1
|
Xie SY, Liu SQ, Zhang T, Shi WK, Xing Y, Fang WX, Zhang M, Chen MY, Xu SC, Fan MQ, Li LL, Zhang H, Zhao N, Zeng ZX, Chen S, Zeng XF, Deng W, Tang QZ. USP28 Serves as a Key Suppressor of Mitochondrial Morphofunctional Defects and Cardiac Dysfunction in the Diabetic Heart. Circulation 2024; 149:684-706. [PMID: 37994595 DOI: 10.1161/circulationaha.123.065603] [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: 05/17/2023] [Accepted: 10/30/2023] [Indexed: 11/24/2023]
Abstract
BACKGROUND The majority of people with diabetes are susceptible to cardiac dysfunction and heart failure, and conventional drug therapy cannot correct diabetic cardiomyopathy progression. Herein, we assessed the potential role and therapeutic value of USP28 (ubiquitin-specific protease 28) on the metabolic vulnerability of diabetic cardiomyopathy. METHODS The type 2 diabetes mouse model was established using db/db leptin receptor-deficient mice and high-fat diet/streptozotocin-induced mice. Cardiac-specific knockout of USP28 in the db/db background mice was generated by crossbreeding db/m and Myh6-Cre+/USP28fl/fl mice. Recombinant adeno-associated virus serotype 9 carrying USP28 under cardiac troponin T promoter was injected into db/db mice. High glucose plus palmitic acid-incubated neonatal rat ventricular myocytes and human induced pluripotent stem cell-derived cardiomyocytes were used to imitate diabetic cardiomyopathy in vitro. The molecular mechanism was explored through RNA sequencing, immunoprecipitation and mass spectrometry analysis, protein pull-down, chromatin immunoprecipitation sequencing, and chromatin immunoprecipitation assay. RESULTS Microarray profiling of the UPS (ubiquitin-proteasome system) on the basis of db/db mouse hearts and diabetic patients' hearts demonstrated that the diabetic ventricle presented a significant reduction in USP28 expression. Diabetic Myh6-Cre+/USP28fl/fl mice exhibited more severe progressive cardiac dysfunction, lipid accumulation, and mitochondrial disarrangement, compared with their controls. On the other hand, USP28 overexpression improved systolic and diastolic dysfunction and ameliorated cardiac hypertrophy and fibrosis in the diabetic heart. Adeno-associated virus serotype 9-USP28 diabetic mice also exhibited less lipid storage, reduced reactive oxygen species formation, and mitochondrial impairment in heart tissues than adeno-associated virus serotype 9-null diabetic mice. As a result, USP28 overexpression attenuated cardiac remodeling and dysfunction, lipid accumulation, and mitochondrial impairment in high-fat diet/streptozotocin-induced type 2 diabetes mice. These results were also confirmed in neonatal rat ventricular myocytes and human induced pluripotent stem cell-derived cardiomyocytes. RNA sequencing, immunoprecipitation and mass spectrometry analysis, chromatin immunoprecipitation assays, chromatin immunoprecipitation sequencing, and protein pull-down assay mechanistically revealed that USP28 directly interacted with PPARα (peroxisome proliferator-activated receptor α), deubiquitinating and stabilizing PPARα (Lys152) to promote Mfn2 (mitofusin 2) transcription, thereby impeding mitochondrial morphofunctional defects. However, such cardioprotective benefits of USP28 were largely abrogated in db/db mice with PPARα deletion and conditional loss-of-function of Mfn2. CONCLUSIONS Our findings provide a USP28-modulated mitochondria homeostasis mechanism that involves the PPARα-Mfn2 axis in diabetic hearts, suggesting that USP28 activation or adeno-associated virus therapy targeting USP28 represents a potential therapeutic strategy for diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Sai-Yang Xie
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Shi-Qiang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Tong Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Wen-Ke Shi
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Yun Xing
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Wen-Xi Fang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Min Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Meng-Ya Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Si-Chi Xu
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, P.R. China (S.-c.X.)
| | - Meng-Qi Fan
- College of Life Sciences, Wuhan University, P.R. China (M.-q.F.)
| | - Lan-Lan Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Heng Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Nan Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Zhao-Xiang Zeng
- Department of Vascular Surgery, Shanghai General Hospital, Shanghai Jiaotong University, P.R. China (Z.-x.Z)
- Department of Cardiac Surgery, Changhai Hospital, Navy Medical University, Shanghai, P.R. China (Z.-x.Z)
| | - Si Chen
- Cardiovascular Research Institute of Wuhan University, P.R. China (S.C., X.-f.Z.)
