention for their role in GC drug resistance, relapse, and metastasis. Therapies targeting GCSCs seem to be one of the most promising methods to improve the outcomes of GC patients. Extensive investigations have attempted to outline the regulatory mechanisms in GCSCs and to develop GCSCs-targeting therapies with which to diminish GC drug resistance, metastasis and relapse. To the best of our knowledge, there is a lack of reviews summarizing these studies. In this review, we systematically recapitulated findings regarding the regulatory mechanisms of GCSCs, as well as therapies that target GCSCs, hoping to support the development of prognostic biomarkers and GCSCs-targeting anticancer therapies in GC.
Collapse
Affiliation(s)
- Yan Fu
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Peizhun Du
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Jing Zhao
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
- Cancer Metastasis Institute, Fudan University, Shanghai, China
| | - Cheng'en Hu
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yunyun Qin
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Guangjian Huang
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
31
|
Control of CCND1 ubiquitylation by the catalytic SAGA subunit USP22 is essential for cell cycle progression through G1 in cancer cells. Proc Natl Acad Sci U S A 2018; 115:E9298-E9307. [PMID: 30224477 PMCID: PMC6176615 DOI: 10.1073/pnas.1807704115] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Overexpression of the deubiquitylase ubiquitin-specific peptidase 22 (USP22) is a marker of aggressive cancer phenotypes like metastasis, therapy resistance, and poor survival. Functionally, this overexpression of USP22 actively contributes to tumorigenesis, as USP22 depletion blocks cancer cell cycle progression in vitro, and inhibits tumor progression in animal models of lung, breast, bladder, ovarian, and liver cancer, among others. Current models suggest that USP22 mediates these biological effects via its role in epigenetic regulation as a subunit of the Spt-Ada-Gcn5-acetyltransferase (SAGA) transcriptional cofactor complex. Challenging the dogma, we report here a nontranscriptional role for USP22 via a direct effect on the core cell cycle machinery: that is, the deubiquitylation of the G1 cyclin D1 (CCND1). Deubiquitylation by USP22 protects CCND1 from proteasome-mediated degradation and occurs separately from the canonical phosphorylation/ubiquitylation mechanism previously shown to regulate CCND1 stability. We demonstrate that control of CCND1 is a key mechanism by which USP22 mediates its known role in cell cycle progression. Finally, USP22 and CCND1 levels correlate in patient lung and colorectal cancer samples and our preclinical studies indicate that targeting USP22 in combination with CDK inhibitors may offer an approach for treating cancer patients whose tumors exhibit elevated CCND1.
Collapse
|
32
|
Li F, Zhang Q, Gong Y, Yu J. The lncKLF6/KLF6 feedback loop regulates the growth of non-small cell lung cancer. Am J Cancer Res 2018; 8:1427-1439. [PMID: 30210914 PMCID: PMC6129497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 07/30/2018] [Indexed: 06/08/2023] Open
Abstract
Non-small lung cancer (NSCLC) is one of the most common causes of cancer-associated death worldwide. Long noncoding RNAs (lncRNAs) regulate cancer initiation and progression through different mechanisms. In the present study, we characterized a novel lncRNA named lncKLF6, which was upregulated in NSCLC and associated with poor clinical outcomes. lncKLF6 inhibited Kruppel-like factor 6 (KLF6) transcription and then facilitated NSCLC growth. lncKLF6 is associated with the epigenetic repressor BMI1 and regulates its stability via recruiting deubiquitinase USP22. Moreover, it was revealed that lncKLF6 was a KLF6-responsive lncRNA, as KLF6 could occupy the lncKLF6 promoter to facilitate its transcription. The negative feedback loop of lncKLF6 and KLF6 continuously enhanced the oncogenic effects. Thus, our study elucidates the mechanism of lncKLF6-mediated growth via suppression of KLF6, which provides the promising target for developing new therapeutic strategy in NSCLC.
