1
|
Guo H, Tang H, Yang Y, Xu H, Fan J, Chen S, Hou L, Yuan Y, Zhang G. NEDD4 facilitates the progression of endometrial carcinoma by enhancing PAMR1 protein degradation through ubiquitination. Funct Integr Genomics 2025; 25:65. [PMID: 40087191 DOI: 10.1007/s10142-025-01567-4] [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: 10/08/2024] [Revised: 01/28/2025] [Accepted: 02/23/2025] [Indexed: 03/17/2025]
Abstract
Endometrial carcinoma (EC) is an epithelial malignant neoplasm that frequently appears in postmenopausal and perimenopausal women. PAMR1 is related to the prognosis of EC. Here, we probed into the significance of PAMR1 in EC progression and its acting mechanism. In silico analysis was conducted to identify the differentially expressed gene PAMR1 and its upstream gene NEDD4 in EC, followed by the determination of their expression in EC tissues and cells. The gene expression, cell proliferation, angiogenesis, migration, invasion, and apoptosis were examined after ectopic expression or knockdown experiments. The interaction between NEDD4 and PAMR1 and the level of PAMR1 ubiquitination were examined. The injection of Ishikawa cell suspensions into nude mice was carried out to establish a tumor xenograft model, validating the roles of PAMR1 and NEDD4 in EC. EC cells exhibited high NEDD4 expression and low PAMR1 expression. NEDD4 knockdown or PAMR1 overexpression suppressed the invasive, migrating, angiogenic, and proliferative properties of EC cells while promoting apoptosis. NEDD4 facilitated PAMR1 protein degradation through ubiquitination. Deletion of PAMR1 abolished the inhibitory effects of NEDD4 knockdown on the malignant behaviors of EC cells. Furthermore, NEDD4 knockdown restrained EC growth in nude mice by increasing PAMR1 protein expression. NEDD4 facilitated EC progression by enhancing PAMR1 protein degradation through ubiquitination.
Collapse
Affiliation(s)
- Hongbo Guo
- Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong Province, 518038, China
- Women and Children's Medical Center, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, No.2004, Hongli Road, Futian District, Shenzhen, Guangdong Province, 518038, China
| | - Hongping Tang
- Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong Province, 518038, China
- Women and Children's Medical Center, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, No.2004, Hongli Road, Futian District, Shenzhen, Guangdong Province, 518038, China
| | - Yihui Yang
- Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong Province, 518038, China
- Women and Children's Medical Center, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, No.2004, Hongli Road, Futian District, Shenzhen, Guangdong Province, 518038, China
| | - Hui Xu
- Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong Province, 518038, China
- Women and Children's Medical Center, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, No.2004, Hongli Road, Futian District, Shenzhen, Guangdong Province, 518038, China
| | - Jiaqi Fan
- Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong Province, 518038, China
- Women and Children's Medical Center, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, No.2004, Hongli Road, Futian District, Shenzhen, Guangdong Province, 518038, China
| | - Shuxia Chen
- Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong Province, 518038, China
- Women and Children's Medical Center, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, No.2004, Hongli Road, Futian District, Shenzhen, Guangdong Province, 518038, China
| | - Lingxiu Hou
- Department of Ultrasound, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, 541001, China
| | - Ying Yuan
- Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong Province, 518038, China.
- Women and Children's Medical Center, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, No.2004, Hongli Road, Futian District, Shenzhen, Guangdong Province, 518038, China.
| | - Guangwu Zhang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, No.1068, Xueyuan Avenue, Nanshan District, Shenzhen, Guangdong Province, 518055, China.
| |
Collapse
|
2
|
Li J, Peng Y, Zhen D, Guo C, Peng W. NEDD4 enhances bone‑tendon healing in rotator cuff tears by reducing fatty infiltration. Mol Med Rep 2025; 31:55. [PMID: 39704218 DOI: 10.3892/mmr.2024.13420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 11/14/2024] [Indexed: 12/21/2024] Open
Abstract
Rotator cuff tears (RCT) can cause shoulder pain, weakness and stiffness, significantly affecting daily life. Analysis of the GSE103266 dataset revealed significant changes in the mTOR/PI3K/Akt signaling pathway and lipid metabolism‑related pathways, suggesting that fatty infiltration may affect RCT. The analysis indicated that the ubiquitin ligase NEDD4 plays a critical role in RCT. NEDD4 was found to be highly associated with the mTOR/PI3K/Akt signaling pathway. An RCT model in Sprague‑Dawley (SD) rats was established to study the role of NEDD4 in regulating the mTOR pathway and investigate its effects on fatty infiltration. SD rats were divided into NEDD4 overexpression and knockout groups. Tissue recovery, apoptosis and fat deposition were measured through histological staining, reverse transcription‑quantitative PCR and western blotting. Additionally, cell culture of fibro‑adipogenic progenitors and lentiviral transfection were conducted to investigate the effect of NEDD4 on adipocyte differentiation. NEDD4 overexpression significantly reduced lipid accumulation, whereas NEDD4 knockdown enhanced lipid accumulation. NEDD4 was found to regulate the mTOR pathway and the expression of adipogenesis‑related genes, promoting fat metabolism and inhibiting adipocyte differentiation. Histological analysis indicated that NEDD4 overexpression improved tissue recovery and reduced apoptosis. Targeting NEDD4 offers a potential therapeutic strategy to improve the clinical outcomes of patients with RCT by modulating the mTOR pathway and fat metabolism.
Collapse
Affiliation(s)
- Jian Li
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Ying Peng
- Thyroid Disease Diagnosis and Treatment Center, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Dong Zhen
- Department of Sports Medicine, The Beijing Jishuitan Hospital Guizhou Hospital, Guiyang, Guizhou 550014, P.R. China
| | - Caifen Guo
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Wuxun Peng
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| |
Collapse
|
3
|
Tang X, Liu X, Sha X, Zhang Y, Zu Y, Fan Q, Hu L, Sun S, Zhang Z, Chen F, Yan C, Chen X, Xu Y, Chen W, Shao Y, Gu J, Pu J, Yu B, Han Y, Xie L, Han Y, Ji Y. NEDD4-Mediated GSNOR Degradation Aggravates Cardiac Hypertrophy and Dysfunction. Circ Res 2025; 136:422-438. [PMID: 39846173 DOI: 10.1161/circresaha.124.324872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 01/09/2025] [Accepted: 01/11/2025] [Indexed: 01/24/2025]
Abstract
BACKGROUND The decrease in S-nitrosoglutathione reductase (GSNOR) leads to an elevation of S-nitrosylation, thereby exacerbating the progression of cardiomyopathy in response to hemodynamic stress. However, the mechanisms under GSNOR decrease remain unclear. Here, we identify NEDD4 (neuronal precursor cell expressed developmentally downregulated 4) as a novel molecule that plays a crucial role in the pathogenesis of pressure overload-induced cardiac hypertrophy, by modulating GSNOR levels, thereby demonstrating significant therapeutic potential. METHODS Protein synthesis and degradation inhibitors were used to verify the reasons for the decrease in GSNOR. Mass spectrometry and database filtering were used to uncover NEDD4, the E3 Ub (ubiquitin) ligase, involved in GSNOR decrease. NEDD4 cardiomyocyte-specific deficiency mice were used to evaluate the role of NEDD4 and NEDD4-induced ubiquitination of GSNOR in cardiac hypertrophy in vivo. Both IBM (indolebutenate methyl ester derivatives), a highly specific NEDD4 inhibitor, and indole-3-carbinol, a NEDD4 inhibitor currently undergoing phase 2 clinical trial, were used to effectively suppress the NEDD4/GSNOR axis. RESULTS GSNOR protein levels were reduced, while mRNA levels remained unchanged in myocardium samples from hypertrophic patients and transverse aortic constriction-induced mice, indicating GSNOR is regulated by ubiquitination. NEDD4, an E3 Ub ligase, was associated with GSNOR ubiquitination, which exhibited significantly higher expression levels in hypertrophic myocardial samples. Moreover, either the NEDD4 enzyme-dead mutant or GSNOR nonubiquitylated mutant decreased GSNOR ubiquitination and inhibited cardiac hypertrophic growth. Cardiomyocyte-specific NEDD4 deficiency inhibited cardiac hypertrophy in vitro and in vivo. NEDD4 inhibitor IBM effectively suppressed GSNOR ubiquitination and cardiac hypertrophy. Clinically, indole-3-carbinol, a NEDD4 inhibitor in phase II clinical trials used as an antitumor drug, demonstrated comparable efficacy. CONCLUSIONS Our findings showed that upregulated NEDD4 leads to GSNOR ubiquitination and subsequent degradation, thereby facilitating the progression of cardiac hypertrophy. NEDD4 inhibitors may serve as a potential therapeutic strategy for the treatment of cardiac hypertrophy and heart failure.
Collapse
Affiliation(s)
- Xin Tang
- Key Laboratory of Drug Targets and Translational Medicine for Cardio-cerebrovascular Diseases, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Jiangsu, China (X.T., X.L., X.S., Y. Zhang, Y. Zu, Q.F., L.H., S.S., F.C., L.X., Y.J.)
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Nanjing Medical University, Nanjing, Jiangsu, China (X.T., X.L., X.S., Y. Zhang, Y. Zu, Q.F., L.H., S.S., F.C., L.X., Y.J.)
| | - Xiameng Liu
- Key Laboratory of Drug Targets and Translational Medicine for Cardio-cerebrovascular Diseases, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Jiangsu, China (X.T., X.L., X.S., Y. Zhang, Y. Zu, Q.F., L.H., S.S., F.C., L.X., Y.J.)
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Nanjing Medical University, Nanjing, Jiangsu, China (X.T., X.L., X.S., Y. Zhang, Y. Zu, Q.F., L.H., S.S., F.C., L.X., Y.J.)
| | - Xinqi Sha
- Key Laboratory of Drug Targets and Translational Medicine for Cardio-cerebrovascular Diseases, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Jiangsu, China (X.T., X.L., X.S., Y. Zhang, Y. Zu, Q.F., L.H., S.S., F.C., L.X., Y.J.)
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Nanjing Medical University, Nanjing, Jiangsu, China (X.T., X.L., X.S., Y. Zhang, Y. Zu, Q.F., L.H., S.S., F.C., L.X., Y.J.)
| | - Yan Zhang
- Key Laboratory of Drug Targets and Translational Medicine for Cardio-cerebrovascular Diseases, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Jiangsu, China (X.T., X.L., X.S., Y. Zhang, Y. Zu, Q.F., L.H., S.S., F.C., L.X., Y.J.)
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Nanjing Medical University, Nanjing, Jiangsu, China (X.T., X.L., X.S., Y. Zhang, Y. Zu, Q.F., L.H., S.S., F.C., L.X., Y.J.)
| | - Yan Zu
- Key Laboratory of Drug Targets and Translational Medicine for Cardio-cerebrovascular Diseases, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Jiangsu, China (X.T., X.L., X.S., Y. Zhang, Y. Zu, Q.F., L.H., S.S., F.C., L.X., Y.J.)
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Nanjing Medical University, Nanjing, Jiangsu, China (X.T., X.L., X.S., Y. Zhang, Y. Zu, Q.F., L.H., S.S., F.C., L.X., Y.J.)
| | - Qiyao Fan
- Key Laboratory of Drug Targets and Translational Medicine for Cardio-cerebrovascular Diseases, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Jiangsu, China (X.T., X.L., X.S., Y. Zhang, Y. Zu, Q.F., L.H., S.S., F.C., L.X., Y.J.)
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Nanjing Medical University, Nanjing, Jiangsu, China (X.T., X.L., X.S., Y. Zhang, Y. Zu, Q.F., L.H., S.S., F.C., L.X., Y.J.)
| | - Lulu Hu
- Key Laboratory of Drug Targets and Translational Medicine for Cardio-cerebrovascular Diseases, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Jiangsu, China (X.T., X.L., X.S., Y. Zhang, Y. Zu, Q.F., L.H., S.S., F.C., L.X., Y.J.)
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Nanjing Medical University, Nanjing, Jiangsu, China (X.T., X.L., X.S., Y. Zhang, Y. Zu, Q.F., L.H., S.S., F.C., L.X., Y.J.)
| | - Shixiu Sun
- Key Laboratory of Drug Targets and Translational Medicine for Cardio-cerebrovascular Diseases, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Jiangsu, China (X.T., X.L., X.S., Y. Zhang, Y. Zu, Q.F., L.H., S.S., F.C., L.X., Y.J.)
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Nanjing Medical University, Nanjing, Jiangsu, China (X.T., X.L., X.S., Y. Zhang, Y. Zu, Q.F., L.H., S.S., F.C., L.X., Y.J.)
| | - Zhiren Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Key Laboratory of Cardiovascular Medicine Research and Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, NHC Key Laboratory of Cell Transplantation, the Central Laboratory of the First Affiliated Hospital, Harbin Medical University, Heilongjiang, China (Z.Z., Yi Han, Y.J.)
| | - Feng Chen
- Key Laboratory of Drug Targets and Translational Medicine for Cardio-cerebrovascular Diseases, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Jiangsu, China (X.T., X.L., X.S., Y. Zhang, Y. Zu, Q.F., L.H., S.S., F.C., L.X., Y.J.)
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Nanjing Medical University, Nanjing, Jiangsu, China (X.T., X.L., X.S., Y. Zhang, Y. Zu, Q.F., L.H., S.S., F.C., L.X., Y.J.)
