1
|
Li F, Zhou Y, Lin X, Zhang Y, Hu Q, Zhao E, Li H, Pan X, Shu F, Zhang K, Huang C, Tang N, Liao W. A novel USP4 inhibitor that suppresses colorectal cancer stemness by promoting β-catenin and Twist1 degradation. J Transl Med 2025; 23:114. [PMID: 39856683 PMCID: PMC11762077 DOI: 10.1186/s12967-024-06001-0] [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: 09/17/2024] [Accepted: 12/13/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND The high mortality rate of metastatic colorectal cancer (CRC) is primarily attributed to resistance to chemotherapy, where cancer stem cells (CSCs) play a crucial role. Deubiquitinating enzymes are essential regulators of CSC maintenance, making them potential targets for eliminating CSCs and overcoming chemotherapy resistance. This study aims to identify key deubiquitinating enzymes regulating CSCs and drug resistance of CRC. METHODS RNA sequencing was performed to examine the mRNA expression of known deubiquitinating enzymes in CRC tissues from patients with alternate response to chemotherapy. Gain- and loss-of-function experiments were performed to evaluate the function of USP4 in regulation of stemness and drug sensitivity in CRC. High-throughput virtual screening and target management assays were conducted to identify small molecule inhibitor targeting USP4. Cell lines, organoids and animal models were used to evaluate the function of USP4 and its small molecule inhibitor in stemness and chemotherapy response. RESULTS The expression of USP4 was significantly elevated in CRC samples from progressive disease (PD) or stable disease (SD) patients compared to partial response (PR) specimen. USP4 promoted stemness by stabilizing the β-catenin and Twist1 proteins in CRC cells. A natural small molecule product U4-I05 diminished the stem-like features of CSCs and enhanced their sensitivity to oxaliplatin and 5-fluorouracil by targeting inhibition of its deubiquitinating enzyme activity through binding the catalytic domain of USP4 (311 cysteine site) at nanomolar concentrations, triggering proteasome-mediated degradation of β-catenin and Twist1. Treatment with U4-I05 also inhibited tumor metastasis and extended survival in a genetically engineered CRC mouse model. CONCLUSIONS This study identifies U4-I05 as a USP4 inhibitor with significant therapeutic efficacy against CRC, offering a promising avenue for the development of new treatments targeting cancer stemness and chemotherapy resistance.
Collapse
Affiliation(s)
- Fengtian Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- School of Biosciences and Technology, Chengdu Medical College, Chengdu, Sichuan, China
- The Second Affiliated Hospital of Chengdu Medical College, National Nuclear Corporation 416 Hospital, Chengdu, 610051, China
| | - Yi Zhou
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Xinyue Lin
- School of Biosciences and Technology, Chengdu Medical College, Chengdu, Sichuan, China
- The Second Affiliated Hospital of Chengdu Medical College, National Nuclear Corporation 416 Hospital, Chengdu, 610051, China
| | - Yaxin Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Qingyong Hu
- School of Basic Medical Sciences and Forensic Medicine, North Sichuan Medical College, Nanchong, 637000, Sichuan, China
| | - Enen Zhao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Huali Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Xingyan Pan
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Feng Shu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Kun Zhang
- School of Biosciences and Technology, Chengdu Medical College, Chengdu, Sichuan, China
- The Second Affiliated Hospital of Chengdu Medical College, National Nuclear Corporation 416 Hospital, Chengdu, 610051, China
| | - Chengmei Huang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| | - Na Tang
- Department of Pathology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
| | - Wenting Liao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| |
Collapse
|
2
|
Chen ZG, Liu GQ, Liu WM, Lu PR. Inhibition of viability of human retinal microvascular endothelial cells by vialinin A under high glucose condition. Int J Ophthalmol 2024; 17:1809-1815. [PMID: 39430017 PMCID: PMC11422364 DOI: 10.18240/ijo.2024.10.06] [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: 04/10/2024] [Accepted: 06/24/2024] [Indexed: 10/22/2024] Open
Abstract
AIM To investigate the effects of vialinin A on viability of human retinal endothelial cells (HRECs) under high glucose condition and its potential mechanism. METHODS The HRECs were divided into four groups: normal glucose control group (NG, 5 mmol/L D-glucose), high glucose group (HG, 30 mmol/L D-glucose), HG+1 µmol/L vialinin A group, and HG+5 µmol/L vialinin A group. The cell viabilities were measured with cell counting kit-8 (CCK-8) assay for proliferation, with scratch assay for migration, and tube formation, for evaluation of the impact of vialinin A on cellular behaviour. Real-time PCR and Western blotting were used to determine the expression level of vascular endothelial growth factor (VEGF). RESULTS The proliferative capacity and migration of HRECs was reduced by 5 µmol/L vialinin A in high glucose environment (both P<0.05). Vialinin A also inhibited high-glucose-induced tube formation of HRECs. The expression level of VEGF and PI3K in HRECs was also significantly decreased by vialinin A (P<0.05). CONCLUSION Vialinin A inhibits the cell viability of HRECs. It may serve as a potential target for anti-angiogenic therapy.