| | - Xiao-Feng Zeng
- Cardiovascular Research Institute of Wuhan University, P.R. China (S.C., X.-f.Z.)
| | - Wei Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, P.R. China (S.-y.X, S.-q.L., T.Z., W.-k.S., Y.X., W.-x.F., M.Z., M.-Y.C., L.-l.L., H.Z., N.Z., W.D., Q.z.T.)
| |
Collapse
|
2
|
Karlowitz R, Stanifer ML, Roedig J, Andrieux G, Bojkova D, Bechtel M, Smith S, Kowald L, Schubert R, Boerries M, Cinatl J, Boulant S, van Wijk SJL. USP22 controls type III interferon signaling and SARS-CoV-2 infection through activation of STING. Cell Death Dis 2022; 13:684. [PMID: 35933402 PMCID: PMC9357023 DOI: 10.1038/s41419-022-05124-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 07/21/2022] [Accepted: 07/21/2022] [Indexed: 01/21/2023]
Abstract
Pattern recognition receptors (PRRs) and interferons (IFNs) serve as essential antiviral defense against SARS-CoV-2, the causative agent of the COVID-19 pandemic. Type III IFNs (IFN-λ) exhibit cell-type specific and long-lasting functions in auto-inflammation, tumorigenesis, and antiviral defense. Here, we identify the deubiquitinating enzyme USP22 as central regulator of basal IFN-λ secretion and SARS-CoV-2 infections in human intestinal epithelial cells (hIECs). USP22-deficient hIECs strongly upregulate genes involved in IFN signaling and viral defense, including numerous IFN-stimulated genes (ISGs), with increased secretion of IFN-λ and enhanced STAT1 signaling, even in the absence of exogenous IFNs or viral infection. Interestingly, USP22 controls basal and 2'3'-cGAMP-induced STING activation and loss of STING reversed STAT activation and ISG and IFN-λ expression. Intriguingly, USP22-deficient hIECs are protected against SARS-CoV-2 infection, viral replication, and the formation of de novo infectious particles, in a STING-dependent manner. These findings reveal USP22 as central host regulator of STING and type III IFN signaling, with important implications for SARS-CoV-2 infection and antiviral defense.
Collapse
Affiliation(s)
- Rebekka Karlowitz
- grid.7839.50000 0004 1936 9721Institute for Experimental Cancer Research in Pediatrics, Goethe University Frankfurt, Komturstrasse 3a, 60528 Frankfurt am Main, Germany
| | - Megan L. Stanifer
- grid.7700.00000 0001 2190 4373Department of Infectious Diseases/Molecular Virology, Medical Faculty, Center for Integrative Infectious Diseases Research (CIID), University of Heidelberg, 69120 Heidelberg, Germany ,grid.15276.370000 0004 1936 8091Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, FL USA
| | - Jens Roedig
- grid.7839.50000 0004 1936 9721Institute for Experimental Cancer Research in Pediatrics, Goethe University Frankfurt, Komturstrasse 3a, 60528 Frankfurt am Main, Germany
| | - Geoffroy Andrieux
- grid.5963.9Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - Denisa Bojkova
- grid.411088.40000 0004 0578 8220Institute of Medical Virology, University Hospital Frankfurt, Goethe University, 60596 Frankfurt am Main, Germany
| | - Marco Bechtel
- grid.411088.40000 0004 0578 8220Institute of Medical Virology, University Hospital Frankfurt, Goethe University, 60596 Frankfurt am Main, Germany
| | - Sonja Smith
- grid.7839.50000 0004 1936 9721Institute for Experimental Cancer Research in Pediatrics, Goethe University Frankfurt, Komturstrasse 3a, 60528 Frankfurt am Main, Germany
| | - Lisa Kowald
- grid.7839.50000 0004 1936 9721Institute for Experimental Cancer Research in Pediatrics, Goethe University Frankfurt, Komturstrasse 3a, 60528 Frankfurt am Main, Germany
| | - Ralf Schubert
- grid.411088.40000 0004 0578 8220Division for Allergy, Pneumology and Cystic Fibrosis, Department for Children and Adolescents, University Hospital Frankfurt, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Melanie Boerries
- grid.5963.9Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany ,grid.7497.d0000 0004 0492 0584German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), partner site Freiburg, 79110 Freiburg, Germany
| | - Jindrich Cinatl
- grid.411088.40000 0004 0578 8220Institute of Medical Virology, University Hospital Frankfurt, Goethe University, 60596 Frankfurt am Main, Germany
| | - Steeve Boulant
- grid.15276.370000 0004 1936 8091Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, FL USA ,grid.7700.00000 0001 2190 4373Department of Infectious Diseases, Virology, Medical Faculty, Center for Integrative Infectious Diseases Research (CIID), University of Heidelberg, 69120 Heidelberg, Germany