- Department of Forensic Medicine, Nanjing Medical University, China (F.C.)
| | - ChengHui Yan
- State Key Laboratory of Frigid Zone Cardiovascular Diseases, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China (C.H.Y., Yaling Han)
| | - Xin Chen
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, China (X.C., Y.X., W.C.)
| | - Yueyue Xu
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, China (X.C., Y.X., W.C.)
| | - Wen Chen
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, China (X.C., Y.X., W.C.)
| | - Yongfeng Shao
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Nanjing Medical University, China (Y.S., J.G.)
| | - Jiaxi Gu
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Nanjing Medical University, China (Y.S., J.G.)
| | - Jun Pu
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China (J.P.)
| | - Bo Yu
- Department of Cardiology, the 2nd Affiliated Hospital of Harbin Medical University, The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, China (B.Y.)
| | - Yaling Han
- State Key Laboratory of Frigid Zone Cardiovascular Diseases, Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China (C.H.Y., Yaling Han)
| | - Liping Xie
- Key Laboratory of Drug Targets and Translational Medicine for Cardio-cerebrovascular Diseases, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Jiangsu, China (X.T., X.L., X.S., Y. Zhang, Y. Zu, Q.F., L.H., S.S., F.C., L.X., Y.J.)
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Nanjing Medical University, Nanjing, Jiangsu, China (X.T., X.L., X.S., Y. Zhang, Y. Zu, Q.F., L.H., S.S., F.C., L.X., Y.J.)
| | - Yi Han
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Key Laboratory of Cardiovascular Medicine Research and Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, NHC Key Laboratory of Cell Transplantation, the Central Laboratory of the First Affiliated Hospital, Harbin Medical University, Heilongjiang, China (Z.Z., Yi Han, Y.J.)
- Department of Critical Care Medicine, the 2nd Affiliated Hospital, Harbin Medical University, China (Yi Han)
| | - Yong Ji
- Key Laboratory of Drug Targets and Translational Medicine for Cardio-cerebrovascular Diseases, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Jiangsu, China (X.T., X.L., X.S., Y. Zhang, Y. Zu, Q.F., L.H., S.S., F.C., L.X., Y.J.)
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Nanjing Medical University, Nanjing, Jiangsu, China (X.T., X.L., X.S., Y. Zhang, Y. Zu, Q.F., L.H., S.S., F.C., L.X., Y.J.)
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Key Laboratory of Cardiovascular Medicine Research and Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, NHC Key Laboratory of Cell Transplantation, the Central Laboratory of the First Affiliated Hospital, Harbin Medical University, Heilongjiang, China (Z.Z., Yi Han, Y.J.)
| |
Collapse
|
4
|
Han J, Shin YH, Kim E, Park HM, Kim JY. Proteomic Characterization of NEDD4 Unveils Its Potential Novel Downstream Effectors in Gastric Cancer. J Proteome Res 2025; 24:891-902. [PMID: 39874481 DOI: 10.1021/acs.jproteome.4c01109] [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] [Indexed: 01/30/2025]
Abstract
The E3 ubiquitin ligase neural precursor cell-expressed developmentally down-regulated 4 (NEDD4) is involved in various cancer signaling pathways, including PTEN/AKT. However, its role in promoting gastric cancer (GC) progression is unclear. This study was conducted to elucidate the role of NEDD4 in GC progression. We found that the inhibition of NEDD4 expression significantly reduced the migratory and proliferative abilities of GC cells, with minimal impact on the PTEN expression or p-AKT activation, suggesting that NEDD4 may exert its GC-promoting effects through alternative pathways. To gain novel insights into the role of NEDD4 in GC, we performed a comprehensive proteomic analysis to search for proteins with altered expression levels following NEDD4 gene knockdown, identifying a total of 3916 proteins. Pathway analysis of differentially expressed proteins (DEPs) indicated the potential involvement of NEDD4 in cancer-related metabolic pathways. Furthermore, the protein-protein interaction network of the DEPs revealed enriched core modules, highlighting key cellular processes and signaling pathways regulated by NEDD4 in GC. Additionally, we identified proteins whose expression was altered by NEDD4 inhibition, some of which were associated with poor prognosis in GC. These findings suggest that these proteins may act as downstream effectors that contribute to NEDD4-mediated GC progression.
Collapse
Affiliation(s)
- Jisoo Han
- Graduate School of Analytical Science and Technology (GRAST), Chungnam National University, Daejeon 34134, Republic of Korea
| | - Yoon-Hee Shin
- Advanced Analysis and Data Center, Korea Institute of Science and Technology (KIST), Seoul 02456, Republic of Korea
| | - Eunjung Kim
- Natural Product Systems Biology Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea
| | - Hyun-Mee Park
- Advanced Analysis and Data Center, Korea Institute of Science and Technology (KIST), Seoul 02456, Republic of Korea
| | - Jae-Young Kim
- Graduate School of Analytical Science and Technology (GRAST), Chungnam National University, Daejeon 34134, Republic of Korea
| |
Collapse
|
5
|
Choi KM, Kim SJ, Ji MJ, Kim E, Kim JS, Park HM, Kim JY. Activity-based protein profiling and global proteome analysis reveal MASTL as a potential therapeutic target in gastric cancer. Cell Commun Signal 2024; 22:397. [PMID: 39138495 PMCID: PMC11323684 DOI: 10.1186/s12964-024-01783-8] [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: 05/24/2024] [Accepted: 08/08/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is a prevalent malignancy with limited therapeutic options for advanced stages. This study aimed to identify novel therapeutic targets for GC by profiling HSP90 client kinases. METHODS We used mass spectrometry-based activity-based protein profiling (ABPP) with a desthiobiotin-ATP probe, combined with sensitivity analysis of HSP90 inhibitors, to profile kinases in a panel of GC cell lines. We identified kinases regulated by HSP90 in inhibitor-sensitive cells and investigated the impact of MASTL knockdown on GC cell behavior. Global proteomic analysis following MASTL knockdown was performed, and bioinformatics tools were used to analyze the resulting data. RESULTS Four kinases-MASTL, STK11, CHEK1, and MET-were identified as HSP90-regulated in HSP90 inhibitor-sensitive cells. Among these, microtubule-associated serine/threonine kinase-like (MASTL) was upregulated in GC and associated with poor prognosis. MASTL knockdown decreased migration, invasion, and proliferation of GC cells. Global proteomic profiling following MASTL knockdown revealed NEDD4-1 as a potential downstream mediator of MASTL in GC progression. NEDD4-1 was also upregulated in GC and associated with poor prognosis. Similar to MASTL inhibition, NEDD4-1 knockdown suppressed migration, invasion, and proliferation of GC cells. CONCLUSIONS Our multi-proteomic analyses suggest that targeting MASTL could be a promising therapy for advanced gastric cancer, potentially through the reduction of tumor-promoting proteins including NEDD4-1. This study enhances our understanding of kinase signaling pathways in GC and provides new insights for potential treatment strategies.
Collapse
Affiliation(s)
- Kyoung-Min Choi
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Sung-Jin Kim
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Mi-Jung Ji
- Advanced Analysis and Data Center, Korea Institute of Science and Technology (KIST), Seoul, 02456, Republic of Korea
| | - Eunjung Kim
- Natural Product Informatics Center, Korea Institute of Science and Technology (KIST), Gangneung, 25451, Republic of Korea
| | - Jae-Sung Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, 01812, Republic of Korea
| | - Hyun-Mee Park
- Advanced Analysis and Data Center, Korea Institute of Science and Technology (KIST), Seoul, 02456, Republic of Korea
| | - Jae-Young Kim
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon, 34134, Republic of Korea.
| |
Collapse
|
6
|
Jan K, Hassan N, James A, Hussain I, Rashid SM. Exploring molecular targets in cancer: Unveiling the anticancer potential of Paeoniflorin through a comprehensive analysis of diverse signaling pathways and recent advances. J Biol Methods 2024; 11:e99010014. [PMID: 39323487 PMCID: PMC11423941 DOI: 10.14440/jbm.2024.0003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/14/2024] [Accepted: 06/20/2024] [Indexed: 09/27/2024] Open
Abstract
Tumors have posed significant threats to human health for over 250 years, emerging as the foremost cause of death. While chemotherapeutic drugs are effective in treating tumors, their side effects can sometimes be challenging to manage during therapy. Nonetheless, there is growing interest in exploring natural compounds as alternatives, which potentially achieve therapeutic outcomes comparable to conventional chemotherapeutics with fewer adverse effects. Paeoniflorin (PF), a monoterpene glycoside derived from the root of Paeonia lactiflora, has garnered significant attention lately due to its promising anti-cancer properties. This review offers an updated outline of the molecular mechanisms underlying PF's anti-tumor function, with a focus on its modulation of various signaling pathways. PF exerts its anti-tumor activity by regulating crucial cellular processes including apoptosis, angiogenesis, proliferation, and metastasis. We explored the multifaceted impact of PF while modulating through signaling pathways, encompassing nuclear factor kappa B, NOTCH, caspase cascade, transforming growth factor-β, NEDD4, P53/14-3-3, STAT 3, MAPK, MMP-9, and SKP2 signaling pathways, highlighting its versatility in targeting diverse malignancies. Furthermore, we discuss future research directions aimed at exploring innovative and targeted cancer therapies facilitated by PF.
Collapse
Affiliation(s)
- Kounser Jan
- Division of Veterinary Biochemistry, Faculty of Veterinary Science and Animal Husbandry, Srinagar, Jammu and Kashmir, 190006, India
| | - Neelofar Hassan
- Division of Veterinary Biochemistry, Faculty of Veterinary Science and Animal Husbandry, Srinagar, Jammu and Kashmir, 190006, India
| | - Antonisamy James
- Departments of Medicinal and Biological Chemistry, The University of Toledo, Toledo, Ohio, 43614, United States of America
| | - Ishraq Hussain
- Division of Veterinary Biochemistry, Faculty of Veterinary Science and Animal Husbandry, Srinagar, Jammu and Kashmir, 190006, India
| | - Shahzada Mudasir Rashid
- Division of Veterinary Biochemistry, Faculty of Veterinary Science and Animal Husbandry, Srinagar, Jammu and Kashmir, 190006, India
- Departments of Medicinal and Biological Chemistry, The University of Toledo, Toledo, Ohio, 43614, United States of America
| |
Collapse
|
7
|
Fan S, Yan X, Hu X, Liu X, Zhao S, Zhang Y, Zhou X, Shen X, Qi Q, Chen Y. Shikonin blocks CAF-induced TNBC metastasis by suppressing mitochondrial biogenesis through GSK-3β/NEDD4-1 mediated phosphorylation-dependent degradation of PGC-1α. J Exp Clin Cancer Res 2024; 43:180. [PMID: 38937832 PMCID: PMC11210116 DOI: 10.1186/s13046-024-03101-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/14/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is characterized by its high metastatic potential, which results in poor patient survival. Cancer-associated fibroblasts (CAFs) are crucial in facilitating TNBC metastasis via induction of mitochondrial biogenesis. However, how to inhibit CAF-conferred mitochondrial biogenesis is still needed to explore. METHODS We investigated metastasis using wound healing and cell invasion assays, 3D-culture, anoikis detection, and NOD/SCID mice. Mitochondrial biogenesis was detected by MitoTracker green FM staining, quantification of mitochondrial DNA levels, and blue-native polyacrylamide gel electrophoresis. The expression, transcription, and phosphorylation of peroxisome-proliferator activated receptor coactivator 1α (PGC-1α) were detected by western blotting, chromatin immunoprecipitation, dual-luciferase reporter assay, quantitative polymerase chain reaction, immunoprecipitation, and liquid chromatography-tandem mass spectrometry. The prognostic role of PGC-1α in TNBC was evaluated using the Kaplan-Meier plotter database and clinical breast cancer tissue samples. RESULTS We demonstrated that PGC-1α indicated lymph node metastasis, tumor thrombus formation, and poor survival in TNBC patients, and it was induced by CAFs, which functioned as an inducer of mitochondrial biogenesis and metastasis in TNBC. Shikonin impeded the CAF-induced PGC-1α expression, nuclear localization, and interaction with estrogen-related receptor alpha (ERRα), thereby inhibiting PGC-1α/ERRα-targeted mitochondrial genes. Mechanistically, the downregulation of PGC-1α was mediated by synthase kinase 3β-induced phosphorylation of PGC-1α at Thr295, which associated with neural precursor cell expressed developmentally downregulated 4e1 recognition and subsequent degradation by ubiquitin proteolysis. Mutation of PGC-1α at Thr295 negated the suppressive effects of shikonin on CAF-stimulated TNBC mitochondrial biogenesis and metastasis in vitro and in vivo. CONCLUSIONS Our findings indicate that PGC-1α is a viable target for blocking TNBC metastasis by disrupting mitochondrial biogenesis, and that shikonin merits potential for treatment of TNBC metastasis as an inhibitor of mitochondrial biogenesis through targeting PGC-1α.
