1
|
Zhuang Z, Zhang L, Wang Y. USP2 alleviates MPP +-induced neuronal injury by stabilizing FOXC1 in SK-N-SH cells. Brain Res 2025; 1862:149689. [PMID: 40355038 DOI: 10.1016/j.brainres.2025.149689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 04/08/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Forkhead box transcription factors play a crucial role in the development of various organ systems, and exhibit neuroprotective properties in many neurodegenerative diseases, including Parkinson's disease (PD). However, the role and mechanism of Forkhead box C1 (FOXC1) in the pathogenesis of PD is poorly defined. METHODS Human neuroblastoma SK-N-SH cells were treated with 1-methyl-4-phenylpyridinium (MPP+) to establish an in vitro model of PD. FOXC1 and Ubiquitin-specific peptidase 2 (USP2) mRNA levels were detected using real-time quantitative polymerase chain reaction (RT-qPCR). FOXC1, B-cell lymphoma-2 (Bcl-2), Bcl-2 related X protein (Bax), Cleaved caspase-3, and USP2 protein levels were determined using Western blot. Cell viability and apoptosis were analyzed using CCK-8 assay and flow cytometry. Tumor necrosis factor α (TNF-α), Interleukin-1β (IL-1β), and IL-6 levels were analyzed using ELISA. Reactive oxygen species (ROS), malondialdehyde (MDA), superoxide dismutase (SOD), catalase (CAT), and Glutathione (GSH) products were examined using special assay kits. After Ubibrowser online database prediction, the interaction between USP2 and FOXC1 was verified using Co-immunoprecipitation (CoIP) assay. RESULTS FOXC1 and USP2 expression were decreased in PD patients and MPP+-treated SK-N-SH cells. MPP+ treatment could elicit SK-N-SH cell viability inhibition, inflammatory response, oxidative stress, and apoptosis promotion in vitro. Furthermore, overexpressing FOXC1 relieved MPP+-induced SK-N-SH cell injury in vitro. Mechanistically, USP2 directly interacted with FOXC1 and deubiquitinated FOXC1, therefore enhancing FOXC1 protein stability. CONCLUSION USP2 attenuated MPP+-triggered SK-N-SH cell injury through stabilizing FOXC1, providing a promising therapeutic target for PD treatment.
Collapse
Affiliation(s)
- Zhijiang Zhuang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou City 450052 Henan, China.
| | - Lihong Zhang
- Department of Integrated Chinese and Western Medicine, Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou City Henan, China
| | - Yizhao Wang
- Department of Rehabilitation, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou City 450052 Henan, China
| |
Collapse
|
2
|
Yang S, Xiong L, Liao T, Li L, Li Y, Kang L, Yang G, Liang Z. Deubiquitinating Enzyme USP2 Alleviates Muscle Atrophy by Stabilizing PPAR-γ. Diabetes 2025; 74:773-786. [PMID: 39874418 PMCID: PMC12015143 DOI: 10.2337/db24-0375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 01/23/2025] [Indexed: 01/30/2025]
Abstract
Insulin resistance, a hallmark of type 2 diabetes, accelerates muscle breakdown and impairs energy metabolism. However, the role of ubiquitin specific peptidase 2 (USP2), a key regulator of insulin resistance, in sarcopenia remains unclear. Peroxisome proliferator-activated receptor γ (PPAR-γ) plays a critical role in regulating muscle atrophy. The role of deubiquitinase USP2 in mitigating muscle atrophy was investigated. Our findings revealed reduced USP2 expression in skeletal muscles of patients with type 2 diabetes. In mouse models of diabetes- and dexamethasone (DEX)-induced muscle atrophy, USP2 expression was downregulated in skeletal muscles. Usp2 knockout exacerbated muscle loss and functional impairment induced by diabetes or DEX. Moreover, skeletal muscle-specific Usp2 knockout further aggravated muscle loss and functional impairment induced by diabetes. Local injection of adeno-associated virus-Usp2 into the gastrocnemius muscles of diabetic mice increased muscle mass and improved skeletal muscle performance and endurance. It enhanced insulin sensitivity in diabetic mice, shown by lower fasting serum glucose and insulin levels and better glucose tolerance. Mechanistic analysis showed USP2 directly interacted with PPAR-γ by deubiquitinating it, stabilizing its protein levels, enhancing insulin signaling and sensitivity, and maintaining muscle mass. Loss of PPAR-γ abolishes the regulatory effects of USP2 on insulin sensitivity and muscle atrophy. MYOD1 activates USP2 transcription by binding to its promoter region. This study demonstrates the protective role of USP2 in mitigating muscle atrophy by stabilizing PPAR-γ through deubiquitination, particularly in models of diabetic and DEX-induced muscle atrophy. Targeting the USP2-PPAR-γ axis may offer promising therapeutic strategies for metabolic disorders and sarcopenia. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Shu Yang
- Department of Geriatrics, The First Affiliated Hospital (Shenzhen People’s Hospital), Southern University of Science and Technology, Shenzhen, Guangdong, China
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Lijiao Xiong
- Department of Geriatrics, The First Affiliated Hospital (Shenzhen People’s Hospital), Southern University of Science and Technology, Shenzhen, Guangdong, China