Collapse
Affiliation(s)
- Zhi-Gang Chen
- Department of Ophthalmology, the First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Gao-Qin Liu
- Department of Ophthalmology, the First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
- Jiangsu Clinical Immunology Institute, the First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Wei-Ming Liu
- Department of Ophthalmology, the First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Pei-Rong Lu
- Department of Ophthalmology, the First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
- Jiangsu Clinical Immunology Institute, the First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| |
Collapse
|
3
|
Lin J, Lin HW, Wang YX, Fang Y, Jiang HM, Li T, Huang J, Zhang HD, Chen DZ, Chen YP. FGF4 ameliorates the liver inflammation by reducing M1 macrophage polarization in experimental autoimmune hepatitis. J Transl Med 2024; 22:717. [PMID: 39095789 PMCID: PMC11295337 DOI: 10.1186/s12967-024-05219-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 04/19/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND The global prevalence of autoimmune hepatitis (AIH) is increasing due in part to the lack of effective pharmacotherapies. Growing evidence suggests that fibroblast growth factor 4 (FGF4) is crucial for diverse aspects of liver pathophysiology. However, its role in AIH remains unknown. Therefore, we investigated whether FGF4 can regulate M1 macrophage and thereby help treat liver inflammation in AIH. METHODS We obtained transcriptome-sequencing and clinical data for patients with AIH. Mice were injected with concanavalin A to induce experimental autoimmune hepatitis (EAH). The mechanism of action of FGF4 was examined using macrophage cell lines and bone marrow-derived macrophages. RESULTS We observed higher expression of markers associated with M1 and M2 macrophages in patients with AIH than that in individuals without AIH. EAH mice showed greater M1-macrophage polarization than control mice. The expression of M1-macrophage markers correlated positively with FGF4 expression. The loss of hepatic Fgf4 aggravated hepatic inflammation by increasing the abundance of M1 macrophages. In contrast, the pharmacological administration of FGF4 mitigated hepatic inflammation by reducing M1-macrophage levels. The efficacy of FGF4 treatment was compromised following the in vivo clearance of macrophage populations. Mechanistically, FGF4 treatment activated the phosphatidylinositol 3-kinase (PI3K)-protein kinase B (AKT)-signal pathway in macrophages, which led to reduced M1 macrophages and hepatic inflammation. CONCLUSION We identified FGF4 as a novel M1/M2 macrophage-phenotype regulator that acts through the PI3K-AKT-signaling pathway, suggesting that FGF4 may represent a novel target for treating inflammation in patients with AIH.