Collapse
Affiliation(s)
- Shuangqin Fan
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou, 561113, China
- Key Laboratory of Novel Anti-Cancer Drug Targets Discovery and Application, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, 561113, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou, 561113, China
| | - Xiaomin Yan
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou, 561113, China
- Key Laboratory of Novel Anti-Cancer Drug Targets Discovery and Application, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, 561113, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou, 561113, China
| | - Xiaoxia Hu
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou, 561113, China
- Key Laboratory of Novel Anti-Cancer Drug Targets Discovery and Application, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, 561113, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou, 561113, China
| | - Xing Liu
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou, 561113, China
- Key Laboratory of Novel Anti-Cancer Drug Targets Discovery and Application, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, 561113, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou, 561113, China
| | - Shijie Zhao
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou, 561113, China
- Key Laboratory of Novel Anti-Cancer Drug Targets Discovery and Application, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, 561113, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou, 561113, China
| | - Yue Zhang
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou, 561113, China
- Key Laboratory of Novel Anti-Cancer Drug Targets Discovery and Application, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, 561113, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou, 561113, China
| | - Xiaofeng Zhou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Xiangchun Shen
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou, 561113, China.
- Key Laboratory of Novel Anti-Cancer Drug Targets Discovery and Application, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, 561113, China.
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou, 561113, China.
| | - Qi Qi
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, China.
| | - Yan Chen
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou, 561113, China.
- Key Laboratory of Novel Anti-Cancer Drug Targets Discovery and Application, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, 561113, China.
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, No.6 Ankang Avenue, Guian New District, Guizhou, 561113, China.
| |
Collapse
|
8
|
Ling X, Xu W, Tang J, Cao Q, Luo G, Chen X, Yang S, Reinach PS, Yan D. The Role of Ubiquitination and the E3 Ligase Nedd4 in Regulating Corneal Epithelial Wound Healing. Invest Ophthalmol Vis Sci 2024; 65:29. [PMID: 38888282 PMCID: PMC11186577 DOI: 10.1167/iovs.65.6.29] [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: 11/25/2023] [Accepted: 05/25/2024] [Indexed: 06/20/2024] Open
Abstract
Purpose Ubiquitination serves as a fundamental post-translational modification in numerous cellular events. Yet, its role in regulating corneal epithelial wound healing (CEWH) remains elusive. This study endeavored to determine the function and mechanism of ubiquitination in CEWH. Methods Western blot and immunoprecipitation were used to discern ubiquitination alterations during CEWH in mice. Interventions, including neuronally expressed developmentally downregulated 4 (Nedd4) siRNA and proteasome/lysosome inhibitor, assessed their impact on CEWH. In vitro analyses, such as the scratch wound assay, MTS assay, and EdU staining, were conducted to gauge cell migration and proliferation in human corneal epithelial cells (HCECs). Moreover, transfection of miR-30/200 coupled with a luciferase activity assay ascertained their regulatory mechanism on Nedd4. Results Global ubiquitination levels were markedly increased during the mouse CEWH. Importantly, the application of either proteasomal or lysosomal inhibitors notably impeded the healing process both in vivo and in vitro. Furthermore, Nedd4 was identified as an essential E3 ligase for CEWH. Nedd4 expression was significantly upregulated during CEWH. In vivo studies revealed that downregulation of Nedd4 substantially delayed CEWH, whereas further investigations underscored its role in regulating cell proliferation and migration, through the Stat3 pathway by targeting phosphatase and tensin homolog (PTEN). Notably, our findings pinpointed miR-30/200 family members as direct regulators of Nedd4. Conclusions Ubiquitination holds pivotal significance in orchestrating CEWH. The critical E3 ligase Nedd4, under the regulatory purview of miR-30 and miR-200, facilitates CEWH through PTEN-mediated Stat3 signaling. This revelation sheds light on a prospective therapeutic target within the realm of CEWH.
Collapse
Affiliation(s)
- Xuemei Ling
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Weiwei Xu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jingjing Tang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Qiongjie Cao
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Guangying Luo
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xiaoyan Chen
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Shuai Yang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Peter Sol Reinach
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Dongsheng Yan
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
9
|
Zhang W, Du D, Lu H, Zhang D, Liu L, Li J, Chen Z, Yu X, Ye M, Wang W, Chen L, Shao J. FAT10 mediates the sorafenib-resistance of hepatocellular carcinoma cells by stabilizing E3 ligase NEDD4 to enhance PTEN/AKT pathway-induced autophagy. Am J Cancer Res 2024; 14:1523-1544. [PMID: 38726263 PMCID: PMC11076247 DOI: 10.62347/epit4481] [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: 11/14/2023] [Accepted: 03/31/2024] [Indexed: 05/12/2024] Open
Abstract
Although sorafenib is the first-line therapeutic agent for advanced hepatocellular carcinoma (HCC), the development of drug resistance in HCC cells limits its clinical efficacy. However, the key factors involved in mediating the sorafenib resistance of HCC cells and the underlying mechanisms have not been elucidated. In this study, we generated sorafenib-resistant HCC cell lines, and our data demonstrate that HLA-F locus-adjacent transcript 10 (FAT10), a ubiquitin-like protein, is markedly upregulated in sorafenib-resistant HCC cells and that reducing the expression of FAT10 in sorafenib-resistant HCC cells increases sensitivity to sorafenib. Mechanistically, FAT10 stabilizes the expression of the PTEN-specific E3 ubiquitin ligase NEDD4 that causes downregulation of PTEN, thereby inducing AKT-mediated autophagy and promoting the resistance of HCC cells to sorafenib. Moreover, we screened the small molecule Compound 7695-0983, which increases the sensitivity of sorafenib-resistant HCC cells to sorafenib by inhibiting the expression of FAT10 to inhibit NEDD4-PTEN/AKT axis-mediated autophagy. Collectively, our preclinical findings identify FAT10 as a key factor in the sorafenib resistance of HCC cells and elucidate its underlying mechanism. This study provides new mechanistic insight for the exploitation of novel sorafenib-based tyrosine kinase inhibitor (TKI)-targeted drugs for treating advanced HCC.
Collapse
Affiliation(s)
- Wenming Zhang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
- Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
- Liver Cancer Institute, Nanchang UniversityNanchang 330000, Jiangxi, China
- Jiangxi Province Clinical Research Center of General Surgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
| | - Dongnian Du
- Department of General Surgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
- Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
- Liver Cancer Institute, Nanchang UniversityNanchang 330000, Jiangxi, China
- Jiangxi Province Clinical Research Center of General Surgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
| | - Hongcheng Lu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
- Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
- Liver Cancer Institute, Nanchang UniversityNanchang 330000, Jiangxi, China
- Jiangxi Province Clinical Research Center of General Surgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
| | - Dandan Zhang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
- Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
- Liver Cancer Institute, Nanchang UniversityNanchang 330000, Jiangxi, China
- Jiangxi Province Clinical Research Center of General Surgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
| | - Lingpeng Liu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
- Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
- Liver Cancer Institute, Nanchang UniversityNanchang 330000, Jiangxi, China
- Jiangxi Province Clinical Research Center of General Surgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
| | - Jiajuan Li
- Department of General Surgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
- Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
- Liver Cancer Institute, Nanchang UniversityNanchang 330000, Jiangxi, China
- Jiangxi Province Clinical Research Center of General Surgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
| | - Zehao Chen
- Department of General Surgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
- Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
- Liver Cancer Institute, Nanchang UniversityNanchang 330000, Jiangxi, China
- Jiangxi Province Clinical Research Center of General Surgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
| | - Xuzhe Yu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
- Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
- Liver Cancer Institute, Nanchang UniversityNanchang 330000, Jiangxi, China
- Jiangxi Province Clinical Research Center of General Surgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
| | - Miao Ye
- Department of General Surgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
- Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
- Liver Cancer Institute, Nanchang UniversityNanchang 330000, Jiangxi, China
- Jiangxi Province Clinical Research Center of General Surgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
| | - Wei Wang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
- Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
- Liver Cancer Institute, Nanchang UniversityNanchang 330000, Jiangxi, China
- Jiangxi Province Clinical Research Center of General Surgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
| | - Leifeng Chen
- Department of General Surgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
- Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
- Liver Cancer Institute, Nanchang UniversityNanchang 330000, Jiangxi, China
- Jiangxi Province Clinical Research Center of General Surgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
| | - Jianghua Shao
- Department of General Surgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
- Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
- Liver Cancer Institute, Nanchang UniversityNanchang 330000, Jiangxi, China
- Jiangxi Province Clinical Research Center of General Surgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330000, Jiangxi, China
| |
Collapse
|
10
|
Kaushik A, Parashar S, Ambasta RK, Kumar P. Ubiquitin E3 ligases assisted technologies in protein degradation: Sharing pathways in neurodegenerative disorders and cancer. Ageing Res Rev 2024; 96:102279. [PMID: 38521359 DOI: 10.1016/j.arr.2024.102279] [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: 01/10/2024] [Revised: 03/08/2024] [Accepted: 03/18/2024] [Indexed: 03/25/2024]
Abstract
E3 ligases, essential components of the ubiquitin-proteasome-mediated protein degradation system, play a critical role in cellular regulation. By covalently attaching ubiquitin (Ub) molecules to target proteins, these ligases mark them for degradation, influencing various bioprocesses. With over 600 E3 ligases identified, there is a growing realization of their potential as therapeutic candidates for addressing proteinopathies in cancer and neurodegenerative disorders (NDDs). Recent research has highlighted the need to delve deeper into the intricate roles of E3 ligases as nexus points in the pathogenesis of both cancer and NDDs. Their dysregulation is emerging as a common thread linking these seemingly disparate diseases, necessitating a comprehensive understanding of their molecular intricacies. Herein, we have discussed (i) the fundamental mechanisms through which different types of E3 ligases actively participate in selective protein degradation in cancer and NDDs, followed by an examination of common E3 ligases playing pivotal roles in both situations, emphasising common players. Moving to, (ii) the functional domains and motifs of E3 ligases involved in ubiquitination, we have explored their interactions with specific substrates in NDDs and cancer. Additionally, (iii) we have explored techniques like PROTAC, molecular glues, and other state-of-the-art methods for hijacking neurotoxic and oncoproteins. Lastly, (iv) we have provided insights into ongoing clinical trials, offering a glimpse into the evolving landscape of E3-based therapeutics for cancer and NDDs. Unravelling the intricate network of E3 ligase-mediated regulation holds the key to unlocking targeted therapies that address the specific molecular signatures of individual patients, heralding a new era in personalized medicines.
Collapse
Affiliation(s)
- Aastha Kaushik
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India
| | - Somya Parashar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India
| | - Rashmi K Ambasta
- Department of Biotechnology and Microbiology, SRM University-Sonepat, Haryana, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi 110042, India.
| |
Collapse
|
11
|
Lv B, Fu P, Wang M, Cui L, Bao L, Wang X, Yu L, Zhou C, Zhu M, Wang F, Pang Y, Qi S, Zhang Z, Cui G. DMT1 ubiquitination by Nedd4 protects against ferroptosis after intracerebral hemorrhage. CNS Neurosci Ther 2024; 30:e14685. [PMID: 38634270 PMCID: PMC11024684 DOI: 10.1111/cns.14685] [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: 10/07/2023] [Revised: 01/20/2024] [Accepted: 02/06/2024] [Indexed: 04/19/2024] Open
Abstract
OBJECTIVE Neuronal precursor cells expressed developmentally down-regulated 4 (Nedd4) are believed to play a critical role in promoting the degradation of substrate proteins and are involved in numerous biological processes. However, the role of Nedd4 in intracerebral hemorrhage (ICH) remains unknown. This study aims to investigate the regulatory role of Nedd4 in the ICH model. METHODS Male C57BL/6J mice were induced with ICH. Subsequently, the levels of glutathione peroxidase 4 (GPX4), malondialdehyde (MDA) concentration, iron content, mitochondrial morphology, as well as the expression of divalent metal transporter 1 (DMT1) and Nedd4 were assessed after ICH. Furthermore, the impact of Nedd4 overexpression was evaluated through analyses of hematoma area, ferroptosis, and neurobehavioral function. The mechanism underlying Nedd4-mediated degradation of DMT1 was elecidated using immunoprecipitation (IP) after ICH. RESULTS Upon ICH, the level of DMT1 in the brain increased, but decreased when Nedd4 was overexpressed using Lentivirus, suggesting a negative correlation between Nedd4 and DMT1. Additionally, the degradation of DMT1 was inhibited after ICH. Furthermore, it was found that Nedd4 can interact with and ubiquitinate DMT1 at lysine residues 6, 69, and 277, facilitating the degradation of DMT1. Functional analysis indicated that overexpression of Nedd4 can alleviate ferroptosis and promote recovery following ICH. CONCLUSION The results demonstrated that ferroptosis occurs via the Nedd4/DMT1 pathway during ICH, suggesting it potential as a valuable target to inhibit ferroptosis for the treatment of ICH.