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Tingfeng Liao
- Department of Geriatrics, The First Affiliated Hospital (Shenzhen People’s Hospital), Southern University of Science and Technology, Shenzhen, Guangdong, China
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Lixing Li
- Department of Geriatrics, The First Affiliated Hospital (Shenzhen People’s Hospital), Southern University of Science and Technology, Shenzhen, Guangdong, China
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Yanchun Li
- Department of Geriatrics, The First Affiliated Hospital (Shenzhen People’s Hospital), Southern University of Science and Technology, Shenzhen, Guangdong, China
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Lin Kang
- Department of Geriatrics, The First Affiliated Hospital (Shenzhen People’s Hospital), Southern University of Science and Technology, Shenzhen, Guangdong, China
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
- The Biobank of National Innovation Center for Advanced Medical Devices, Shenzhen People’s Hospital, Shenzhen, Guangdong, China
| | - Guangyan Yang
- Department of Geriatrics, The First Affiliated Hospital (Shenzhen People’s Hospital), Southern University of Science and Technology, Shenzhen, Guangdong, China
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
- Shantou University Medical College, Shantou, Guangdong, China
| | - Zhen Liang
- Department of Geriatrics, Peking University Shenzhen Hospital, Shenzhen, China
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| |
Collapse
|
3
|
Fu Q, Qiu R, Yao T, Liu L, Li Y, Li X, Qi W, Chen Y, Cheng Y. Music therapy as a preventive intervention for postpartum depression: modulation of synaptic plasticity, oxidative stress, and inflammation in a mouse model. Transl Psychiatry 2025; 15:143. [PMID: 40216751 PMCID: PMC11992210 DOI: 10.1038/s41398-025-03370-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 03/17/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
Postpartum depression (PPD) significantly impacts women's mental health and social functioning, yet effective therapies remain limited. This study investigates the preventive effects of music therapy on PPD-like behaviors and the underlying neurobiological mechanisms in a mouse model subjected to ovarian hormone withdrawal (HW). Mice exposed to daily music sessions exhibited markedly reduced depression- and anxiety-like behaviors, as evidenced by enhanced performance in behavioral tests such as the open field test (OFT), forced swim test (FST), elevated plus maze test (EPM), sucrose preference test (SPT), novelty-suppressed feeding (NSF) test, and tail suspension test (TST). Furthermore, music therapy normalized oxidative stress indicators (NO, MDA, SOD, CAT, GSH-Px, T-AOC, ATP, and glutamate) in the serum, hippocampus, and prefrontal cortex. Additionally, music exposure reduced levels of proinflammatory factors (IL-6, IL-1β, iNOS, TNF-α, and TGF-β) and the activation of microglia and astrocytes in these brain regions. Notably, music therapy preserved neuronal integrity, promoted neurogenesis, and maintained synaptic plasticity, evidenced by the restoration of dendritic spines. Transcriptome sequencing identified differential gene expression in pathways related to synaptic plasticity, inflammation, and oxidative stress. These findings suggest that music therapy prevents PPD by modulating oxidative stress, inflammation, and synaptic integrity, providing robust preclinical evidence for its potential as a natural preventive intervention for PPD. This study underscores the need for further clinical research to validate the therapeutic efficacy of music in preventing PPD in humans, highlighting its promise as a non-invasive and accessible treatment modality.
Collapse
Affiliation(s)
- Qiang Fu
- Institute of National Security, Center on Translational Neuroscience, Minzu University of China, Beijing, China
- School of Ethnology and Sociology, Minzu University of China, Beijing, China
| | - Rui Qiu
- Institute of National Security, Center on Translational Neuroscience, Minzu University of China, Beijing, China
- School of Ethnology and Sociology, Minzu University of China, Beijing, China
| | - Tongtong Yao
- Institute of National Security, Center on Translational Neuroscience, Minzu University of China, Beijing, China
- School of Ethnology and Sociology, Minzu University of China, Beijing, China
| | - Liming Liu
- Institute of National Security, Center on Translational Neuroscience, Minzu University of China, Beijing, China
- School of Ethnology and Sociology, Minzu University of China, Beijing, China
| | - Yaobo Li
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Xiaodong Li
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Wen Qi
- College of Dance, Minzu University of China, Beijing, China.
| | - Yuewen Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, Guangdong, China.
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, Guangdong, China.
- SIAT-HKUST Joint Laboratory for Brain Science, Chinese Academy of Sciences, Shenzhen, Guangdong, China.
| | - Yong Cheng
- Institute of National Security, Center on Translational Neuroscience, Minzu University of China, Beijing, China.