Collapse
Affiliation(s)
- Jing Lin
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Hepatology Institute of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
- Department of Infectious Diseases, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| | - Hong-Wei Lin
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Hepatology Institute of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yu-Xing Wang
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Hepatology Institute of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yan Fang
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Hepatology Institute of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Hui-Mian Jiang
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Hepatology Institute of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Ting Li
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Hepatology Institute of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Jia Huang
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Hepatology Institute of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Hua-Dong Zhang
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Hepatology Institute of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Da-Zhi Chen
- Department of Clinical Medicine, Hangzhou Medical College, Hangzhou, 310053, China.
| | - Yong-Ping Chen
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Hepatology Institute of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
4
|
Mao M, Xia Q, Zhan G, Bing H, Zhang C, Wang J, Tian W, Lian H, Li X, Chu Q. Vialinin A alleviates oxidative stress and neuronal injuries after ischaemic stroke by accelerating Keap1 degradation through inhibiting USP4-mediated deubiquitination. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 124:155304. [PMID: 38176274 DOI: 10.1016/j.phymed.2023.155304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/02/2023] [Accepted: 12/18/2023] [Indexed: 01/06/2024]
Abstract
BACKGROUND Oxidative stress is known as a hallmark of cerebral ischaemia‒reperfusion injury and it exacerbates the pathologic progression of ischaemic brain damage. Vialinin A, derived from a Chinese edible mushroom, possesses multiple pharmacological activities in cancer, Kawasaki disease, asthma and pathological scarring. Notably, vialinin A is an inhibitor of ubiquitin-specific peptidase 4 (USP4) that shows anti-inflammatory and antioxidative properties. However, the precise effect of vialinin A in ischaemic stroke, as well as its underlying mechanisms, remains largely unexplored. PURPOSE The present research focuses on the impacts of vialinin A on oxidative stress and explores the underlying mechanisms involved while also examining its potentiality as a therapeutic candidate for ischaemic stroke. METHODS Mouse ischaemic stroke was conducted by MCAO surgery. Vialinin A was administered via lateral ventricular injection at a dose of 2 mg/kg after reperfusion. Subsequent experiments were meticulously conducted at the appropriate time points. Stroke outcomes were evaluated by TTC staining, neurological score, Nissl staining and behavioural analysis. Co-IP assays were operated to examine the protein-protein interactions. Immunoblot analysis, qRT-PCR, and luciferase reporter assays were conducted to further investigate its underlying mechanisms. RESULTS In this study, we initially showed that administration of vialinin A alleviated cerebral ischaemia‒reperfusion injury-induced neurological deficits and neuronal apoptosis. Furthermore, vialinin A, which is an antioxidant, reduced oxidative stress injury, promoted the activation of the Keap1-Nrf2-ARE signaling pathway and increased the protein degradation of Keap1. The substantial neuroprotective effects of vialinin A against ischaemic stroke were compromised by the overexpression of USP4. Mechanistically, vialinin A inhibited the deubiquitinating enzymatic activity of USP4, leading to enhanced ubiquitination of Keap1 and subsequently promoting its degradation. This cascade caused the activation of Nrf2-dependent antioxidant response, culminating in a reduction of neuronal apoptosis and the amelioration of neurological dysfunction following ischaemic stroke. CONCLUSIONS This study demonstrates that inhibition of USP4 to activate Keap1-Nrf2-ARE signaling pathway may represent a mechanism by which vialinin A conferred protection against cerebral ischaemia‒reperfusion injury and sheds light on its promising prospects as a therapeutic intervention for ischaemic stroke.
Collapse
Affiliation(s)
- Meng Mao
- Department of Anesthesiology and Perioperative Medicine, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, China; Trauma Research Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, China; Center for Advanced Medicine, College of Medicine, Zhengzhou University, Zhengzhou 450007, China
| | - Qian Xia
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Gaofeng Zhan
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hailong Bing
- Department of Anesthesiology and Perioperative Medicine, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, China
| | - Chenxi Zhang
- Department of Anesthesiology and Perioperative Medicine, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, China
| | - Jie Wang
- Department of Anesthesiology and Perioperative Medicine, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, China
| | - Wangli Tian
- Department of Anesthesiology and Perioperative Medicine, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, China
| | - Hongkai Lian
- Trauma Research Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, China; Center for Advanced Medicine, College of Medicine, Zhengzhou University, Zhengzhou 450007, China
| | - Xing Li
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Qinjun Chu
- Department of Anesthesiology and Perioperative Medicine, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou 450007, China.