Collapse
Affiliation(s)
- Bingchen Lv
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
- Institute of Stroke Research, Xuzhou Medical UniversityXuzhouChina
| | - Ping Fu
- School of Life Sciences, Nanjing UniversityNanjingChina
| | - Miao Wang
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
- Department of GeriatricsThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| | - Likun Cui
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
- Institute of Stroke Research, Xuzhou Medical UniversityXuzhouChina
| | - Lei Bao
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
- Institute of Stroke Research, Xuzhou Medical UniversityXuzhouChina
| | - Xingzhi Wang
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
- Institute of Stroke Research, Xuzhou Medical UniversityXuzhouChina
| | - Lu Yu
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
- Institute of Stroke Research, Xuzhou Medical UniversityXuzhouChina
| | - Chao Zhou
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
- Institute of Stroke Research, Xuzhou Medical UniversityXuzhouChina
| | - Mengxin Zhu
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
- Institute of Stroke Research, Xuzhou Medical UniversityXuzhouChina
| | - Fei Wang
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
- Institute of Stroke Research, Xuzhou Medical UniversityXuzhouChina
| | - Ye Pang
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
- Institute of Stroke Research, Xuzhou Medical UniversityXuzhouChina
| | - Suhua Qi
- School of Medical Technology, Xuzhou Medical UniversityXuzhouChina
| | - Zuohui Zhang
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
- Institute of Stroke Research, Xuzhou Medical UniversityXuzhouChina
| | - Guiyun Cui
- Department of NeurologyThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
- Institute of Stroke Research, Xuzhou Medical UniversityXuzhouChina
| |
Collapse
|
12
|
Sakurai Y, Kubota N, Takamoto I, Wada N, Aihara M, Hayashi T, Kubota T, Hiraike Y, Sasako T, Nakao H, Aiba A, Chikaoka Y, Kawamura T, Kadowaki T, Yamauchi T. Overexpression of UBE2E2 in Mouse Pancreatic β-Cells Leads to Glucose Intolerance via Reduction of β-Cell Mass. Diabetes 2024; 73:474-489. [PMID: 38064504 DOI: 10.2337/db23-0150] [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: 02/22/2023] [Accepted: 12/03/2023] [Indexed: 02/22/2024]
Abstract
Genome-wide association studies have identified several gene polymorphisms, including UBE2E2, associated with type 2 diabetes. Although UBE2E2 is one of the ubiquitin-conjugating enzymes involved in the process of ubiquitin modifications, the pathophysiological roles of UBE2E2 in metabolic dysfunction are not yet understood. Here, we showed upregulated UBE2E2 expression in the islets of a mouse model of diet-induced obesity. The diabetes risk allele of UBE2E2 (rs13094957) in noncoding regions was associated with upregulation of UBE2E2 mRNA in the human pancreas. Although glucose-stimulated insulin secretion was intact in the isolated islets, pancreatic β-cell-specific UBE2E2-transgenic (TG) mice exhibited reduced insulin secretion and decreased β-cell mass. In TG mice, suppressed proliferation of β-cells before the weaning period and while receiving a high-fat diet was accompanied by elevated gene expression levels of p21, resulting in decreased postnatal β-cell mass expansion and compensatory β-cell hyperplasia, respectively. In TG islets, proteomic analysis identified enhanced formation of various types of polyubiquitin chains, accompanied by increased expression of Nedd4 E3 ubiquitin protein ligase. Ubiquitination assays showed that UBE2E2 mediated the elongation of ubiquitin chains by Nedd4. The data suggest that UBE2E2-mediated ubiquitin modifications in β-cells play an important role in regulating glucose homeostasis and β-cell mass.
Collapse
Affiliation(s)
- Yoshitaka Sakurai
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Naoto Kubota
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
- Department of Metabolic Medicine, Faculty of Life Science, Kumamoto University, Kumamoto, Japan
- Clinical Nutrition Program, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Iseki Takamoto
- Department of Metabolism and Endocrinology, Ibaraki Medical Center, Tokyo Medical University, Tokyo, Japan
| | - Nobuhiro Wada
- Department of Anatomy I, School of Medicine, Sapporo Medical University, Sapporo, Japan
| | - Masakazu Aihara
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Takanori Hayashi
- Clinical Nutrition Program, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Tetsuya Kubota
- Clinical Nutrition Program, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
- Division of Diabetes and Metabolism, Institute of Medical Science, Asahi Life Foundation, Tokyo, Japan
| | - Yuta Hiraike
- Division for Health Service Promotion, The University of Tokyo, Tokyo, Japan
| | - Takayoshi Sasako
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Harumi Nakao
- Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Atsu Aiba
- Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoko Chikaoka
- Isotope Science Center, The University of Tokyo, Tokyo, Japan
| | | | | | - Toshimasa Yamauchi
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| |
Collapse
|
13
|
Bolhuis DL, Emanuele MJ, Brown NG. Friend or foe? Reciprocal regulation between E3 ubiquitin ligases and deubiquitinases. Biochem Soc Trans 2024; 52:241-267. [PMID: 38414432 PMCID: PMC11349938 DOI: 10.1042/bst20230454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/31/2024] [Accepted: 02/06/2024] [Indexed: 02/29/2024]
Abstract
Protein ubiquitination is a post-translational modification that entails the covalent attachment of the small protein ubiquitin (Ub), which acts as a signal to direct protein stability, localization, or interactions. The Ub code is written by a family of enzymes called E3 Ub ligases (∼600 members in humans), which can catalyze the transfer of either a single ubiquitin or the formation of a diverse array of polyubiquitin chains. This code can be edited or erased by a different set of enzymes termed deubiquitinases (DUBs; ∼100 members in humans). While enzymes from these distinct families have seemingly opposing activities, certain E3-DUB pairings can also synergize to regulate vital cellular processes like gene expression, autophagy, innate immunity, and cell proliferation. In this review, we highlight recent studies describing Ub ligase-DUB interactions and focus on their relationships.
Collapse
Affiliation(s)
- Derek L Bolhuis
- Department of Biochemistry and Biophysics, UNC Chapel Hill School of Medicine, Chapel Hill, NC, 27599
| | - Michael J Emanuele
- Department of Pharmacology and Lineberger Comprehensive Care Center, UNC Chapel Hill School of Medicine, Chapel Hill, NC, 27599
| | - Nicholas G Brown
- Department of Pharmacology and Lineberger Comprehensive Care Center, UNC Chapel Hill School of Medicine, Chapel Hill, NC, 27599
| |
Collapse
|
14
|
Li Q, Zhang F, Wang H, Tong Y, Fu Y, Wu K, Li J, Wang C, Wang Z, Jia Y, Chen R, Wu Y, Cui R, Wu Y, Qi Y, Qu K, Liu C, Zhang J. NEDD4 lactylation promotes APAP induced liver injury through Caspase11 dependent non-canonical pyroptosis. Int J Biol Sci 2024; 20:1413-1435. [PMID: 38385085 PMCID: PMC10878146 DOI: 10.7150/ijbs.91284] [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: 10/19/2023] [Accepted: 01/07/2024] [Indexed: 02/23/2024] Open
Abstract
Caspase-11 detection of intracellular lipopolysaccharide mediates non-canonical pyroptosis, which could result in inflammatory damage and organ lesions in various diseases such as sepsis. Our research found that lactate from the microenvironment of acetaminophen-induced acute liver injury increased Caspase-11 levels, enhanced gasdermin D activation and accelerated macrophage pyroptosis, which lead to exacerbation of liver injury. Further experiments unveiled that lactate inhibits Caspase-11 ubiquitination by reducing its binding to NEDD4, a negative regulator of Caspase-11. We also identified that lactates regulated NEDD4 K33 lactylation, which inhibits protein interactions between Caspase-11 and NEDD4. Moreover, restraining lactylation reduces non-canonical pyroptosis in macrophages and ameliorates liver injury. Our work links lactate to the exquisite regulation of the non-canonical inflammasome, and provides a basis for targeting lactylation signaling to combat Caspase-11-mediated non-canonical pyroptosis and acetaminophen-induced liver injury.
Collapse
Affiliation(s)
- Qinglin Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi 710061, People's Republic of China
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, People's Republic of China
- Department of Vascular Surgery, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Fengping Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi 710061, People's Republic of China
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, People's Republic of China
| | - Hai Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi 710061, People's Republic of China
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, People's Republic of China
| | - Yingmu Tong
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, People's Republic of China
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi 710061, People's Republic of China
| | - Yunong Fu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi 710061, People's Republic of China
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, People's Republic of China
| | - Kunjin Wu
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, People's Republic of China
- Department of Hepatobiliary Surgery and Liver Transplantation, The Second Affiliated Hospital of Xi'an Jiaotong University, People's Republic of China
| | - Jing Li
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, People's Republic of China
- Department of Hepatobiliary Surgery and Liver Transplantation, The Second Affiliated Hospital of Xi'an Jiaotong University, People's Republic of China
| | - Cong Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi 710061, People's Republic of China
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, People's Republic of China
| | - Zi Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi 710061, People's Republic of China
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, People's Republic of China
| | - Yifan Jia
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, People's Republic of China
- Department of Vascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi 710061, People's Republic of China
| | - Rui Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi 710061, People's Republic of China
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, People's Republic of China
| | - Yang Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi 710061, People's Republic of China
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, People's Republic of China
| | - Ruixia Cui
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi 710061, People's Republic of China
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, People's Republic of China
| | - Yi Wu
- MOE Key Laboratory of Environment and Genes Related to Diseases, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an Shaanxi 710061, People's Republic of China
| | - Yun Qi
- Department of Ophthalmology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi 710061, People's Republic of China
| | - Kai Qu
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, People's Republic of China
- Department of Hepatobiliary Surgery and Liver Transplantation, The Second Affiliated Hospital of Xi'an Jiaotong University, People's Republic of China
| | - Chang Liu
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, People's Republic of China
- Department of Hepatobiliary Surgery and Liver Transplantation, The Second Affiliated Hospital of Xi'an Jiaotong University, People's Republic of China
| | - Jingyao Zhang
- Key Laboratory of Surgical Critical Care and Life Support (Xi'an Jiaotong University), Ministry of Education, People's Republic of China
- Department of SICU, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Shaanxi 710061, People's Republic of China
| |
Collapse
|
15
|
Chandrasekaran K, Najimi N, Sagi AR, Yarlagadda S, Salimian M, Arvas MI, Hedayat AF, Kevas Y, Kadakia A, Kristian T, Russell JW. NAD + Precursors Reverse Experimental Diabetic Neuropathy in Mice. Int J Mol Sci 2024; 25:1102. [PMID: 38256175 PMCID: PMC10816262 DOI: 10.3390/ijms25021102] [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: 11/16/2023] [Revised: 01/05/2024] [Accepted: 01/13/2024] [Indexed: 01/24/2024] Open
Abstract
Abnormal NAD+ signaling has been implicated in axonal degeneration in diabetic peripheral neuropathy (DPN). We hypothesized that supplementing NAD+ precursors could alleviate DPN symptoms through increasing the NAD+ levels and activating the sirtuin-1 (SIRT1) protein. To test this, we exposed cultured Dorsal Root Ganglion neurons (DRGs) to Nicotinamide Riboside (NR) or Nicotinamide Mononucleotide (NMN), which increased the levels of NAD+, the SIRT1 protein, and the deacetylation activity that is associated with increased neurite growth. A SIRT1 inhibitor blocked the neurite growth induced via NR or NMN. We then induced neuropathy in C57BL6 mice with streptozotocin (STZ) or a high fat diet (HFD) and administered NR or NMN for two months. Both the STZ and HFD mice developed neuropathy, which was reversed through the NR or NMN administration: sensory function improved, nerve conduction velocities normalized, and intraepidermal nerve fibers were restored. The NAD+ levels and SIRT1 activity were reduced in the DRGs from diabetic mice but were preserved with the NR or NMN treatment. We also tested the effect of NR or NMN administration in mice that overexpress the SIRT1 protein in neurons (nSIRT1 OE) and found no additional benefit from the addition of the drug. These findings suggest that supplementing with NAD+ precursors or activating SIRT1 may be a promising treatment for DPN.
Collapse
Affiliation(s)
- Krish Chandrasekaran
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (S.Y.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Neda Najimi
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (S.Y.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Avinash R. Sagi
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (S.Y.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Sushuma Yarlagadda
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (S.Y.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Mohammad Salimian
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (S.Y.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Muhammed Ikbal Arvas
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (S.Y.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Ahmad F. Hedayat
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (S.Y.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Yanni Kevas
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (S.Y.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Anand Kadakia
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (S.Y.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
| | - Tibor Kristian
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
- Veterans Affairs Medical Center, Baltimore, MD 21201, USA
| | - James W. Russell
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.C.); (N.N.); (S.Y.); (M.I.A.); (A.F.H.); (Y.K.); (A.K.)
- Veterans Affairs Medical Center, Baltimore, MD 21201, USA
- CAMC Institute for Academic Medicine, 415 Morris Street Suite 300, Charleston, WV 25301, USA
| |
Collapse
|
16
|
Miyauchi S, Arimoto KI, Liu M, Zhang Y, Zhang DE. Reprogramming of tumor-associated macrophages via NEDD4-mediated CSF1R degradation by targeting USP18. Cell Rep 2023; 42:113560. [PMID: 38100351 PMCID: PMC10822669 DOI: 10.1016/j.celrep.2023.113560] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 07/25/2023] [Accepted: 11/23/2023] [Indexed: 12/17/2023] Open
Abstract
Tumor-associated myeloid cells modulate the tumor microenvironment and affect tumor progression. Type I interferon (IFN-I) has multiple effects on tumors and immune response, and ubiquitin-specific peptidase 18 (USP18) functions as a negative regulator of IFN-I signal transduction. This study aims to examine the function of IFN-I in myeloid cells during tumor progression. Here, we show that deletion of USP18 in myeloid cells suppresses tumor progression. Enhanced IFN-I signaling and blocked USP18 expression prompt downregulation of colony stimulating factor 1 receptor (CSF1R) and polarization of tumor-associated macrophages toward pro-inflammatory phenotypes. Further in vitro experiments reveal that downregulation of CSF1R is mediated by ubiquitin-proteasome degradation via E3 ligase neural precursor cell-expressed, developmentaly downregulated 4 (NEDD4) and the IFN-induced increase in ubiquitin E2 ubiquitin-conjugating enzyme H5. USP18 impairs ubiquitination and subsequent degradation of CSF1R by interrupting NEDD4 binding to CSF1R. These results reveal a previously unappreciated role of IFN-I in macrophage polarization by regulating CSF1R via USP18 and suggest targeting USP18 in myeloid-lineage cells as an effective strategy for IFN-based therapies.