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China.
| |
Collapse
|
4
|
Liu X, Zhang G, Liu L, Xiong G, Liu J, Wei W. USP2 Promotes the Proliferation and Inflammation of Fibroblast-Like Synovial Cells in Rheumatoid Arthritis Through Deubiquitination of TRAF2. Biochem Genet 2025; 63:592-605. [PMID: 38480669 DOI: 10.1007/s10528-024-10737-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 02/09/2024] [Indexed: 02/19/2025]
Abstract
Rheumatoid arthritis (RA) is a prevalent inflammatory disorder affecting about 1% of the global population. The ubiquitin-specific protease 2 (USP2) is known to have a substantial influence on the regulation of several cellular processes. Both in vivo (using rats with collagen-induced arthritis, CIA) and in vitro (using human fibroblast-like synoviocytes, HFLS-RA) models of RA were used to examine the role of USP2 in RA. The proliferation of HFLS-RA cells was assessed using the cell counting kit 8 test and EdU staining. The technique used for the assessment of gene expression was quantitative real-time PCR. Protein expression was quantified using Western blot (WB) analysis, while the quantities of inflammatory factors and matrix metalloproteinases were assessed using an ELISA test. The co-immunoprecipitation and ubiquitination tests investigated the relationships between proteins and the underlying molecular pathways. The results of this study demonstrate an upregulation of USP2 expression in both vivo and vitro models of RA. In addition, our findings indicate that the overexpression of USP2 notably exacerbates both proliferation and inflammation. The consistent downregulation of USP2 resulted in a reduction in the secretion of inflammatory cytokines and a suppression of cellular proliferation. Furthermore, it was shown that USP2 interacts with tumor necrosis factor receptor-associated factor 2 (TRAF2) and facilitates the removal of ubiquitination chains from TRAF2, enhancing its stability. Our findings propose that USP2 functions as a favorable modulator of proliferation and inflammatory reactions in HFLS-RA, thereby indicating its potential as a therapeutic target for the treatment of RA.
Collapse
Affiliation(s)
- Xiuchan Liu
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China
- Department of Infectious Diseases, Tianjin University Tianjin Hospital, Tianjin, China
| | - Geng Zhang
- Department of Infectious Diseases, Tianjin Medical University Baodi Clinical College, Tianjin, China
| | - Lei Liu
- Department of Infectious Diseases, Tianjin Medical University Baodi Clinical College, Tianjin, China
| | - Guangyi Xiong
- Department of Pathology, Tianjin University Tianjin Hospital, Tianjin, China
| | - Jun Liu
- Department of Joint Surgery, Tianjin University Tianjin Hospital, Tianjin, China.
| | - Wei Wei
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
5
|
Fu Q, Qiu R, Liang J, Wu S, Huang D, Qin Y, Li Q, Shi X, Xiong X, Jiang Z, Chen Y, Cheng Y. Sugemule-7 alleviates oxidative stress, neuroinflammation, and cell death, promoting synaptic plasticity recovery in mice with postpartum depression. Sci Rep 2025; 15:1426. [PMID: 39789071 PMCID: PMC11718020 DOI: 10.1038/s41598-025-85276-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/01/2025] [Indexed: 01/12/2025] Open
Abstract
Postpartum depression (PPD) profoundly impacts the mental and physical health of women globally and is an incurable psychological disorder. Traditional pharmacological treatments often have strong side effects and may adversely affect infant health through breastfeeding, underscoring the critical need for natural and gentle treatment strategies. Sugemule-7, a traditional Chinese medicine comprising multiple natural plant ingredients, represents a potentially safer and more effective alternative. To investigate its preventive effects on PPD, we established an animal model and administered the drug Sugemule-7. Our study demonstrated that varying doses of Sugemule-7 effectively alleviated depressive and anxiety-like behaviors in PPD mice, as assessed through a battery of tests, including the open field test, tail suspension test, sucrose preference test, forced swim test, novelty-suppressed feeding test, and elevated plus maze test. Furthermore, Sugemule-7 significantly improved oxidative stress levels in the serum, prefrontal cortex, and hippocampus of PPD-induced mice while also suppressing inflammatory responses and abnormal neuronal death in these brain regions. Transcriptomic sequencing of hippocampal and prefrontal cortex tissues supported our findings, revealing that differential gene expression is primarily involved in regulating synaptic plasticity. Overall, our study confirms the efficacy of Sugemule-7 in treating PPD at different concentrations, potentially alleviating depressive behaviors by enhancing synaptic plasticity, mitigating oxidative stress, reducing inflammation, and protecting neurons.
Collapse
Affiliation(s)
- Qiang Fu
- Center on Translational Neuroscience, Institute of National Security, Minzu University of China, Beijing, China
- School of Ethnology and Sociology, Minzu University of China, Beijing, China
| | - Rui Qiu
- Center on Translational Neuroscience, Institute of National Security, Minzu University of China, Beijing, China
- School of Ethnology and Sociology, Minzu University of China, Beijing, China
| | - Jiaquan Liang
- Center on Translational Neuroscience, Institute of National Security, Minzu University of China, Beijing, China
| | - Shuai Wu
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Dezhi Huang
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Yuxiang Qin
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Qiaosheng Li
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Xiaojie Shi
- School of Pharmacy, Minzu University of China, Beijing, China
| | - Xiyue Xiong
- NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China
| | - Zhongyong Jiang
- Department of Medical Laboratory, Affiliated Cancer Hospital of Chengdu Medical College, Chengdu Seventh People's Hospital, Chengdu, Sichuan, China
| | - Yuewen Chen
- Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Chinese Academy of Sciences, Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, Guangdong, China.