Collapse
Affiliation(s)
- Fei Li
- Department of Pulmonary and Critical Care Medicine (PCCM) ward II, Hebei Cangzhou Central Hospital Hebei Province, China
| | - Qianyun Zhang
- Department of Pulmonary and Critical Care Medicine (PCCM) ward II, Hebei Cangzhou Central Hospital Hebei Province, China
| | - Yange Gong
- Department of Pulmonary and Critical Care Medicine (PCCM) ward II, Hebei Cangzhou Central Hospital Hebei Province, China
| | - Jinxiang Yu
- Department of Pulmonary and Critical Care Medicine (PCCM) ward II, Hebei Cangzhou Central Hospital Hebei Province, China
| |
Collapse
|
33
|
Qiu GZ, Mao XY, Ma Y, Gao XC, Wang Z, Jin MZ, Sun W, Zou YX, Lin J, Fu HL, Jin WL. Ubiquitin-specific protease 22 acts as an oncoprotein to maintain glioma malignancy through deubiquitinating B cell-specific Moloney murine leukemia virus integration site 1 for stabilization. Cancer Sci 2018; 109:2199-2210. [PMID: 29788550 PMCID: PMC6029839 DOI: 10.1111/cas.13646] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 05/08/2018] [Accepted: 05/16/2018] [Indexed: 12/13/2022] Open
Abstract
Ubiquitin‐specific protease 22 (USP22) is a member of the “death‐from‐cancer” signature, which plays a key role in cancer progression. Previous evidence has shown that USP22 is overexpressed and correlates with poor prognosis in glioma. The effect and mechanism of USP22 in glioma malignancy, especially cancer stemness, remain elusive. Herein, we find USP22 is more enriched in stem‐like tumorspheres than differentiated glioma cells. USP22 knockdown inhibits cancer stemness in glioma cell lines. With a cell‐penetrating TAT‐tag protein, B cell‐specific Moloney murine leukemia virus integration site 1 (BMI1), a robust glioma stem‐cell marker, is found to mediate the effect of USP22 on glioma stemness. By immunofluorescence, USP22 and BMI1 are found to share similar intranuclear expression in glioma cells. By analysis with immunohistochemistry and bioinformatics, USP22 is found to positively correlate with BMI1 at the post‐translational level only rather than at the transcriptional level. By immunoprecipitation and in vivo deubiquitination assay, USP22 is found to interact with and deubiquitinate BMI1 for protein stabilization. Microarray analysis shows that USP22 and BMI1 mutually regulate a series of genes involved in glioma stemness such as POSTN,HEY2,PDGFRA and ATF3. In vivo study with nude mice confirms the role of USP22 in promoting glioma tumorigenesis by regulating BMI1. All these findings indicate USP22 as a novel deubiquitinase of BMI1 in glioma. We propose a working model of the USP22‐BMI1 axis, which promotes glioma stemness and tumorigenesis through oncogenic activation. Thus, targeting USP22 might be an effective strategy to treat glioma especially in those with elevated BMI1 expression.
Collapse
Affiliation(s)
- Guan-Zhong Qiu
- Department of Instrument Science and Engineering, Key Lab. for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Institute of Nano Biomedicine and Engineering, Shanghai Jiao Tong University, Shanghai, China.,Department of Neurosurgery, General Hospital of Jinan Military Command, Jinan, China
| | - Xiao-Yuan Mao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Central South University, Changsha, China.,Human Key Laboratory of Pharmacogenetics, Changsha, China
| | - Yue Ma
- Department of Instrument Science and Engineering, Key Lab. for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Institute of Nano Biomedicine and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Xing-Chun Gao
- Shanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic Medical Medicine, Xi'an Medical University, Xi'an, China
| | - Zhen Wang
- Department of Instrument Science and Engineering, Key Lab. for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Institute of Nano Biomedicine and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Ming-Zhu Jin
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Sun
- Department of Neurosurgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Yong-Xiang Zou
- Department of Neurosurgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Jing Lin
- Department of Neurosurgery, The General Hospital of Western Air Force, Chengdu, China
| | - Hua-Lin Fu
- Department of Instrument Science and Engineering, Key Lab. for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Institute of Nano Biomedicine and Engineering, Shanghai Jiao Tong University, Shanghai, China.,National Centers for Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei-Lin Jin
- Department of Instrument Science and Engineering, Key Lab. for Thin Film and Microfabrication Technology of Ministry of Education, School of Electronic Information and Electronic Engineering, Institute of Nano Biomedicine and Engineering, Shanghai Jiao Tong University, Shanghai, China.,National Centers for Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|