Collapse
Affiliation(s)
- Sayuri Miyauchi
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92037, USA
| | - Kei-Ichiro Arimoto
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92037, USA
| | - Mengdan Liu
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92037, USA; School of Biological Sciences, University of California San Diego, La Jolla, CA 92037, USA
| | - Yue Zhang
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92037, USA; School of Biological Sciences, University of California San Diego, La Jolla, CA 92037, USA
| | - Dong-Er Zhang
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92037, USA; School of Biological Sciences, University of California San Diego, La Jolla, CA 92037, USA; Department of Pathology, University of California San Diego, La Jolla, CA 92037, USA.
| |
Collapse
|
17
|
Anand S, Nedeva C, Chitti SV, Fonseka P, Kang T, Gangoda L, Tabassum NI, Abdirahman S, Arumugam TV, Putoczki TL, Kumar S, Mathivanan S. The E3 ubiquitin ligase NEDD4 regulates chemoresistance to 5-fluorouracil in colorectal cancer cells by altering JNK signalling. Cell Death Dis 2023; 14:828. [PMID: 38097550 PMCID: PMC10721789 DOI: 10.1038/s41419-023-06349-z] [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: 06/05/2023] [Revised: 11/12/2023] [Accepted: 11/29/2023] [Indexed: 12/17/2023]
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer deaths. Though chemotherapy is the main treatment option for advanced CRC, patients invariably acquire resistance to chemotherapeutic drugs and fail to respond to the therapy. Although understanding the mechanisms regulating chemoresistance has been a focus of intense research to manage this challenge, the pathways governing resistance to drugs are poorly understood. In this study, we provide evidence for the role of ubiquitin ligase NEDD4 in resistance developed against the most commonly used CRC chemotherapeutic drug 5-fluorouracil (5-FU). A marked reduction in NEDD4 protein abundance was observed in a panel of CRC cell lines and patient-derived xenograft samples that were resistant to 5-FU. Knockout of NEDD4 in CRC cells protected them from 5-FU-mediated apoptosis but not oxaliplatin or irinotecan. Furthermore, NEDD4 depletion in CRC cells reduced proliferation, colony-forming abilities and tumour growth in mice. Follow-up biochemical analysis highlighted the inhibition of the JNK signalling pathway in NEDD4-deficient cells. Treatment with the JNK activator hesperidin in NEDD4 knockout cells sensitised the CRC cells against 5-FU. Overall, we show that NEDD4 regulates cell proliferation, colony formation, tumour growth and 5-FU chemoresistance in CRC cells.
Collapse
Affiliation(s)
- Sushma Anand
- Department of Biochemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Christina Nedeva
- Department of Biochemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Sai V Chitti
- Department of Biochemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Pamali Fonseka
- Department of Biochemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Taeyoung Kang
- Department of Biochemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Lahiru Gangoda
- Department of Biochemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Nishat I Tabassum
- Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, Centre for Cardiovascular Biology and Disease Research, La Trobe University, Melbourne, Australia
| | - Suad Abdirahman
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, 3052, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3052, Australia
| | - Thiruma V Arumugam
- Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, Centre for Cardiovascular Biology and Disease Research, La Trobe University, Melbourne, Australia
| | - Tracy L Putoczki
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia, Adelaide, SA, 5001, Australia
| | - Suresh Mathivanan
- Department of Biochemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia.
| |
Collapse
|
18
|
Hu S, Zhu Y, Zhao X, Li R, Shao G, Gong D, Hu C, Liu H, Xu K, Liu C, Xu M, Zhao Z, Li T, Hu Z, Shao M, Liu J, Li X, Wu H, Li J, Xu Y. Hepatocytic lipocalin-2 controls HDL metabolism and atherosclerosis via Nedd4-1-SR-BI axis in mice. Dev Cell 2023; 58:2326-2337.e5. [PMID: 37863040 DOI: 10.1016/j.devcel.2023.09.007] [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: 02/28/2023] [Revised: 07/03/2023] [Accepted: 09/25/2023] [Indexed: 10/22/2023]
Abstract
High-density lipoprotein (HDL) metabolism is regulated by complex interplay between the scavenger receptor group B type 1 (SR-BI) and multiple signaling molecules in the liver. Here, we show that lipocalin-2 (Lcn2) is a key regulator of hepatic SR-BI, HDL metabolism, and atherosclerosis. Overexpression of human Lcn2 in hepatocytes attenuates the development of atherosclerosis via SR-BI in western-diet-fed Ldlr-/- mice, whereas hepatocyte-specific ablation of Lcn2 has the opposite effect. Mechanistically, hepatocyte Lcn2 improves HDL metabolism and alleviates atherogenesis by blocking Nedd4-1-mediated SR-BI ubiquitination at K500 and K508. The Lcn2-improved HDL metabolism is abolished in mice with hepatocyte-specific Nedd4-1 or SR-BI deletion and in SR-BI (K500A/K508A) mutation mice. This study identifies a regulatory axis from Lcn2 to HDL via blocking Nedd4-1-mediated SR-BI ubiquitination and demonstrates that hepatocyte Lcn2 may be a promising target to improve HDL metabolism to treat atherosclerotic cardiovascular diseases.
Collapse
Affiliation(s)
- Shuwei Hu
- School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China; Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Yingdong Zhu
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Xiaojie Zhao
- Department of Pathology, School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China
| | - Rui Li
- Department of Pathology, School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China
| | - Guangze Shao
- Department of Pathology, School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China
| | - Dongxu Gong
- Department of Pathology, School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China
| | - Chencheng Hu
- Department of Pathology, School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China
| | - Hongjun Liu
- Department of Pathology, School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China
| | - Kexin Xu
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Chenxi Liu
- Department of Pathology, School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China
| | - Minghuan Xu
- Department of Pathology, School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China
| | - Zhonghua Zhao
- Department of Pathology, School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China
| | - Tao Li
- Department of Hepatobiliary Surgery, Peking University People's Hospital Xizhimen South Street, West District, Beijing 100044, China
| | - Zhigang Hu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Mengle Shao
- Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jun- Liu
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Xinwei Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Huijuan Wu
- Department of Pathology, School of Basic Medical Sciences, Fudan University Shanghai, Shanghai 200032, China
| | - Jing Li
- Department of Endocrinology, Beijing Chao-yang Hospital, Capital Medical University, Beijing 100020, China.
| | - Yanyong Xu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Pathology of School of Basic Medical Sciences, Frontier Innovation Center, Fudan University Shanghai, Shanghai 200032, China; Diabetes, Obesity and Metabolism Research Center, Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA.
| |
Collapse
|
19
|
Yong D, Green SR, Ghiabi P, Santhakumar V, Vedadi M. Discovery of Nedd4 auto-ubiquitination inhibitors. Sci Rep 2023; 13:16057. [PMID: 37749144 PMCID: PMC10520017 DOI: 10.1038/s41598-023-42997-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 09/18/2023] [Indexed: 09/27/2023] Open
Abstract
E3 ubiquitin ligases are critical to the protein degradation pathway by catalyzing the final step in protein ubiquitination by mediating ubiquitin transfer from E2 enzymes to target proteins. Nedd4 is a HECT domain-containing E3 ubiquitin ligase with a wide range of protein targets, the dysregulation of which has been implicated in myriad pathologies, including cancer and Parkinson's disease. Towards the discovery of compounds disrupting the auto-ubiquitination activity of Nedd4, we developed and optimized a TR-FRET assay for high-throughput screening. Through selective screening of a library of potentially covalent compounds, compounds 25 and 81 demonstrated apparent IC50 values of 52 µM and 31 µM, respectively. Tandem mass spectrometry (MS/MS) analysis confirmed that 25 and 81 were covalently bound to Nedd4 cysteine residues (Cys182 and Cys867). In addition, 81 also adducted to Cys627. Auto-ubiquitination assays of Nedd4 mutants featuring alanine substitutions for each of these cysteines suggested that the mode of inhibition of these compounds occurs through blocking the catalytic Cys867. The discovery of these inhibitors could enable the development of therapeutics for various diseases caused by Nedd4 E3 ligase dysregulation.
Collapse
Affiliation(s)
- Darren Yong
- Structural Genomics Consortium, University of Toronto, Toronto, ON, M5G 1L7, Canada
| | - Stuart R Green
- Structural Genomics Consortium, University of Toronto, Toronto, ON, M5G 1L7, Canada
| | - Pegah Ghiabi
- Structural Genomics Consortium, University of Toronto, Toronto, ON, M5G 1L7, Canada
| | | | - Masoud Vedadi
- Structural Genomics Consortium, University of Toronto, Toronto, ON, M5G 1L7, Canada.
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, M5S 1A8, Canada.
- Drug Discovery Program, Ontario Institute for Cancer Research, Toronto, ON, M5G 0A3, Canada.
| |
Collapse
|
20
|
Datkhayev UM, Rakhmetova V, Shepetov AM, Kodasbayev A, Datkayeva GM, Pazilov SB, Farooqi AA. Unraveling the Complex Web of Mechanistic Regulation of Versatile NEDD4 Family by Non-Coding RNAs in Carcinogenesis and Metastasis: From Cell Culture Studies to Animal Models. Cancers (Basel) 2023; 15:3971. [PMID: 37568787 PMCID: PMC10417118 DOI: 10.3390/cancers15153971] [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: 04/23/2023] [Revised: 07/03/2023] [Accepted: 07/07/2023] [Indexed: 08/13/2023] Open
Abstract
Discoveries related to an intriguing feature of ubiquitination have prompted a detailed analysis of the ubiquitination patterns in malignant cells. How the "ubiquitinome" is reshaped during multistage carcinogenesis has garnered significant attention. Seminal studies related to the structural and functional characterization of NEDD4 (Neuronal precursor cell-expressed developmentally downregulated-4) have consolidated our understanding at a new level of maturity. Additionally, regulatory roles of non-coding RNAs have further complicated the complex interplay between non-coding RNAs and the members of NEDD4 family. These mechanisms range from the miRNA-mediated targeting of NEDD4 family members to the regulation of transcriptional factors for a broader range of non-coding RNAs. Additionally, the NEDD4-mediated degradation of different proteins is modulated by lncRNAs and circRNAs. The miRNA-mediated targeting of NEDD4 family members is also regulated by circRNAs. Tremendous advancements have been made in the identification of different substrates of NEDD4 family and in the comprehensive analysis of the molecular mechanisms by which various members of NEDD4 family catalyze the ubiquitination of substrates. In this review, we have attempted to summarize the multifunctional roles of the NEDD4 family in cancer biology, and how different non-coding RNAs modulate these NEDD4 family members in the regulation of cancer. Future molecular studies should focus on the investigation of a broader drug design space and expand the scope of accessible targets for the inhibition/prevention of metastasis.
Collapse
Affiliation(s)
- Ubaidilla M. Datkhayev
- Asfendiyarov Kazakh National Medical University, Tole Bi St 94, Almaty 050000, Kazakhstan
| | | | - Abay M. Shepetov
- Department of Nephrology, Asfendiyarov Kazakh National Medical University, Tole Bi St 94, Almaty 050000, Kazakhstan;
| | - Almat Kodasbayev
- Department of Cardiovascular Surgery, Asfendiyarov Kazakh National Medical University, Tole Bi St 94, Almaty 050000, Kazakhstan
| | | | - Sabit B. Pazilov
- Department of Healthcare of Kyzylorda Region, Kyzylorda, Abay Avenue, 27, Kyzylorda 120008, Kazakhstan;
| | - Ammad Ahmad Farooqi
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad 44000, Pakistan
| |
Collapse
|
21
|
Cao L, Li H, Liu X, Wang Y, Zheng B, Xing C, Zhang N, Liu J. Expression and regulatory network of E3 ubiquitin ligase NEDD4 family in cancers. BMC Cancer 2023; 23:526. [PMID: 37291499 DOI: 10.1186/s12885-023-11007-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 05/25/2023] [Indexed: 06/10/2023] Open
Abstract
NEDD4 family represent an important group of E3 ligases, which regulate various cellular pathways of cell proliferation, cell junction and inflammation. Emerging evidence suggested that NEDD4 family members participate in the initiation and development of tumor. In this study, we systematically investigated the molecular alterations as well as the clinical relevance regarding NEDD4 family genes in 33 cancer types. Finally, we found that NEDD4 members showed increased expression in pancreas cancer and decreased expression in thyroid cancer. NEDD4 E3 ligase family genes had an average mutation frequency in the range of 0-32.1%, of which HECW1 and HECW2 demonstrated relatively high mutation rate. Breast cancer harbors large amount of NEDD4 copy number amplification. NEDD4 family members interacted proteins were enriched in various pathways including p53, Akt, apoptosis and autophagy, which were confirmed by further western blot and flow cytometric analysis in A549 and H1299 lung cancer cells. In addition, expression of NEDD4 family genes were associated with survival of cancer patients. Our findings provide novel insight into the effect of NEDD4 E3 ligase genes on cancer progression and treatment in the future.