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, 518057, Guangdong, China.
- Xili Shenzhen University Town, No.1068 Xueyuan Avenue, Nanshan District, Shenzhen, 518055, China.
| | - Yong Cheng
- Center on Translational Neuroscience, Institute of National Security, Minzu University of China, Beijing, China.
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China.
- , 27th South Zhongguancun Avenue, Beijing, 100081, China.
| |
Collapse
|
6
|
Wang YH, Yang X, Liu CC, Wang X, Yu KD. Unraveling the peripheral nervous System's role in tumor: A Double-edged Sword. Cancer Lett 2025; 611:217451. [PMID: 39793755 DOI: 10.1016/j.canlet.2025.217451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 01/01/2025] [Accepted: 01/07/2025] [Indexed: 01/13/2025]
Abstract
The peripheral nervous system (PNS) includes all nerves outside the brain and spinal cord, comprising various cells like neurons and glial cells, such as schwann and satellite cells. The PNS is increasingly recognized for its bidirectional interactions with tumors, exhibiting both pro- and anti-tumor effects. Our review delves into the complex mechanisms underlying these interactions, highlighting recent findings that challenge the conventional understanding of PNS's role in tumorigenesis. We emphasize the contradictory results in the literature and propose novel perspectives on how these discrepancies can be resolved. By focusing on the PNS's influence on tumor initiation, progression, and microenvironment remodeling, we provide a comprehensive analysis that goes beyond the structural description of the PNS. Our review suggests that a deeper comprehension of the PNS-tumor crosstalk is pivotal for developing targeted anticancer strategies. We conclude by emphasizing the need for future research to unravel the intricate dynamics of the PNS in cancer, which may lead to innovative diagnostic tools and therapeutic approaches.
Collapse
Affiliation(s)
- Yan-Hao Wang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center and Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China; Key Laboratory of Breast Cancer in Shanghai, Shanghai, 200032, PR China
| | - Xuan Yang
- Department of General Surgery, Shanxi Provincial People's Hospital, Taiyuan, 030000, PR China
| | - Cui-Cui Liu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center and Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China; Key Laboratory of Breast Cancer in Shanghai, Shanghai, 200032, PR China
| | - Xin Wang
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China
| | - Ke-Da Yu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center and Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China; Key Laboratory of Breast Cancer in Shanghai, Shanghai, 200032, PR China.
| |
Collapse
|
7
|
Kitamura H, Fujimoto M, Hashimoto M, Yasui H, Inanami O. USP2 Mitigates Reactive Oxygen Species-Induced Mitochondrial Damage via UCP2 Expression in Myoblasts. Int J Mol Sci 2024; 25:11936. [PMID: 39596006 PMCID: PMC11593688 DOI: 10.3390/ijms252211936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/22/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Ubiquitin-specific protease 2 (USP2) maintains mitochondrial integrity in culture myoblasts. In this study, we investigated the molecular mechanisms underlying the protective role of USP2 in mitochondria. The knockout (KO) of the Usp2 gene or the chemical inhibition of USP2 induced a robust accumulation of mitochondrial reactive oxygen species (ROS), accompanied by defects in mitochondrial membrane potential, in C2C12 myoblasts. ROS removal by N-acetyl-L-cysteine restored the mitochondrial dysfunction induced by USP2 deficiency. Comprehensive RT-qPCR screening and following protein analysis indicated that both the genetic and chemical inhibition of USP2 elicited a decrease in uncoupling protein 2 (UCP2) at mRNA and protein levels. Accordingly, the introduction of a Ucp2-expressing construct effectively recovered the mitochondrial membrane potential, entailing an increment in the intracellular ATP level in Usp2KO C2C12 cells. In contrast, USP2 deficiency also decreased peroxisome proliferator-activated receptor γ coactivator 1α (PGC1α) protein in C2C12 cells, while it upregulated Ppargc1a mRNA. Overexpression studies indicated that USP2 potentially stabilizes PGC1α in an isopeptidase-dependent manner. Given that PGC1α is an inducer of UCP2 in C2C12 cells, USP2 might ameliorate mitochondrial ROS by maintaining the PGC1α-UCP2 axis in myoblasts.
Collapse
Affiliation(s)
- Hiroshi Kitamura
- Laboratory of Disease Models, School of Veterinary Medicine, Rakuno Gakuen University, Ebestsu 069-8501, Japan;
| | - Masaki Fujimoto
- Laboratory of Disease Models, School of Veterinary Medicine, Rakuno Gakuen University, Ebestsu 069-8501, Japan;
| | - Mayuko Hashimoto
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Osaka 584-8540, Japan;
| | - Hironobu Yasui
- Laboratory of Radiation Biology, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan; (H.Y.); (O.I.)
| | - Osamu Inanami
- Laboratory of Radiation Biology, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan; (H.Y.); (O.I.)