| |
Collapse
|
5
|
Chen R, Zhang H, Li L, Li J, Xie J, Weng J, Tan H, Liu Y, Guo T, Wang M. Roles of ubiquitin-specific proteases in inflammatory diseases. Front Immunol 2024; 15:1258740. [PMID: 38322269 PMCID: PMC10844489 DOI: 10.3389/fimmu.2024.1258740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 01/02/2024] [Indexed: 02/08/2024] Open
Abstract
Ubiquitin-specific proteases (USPs), as one of the deubiquitinating enzymes (DUBs) families, regulate the fate of proteins and signaling pathway transduction by removing ubiquitin chains from the target proteins. USPs are essential for the modulation of a variety of physiological processes, such as DNA repair, cell metabolism and differentiation, epigenetic modulations as well as protein stability. Recently, extensive research has demonstrated that USPs exert a significant impact on innate and adaptive immune reactions, metabolic syndromes, inflammatory disorders, and infection via post-translational modification processes. This review summarizes the important roles of the USPs in the onset and progression of inflammatory diseases, including periodontitis, pneumonia, atherosclerosis, inflammatory bowel disease, sepsis, hepatitis, diabetes, and obesity. Moreover, we highlight a comprehensive overview of the pathogenesis of USPs in these inflammatory diseases as well as post-translational modifications in the inflammatory responses and pave the way for future prospect of targeted therapies in these inflammatory diseases.
Collapse
Affiliation(s)
- Rui Chen
- Center of Obesity and Metabolic Diseases, Department of General Surgery, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
- Department of Stomatology, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, China
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Hui Zhang
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
- College of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Linke Li
- Center of Obesity and Metabolic Diseases, Department of General Surgery, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
- Department of Stomatology, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Jinsheng Li
- College of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Jiang Xie
- Department of Pediatrics, Chengdu Third People's Hospital, Chengdu, Sichuan, China
| | - Jie Weng
- College of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Huan Tan
- Center of Obesity and Metabolic Diseases, Department of General Surgery, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Yanjun Liu
- Center of Obesity and Metabolic Diseases, Department of General Surgery, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Tailin Guo
- Center of Obesity and Metabolic Diseases, Department of General Surgery, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Mengyuan Wang
- Center of Obesity and Metabolic Diseases, Department of General Surgery, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
- Department of Stomatology, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan, China
- College of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
| |
Collapse
|
6
|
Wei X, Cheng X, Luo Y, Li X. Umbilical Cord-Derived Mesenchymal Stem Cells Attenuate S100-Induced Autoimmune Hepatitis via Modulating Th1 and Th17 Cell Responses in Mice. Stem Cells Int 2023; 2023:9992207. [PMID: 37881518 PMCID: PMC10597736 DOI: 10.1155/2023/9992207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 09/08/2023] [Accepted: 09/19/2023] [Indexed: 10/27/2023] Open
Abstract
Currently, the first-line treatment for autoimmune hepatitis (AIH) is still the combination of glucocorticoids or immunosuppressants. However, hormone and immunosuppressive therapy can cause serious side effects, such as Cushing syndrome and bone marrow suppression. Previous studies reported on the applicability and safety of mesenchymal stem cells (MSCs) to ameliorate liver inflammation and fibrosis. However, the characteristics of MSCs sources directly contribute to the different conclusions on the mechanisms underlying MSC-mediated immunoregulation. Bone marrow-derived MSCs can exert an immunosuppression effect to ameliorate the S100-induced AIH model by inhibiting several proinflammatory cytokines and upregulating of PD-L1 in liver tissue. It is not clear whether human umbilical cord-derived MSCs (hUC-MSCs) could directly inhibit liver inflammation and ultimately alleviate the dysfunction of hepatocytes in the AIH model. First, hUC-MSCs were extracted from umbilical cord tissue, and the basic biological properties and multilineage differentiation potential were examined. Second, 1 × 106 hUC-MSCs were administered intravenously to AIH mice. At the peak of the disease, serum levels of alanine aminotransferase and aspartate aminotransferase and pathologic damage to liver tissue were measured to evaluate liver function and degree of inflammation. We also observed that the infiltration of CD4+ T cells in the liver was significantly reduced. Furthermore, the frequency of the splenic IFNγ- and IL-17A- producing CD4+ T cells were also significantly decreased, while we only observed an increasing trend in Treg cells in liver tissue. Third, an RNA sequencing analysis of liver tissue was performed, which showed that in the UC-MSC-treated group, the transcriptional profiles of inflammation-related signaling pathways were significantly negatively regulated compared to those of phosphate-buffered saline-treated mice. Collectively, these findings indicated the potential of hUC-MSC to suppress immune responses in immune anomaly mediated liver disease, thus offering a potential clinical option to improve AIH.