Collapse
Affiliation(s)
- Liangzi Cao
- Department of Anus and Intestine Surgery, the First Affiliated Hospital of China Medical University, 155# North Nanjing Street, Heping District, Shenyang City, 110001, Liaoning Province, China
| | - Hao Li
- Department of Clinical Laboratory, The First Hospital of China Medical University, Shenyang, China
| | - Xiaofang Liu
- Department of Anus and Intestine Surgery, the First Affiliated Hospital of China Medical University, 155# North Nanjing Street, Heping District, Shenyang City, 110001, Liaoning Province, China
| | - Yubang Wang
- Department of Anus and Intestine Surgery, the First Affiliated Hospital of China Medical University, 155# North Nanjing Street, Heping District, Shenyang City, 110001, Liaoning Province, China
| | - Bowen Zheng
- Department of Anus and Intestine Surgery, the First Affiliated Hospital of China Medical University, 155# North Nanjing Street, Heping District, Shenyang City, 110001, Liaoning Province, China
| | - Chengzhong Xing
- Department of Anus and Intestine Surgery, the First Affiliated Hospital of China Medical University, 155# North Nanjing Street, Heping District, Shenyang City, 110001, Liaoning Province, China.
| | - Naijin Zhang
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Jingwei Liu
- Department of Anus and Intestine Surgery, the First Affiliated Hospital of China Medical University, 155# North Nanjing Street, Heping District, Shenyang City, 110001, Liaoning Province, China.
| |
Collapse
|
22
|
Almeida Lima K, Osawa IYA, Ramalho MCC, de Souza I, Guedes CB, Souza Filho CHDD, Monteiro LKS, Latancia MT, Rocha CRR. Temozolomide Resistance in Glioblastoma by NRF2: Protecting the Evil. Biomedicines 2023; 11:biomedicines11041081. [PMID: 37189700 DOI: 10.3390/biomedicines11041081] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/26/2023] [Accepted: 03/29/2023] [Indexed: 04/07/2023] Open
Abstract
The transcription factor NRF2 is constitutively active in glioblastoma, a highly aggressive brain tumor subtype with poor prognosis. Temozolomide (TMZ) is the primary chemotherapeutic agent for this type of tumor treatment, but resistance to this drug is often observed. This review highlights the research that is demonstrating how NRF2 hyperactivation creates an environment that favors the survival of malignant cells and protects against oxidative stress and TMZ. Mechanistically, NRF2 increases drug detoxification, autophagy, DNA repair, and decreases drug accumulation and apoptotic signaling. Our review also presents potential strategies for targeting NRF2 as an adjuvant therapy to overcome TMZ chemoresistance in glioblastoma. Specific molecular pathways, including MAPKs, GSK3β, βTRCP, PI3K, AKT, and GBP, that modulate NRF2 expression leading to TMZ resistance are discussed, along with the importance of identifying NRF2 modulators to reverse TMZ resistance and develop new therapeutic targets. Despite the significant progress in understanding the role of NRF2 in GBM, there are still unanswered questions regarding its regulation and downstream effects. Future research should focus on elucidating the precise mechanisms by which NRF2 mediates resistance to TMZ, and identifying potential novel targets for therapeutic intervention.
Collapse
Affiliation(s)
- Karoline Almeida Lima
- Department of Clinical and Experimental Oncology, Federal University of Sao Paulo (UNIFESP), Sao Paulo 04037-003, Brazil
| | - Isabeli Yumi Araújo Osawa
- Department of Clinical and Experimental Oncology, Federal University of Sao Paulo (UNIFESP), Sao Paulo 04037-003, Brazil
| | - Maria Carolina Clares Ramalho
- Department of Clinical and Experimental Oncology, Federal University of Sao Paulo (UNIFESP), Sao Paulo 04037-003, Brazil
| | - Izadora de Souza
- Department of Clinical and Experimental Oncology, Federal University of Sao Paulo (UNIFESP), Sao Paulo 04037-003, Brazil
| | - Camila Banca Guedes
- Department of Clinical and Experimental Oncology, Federal University of Sao Paulo (UNIFESP), Sao Paulo 04037-003, Brazil
| | | | | | - Marcela Teatin Latancia
- Laboratory of Genomic Integrity, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-3371, USA
| | - Clarissa Ribeiro Reily Rocha
- Department of Clinical and Experimental Oncology, Federal University of Sao Paulo (UNIFESP), Sao Paulo 04037-003, Brazil
| |
Collapse
|
23
|
Saito M, Tokunaga N, Saito T, Hatakenaka T, Sasaki T, Matsuki N, Minagawa S. Connexin 45 is a novel suppressor of melanoma metastasis. Cytotechnology 2023; 75:103-113. [PMID: 36969569 PMCID: PMC10030756 DOI: 10.1007/s10616-022-00563-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
The expression spectra of connexin (Cx) isoforms were investigated in three mouse melanoma cell lines: B16-F1 (F1), B16-F10 (F10), and B16-BL6 (BL6). Metastatic potential intensity was higher in the order of F1, F10, and BL6. A remarkable behavior of Cx45 was found among 20 isoforms. The expression level of Cx45 was highest in F1 and lowest in BL6. It was inductively predicted that Cx45 might be a novel suppressor of metastasis. A Cx45-overexpressing BL6 cell line (Cx45 +BL6) was developed and its properties were compared with those of a wild-type cell line of BL6 (W-BL6). Compared to W-BL6, Cx45 +BL6 showed reduced wound healing, Transwell® permeability, and matrix metalloproteinase 9 expression, suggesting the suppression of cellular migration and invasion. The expression of E-cadherin and integrin β1 in Cx45 +BL6 was also lower than in W-BL6, suggesting reduced cell adhesion. The decrease in cell adhesion was supported by the cell washing-out assay. In contrast, no difference between W-BL6 and Cx45 +BL6 was observed in cell proliferation, suggesting no effect on cell-cycle regulating factors. Finally, an in vivo assay revealed a significant decrease in the number of metastatic colonies of Cx45 +BL6 (176 ± 25/lung) in comparison with those of W-BL6 (252 ± 23/lung) in a mouse model. In conclusion, Cx45 is a novel suppressor of melanoma metastasis. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-022-00563-x.
Collapse
Affiliation(s)
- Mikako Saito
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, 184-8588 Tokyo, Japan
| | - Naruwa Tokunaga
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, 184-8588 Tokyo, Japan
| | - Toshiki Saito
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, 184-8588 Tokyo, Japan
| | - Tomohiro Hatakenaka
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, 184-8588 Tokyo, Japan
| | - Tomonori Sasaki
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, 184-8588 Tokyo, Japan
| | - Nahoko Matsuki
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, 184-8588 Tokyo, Japan
| | - Seiya Minagawa
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, 184-8588 Tokyo, Japan
| |
Collapse
|
24
|
Zhang R, Shi S. The role of NEDD4 related HECT-type E3 ubiquitin ligases in defective autophagy in cancer cells: molecular mechanisms and therapeutic perspectives. Mol Med 2023; 29:34. [PMID: 36918822 PMCID: PMC10015828 DOI: 10.1186/s10020-023-00628-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 02/21/2023] [Indexed: 03/15/2023] Open
Abstract
The homologous to the E6-AP carboxyl terminus (HECT)-type E3 ubiquitin ligases are the selective executers in the protein ubiquitination, playing a vital role in modulation of the protein function and stability. Evidence shows the regulatory role of HECT-type E3 ligases in various steps of the autophagic process. Autophagy is an intracellular digestive and recycling process that controls the cellular hemostasis. Defective autophagy is involved in tumorigenesis and has been detected in various types of cancer cells. A growing body of findings indicates that HECT-type E3 ligases, in particular members of the neural precursor cell expressed developmentally downregulated protein 4 (NEDD4) including NEDD4-1, NEDD4-L, SMURFs, WWPs, and ITCH, play critical roles in dysregulation or dysfunction of autophagy in cancer cells. The present review focuses on NEDD4 E3 ligases involved in defective autophagy in cancer cells and discusses their autophagic function in different cancer cells as well as substrates and the signaling pathways in which they participate, conferring a basis for the cancer treatment through the modulating of these E3 ligases.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Thoracic Surgery, The Seventh People's Hospital of Chengdu, Chengdu, 610021, Sichuan, People's Republic of China
| | - Shaoqing Shi
- Scientific Research Laboratory Center, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, People's Republic of China.
| |
Collapse
|
25
|
Michaelides IN, Collie GW. E3 Ligases Meet Their Match: Fragment-Based Approaches to Discover New E3 Ligands and to Unravel E3 Biology. J Med Chem 2023; 66:3173-3194. [PMID: 36821822 PMCID: PMC10009759 DOI: 10.1021/acs.jmedchem.2c01882] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Indexed: 02/25/2023]
Abstract
Ubiquitination is a key post-translational modification of proteins, affecting the regulation of multiple cellular processes. Cells are equipped with over 600 ubiquitin orchestrators, called E3 ubiquitin ligases, responsible for directing the covalent attachment of ubiquitin to substrate proteins. Due to their regulatory role in cells, significant efforts have been made to discover ligands for E3 ligases. The recent emergence of the proteolysis targeting chimera (PROTAC) and molecular glue degrader (MGD) modalities has further increased interest in E3 ligases as drug targets. This perspective focuses on how fragment based lead discovery (FBLD) methods have been used to discover new ligands for this important target class. In some cases these efforts have led to clinical candidates; in others, they have provided tools for deepening our understanding of E3 ligase biology. Recently, FBLD-derived ligands have inspired the design of PROTACs that are able to artificially modulate protein levels in cells.
Collapse
Affiliation(s)
- Iacovos N. Michaelides
- Discovery Sciences, BioPharmaceuticals
R&D, AstraZeneca, Cambridge, CB4 0WG, United
Kingdom
| | - Gavin W. Collie
- Discovery Sciences, BioPharmaceuticals
R&D, AstraZeneca, Cambridge, CB4 0WG, United
Kingdom
| |
Collapse
|
26
|
Canever JB, Soares ES, de Avelar NCP, Cimarosti HI. Targeting α-synuclein post-translational modifications in Parkinson's disease. Behav Brain Res 2023; 439:114204. [PMID: 36372243 DOI: 10.1016/j.bbr.2022.114204] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 10/25/2022] [Accepted: 11/04/2022] [Indexed: 11/13/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterized by the progressive loss of dopaminergic neurons in the nigrostriatal pathway. Although the exact mechanisms underlying PD are still not completely understood, it is well accepted that α-synuclein plays key pathophysiological roles as the main constituent of the cytoplasmic inclusions known as Lewy bodies. Several post-translational modifications (PTMs), such as the best-known phosphorylation, target α-synuclein and are thus implicated in its physiological and pathological functions. In this review, we present (1) an overview of the pathophysiological roles of α-synuclein, (2) a descriptive analysis of α-synuclein PTMs, including phosphorylation, ubiquitination, SUMOylation, acetylation, glycation, truncation, and O-GlcNAcylation, as well as (3) a brief summary on α-synuclein PTMs as potential biomarkers for PD. A better understanding of α-synuclein PTMs is of paramount importance for elucidating the mechanisms underlying PD and can thus be expected to improve early detection and monitoring disease progression, as well as identify promising new therapeutic targets.
Collapse
Affiliation(s)
- Jaquelini B Canever
- Post-Graduate Program in Neuroscience, Federal University of Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil; Laboratory of Aging, Resources and Rheumatology, UFSC, Araranguá, Santa Catarina, Brazil
| | - Ericks Sousa Soares
- Post-Graduate Program in Pharmacology, UFSC, Florianópolis, Santa Catarina, Brazil
| | - Núbia C P de Avelar
- Laboratory of Aging, Resources and Rheumatology, UFSC, Araranguá, Santa Catarina, Brazil
| | - Helena I Cimarosti
- Post-Graduate Program in Neuroscience, Federal University of Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil; Post-Graduate Program in Pharmacology, UFSC, Florianópolis, Santa Catarina, Brazil.
| |
Collapse
|
27
|
Sharma S, Kumar P. Decoding the Role of MDM2 as a Potential Ubiquitin E3 Ligase and Identifying the Therapeutic Efficiency of Alkaloids against MDM2 in Combating Glioblastoma. ACS OMEGA 2023; 8:5072-5087. [PMID: 36777618 PMCID: PMC9910072 DOI: 10.1021/acsomega.2c07904] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/17/2023] [Indexed: 05/28/2023]
Abstract
Glioblastomas (GBMs) represent the most aggressive form of brain tumor arising from the malignant transformation of astrocytes. Despite various advancements, treatment options remain limited to chemotherapy and radiotherapy followed by surgery giving an overall survival of 14-15 months. These therapies are somewhere restricted in giving a better survival and cure. There is a need for new therapeutics that could potentially target GBM based on molecular pathways and pathology. Here, ubiquitin E3 ligases can be used as targets as they bind a wide array of substrates and therefore can be attractive targets for new inhibitors. Through this study, we have tried to sort various ubiquitin E3 ligases based on their expression, pathways to which these ligases are associated, and mutational frequencies, and then we tried to screen potent inhibitors against the most favorable E3 ligase as very few studies are available concerning inhibition of E3 ligase in GBM. Our study found MDM2 to be the most ideal E3 ligase and further we tried to target MDM2 against various compounds under the alkaloid class. Molecular Docking and MD simulations combined with ADMET properties and BBB scores revealed that only evodiamine and sanguinarine were effective in inhibiting MDM2. We also tried to give a proposed mechanism of how these inhibitors mediate the p53 signaling in GBM. Therefore, the new scaffolds predicted by the computational approach could help in designing promising therapeutic agents targeting MDM2 in glioblastoma.