| | - Sjoerd J. L. van Wijk
- grid.7839.50000 0004 1936 9721Institute for Experimental Cancer Research in Pediatrics, Goethe University Frankfurt, Komturstrasse 3a, 60528 Frankfurt am Main, Germany ,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) partner site Frankfurt/Mainz, Frankfurt am Main, Germany
| |
Collapse
|
3
|
Song S, Wang Y, Wang HY, Guo LL. Role of sevoflurane in myocardial ischemia-reperfusion injury via the ubiquitin-specific protease 22/lysine-specific demethylase 3A axis. Bioengineered 2022; 13:13366-13383. [PMID: 36700466 PMCID: PMC9275884 DOI: 10.1080/21655979.2022.2062535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Myocardial ischemia-reperfusion injury (MIRI) represents a coronary artery disease, accompanied by high morbidity and mortality. Sevoflurane post-conditioning (SPC) is importantly reported in myocardial disease. Accordingly, the current study sought to evaluate the role of Sevo in MI/RI. Firstly, MI/RI models were established and subjected to SPC. Subsequently, pathological injury in the myocardium, myocardial infarction areas, H9c2 cell viability, apoptosis, and levels of creatine kinase-MB (CK-MB), cardiac troponin I (cTnI), and lactate dehydrogenase (LDH) were all measured. Ubiquitin-specific peptidase (22USP22), lysine-specific demethylase 3A (KDM3A), and Yes1 associated transcriptional regulator (YAP1) were down-regulated in H9c2 cells using cell transfection to verify their roles. The interaction between USP22 and KDM3A and between KDM3A and YAP1 was further validated. USP 22, KDM3A, and YAP1 were found to be down-regulated in MI/RI and SPC protected MI/RI rats, as evidenced by up-regulated expressions of USP22, KDM3A, and YAP1, reduced pathological injury in the myocardium, myocardial infarction areas, apoptosis, and levels of CK-MB, cTnI, and LDH, and enhanced H9c2 cell viability; while the protective effects of Sevo were counteracted by silencing of USP22, KDM3A, and SPC upregulated USP22, which stabilized KDM3A protein levels via deubiquitination, and KDM3A inhibited histone 3 lysine 9 di-methylation (H3K9me2) levels in the YAP1 promoter to encourage YAP1 transcription, to reduce MI/RI.
Collapse
Affiliation(s)
- Shan Song
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Yang Wang
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Hai-Yan Wang
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China,Hai-Yan Wang Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, No. 20 Yuhuangding East Road, Zhifu District, Yantai City264000, Shandong Province, China
| | - Long-Long Guo
- Department of Anesthesiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China,CONTACT Long-Long Guo
| |
Collapse
|
4
|
Yang Y, Zhou X, Liu X, Song R, Gao Y, Wang S. Implications of FBXW7 in Neurodevelopment and Neurodegeneration: Molecular Mechanisms and Therapeutic Potential. Front Cell Neurosci 2021; 15:736008. [PMID: 34512273 PMCID: PMC8424092 DOI: 10.3389/fncel.2021.736008] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 08/04/2021] [Indexed: 11/25/2022] Open
Abstract
The ubiquitin-proteasome system (UPS) mediated protein degradation is crucial to maintain quantitive and functional homeostasis of diverse proteins. Balanced cellular protein homeostasis controlled by UPS is fundamental to normal neurological functions while impairment of UPS can also lead to some neurodevelopmental and neurodegenerative disorders. Functioning as the substrate recognition component of the SCF-type E3 ubiquitin ligase, FBXW7 is essential to multiple aspects of cellular processes via targeting a wide range of substrates for proteasome-mediated degradation. Accumulated evidence shows that FBXW7 is fundamental to neurological functions and especially implicated in neurodevelopment and the nosogenesis of neurodegeneration. In this review, we describe general features of FBXW7 gene and proteins, and mainly present recent findings that highlight the vital roles and molecular mechanisms of FBXW7 in neurodevelopment such as neurogenesis, myelination and cerebral vasculogenesis and in the pathogenesis of some typical neurodegenerative disorders such as Alzheimer’s disease, Parkinson’s disease and Huntington’s disease. Additionally, we also provide a prospect on focusing FBXW7 as a potential therapeutic target to rescue neurodevelopmental and neurodegenerative impairment.