Collapse
Affiliation(s)
- Xiaofeng Wei
- The First Hospital of Lanzhou University, Lanzhou City, Gansu Province, China
- Key Laboratory of Biotherapy and Regenerative Medicine, The First Hospital of Lanzhou University, Lanzhou City, Gansu Province 730000, China
| | - Xinhong Cheng
- The First School of Clinical Medicine, Lanzhou University, Lanzhou City, Gansu Province, China
| | - Yang Luo
- The First Hospital of Lanzhou University, Lanzhou City, Gansu Province, China
- Key Laboratory of Biotherapy and Regenerative Medicine, The First Hospital of Lanzhou University, Lanzhou City, Gansu Province 730000, China
| | - Xun Li
- The First Hospital of Lanzhou University, Lanzhou City, Gansu Province, China
- Key Laboratory of Biotherapy and Regenerative Medicine, The First Hospital of Lanzhou University, Lanzhou City, Gansu Province 730000, China
| |
Collapse
|
7
|
Guo Y, Cui S, Chen Y, Guo S, Chen D. Ubiquitin specific peptidases and prostate cancer. PeerJ 2023; 11:e14799. [PMID: 36811009 PMCID: PMC9939025 DOI: 10.7717/peerj.14799] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/04/2023] [Indexed: 02/18/2023] Open
Abstract
Protein ubiquitination is an important post-translational modification mechanism, which regulates protein stability and activity. The ubiquitination of proteins can be reversed by deubiquitinating enzymes (DUBs). Ubiquitin-specific proteases (USPs), the largest DUB subfamily, can regulate cellular functions by removing ubiquitin(s) from the target proteins. Prostate cancer (PCa) is the second leading type of cancer and the most common cause of cancer-related deaths in men worldwide. Numerous studies have demonstrated that the development of PCa is highly correlated with USPs. The expression of USPs is either high or low in PCa cells, thereby regulating the downstream signaling pathways and causing the development or suppression of PCa. This review summarized the functional roles of USPs in the development PCa and explored their potential applications as therapeutic targets for PCa.