Collapse
|
28
|
E3 Ubiquitin Ligase NEDD4 Affects Estrogen Receptor α Expression and the Prognosis of Patients with Hormone Receptor-Positive Breast Cancer. Cancers (Basel) 2023; 15:cancers15020539. [PMID: 36672488 PMCID: PMC9857178 DOI: 10.3390/cancers15020539] [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: 11/18/2022] [Revised: 01/09/2023] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
Neural precursor cell-expressed developmentally downregulated 4-1 (NEDD4) is an E3 ligase that leads to the degradation of proteins, including estrogen receptor α. We evaluated whether the expression level of NEDD4 affected the outcome of breast cancer patients. We performed a retrospective cohort study enrolling 143 patients with hormone receptor-positive, human epidermal growth factor receptor 2-negative early breast cancer. Of the 66 patients with high NEDD4 mRNA levels (high NEDD4 group) and 77 patients with low NEDD4 mRNA levels (low NEDD4 group), 98.4% and 96.1%, respectively, of the patients had received neoadjuvant/adjuvant hormone therapy. Disease-free survival and overall survival were significantly longer in the low NEDD4 group than in the high NEDD4 group (p = 0.048 and p = 0.022, respectively). Western blotting revealed a high expression of estrogen receptor α in the NEDD4-knockdown culture cells. The proliferation of NEDD4-knockdown cells treated with tamoxifen or estradiol deprivation was suppressed, compared with that of NEDD4-expressing cells. Knockdown of NEDD4 in breast cancer cells induced the accumulation of estrogen receptor α and increased sensitivity to hormone therapy. In summary, this mechanism may lead to a better prognosis in hormone receptor-positive breast cancer patients with a low expression of NEDD4.
Collapse
|
29
|
Sun Y, He P, Li L, Ding X. The significance of the crosstalk between ubiquitination or deubiquitination and ncRNAs in non-small cell lung cancer. Front Oncol 2023; 12:969032. [PMID: 36727069 PMCID: PMC9884829 DOI: 10.3389/fonc.2022.969032] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 12/28/2022] [Indexed: 01/18/2023] Open
Abstract
Lung cancer (LC) remains the leading cause of cancer-related deaths worldwide, with extremely high morbidity and mortality rates. Non-small cell lung cancer (NSCLC) is the most critical type of LC. It seriously threatens the life and health of patients because of its early metastasis, late clinical symptoms, limited early screening methods, and poor treatment outcomes. Non-coding RNAs (ncRNAs), including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), participate in cell proliferation, metastasis, and chemoresistance. Several previous studies have proven that ncRNAs are vital regulators of tumorigenesis. Ubiquitination plays the most crucial role in protein post-translational modification (PTM). Deubiquitination and ubiquitination form a homeostasis. In summary, ubiquitination and deubiquitination play essential roles in mediating the degradation or overexpression of a range of crucial proteins in various cancers. A growing number of researchers have found that interactions between ncRNAs and ubiquitination (or deubiquitination) play a crucial role in NSCLC. This review presents several typical examples of the important effects of ncRNAs and ubiquitination (or deubiquitination) in NSCLC, aiming to provide more creative ideas for exploring the diagnosis and treatment of NSCLC.
Collapse
Affiliation(s)
- Yiyang Sun
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ping He
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, Shenyang, China,*Correspondence: Ping He,
| | - Li Li
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xue Ding
- General Medicine Department, Dalian Friendship Hospital, Dalian, China
| |
Collapse
|
30
|
Vergara IA, Aivazian K, Carlino MS, Guminski AD, Maher NG, Shannon KF, Ch'ng S, Saw RPM, Long GV, Wilmott JS, Scolyer RA. Genomic Profiling of Metastatic Basal cell Carcinoma Reveals Candidate Drivers of Disease and Therapeutic Targets. Mod Pathol 2023; 36:100099. [PMID: 36788083 DOI: 10.1016/j.modpat.2023.100099] [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: 10/26/2022] [Revised: 11/30/2022] [Accepted: 12/26/2022] [Indexed: 01/11/2023]
Abstract
Basal cell carcinomas (BCCs) are human beings' most common malignant tumors. Most are easily managed by surgery or topical therapies, and metastasis is rare. Although BCCs can become locally advanced, metastatic BCCs are very uncommon and may be biologically distinct. We assessed the clinicopathologic characteristics of 17 patients with metastatic BCC and pursued whole-exome sequencing of tumor and germline DNA from 8 patients. Genomic profiling revealed aberrant activation of Hedgehog signaling and alterations in GLI transcriptional regulators and Notch and Hippo signaling. Matched local recurrences of primary BCCs and metastases from 3 patients provided evidence of a clonal origin in all cases. Mutations associated with YAP inhibition were found exclusively in 2 hematogenously-spread lung metastases, and metastatic BCCs were enriched for mutations in the YAP/TAZ-binding domain of TEAD genes. Accordingly, YAP/TAZ nuclear localization was associated with metastatic types and Hippo mutations, suggesting an enhanced oncogenic role in hematogenously-spread metastases. Mutations in RET, HGF, and phosphatidylinositol 3‑kinase (PI3K)/protein kinase B (AKT) signaling were enriched compared with a cohort of low clinical-risk BCCs. Our results implicate Hippo and PI3K/AKT dysregulation in metastatic progression of BCCs, making these potential therapeutic targets in metastatic disease. The common clonal origin of matched recurrent and metastatic BCCs suggests that molecular profiling can assist in determining the nature/origin of poorly differentiated metastatic tumors of uncertain type. Genes and pathways enriched for mutations in this cohort are candidate drivers of metastasis and can be used to identify patients at high risk of metastasis who may benefit from aggressive local treatment and careful clinical follow-up.
Collapse
Affiliation(s)
- Ismael A Vergara
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia; Charles Perkin Centre, The University of Sydney, Sydney, NSW, Australia
| | - Karina Aivazian
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia; Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia
| | - Matteo S Carlino
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia; Department of Medicine, Blacktown Hospital, Blacktown, New South Wales, Australia; Department of Medicine, Crown Princess Mary Cancer Centre, Westmead Hospital, Sydney, New South Wales, Australia
| | - Alexander D Guminski
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia; Royal North Shore and Mater Hospitals, Sydney, NSW, Australia
| | - Nigel G Maher
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia; Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia
| | - Kerwin F Shannon
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia; Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Sydney Ch'ng
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia; Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Robyn P M Saw
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia; Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia; Charles Perkin Centre, The University of Sydney, Sydney, NSW, Australia; Royal North Shore and Mater Hospitals, Sydney, NSW, Australia
| | - James S Wilmott
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia; Charles Perkin Centre, The University of Sydney, Sydney, NSW, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia; Charles Perkin Centre, The University of Sydney, Sydney, NSW, Australia; Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia.
| |
Collapse
|
31
|
The E3 ubiquitin ligase NEDD4-1 protects against acetaminophen-induced liver injury by targeting VDAC1 for degradation. Acta Pharm Sin B 2023; 13:1616-1630. [PMID: 37139424 PMCID: PMC10150139 DOI: 10.1016/j.apsb.2023.01.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/27/2022] [Accepted: 12/15/2022] [Indexed: 01/30/2023] Open
Abstract
Acetaminophen (APAP) overdose is a major cause of liver injury. Neural precursor cell expressed developmentally downregulated 4-1 (NEDD4-1) is an E3 ubiquitin ligase that has been implicated in the pathogenesis of numerous liver diseases; however, its role in APAP-induced liver injury (AILI) is unclear. Thus, this study aimed to investigate the role of NEDD4-1 in the pathogenesis of AILI. We found that NEDD4-1 was dramatically downregulated in response to APAP treatment in mouse livers and isolated mouse hepatocytes. Hepatocyte-specific NEDD4-1 knockout exacerbated APAP-induced mitochondrial damage and the resultant hepatocyte necrosis and liver injury, while hepatocyte-specific NEDD4-1 overexpression mitigated these pathological events both in vivo and in vitro. Additionally, hepatocyte NEDD4-1 deficiency led to marked accumulation of voltage-dependent anion channel 1 (VDAC1) and increased VDAC1 oligomerization. Furthermore, VDAC1 knockdown alleviated AILI and weakened the exacerbation of AILI caused by hepatocyte NEDD4-1 deficiency. Mechanistically, NEDD4-1 was found to interact with the PPTY motif of VDAC1 through its WW domain and regulate K48-linked ubiquitination and degradation of VDAC1. Our present study indicates that NEDD4-1 is a suppressor of AILI and functions by regulating the degradation of VDAC1.
Collapse
|
32
|
Kang D, Baek Y, Lee JS. Mechanisms of RNA and Protein Quality Control and Their Roles in Cellular Senescence and Age-Related Diseases. Cells 2022; 11:cells11244062. [PMID: 36552825 PMCID: PMC9777292 DOI: 10.3390/cells11244062] [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: 11/10/2022] [Revised: 12/04/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Cellular senescence, a hallmark of aging, is defined as irreversible cell cycle arrest in response to various stimuli. It plays both beneficial and detrimental roles in cellular homeostasis and diseases. Quality control (QC) is important for the proper maintenance of cellular homeostasis. The QC machineries regulate the integrity of RNA and protein by repairing or degrading them, and are dysregulated during cellular senescence. QC dysfunction also contributes to multiple age-related diseases, including cancers and neurodegenerative, muscle, and cardiovascular diseases. In this review, we describe the characters of cellular senescence, discuss the major mechanisms of RNA and protein QC in cellular senescence and aging, and comprehensively describe the involvement of these QC machineries in age-related diseases. There are many open questions regarding RNA and protein QC in cellular senescence and aging. We believe that a better understanding of these topics could propel the development of new strategies for addressing age-related diseases.
Collapse
Affiliation(s)
- Donghee Kang
- Research Center for Controlling Intercellular Communication (RCIC), College of Medicine, Inha University, Incheon 22212, Republic of Korea
- Department of Molecular Medicine, College of Medicine, Inha University, Incheon 22212, Republic of Korea
| | - Yurim Baek
- Research Center for Controlling Intercellular Communication (RCIC), College of Medicine, Inha University, Incheon 22212, Republic of Korea
- Department of Molecular Medicine, College of Medicine, Inha University, Incheon 22212, Republic of Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon 22212, Republic of Korea
| | - Jae-Seon Lee
- Research Center for Controlling Intercellular Communication (RCIC), College of Medicine, Inha University, Incheon 22212, Republic of Korea
- Department of Molecular Medicine, College of Medicine, Inha University, Incheon 22212, Republic of Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon 22212, Republic of Korea
- Correspondence: ; Tel.: +82-32-860-9832; Fax: +82-32-885-8302
| |
Collapse
|
33
|
Xia H, Hu H, Wang Z, Xia L, Chen W, Long M, Gan Z, Fan H, Yu D, Lu Y. Molecular cloning, expression analysis and functional characterization of NEDD4 from Nile tilapia (Oreochromis niloticus). FISH & SHELLFISH IMMUNOLOGY 2022; 131:257-263. [PMID: 36183983 DOI: 10.1016/j.fsi.2022.09.032] [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: 05/12/2022] [Revised: 09/09/2022] [Accepted: 09/13/2022] [Indexed: 06/16/2023]
Abstract
Neural precursor cell-expressed developmentally downregulated gene 4 (NEDD4) was a member of HECT E3 ubiquitin ligases, which participated in various biological processes. In this study, a NEDD4 was identified and analyzed in Nile tilapia, Oreochromis niloticus (OnNEDD4) and its open reading frame was 2781 bp, encoding 926 amino acids. Three conserved structure features were found in OnNEDD4, including C2 domain, WW domains and HECT domain. OnNEDD4 was constitutively expressed in all examined tissues and the highest expression level was observed in thymus. After Streptococcus agalactiae stimulation, OnNEDD4 was significantly induced in several tissues, including thymus, intestine, blood and gill. Moreover, yeast two-hybrid assay shown OnNEDD4 could interact with extracellular region of OnCD40, but this interaction didn't affect the phagocytosis of monocytes/macrophages (MO/MΦ) to S. agalactiae and A. hydrophila. Taken together, the present study suggested that OnNEDD4 participate in CD40-mediated immune response excluding phagocytosis.