| |
Collapse
|
8
|
Liao Y, Zhang W, Liu Y, Zhu C, Zou Z. The role of ubiquitination in health and disease. MedComm (Beijing) 2024; 5:e736. [PMID: 39329019 PMCID: PMC11424685 DOI: 10.1002/mco2.736] [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: 02/11/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/28/2024] Open
Abstract
Ubiquitination is an enzymatic process characterized by the covalent attachment of ubiquitin to target proteins, thereby modulating their degradation, transportation, and signal transduction. By precisely regulating protein quality and quantity, ubiquitination is essential for maintaining protein homeostasis, DNA repair, cell cycle regulation, and immune responses. Nevertheless, the diversity of ubiquitin enzymes and their extensive involvement in numerous biological processes contribute to the complexity and variety of diseases resulting from their dysregulation. The ubiquitination process relies on a sophisticated enzymatic system, ubiquitin domains, and ubiquitin receptors, which collectively impart versatility to the ubiquitination pathway. The widespread presence of ubiquitin highlights its potential to induce pathological conditions. Ubiquitinated proteins are predominantly degraded through the proteasomal system, which also plays a key role in regulating protein localization and transport, as well as involvement in inflammatory pathways. This review systematically delineates the roles of ubiquitination in maintaining protein homeostasis, DNA repair, genomic stability, cell cycle regulation, cellular proliferation, and immune and inflammatory responses. Furthermore, the mechanisms by which ubiquitination is implicated in various pathologies, alongside current modulators of ubiquitination are discussed. Enhancing our comprehension of ubiquitination aims to provide novel insights into diseases involving ubiquitination and to propose innovative therapeutic strategies for clinical conditions.
Collapse
Affiliation(s)
- Yan Liao
- Faculty of Anesthesiology Changhai Hospital Naval Medical University Shanghai China
- School of Anesthesiology Naval Medical University Shanghai China
| | - Wangzheqi Zhang
- Faculty of Anesthesiology Changhai Hospital Naval Medical University Shanghai China
- School of Anesthesiology Naval Medical University Shanghai China
| | - Yang Liu
- Faculty of Anesthesiology Changhai Hospital Naval Medical University Shanghai China
- School of Anesthesiology Naval Medical University Shanghai China
| | - Chenglong Zhu
- Faculty of Anesthesiology Changhai Hospital Naval Medical University Shanghai China
- School of Anesthesiology Naval Medical University Shanghai China
| | - Zui Zou
- Faculty of Anesthesiology Changhai Hospital Naval Medical University Shanghai China
- School of Anesthesiology Naval Medical University Shanghai China
| |
Collapse
|
9
|
Son Y, Su Yang J, Chul Shin S, Kyoung Park S, Kim Y, Park J, Yu J. Structural optimization and biological evaluation of ML364 based derivatives as USP2a inhibitors. Bioorg Chem 2024; 145:107222. [PMID: 38401359 DOI: 10.1016/j.bioorg.2024.107222] [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: 11/30/2023] [Revised: 01/29/2024] [Accepted: 02/16/2024] [Indexed: 02/26/2024]
Abstract
Ubiquitination is a representative post-translational modification that tags target proteins with ubiquitin to induce protein degradation or modify their functions. Deubiquitinating enzymes (DUBs) play a crucial role in reversing this process by removing ubiquitin from target proteins. Among them, USP2a has emerged as a promising target for cancer therapy due to its oncogenic properties in various cancer types, but its inhibitors have been limited. In this study, our aim was to optimize the structure of ML364, a USP2a inhibitor, and synthesize a series of its derivatives to develop potent USP2a inhibitors. Compound 8v emerged as a potential USP2a inhibitor with lower cytotoxicity compared to ML364. Cellular assays demonstrated that compound 8v effectively reduced the levels of USP2a substrates and attenuated cancer cell growth. We confirmed its direct interaction with the catalytic domain of USP2a and its selective inhibitory activity against USP2a over other USP subfamily proteins (USP7, 8, or 15). In conclusion, compound 8v has been identified as a potent USP2a inhibitor with substantial potential for cancer therapy.
Collapse
Affiliation(s)
- Youngchai Son
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Ji Su Yang
- Biomedical Research Division, Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Sang Chul Shin
- Research Resources Division, Technological Convergence Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Seo Kyoung Park
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Yeojin Kim
- Biomedical Research Division, Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Department of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Jinyoung Park
- Biomedical Research Division, Center for Advanced Biomolecular Recognition, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio‑Medical Science and Technology, KIST‑School, University of Science and Technology (UST), Seoul 02792, Republic of Korea.
| | - Jinha Yu
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea.