Collapse
Affiliation(s)
- Yu Yang
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China.,Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China
| | - Xuan Zhou
- Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China.,Research Center for Quality of Life and Applied Psychology, School of Humanities and Management, Guangdong Medical University, Dongguan, China
| | - Xinpeng Liu
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China.,Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China
| | - Ruying Song
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China.,Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China
| | - Yiming Gao
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China.,Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China
| | - Shuai Wang
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China.,Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China
| |
Collapse
|
5
|
Feng T, Ling S, Xu C, Ying L, Su D, Xu X. Ubiquitin-specific peptidase 22 in cancer. Cancer Lett 2021; 514:30-37. [PMID: 33989708 DOI: 10.1016/j.canlet.2021.05.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/21/2021] [Accepted: 05/05/2021] [Indexed: 02/07/2023]
Abstract
Recently, many studies have shown that deubiquitination modification of proteins is of great significance in major physiological processes such as cell proliferation, apoptosis, and differentiation. The ubiquitin-specific peptidase (USP) family is one of the most numerous and structurally diverse of the deubiquitinates known to date. USP22, an important member of the USP family, has been found to be closely associated with tumor cell cycle regulation, stemness maintenance, invasion and metastasis, chemoresistance, and immune regulation. We focus on recent advances regarding USP22's function in cancer and discuss the prospect of USP22 in this review.
Collapse
Affiliation(s)
- Tingting Feng
- Department of Pathology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer(IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Cancer Research Institute, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer(IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Department of Colorectal Medicine, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer(IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Department of Hepatobiliary and Pancreatic Surgery, The Center for Integrated Oncology and Precision Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Sunbin Ling
- Department of Hepatobiliary and Pancreatic Surgery, The Center for Integrated Oncology and Precision Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Chenyang Xu
- Department of Pathology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer(IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Lisha Ying
- Cancer Research Institute, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer(IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Dan Su
- Department of Pathology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer(IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| | - Xiao Xu
- Department of Hepatobiliary and Pancreatic Surgery, The Center for Integrated Oncology and Precision Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| |
Collapse
|
6
|
Roedig J, Kowald L, Juretschke T, Karlowitz R, Ahangarian Abhari B, Roedig H, Fulda S, Beli P, van Wijk SJL. USP22 controls necroptosis by regulating receptor-interacting protein kinase 3 ubiquitination. EMBO Rep 2021; 22:e50163. [PMID: 33369872 PMCID: PMC7857539 DOI: 10.15252/embr.202050163] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 10/21/2020] [Accepted: 11/03/2020] [Indexed: 12/27/2022] Open
Abstract
Dynamic control of ubiquitination by deubiquitinating enzymes is essential for almost all biological processes. Ubiquitin-specific peptidase 22 (USP22) is part of the SAGA complex and catalyzes the removal of mono-ubiquitination from histones H2A and H2B, thereby regulating gene transcription. However, novel roles for USP22 have emerged recently, such as tumor development and cell death. Apart from apoptosis, the relevance of USP22 in other programmed cell death pathways still remains unclear. Here, we describe a novel role for USP22 in controlling necroptotic cell death in human tumor cell lines. Loss of USP22 expression significantly delays TNFα/Smac mimetic/zVAD.fmk (TBZ)-induced necroptosis, without affecting TNFα-mediated NF-κB activation or extrinsic apoptosis. Ubiquitin remnant profiling identified receptor-interacting protein kinase 3 (RIPK3) lysines 42, 351, and 518 as novel, USP22-regulated ubiquitination sites during necroptosis. Importantly, mutation of RIPK3 K518 reduced necroptosis-associated RIPK3 ubiquitination and amplified necrosome formation and necroptotic cell death. In conclusion, we identify a novel role of USP22 in necroptosis and further elucidate the relevance of RIPK3 ubiquitination as crucial regulator of necroptotic cell death.