Collapse
Affiliation(s)
- Yunfei Guo
- Shandong University of Technology, School of Life Sciences and Medicine, Zibo, Shandong, China
| | - Shuaishuai Cui
- Shandong University of Technology, School of Life Sciences and Medicine, Zibo, Shandong, China
| | - Yuanyuan Chen
- Shandong University of Technology, School of Life Sciences and Medicine, Zibo, Shandong, China
| | - Song Guo
- Shandong University of Technology, School of Life Sciences and Medicine, Zibo, Shandong, China
| | - Dahu Chen
- Shandong University of Technology, School of Life Sciences and Medicine, Zibo, Shandong, China
| |
Collapse
|
8
|
Cai T, Xu L, Xia D, Zhu L, Lin Y, Yu S, Zhu K, Wang X, Pan C, Chen Y, Chen D. Polyguanine alleviated autoimmune hepatitis through regulation of macrophage receptor with collagenous structure and TLR4‐TRIF‐NF‐κB signalling. J Cell Mol Med 2022; 26:5690-5701. [PMID: 36282897 PMCID: PMC9667514 DOI: 10.1111/jcmm.17599] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/15/2022] [Accepted: 10/10/2022] [Indexed: 11/30/2022] Open
Abstract
Autoimmune hepatitis (AIH) is a progressive and chronic inflammatory disease in the liver. MARCO is a surface receptor of macrophage involving in tissue inflammation and immune disorders. Moreover, polyguanine (PolyG) is considered to bind to macrophage receptor with collagenous structure (MARCO). However, the role of MARCO and PolyG in the development and treatment of AIH still remains unclear. Therefore, this study explores the expression of MARCO and therapeutic activity of PolyG in both S100‐induced AIH in mouse and Lipopolysaccharide (LPS)‐treated macrophage (RAW264.7 cells). Moreover, there were significant increases in inflammatory factors and MARCO, as well as decrease in I‐kappa‐B‐alpha (Ik‐B) in the liver of AIH mice and LPS‐induced cells. However, PolyG treatment significantly reversed the elevation of inflammatory cytokins, MARCO and reduction of Ik‐B. In addition, PolyG treatment could downregulate the expression of Toll‐like receptor 4 (TLR4) and TIR‐domain‐containing adaptor inducing interferon‐β (TRIF), decrease macrophage M1 polarization and increase macrophage M2 polarization. When hepatocytes were co‐cultured with different treatment of macrophages, similar expression profile of inflammatory cytokines was observed in hepatocytes. The research revealed that MARCO expression was elevated in AIH mice. PolyG treatment and inhibition of MARCO significantly reduced inflammatory cytokines expression in the liver as well as hepatocytes and macrophages. Therefore, MARCO could be a target for the treatment of AIH.
Collapse
Affiliation(s)
- Tingchen Cai
- Department of Infectious Diseases, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University The First Affiliated Hospital of Wenzhou Medical University Wenzhou China
| | - Lanman Xu
- Department of Infectious Diseases and Liver Diseases, Ningbo Medical Centre Lihuili Hospital, Affiliated Lihuili Hospital of Ningbo University Ningbo Institute of Innovation for Combined Medicine and Engineering Ningbo China
| | - Dingchao Xia
- Department of Infectious Diseases, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University The First Affiliated Hospital of Wenzhou Medical University Wenzhou China
| | - Lujian Zhu
- Department of Infectious Diseases, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University The First Affiliated Hospital of Wenzhou Medical University Wenzhou China
| | - Yanhan Lin
- Department of Infectious Diseases, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University The First Affiliated Hospital of Wenzhou Medical University Wenzhou China
| | - Sijie Yu
- Department of Infectious Diseases, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University The First Affiliated Hospital of Wenzhou Medical University Wenzhou China
| | - Kailu Zhu
- Department of Infectious Diseases, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University The First Affiliated Hospital of Wenzhou Medical University Wenzhou China
| | - Xiaodong Wang
- Department of Infectious Diseases, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University The First Affiliated Hospital of Wenzhou Medical University Wenzhou China
| | - Chenwei Pan
- Department of Infectious Diseases The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou China
| | - Yongping Chen
- Department of Infectious Diseases, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University The First Affiliated Hospital of Wenzhou Medical University Wenzhou China