| |
Collapse
|
10
|
Fu Q, Qiu R, Chen L, Chen Y, Qi W, Cheng Y. Music prevents stress-induced depression and anxiety-like behavior in mice. Transl Psychiatry 2023; 13:317. [PMID: 37828015 PMCID: PMC10570293 DOI: 10.1038/s41398-023-02606-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 09/14/2023] [Accepted: 09/20/2023] [Indexed: 10/14/2023] Open
Abstract
Depression is the most prevalent psychiatric disorder worldwide and remains incurable; however, there is little research on its prevention. The leading cause of depression is stress, and music has been hypothesized to alleviate stress. To examine the potential beneficial effects of music on stress and depression, we subjected mice to chronic unpredictable mild stress (CUMS) during the day and music at night. Strikingly, our results indicated that music completely prevented CUMS-induced depression and anxiety-like behaviors in mice, as assessed by the open field, tail suspension, sucrose preference, novelty suppressed feeding, and elevated plus maze tests. We found that listening to music restored serum corticosterone levels in CUMS mice, which may contribute to the beneficial effects of music on the mouse brain, including the restoration of BDNF and Bcl-2 levels. Furthermore, listening to music prevented CUMS-induced oxidative stress in the serum, prefrontal cortex, and hippocampus of mice. Moreover, the CUMS-induced inflammatory responses in the prefrontal cortex and hippocampus of mice were prevented by listening to music. Taken together, we have demonstrated for the first time in mice experiments that listening to music prevents stress-induced depression and anxiety-like behaviors in mice. Music may restore hypothalamus-pituitary-adrenal axis homeostasis, preventing oxidative stress, inflammation, and neurotrophic factor deficits, which had led to the observed phenotypes in CUMS mice.
Collapse
Affiliation(s)
- Qiang Fu
- Institute of National Security, Center on Translational Neuroscience, Minzu University of China, Beijing, China
- School of Ethnology and Sociology, Minzu University of China, Beijing, China
| | - Rui Qiu
- Institute of National Security, Center on Translational Neuroscience, Minzu University of China, Beijing, China
- School of Ethnology and Sociology, Minzu University of China, Beijing, China
| | - Lei Chen
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| | - Yuewen Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, Guangdong, 518055, China.
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, Guangdong, 518057, China.
| | - Wen Qi
- College of Dance, Minzu University of China, Beijing, China.
| | - Yong Cheng
- Institute of National Security, Center on Translational Neuroscience, Minzu University of China, Beijing, China
- College of Life and Environmental Sciences, Minzu University of China, Beijing, China
| |
Collapse
|
11
|
Zhang S, Guo Y, Zhang S, Wang Z, Zhang Y, Zuo S. Targeting the deubiquitinase USP2 for malignant tumor therapy (Review). Oncol Rep 2023; 50:176. [PMID: 37594087 PMCID: PMC10463009 DOI: 10.3892/or.2023.8613] [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: 03/14/2023] [Accepted: 07/24/2023] [Indexed: 08/19/2023] Open
Abstract
The ubiquitin‑proteasome system is a major degradation pathway for >80% of proteins in vivo. Deubiquitylases, which remove ubiquitinated tags to stabilize substrate proteins, are important components involved in regulating the degradation of ubiquitinated proteins. In addition, they serve multiple roles in tumor development by participating in physiological processes such as protein metabolism, cell cycle regulation, DNA damage repair and gene transcription. The present review systematically summarized the role of ubiquitin‑specific protease 2 (USP2) in malignant tumors and the specific molecular mechanisms underlying the involvement of USP2 in tumor‑associated pathways. USP2 reverses ubiquitin‑mediated degradation of proteins and is involved in aberrant proliferation, migration, invasion, apoptosis and drug resistance of tumors. Additionally, the present review summarized studies reporting on the use of USP2 as a therapeutic target for malignancies such as breast, liver, ovarian, colorectal, bladder and prostate cancers and glioblastoma and highlights the current status of pharmacological research on USP2. The clinical significance of USP2 as a therapeutic target for malignant tumors warrants further investigation.
Collapse
Affiliation(s)
- Shilong Zhang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Yi Guo
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Shenjie Zhang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Zhi Wang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Yewei Zhang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Shi Zuo
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
- Precision Medicine Research Institute of Guizhou, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| |
Collapse
|
12
|
Chen Z, Liu XA, Kenny PJ. Central and peripheral actions of nicotine that influence blood glucose homeostasis and the development of diabetes. Pharmacol Res 2023; 194:106860. [PMID: 37482325 DOI: 10.1016/j.phrs.2023.106860] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/06/2023] [Accepted: 07/18/2023] [Indexed: 07/25/2023]
Abstract
Cigarette smoking has long been recognized as a risk factor for type 2 diabetes (T2D), although the precise causal mechanisms underlying this relationship remain poorly understood. Recent evidence suggests that nicotine, the primary reinforcing component in tobacco, may play a pivotal role in connecting cigarette smoking and T2D. Extensive research conducted in both humans and animals has demonstrated that nicotine can elevate blood glucose levels, disrupt glucose homeostasis, and induce insulin resistance. The review aims to elucidate the genetic variants of nicotinic acetylcholine receptors associated with diabetes risk and provide a comprehensive overview of the available data on the mechanisms through which nicotine influences blood glucose homeostasis and the development of diabetes. Here we emphasize the central and peripheral actions of nicotine on the release of glucoregulatory hormones, as well as its effects on glucose tolerance and insulin sensitivity. Notably, the central actions of nicotine within the brain, which encompass both insulin-dependent and independent mechanisms, are highlighted as potential targets for intervention strategies in diabetes management.