Collapse
Affiliation(s)
- Jens Roedig
- Institute for Experimental Cancer Research in PediatricsGoethe‐UniversityFrankfurt am MainGermany
| | - Lisa Kowald
- Institute for Experimental Cancer Research in PediatricsGoethe‐UniversityFrankfurt am MainGermany
| | | | - Rebekka Karlowitz
- Institute for Experimental Cancer Research in PediatricsGoethe‐UniversityFrankfurt am MainGermany
| | - Behnaz Ahangarian Abhari
- Lighthouse Core FacilityZentrum für Translationale ZellforschungUniversitaetsklinikum FreiburgKlinik für Innere Medizin IFreiburgGermany
| | - Heiko Roedig
- Pharmazentrum FrankfurtInstitut für Allgemeine Pharmakologie und ToxikologieGoethe‐UniversityFrankfurt am MainGermany
| | - Simone Fulda
- Institute for Experimental Cancer Research in PediatricsGoethe‐UniversityFrankfurt am MainGermany
| | - Petra Beli
- Institute of Molecular Biology (IMB)MainzGermany
| | - Sjoerd JL van Wijk
- Institute for Experimental Cancer Research in PediatricsGoethe‐UniversityFrankfurt am MainGermany
| |
Collapse
|
7
|
Bai Z, Du Y, Cong L, Cheng Y. The USP22 promotes the growth of cancer cells through the DYRK1A in pancreatic ductal adenocarcinoma. Gene 2020; 758:144960. [PMID: 32687947 DOI: 10.1016/j.gene.2020.144960] [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: 03/03/2020] [Revised: 06/16/2020] [Accepted: 07/13/2020] [Indexed: 01/27/2023]
Abstract
As a member of the ubiquitin-specific protease (USP) family, USP22 could remove ubiquitin moieties from its target proteins to control the function of the target proteins. Accumulating studies show that USP22 essentially participates in diverse types of cancer as an oncogene-like protein. However, the roles of USP22 in human pancreatic ductal adenocarcinoma (PDAC) and the underlying mechanism are unknown. Here we report that USP22 promotes the growth of PDAC cells by promoting the expression of dual-specificity tyrosine regulated kinase 1A (DYRK1A). Our results showed that the expression levels of USP22 were up-regulated in human PDAC tissues and cell lines (BxPC-3, AsPC-1, MIA-PaCa-2, PANC-1, and CAPAN-1). Lentivirus-mediated knockdown of USP22 repressed the rate of proliferation and capacity of colony formation of BxPC3 and CAPAN1 cancer cells and USP22 overexpression promoted the proliferation and capacity of the colony formation of BxPC3 and CAPAN1 cancer cells. The further mechanism study showed that USP22 elevated the expression of the mRNA and protein levels of DYRK1A in PDAC cancer cells. Inhibition of DYRK1A with EHT-5732 or lentivirus-mediated knockdown of DYRK1A blocked the function of USP22 overexpression in the regulation of the proliferation and colony formation of PDAC cells. Taken together, our findings demonstrated that USP22 overexpression in PDAC promoted the growth of the cancer cells partially through upregulating the expression of DYRK1A.
Collapse
Affiliation(s)
- Zhile Bai
- Key Laboratory of Ethnomedicine for Ministry of Education, Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Yang Du
- Key Laboratory of Ethnomedicine for Ministry of Education, Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Lin Cong
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 100730 Beijing, China.
| | - Yong Cheng
- Key Laboratory of Ethnomedicine for Ministry of Education, Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing, China.