| | - Dazhi Chen
- Department of Infectious Diseases, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University The First Affiliated Hospital of Wenzhou Medical University Wenzhou China
- Department of Clinical Medicine Hangzhou Medical College Hangzhou China
| |
Collapse
|
9
|
Inhibition of Ubiquitin-specific Protease 4 Attenuates Epithelial—Mesenchymal Transition of Renal Tubular Epithelial Cells via Transforming Growth Factor Beta Receptor Type I. Curr Med Sci 2022; 42:1000-1006. [DOI: 10.1007/s11596-022-2632-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 05/12/2022] [Indexed: 11/03/2022]
|
10
|
Bailly C, Vergoten G. Binding of Vialinin A and p-Terphenyl Derivatives to Ubiquitin-Specific Protease 4 (USP4): A Molecular Docking Study. Molecules 2022; 27:5909. [PMID: 36144645 PMCID: PMC9505430 DOI: 10.3390/molecules27185909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/06/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
The para-terphenyl derivative vialinin A (Vi-A), isolated from Thelephora fungi, has been characterized as a potent inhibitor of the ubiquitin-specific protease 4 (USP4). Blockade of USP4 contributes to the anti-inflammatory and anticancer properties of the natural product. We have investigated the interaction of Vi-A with USP4 by molecular modeling, to locate the binding site (around residue V98 within the domain in USP segment) and to identify the binding process and interaction contacts. From this model, a series of 32 p-terphenyl compounds were tested as potential USP4 binders, mainly in the vialinin, terrestrin and telephantin series. We identified 11 compounds presenting a satisfactory USP4 binding capacity, including two fungal products, vialinin B and aurantiotinin A, with a more favorable empirical energy of USP4 interaction (ΔE) than the reference product Vi-A. The rare p-terphenyl aurantiotinin A, isolated from the basidiomycete T. aurantiotincta, emerged as a remarkable USP4 binder. Structure-binding relationships have been identified and discussed, to guide the future design of USP4 inhibitors based on the p-terphenyl skeleton. The docking study should help the identification of other protease inhibitors from fungus.
Collapse
Affiliation(s)
- Christian Bailly
- OncoWitan, Consulting Scientific Office, 59290 Lille (Wasquehal), France
| | - Gérard Vergoten
- Institut de Chimie Pharmaceutique Albert Lespagnol, Faculté de Pharmacie, University of Lille, Inserm, INFINITE-U1286, 3 rue du Professeur Laguesse, BP-83, 59006 Lille, France
| |
Collapse
|
11
|
Bailly C. Anti-inflammatory and anticancer p-terphenyl derivatives from fungi of the genus Thelephora. Bioorg Med Chem 2022; 70:116935. [PMID: 35901638 DOI: 10.1016/j.bmc.2022.116935] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/22/2022] [Accepted: 07/11/2022] [Indexed: 02/08/2023]
Abstract
Fungi from the genus Thelephora have been exploited to identify bioactive compounds. The main natural products characterized are para-terphenyl derivatives, chiefly represented by the lead anti-inflammatory compound vialinin A isolated from species T. vialis and T. terrestris. Different series of p-terphenyls have been identified, including vialinins, ganbajunins, terrestrins, telephantins and other products. Their mechanism of action is not always clearly identified, and different potential molecule targets have been proposed. The lead vialinin A functions as a protease inhibitor, efficiently targeting ubiquitin-specific peptidases USP4/5 and sentrin-specific protease SENP1 which are prominent anti-inflammatory and anticancer targets. Protease inhibition is coupled with a powerful inhibition of the cellular production of tumor necrosis factor TNFα. Other mechanisms contributing to the anti-inflammatory or anti-proliferative action of these p-terphenyl compounds have been invoked, including the formation of cytotoxic copper complexes for derivatives bearing a catechol central unit such vialinin A, terrestrin B and telephantin O. These p-terphenyl compounds could be further exploited to design novel anticancer agents, as evidenced with the parent compound terphenyllin (essentially found in Aspergillus species) which has revealed marked antitumor and anti-metastatic effects in xenograft models of gastric and pancreatic cancer. This review shed light on the structural and functional diversity of p-terphenyls compounds isolated from Thelephora species, their molecular targets and pharmacological properties.