Collapse
Affiliation(s)
- Zuxin Chen
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; University of Chinese Academy of Sciences, Beijing, China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Xin-An Liu
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; University of Chinese Academy of Sciences, Beijing, China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.
| | - Paul J Kenny
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, USA.
| |
Collapse
|
13
|
Zhu H, Zhang H, Guo J, Zhang C, Zhang Q, Gao F. Up-regulated oxidized USP2a can increase Mdm2-p60-p53 to promote cell apoptosis. Exp Cell Res 2023; 427:113597. [PMID: 37044314 DOI: 10.1016/j.yexcr.2023.113597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 03/17/2023] [Accepted: 04/08/2023] [Indexed: 04/14/2023]
Abstract
Mdm2 promotes the ubiquitination and degradation of p53, while Mdm2-p60 can bind to p53 and reduce the Mdm2-induced p53 ubiquitination to improve its stability. USP2a can deubiquitinate and stabilize Mdm2, whether USP2a can regulate Mdm2-p60 needs to be further confirmed and elucidated. We found that oxidative stress can up-regulate USP2a at the post-transcriptional level and induce USP2a to be oxidized by forming inter-subunit disulfide bonds. The oxidized USP2a is closely related with cell apoptosis. In apoptotic cells, oxidized USP2a has enhanced protein stability and further stabilizes Mdm2-p60 through deubiquitination, and the USP2a-Mdm2-p60-p53 axis plays a role in cell apoptosis. Altogether USP2a is oxygen sensitive, oxidized USP2a exerts apoptotic effects through the Mdm2-p60-p53 axis, which provides an experimental basis for regulating p53 apoptotic signaling by targeting USP2a.
Collapse
Affiliation(s)
- Hanqing Zhu
- Department of Pulmonary Function Test, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| | - Hongliang Zhang
- Department of Blood Transfusion, Henan Provincial People's Hospital, Department of Blood Transfusion of Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 450053, China.
| | - Jiahui Guo
- Department of Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201999, China.
| | - Chao Zhang
- Department of Geriatrics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201999, China.
| | - Quanwu Zhang
- Department of Pathology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, 450007, China.
| | - Fenghou Gao
- Department of Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201999, China; Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, China.
| |
Collapse
|
14
|
Kamoshita K, Ishii KA, Tahira Y, Kikuchi A, Abuduwaili H, Tajima-Shirasaki N, Li Q, Takayama H, Matsumoto K, Takamura T. Insulin Suppresses Ubiquitination via the Deubiquitinating Enzyme Ubiquitin-Specific Protease 14, Independent of Proteasome Activity in H4IIEC3 Hepatocytes. J Pharmacol Exp Ther 2023; 385:5-16. [PMID: 36328485 DOI: 10.1124/jpet.122.001088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 08/16/2022] [Accepted: 09/09/2022] [Indexed: 11/06/2022] Open
Abstract
Ubiquitin-proteasome dysfunction contributes to obesity-related metabolic disorders, such as diabetes and fatty liver disease. However, the regulation of ubiquitin-proteasome activity by insulin remains to be elucidated. Here, we show that prolonged insulin stimulation activates proteasome function even though it reduces the ubiquitinated proteins in H4IIEC3 hepatocytes. Looking for a pathway by which insulin inhibits ubiquitination, we found that hepatic expression of ubiquitin-specific protease 14 (USP14) was upregulated in the liver of patients with insulin resistance. Indeed, the USP14-specific inhibitor IU1 canceled the insulin-mediated reduction of ubiquitinated proteins. Furthermore, insulin-induced endoplasmic reticulum (ER) stress, which was canceled by IU1, suggesting that USP14 activity is involved in insulin-induced ER stress. Co-stimulation with insulin and IU1 for 2 hours upregulated the nuclear translocation of the lipogenic transcription factor, sterol regulatory element binding protein-1c (SREBP-1c), upregulated the expression of the lipogenic gene, fatty acid synthase (Fasn), and repressed the gluconeogenic genes. In conclusion, insulin activates proteasome function even though it inhibits protein ubiquitination by activating USP14 in hepatocytes. USP14 activation by insulin inhibits mature SREBP-1c while upregulating ER stress and the expression of genes involved in gluconeogenesis. Further understanding mechanisms underlying the USP14 activation and its pleiotropic effects may lead to therapeutic development for obesity-associated metabolic disorders, such as diabetes and fatty liver disease. SIGNIFICANCE STATEMENT: This study shows that insulin stimulation inhibits ubiquitination by activating USP14, independent of its effect on proteasome activity in hepatocytes. USP14 also downregulates the nuclear translocation of the lipogenic transcription factor SREBP-1c and upregulates the expression of genes involved in gluconeogenesis. Since USP14 is upregulated in the liver of insulin-resistant patients, understanding mechanisms underlying the USP14 activation and its pleiotropic effects will help develop treatments for metabolic disorders such as diabetes and fatty liver.