| |
Collapse
|
8
|
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
|
9
|
ERAP1 promotes Hedgehog-dependent tumorigenesis by controlling USP47-mediated degradation of βTrCP. Nat Commun 2019; 10:3304. [PMID: 31341163 PMCID: PMC6656771 DOI: 10.1038/s41467-019-11093-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 06/18/2019] [Indexed: 12/14/2022] Open
Abstract
The Hedgehog (Hh) pathway is essential for embryonic development and tissue homeostasis. Aberrant Hh signaling may occur in a wide range of human cancers, such as medulloblastoma, the most common brain malignancy in childhood. Here, we identify endoplasmic reticulum aminopeptidase 1 (ERAP1), a key regulator of innate and adaptive antitumor immune responses, as a previously unknown player in the Hh signaling pathway. We demonstrate that ERAP1 binds the deubiquitylase enzyme USP47, displaces the USP47-associated βTrCP, the substrate-receptor subunit of the SCFβTrCP ubiquitin ligase, and promotes βTrCP degradation. These events result in the modulation of Gli transcription factors, the final effectors of the Hh pathway, and the enhancement of Hh activity. Remarkably, genetic or pharmacological inhibition of ERAP1 suppresses Hh-dependent tumor growth in vitro and in vivo. Our findings unveil an unexpected role for ERAP1 in cancer and indicate ERAP1 as a promising therapeutic target for Hh-driven tumors. ERAP1 is an endoplasmic reticulum aminopeptidase that trims MHC Class-I peptides for antigen presentation. Here, the authors show that ERAP1 enhances Hedgehog signalling by sequestering USP47 from βTrCP and promoting tumorigenesis through βTrCP degradation and increased Gli protein stability.
Collapse
|
10
|
王 玉, 张 淑, 穆 淑, 张 柏, 马 树. [USP33 suppresses lung adenocarcinoma lung cell invasion and metastasis by down-regulating SLIT2/ROBO1 signaling pathway]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:956-961. [PMID: 30187867 PMCID: PMC6744049 DOI: 10.3969/j.issn.1673-4254.2018.08.09] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To investigate the role of USP33 as an independent prognostic marker in the regulation of SLIT2/ROBO1 signaling pathway to inhibit lung adenocarcinoma invasion and metastasis. METHODS The expression of USP33 in 20 lung adenocarcinoma specimens was detected by qPCR and immunohistochemistry. A549 and SPC-A-1 cells with small interfering RNA (siRNA)-mediated USP33 silencing were examined for changes in invasion and metastasis abilities using scratch assay and Matrigel assay. Western blotting was used to detect the expression of SLIT2 and ROBO1 in the cells after USP33 silencing and the expression of USP33 after interleukin-6 (IL-6) stimulation. RESULTS qPCR and immunohistochemistry showed that USP33 was significantly decreased in lung adenocarcinoma tissues as compared with the adjacent tissues. USP33 silencing in A549 and SPC-A-1 cells significantly promoted the cell migration, invasion and metastasis and obviously down-regulated the expressions of SLIT2 and ROBO1. IL-6 stimulation of the cells obviously enhanced the expression of USP33. CONCLUSIONS USP33 silencing can promote the migration, invasion and metastasis of lung adenocarcinoma cells in vitro, and the mechanism may involve IL-6 and SLIT2/ROBO1 signaling pathways.
Collapse
Affiliation(s)
- 玉环 王
- 南方医科大学南方医院肿瘤科,广东 广州 510515Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 淑华 张
- 南方医科大学南方医院肿瘤科,广东 广州 510515Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 淑坤 穆
- 南方医科大学南方医院肿瘤科,广东 广州 510515Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 柏深 张
- 南方医科大学南方医院肿瘤科,广东 广州 510515Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 树东 马
- 南方医科大学南方医院肿瘤科,广东 广州 510515Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- 新疆喀什地区第一人民医院肿瘤中心,新疆 喀什 844000Cancer Center of the First People's Hospital of Kashi, Kashi 844000, China
| |
Collapse
|
11
|
Melo-Cardenas J, Zhang Y, Zhang DD, Fang D. Ubiquitin-specific peptidase 22 functions and its involvement in disease. Oncotarget 2018; 7:44848-44856. [PMID: 27057639 PMCID: PMC5190139 DOI: 10.18632/oncotarget.8602] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 03/10/2016] [Indexed: 12/24/2022] Open
Abstract
Deubiquitylases remove ubiquitin moieties from different substrates to regulate protein activity and cell homeostasis. Since this posttranslational modification plays a role in several different cellular functions, its deregulation has been associated with different pathologies. Aberrant expression of the Ubiquitin-Specific Peptidase 22 (USP22) has been associated with poor cancer prognosis and neurological disorders. However, little is known about USP22 role in these pathologies or in normal physiology. This review summarizes the current knowledge about USP22 function from yeast to human and provides an overview of the possible mechanisms by which USP22 is emerging as a potential oncogene.