Collapse
Affiliation(s)
- Christian Bailly
- OncoWitan, Scientific Consulting Office, Lille (Wasquehal) 59290, France.
| |
Collapse
|
12
|
Xia D, Chen D, Cai T, Zhu L, Lin Y, Yu S, Zhu K, Wang X, Xu L, Chen Y. Nimbolide attenuated the inflammation in the liver of autoimmune hepatitis's mice through regulation of HDAC3. Toxicol Appl Pharmacol 2021; 434:115795. [PMID: 34780724 DOI: 10.1016/j.taap.2021.115795] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/26/2021] [Accepted: 11/07/2021] [Indexed: 02/07/2023]
Abstract
A chronic liver disease named autoimmune hepatitis (AIH) will carry elevated levels of inflammatory cytokines, but there is currently no effective treatment to cure it. Histone deacetylase 3 (HDAC3) takes an important position in regulating the expression of inflammatory genes. Nimbolide (NIB) is a limonoid extracted from the neem tree (Azadirachta indica) that has been found to be effective against many diseases, including cancer, scleroderma, and acute respiratory distress syndrome. Here, we investigated the protective effect of nimbolide on AIH liver. Mice and AML12 cells were employed to establish AIH model with liver antigen S100 and cell injury model of LPS, and then treated with different concentrations of nimbolide. After the successful establishment of the animal model and cell model, inflammatory cytokines of IL-1β, IL-6 and TNF-α as well as cellular signaling related to inflammation such as STAT3, IκB-α and NF-κB were examined. We observed for the first time about nimbolide can effectively inhibit inflammation in AIH mice's liver and AML12 cells by inhibiting HDAC3 expression. HDAC3 knocked down by siRNA in cells can also effectively alleviate the inflammation in AML12 cells, further confirming that HDAC3 plays an important role in the inflammation of liver cells. These results suggest nimbolide could be a potential new treatment for autoimmune hepatitis, and HDAC3 may become a new target for autoimmune hepatitis.
Collapse
Affiliation(s)
- Dingchao Xia
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, Wenzhou 325006, China
| | - Dazhi Chen
- Department of Gastroenterology, The First Hospital of Peking University, BeiJing 100032, China
| | - Tingchen Cai
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, Wenzhou 325006, China
| | - Lujian Zhu
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, Wenzhou 325006, China
| | - Yanhan Lin
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, Wenzhou 325006, China
| | - Sijie Yu
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, Wenzhou 325006, China
| | - Kailu Zhu
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, Wenzhou 325006, China
| | - Xiaodong Wang
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, Wenzhou 325006, China
| | - Lanman Xu
- Department of Infectious Diseases and Liver Diseases, Ningbo Medical Center Lihuili Hospital, Affiliated Hospital of Ningbo University, Ningbo, 315040, Zhejiang, China.
| | - Yongping Chen
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, Wenzhou 325006, China.
| |
Collapse
|
13
|
Kim SH, Baek KH. Regulation of Cancer Metabolism by Deubiquitinating Enzymes: The Warburg Effect. Int J Mol Sci 2021; 22:ijms22126173. [PMID: 34201062 PMCID: PMC8226939 DOI: 10.3390/ijms22126173] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 05/31/2021] [Accepted: 06/05/2021] [Indexed: 12/21/2022] Open
Abstract
Cancer is a disorder of cell growth and proliferation, characterized by different metabolic pathways within normal cells. The Warburg effect is a major metabolic process in cancer cells that affects the cellular responses, such as proliferation and apoptosis. Various signaling factors down/upregulate factors of the glycolysis pathway in cancer cells, and these signaling factors are ubiquitinated/deubiquitinated via the ubiquitin-proteasome system (UPS). Depending on the target protein, DUBs act as both an oncoprotein and a tumor suppressor. Since the degradation of tumor suppressors and stabilization of oncoproteins by either negative regulation by E3 ligases or positive regulation of DUBs, respectively, promote tumorigenesis, it is necessary to suppress these DUBs by applying appropriate inhibitors or small molecules. Therefore, we propose that the DUBs and their inhibitors related to the Warburg effect are potential anticancer targets.
Collapse
|