Collapse
Affiliation(s)
- Kyoko Kamoshita
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences (K.K., K.A.I., Y.T., A.K., H.A., N.T.S., Q.L., H.T., T.T.); Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences (K.A.I.); Life Sciences Division, Engineering and Technology Department (H.T.); and Division of Tumor Dynamics and Regulation, Cancer Research Institute (K.M.), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Kiyo-Aki Ishii
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences (K.K., K.A.I., Y.T., A.K., H.A., N.T.S., Q.L., H.T., T.T.); Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences (K.A.I.); Life Sciences Division, Engineering and Technology Department (H.T.); and Division of Tumor Dynamics and Regulation, Cancer Research Institute (K.M.), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Yumiko Tahira
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences (K.K., K.A.I., Y.T., A.K., H.A., N.T.S., Q.L., H.T., T.T.); Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences (K.A.I.); Life Sciences Division, Engineering and Technology Department (H.T.); and Division of Tumor Dynamics and Regulation, Cancer Research Institute (K.M.), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Akihiro Kikuchi
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences (K.K., K.A.I., Y.T., A.K., H.A., N.T.S., Q.L., H.T., T.T.); Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences (K.A.I.); Life Sciences Division, Engineering and Technology Department (H.T.); and Division of Tumor Dynamics and Regulation, Cancer Research Institute (K.M.), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Halimulati Abuduwaili
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences (K.K., K.A.I., Y.T., A.K., H.A., N.T.S., Q.L., H.T., T.T.); Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences (K.A.I.); Life Sciences Division, Engineering and Technology Department (H.T.); and Division of Tumor Dynamics and Regulation, Cancer Research Institute (K.M.), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Natsumi Tajima-Shirasaki
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences (K.K., K.A.I., Y.T., A.K., H.A., N.T.S., Q.L., H.T., T.T.); Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences (K.A.I.); Life Sciences Division, Engineering and Technology Department (H.T.); and Division of Tumor Dynamics and Regulation, Cancer Research Institute (K.M.), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Qifang Li
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences (K.K., K.A.I., Y.T., A.K., H.A., N.T.S., Q.L., H.T., T.T.); Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences (K.A.I.); Life Sciences Division, Engineering and Technology Department (H.T.); and Division of Tumor Dynamics and Regulation, Cancer Research Institute (K.M.), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Hiroaki Takayama
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences (K.K., K.A.I., Y.T., A.K., H.A., N.T.S., Q.L., H.T., T.T.); Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences (K.A.I.); Life Sciences Division, Engineering and Technology Department (H.T.); and Division of Tumor Dynamics and Regulation, Cancer Research Institute (K.M.), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Kunio Matsumoto
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences (K.K., K.A.I., Y.T., A.K., H.A., N.T.S., Q.L., H.T., T.T.); Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences (K.A.I.); Life Sciences Division, Engineering and Technology Department (H.T.); and Division of Tumor Dynamics and Regulation, Cancer Research Institute (K.M.), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Toshinari Takamura
- Department of Endocrinology and Metabolism, Graduate School of Medical Sciences (K.K., K.A.I., Y.T., A.K., H.A., N.T.S., Q.L., H.T., T.T.); Department of Integrative Medicine for Longevity, Graduate School of Medical Sciences (K.A.I.); Life Sciences Division, Engineering and Technology Department (H.T.); and Division of Tumor Dynamics and Regulation, Cancer Research Institute (K.M.), Kanazawa University, Kanazawa, Ishikawa, Japan
| |
Collapse
|
15
|
Kitamura H. Ubiquitin-Specific Proteases (USPs) and Metabolic Disorders. Int J Mol Sci 2023; 24:3219. [PMID: 36834633 PMCID: PMC9966627 DOI: 10.3390/ijms24043219] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
Ubiquitination and deubiquitination are reversible processes that modify the characteristics of target proteins, including stability, intracellular localization, and enzymatic activity. Ubiquitin-specific proteases (USPs) constitute the largest deubiquitinating enzyme family. To date, accumulating evidence indicates that several USPs positively and negatively affect metabolic diseases. USP22 in pancreatic β-cells, USP2 in adipose tissue macrophages, USP9X, 20, and 33 in myocytes, USP4, 7, 10, and 18 in hepatocytes, and USP2 in hypothalamus improve hyperglycemia, whereas USP19 in adipocytes, USP21 in myocytes, and USP2, 14, and 20 in hepatocytes promote hyperglycemia. In contrast, USP1, 5, 9X, 14, 15, 22, 36, and 48 modulate the progression of diabetic nephropathy, neuropathy, and/or retinopathy. USP4, 10, and 18 in hepatocytes ameliorates non-alcoholic fatty liver disease (NAFLD), while hepatic USP2, 11, 14, 19, and 20 exacerbate it. The roles of USP7 and 22 in hepatic disorders are controversial. USP9X, 14, 17, and 20 in vascular cells are postulated to be determinants of atherosclerosis. Moreover, mutations in the Usp8 and Usp48 loci in pituitary tumors cause Cushing syndrome. This review summarizes the current knowledge about the modulatory roles of USPs in energy metabolic disorders.
Collapse
Affiliation(s)
- Hiroshi Kitamura
- Laboratory of Comparative Medicine, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu 069-8501, Japan
| |
Collapse
|