Collapse
Affiliation(s)
- Johanna Melo-Cardenas
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yusi Zhang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Donna D Zhang
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA
| | - Deyu Fang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
12
|
Kim D, Hong A, Park HI, Shin WH, Yoo L, Jeon SJ, Chung KC. Deubiquitinating enzyme USP22 positively regulates c-Myc stability and tumorigenic activity in mammalian and breast cancer cells. J Cell Physiol 2017; 232:3664-3676. [PMID: 28160502 DOI: 10.1002/jcp.25841] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 02/03/2017] [Accepted: 02/03/2017] [Indexed: 12/22/2022]
Abstract
The proto-oncogene c-Myc has a pivotal function in growth control, differentiation, and apoptosis and is frequently affected in human cancer, including breast cancer. Ubiquitin-specific protease 22 (USP22), a member of the USP family of deubiquitinating enzymes (DUBs), mediates deubiquitination of target proteins, including histone H2B and H2A, telomeric repeat binding factor 1, and cyclin B1. USP22 is also a component of the mammalian SAGA transcriptional co-activating complex. In this study, we explored the functional role of USP22 in modulating c-Myc stability and its physiological relevance in breast cancer progression. We found that USP22 promotes deubiquitination of c-Myc in several breast cancer cell lines, resulting in increased levels of c-Myc. Consistent with this, USP22 knockdown reduces c-Myc levels. Furthermore, overexpression of USP22 stimulates breast cancer cell growth and colony formation, and increases c-Myc tumorigenic activity. In conclusion, the present study reveals that USP22 in breast cancer cell lines increases c-Myc stability through c-Myc deubiquitination, which is closely correlated with breast cancer progression.
Collapse
Affiliation(s)
- Dongyeon Kim
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Ahyoung Hong
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Hye In Park
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Woo Hyun Shin
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Lang Yoo
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Seo Jeong Jeon
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Kwang Chul Chung
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| |
Collapse
|
13
|
Kosinsky RL, Wegwitz F, Hellbach N, Dobbelstein M, Mansouri A, Vogel T, Begus-Nahrmann Y, Johnsen SA. Usp22 deficiency impairs intestinal epithelial lineage specification in vivo. Oncotarget 2016; 6:37906-18. [PMID: 26431380 PMCID: PMC4741973 DOI: 10.18632/oncotarget.5412] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 09/14/2015] [Indexed: 12/31/2022] Open
Abstract
Epigenetic regulatory mechanisms play a central role in controlling gene expression during development, cell differentiation and tumorigenesis. Monoubiquitination of histone H2B is one epigenetic modification which is dynamically regulated by the opposing activities of specific ubiquitin ligases and deubiquitinating enzymes (DUBs). The Ubiquitin-specific Protease 22 (USP22) is the ubiquitin hydrolase component of the human SAGA complex which deubiquitinates histone H2B during transcription. Recently, many studies have investigated an oncogenic potential of USP22 overexpression. However, its physiological function in organ maintenance, development and its cellular function remain largely unknown. A previous study reported embryonic lethality in Usp22 knockout mice. Here we describe a mouse model with a global reduction of USP22 levels which expresses the LacZ gene under the control of the endogenous Usp22 promoter. Using this reporter we found Usp22 to be ubiquitously expressed in murine embryos. Notably, adult Usp22lacZ/lacZ displayed low residual Usp22 expression levels coupled with a reduced body size and weight. Interestingly, the reduction of Usp22 significantly influenced the frequency of differentiated cells in the small intestine and the brain while H2B and H2Bub1 levels remained constant. Taken together, we provide evidence for a physiological role for USP22 in controlling cell differentiation and lineage specification.
Collapse
Affiliation(s)
- Robyn L Kosinsky
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, 37075 Göttingen, Germany.,Institute of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), Faculty of Medicine, University of Göttingen, 37077 Göttingen, Germany
| | - Florian Wegwitz
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Nicole Hellbach
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Matthias Dobbelstein
- Institute of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), Faculty of Medicine, University of Göttingen, 37077 Göttingen, Germany
| | - Ahmed Mansouri
- Department of Molecular Cell Biology, Max-Planck Institute for Biophysical Chemistry, RG Molecular Cell Differentiation, 37077 Göttingen, Germany
| | - Tanja Vogel
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Yvonne Begus-Nahrmann
- Institute of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), Faculty of Medicine, University of Göttingen, 37077 Göttingen, Germany
| | - Steven A Johnsen
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, 37075 Göttingen, Germany
| |
Collapse
|