Collapse
Affiliation(s)
- Hongli Xia
- Shenzhen Institute of Guangdong Ocean University, Shenzhen, 518120, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, 518120, China; Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen, 518120, China
| | - Huiling Hu
- College of Fishery, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Zhiwen Wang
- Shenzhen Institute of Guangdong Ocean University, Shenzhen, 518120, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, 518120, China; Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen, 518120, China
| | - Liqun Xia
- Shenzhen Institute of Guangdong Ocean University, Shenzhen, 518120, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, 518120, China; Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen, 518120, China; College of Fishery, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Wenjie Chen
- Shenzhen Institute of Guangdong Ocean University, Shenzhen, 518120, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, 518120, China; Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen, 518120, China
| | - Meng Long
- Shenzhen Institute of Guangdong Ocean University, Shenzhen, 518120, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, 518120, China; Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen, 518120, China
| | - Zhen Gan
- Shenzhen Institute of Guangdong Ocean University, Shenzhen, 518120, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, 518120, China; Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen, 518120, China
| | - Huimin Fan
- Shenzhen Institute of Guangdong Ocean University, Shenzhen, 518120, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, 518120, China; Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen, 518120, China
| | - Dapeng Yu
- Shenzhen Institute of Guangdong Ocean University, Shenzhen, 518120, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, 518120, China; Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen, 518120, China
| | - Yishan Lu
- Shenzhen Institute of Guangdong Ocean University, Shenzhen, 518120, China; Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, 518120, China; Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen, 518120, China; College of Fishery, Guangdong Ocean University, Zhanjiang, 524088, China.
| |
Collapse
|
34
|
Chen H, Chew G, Devapragash N, Loh JZ, Huang KY, Guo J, Liu S, Tan ELS, Chen S, Tee NGZ, Mia MM, Singh MK, Zhang A, Behmoaras J, Petretto E. The E3 ubiquitin ligase WWP2 regulates pro-fibrogenic monocyte infiltration and activity in heart fibrosis. Nat Commun 2022; 13:7375. [PMID: 36450710 PMCID: PMC9712659 DOI: 10.1038/s41467-022-34971-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 11/11/2022] [Indexed: 12/03/2022] Open
Abstract
Non-ischemic cardiomyopathy (NICM) can cause left ventricular dysfunction through interstitial fibrosis, which corresponds to the failure of cardiac tissue remodeling. Recent evidence implicates monocytes/macrophages in the etiopathology of cardiac fibrosis, but giving their heterogeneity and the antagonizing roles of macrophage subtypes in fibrosis, targeting these cells has been challenging. Here we focus on WWP2, an E3 ubiquitin ligase that acts as a positive genetic regulator of human and murine cardiac fibrosis, and show that myeloid specific deletion of WWP2 reduces cardiac fibrosis in hypertension-induced NICM. By using single cell RNA sequencing analysis of immune cells in the same model, we establish the functional heterogeneity of macrophages and define an early pro-fibrogenic phase of NICM that is driven by Ccl5-expressing Ly6chigh monocytes. Among cardiac macrophage subtypes, WWP2 dysfunction primarily affects Ly6chigh monocytes via modulating Ccl5, and consequentially macrophage infiltration and activation, which contributes to reduced myofibroblast trans-differentiation. WWP2 interacts with transcription factor IRF7, promoting its non-degradative mono-ubiquitination, nuclear translocation and transcriptional activity, leading to upregulation of Ccl5 at transcriptional level. We identify a pro-fibrogenic macrophage subtype in non-ischemic cardiomyopathy, and demonstrate that WWP2 is a key regulator of IRF7-mediated Ccl5/Ly6chigh monocyte axis in heart fibrosis.
Collapse
Affiliation(s)
- Huimei Chen
- grid.428397.30000 0004 0385 0924Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, 169857 Singapore, Singapore ,grid.254147.10000 0000 9776 7793Institute for Big Data and Artificial Intelligence in Medicine, School of Science, China Pharmaceutical University, Nanjing, 210009 China
| | - Gabriel Chew
- grid.428397.30000 0004 0385 0924Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, 169857 Singapore, Singapore
| | - Nithya Devapragash
- grid.428397.30000 0004 0385 0924Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, 169857 Singapore, Singapore
| | - Jui Zhi Loh
- grid.428397.30000 0004 0385 0924Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, 169857 Singapore, Singapore
| | - Kevin Y. Huang
- grid.428397.30000 0004 0385 0924Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, 169857 Singapore, Singapore
| | - Jing Guo
- grid.428397.30000 0004 0385 0924Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, 169857 Singapore, Singapore
| | - Shiyang Liu
- grid.428397.30000 0004 0385 0924Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, 169857 Singapore, Singapore
| | - Elisabeth Li Sa Tan
- grid.428397.30000 0004 0385 0924Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, 169857 Singapore, Singapore
| | - Shuang Chen
- grid.254147.10000 0000 9776 7793Institute for Big Data and Artificial Intelligence in Medicine, School of Science, China Pharmaceutical University, Nanjing, 210009 China ,grid.452511.6Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, 210008 China
| | - Nicole Gui Zhen Tee
- grid.419385.20000 0004 0620 9905National Heart Centre Singapore, Singapore, 169609 Singapore
| | - Masum M. Mia
- grid.428397.30000 0004 0385 0924Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, 169857 Singapore, Singapore
| | - Manvendra K. Singh
- grid.428397.30000 0004 0385 0924Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, 169857 Singapore, Singapore
| | - Aihua Zhang
- grid.452511.6Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, 210008 China
| | - Jacques Behmoaras
- grid.428397.30000 0004 0385 0924Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, 169857 Singapore, Singapore ,grid.413629.b0000 0001 0705 4923Centre for Inflammatory Disease, Imperial College London, Hammersmith Hospital, London, W12 0NN UK
| | - Enrico Petretto
- grid.428397.30000 0004 0385 0924Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 8 College Road, 169857 Singapore, Singapore ,grid.254147.10000 0000 9776 7793Institute for Big Data and Artificial Intelligence in Medicine, School of Science, China Pharmaceutical University, Nanjing, 210009 China
| |
Collapse
|
35
|
Jayaprakash S, Hegde M, BharathwajChetty B, Girisa S, Alqahtani MS, Abbas M, Sethi G, Kunnumakkara AB. Unraveling the Potential Role of NEDD4-like E3 Ligases in Cancer. Int J Mol Sci 2022; 23:ijms232012380. [PMID: 36293239 PMCID: PMC9604169 DOI: 10.3390/ijms232012380] [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: 07/25/2022] [Revised: 09/28/2022] [Accepted: 10/04/2022] [Indexed: 11/06/2022] Open
Abstract
Cancer is a deadly disease worldwide, with an anticipated 19.3 million new cases and 10.0 million deaths occurring in 2020 according to GLOBOCAN 2020. It is well established that carcinogenesis and cancer development are strongly linked to genetic changes and post-translational modifications (PTMs). An important PTM process, ubiquitination, regulates every aspect of cellular activity, and the crucial enzymes in the ubiquitination process are E3 ubiquitin ligases (E3s) that affect substrate specificity and must therefore be carefully regulated. A surfeit of studies suggests that, among the E3 ubiquitin ligases, neuronal precursor cell-expressed developmentally downregulated 4 (NEDD4)/NEDD4-like E3 ligases show key functions in cellular processes by controlling subsequent protein degradation and substrate ubiquitination. In addition, it was demonstrated that NEDD4 mainly acts as an oncogene in various cancers, but also plays a tumor-suppressive role in some cancers. In this review, to comprehend the proper function of NEDD4 in cancer development, we summarize its function, both its tumor-suppressive and oncogenic role, in multiple types of malignancies. Moreover, we briefly explain the role of NEDD4 in carcinogenesis and progression, including cell survival, cell proliferation, autophagy, cell migration, invasion, metastasis, epithelial-mesenchymal transition (EMT), chemoresistance, and multiple signaling pathways. In addition, we briefly explain the significance of NEDD4 as a possible target for cancer treatment. Therefore, we conclude that targeting NEDD4 as a therapeutic method for treating human tumors could be a practical possibility.
Collapse
Affiliation(s)
- Sujitha Jayaprakash
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Guwahati, Guwahati 781039, Assam, India
| | - Mangala Hegde
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Guwahati, Guwahati 781039, Assam, India
| | - Bandari BharathwajChetty
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Guwahati, Guwahati 781039, Assam, India
| | - Sosmitha Girisa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Guwahati, Guwahati 781039, Assam, India
| | - Mohammed S. Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia
- BioImaging Unit, Space Research Centre, Michael Atiyah Building, University of Leicester, Leicester LE1 7RH, UK
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha 61421, Saudi Arabia
- Electronics and Communications Department, College of Engineering, Delta University for Science and Technology, Gamasa 35712, Egypt
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Correspondence: (G.S.); (A.B.K.)
| | - Ajaikumar B. Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Guwahati, Guwahati 781039, Assam, India
- Correspondence: (G.S.); (A.B.K.)
| |
Collapse
|
36
|
Lu X, Xu H, Xu J, Lu S, You S, Huang X, Zhang N, Zhang L. The regulatory roles of the E3 ubiquitin ligase NEDD4 family in DNA damage response. Front Physiol 2022; 13:968927. [PMID: 36091384 PMCID: PMC9458852 DOI: 10.3389/fphys.2022.968927] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 07/21/2022] [Indexed: 11/24/2022] Open
Abstract
E3 ubiquitin ligases, an important part of ubiquitin proteasome system, catalyze the covalent binding of ubiquitin to target substrates, which plays a role in protein ubiquitination and regulates different biological process. DNA damage response (DDR) is induced in response to DNA damage to maintain genome integrity and stability, and this process has crucial significance to a series of cell activities such as differentiation, apoptosis, cell cycle. The NEDD4 family, belonging to HECT E3 ubiquitin ligases, is reported as regulators that participate in the DDR process by recognizing different substrates. In this review, we summarize recent researches on NEDD4 family members in the DDR and discuss the roles of NEDD4 family members in the cascade reactions induced by DNA damage. This review may contribute to the further study of pathophysiology for certain diseases and pharmacology for targeted drugs.
Collapse
Affiliation(s)
- Xinxin Lu
- Department of Hematology, the First Affiliated Hospital of China Medical University, Shenyang, LN, China
| | - Haiqi Xu
- Department of Hematology, General Hospital of PLA Northern Theater Command, Shenyang, LN, China
| | - Jiaqi Xu
- Department of Cardiology, the First Affiliated Hospital of China Medical University, Shenyang, LN, China
| | - Saien Lu
- Department of Cardiology, the First Affiliated Hospital of China Medical University, Shenyang, LN, China
| | - Shilong You
- Department of Cardiology, the First Affiliated Hospital of China Medical University, Shenyang, LN, China
| | - Xinyue Huang
- Department of Cardiology, the First Affiliated Hospital of China Medical University, Shenyang, LN, China
| | - Naijin Zhang
- Department of Cardiology, the First Affiliated Hospital of China Medical University, Shenyang, LN, China
| | - Lijun Zhang
- Department of Hematology, the First Affiliated Hospital of China Medical University, Shenyang, LN, China
| |
Collapse
|
37
|
Ubiquitin and Ubiquitin-like Proteins in Cancer, Neurodegenerative Disorders, and Heart Diseases. Int J Mol Sci 2022; 23:ijms23095053. [PMID: 35563444 PMCID: PMC9105348 DOI: 10.3390/ijms23095053] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 01/14/2023] Open
Abstract
Post-translational modification (PTM) is an essential mechanism for enhancing the functional diversity of proteins and adjusting their signaling networks. The reversible conjugation of ubiquitin (Ub) and ubiquitin-like proteins (Ubls) to cellular proteins is among the most prevalent PTM, which modulates various cellular and physiological processes by altering the activity, stability, localization, trafficking, or interaction networks of its target molecules. The Ub/Ubl modification is tightly regulated as a multi-step enzymatic process by enzymes specific to this family. There is growing evidence that the dysregulation of Ub/Ubl modifications is associated with various diseases, providing new targets for drug development. In this review, we summarize the recent progress in understanding the roles and therapeutic targets of the Ub and Ubl systems in the onset and progression of human diseases, including cancer, neurodegenerative disorders, and heart diseases.
Collapse
|
38
|
Jiang H, Chiang CY, Chen Z, Nathan S, D'Agostino G, Paulo JA, Song G, Zhu H, Gabelli SB, Cole PA. Enzymatic analysis of WWP2 E3 ubiquitin ligase using protein microarrays identifies autophagy-related substrates. J Biol Chem 2022; 298:101854. [PMID: 35331737 PMCID: PMC9034101 DOI: 10.1016/j.jbc.2022.101854] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 11/30/2022] Open
Abstract
WWP2 is a HECT E3 ligase that targets protein Lys residues for ubiquitination and is comprised of an N-terminal C2 domain, four central WW domains, and a C-terminal catalytic HECT domain. The peptide segment between the middle WW domains, the 2,3-linker, is known to autoinhibit the catalytic domain, and this autoinhibition can be relieved by phosphorylation at Tyr369. Several protein substrates of WWP2 have been identified, including the tumor suppressor lipid phosphatase PTEN, but the full substrate landscape and biological functions of WWP2 remain to be elucidated. Here, we used protein microarray technology and the activated enzyme phosphomimetic mutant WWP2Y369E to identify potential WWP2 substrates. We identified 31 substrate hits for WWP2Y369E using protein microarrays, of which three were known autophagy receptors (NDP52, OPTN, and SQSTM1). These three hits were validated with in vitro and cell-based transfection assays and the Lys ubiquitination sites on these proteins were mapped by mass spectrometry. Among the mapped ubiquitin sites on these autophagy receptors, many had been previously identified in the endogenous proteins. Finally, we observed that WWP2 KO SH-SH5Y neuroblastoma cells using CRISPR-Cas9 showed a defect in mitophagy, which could be rescued by WWP2Y369E transfection. These studies suggest that WWP2-mediated ubiquitination of the autophagy receptors NDP52, OPTN, and SQSTM1 may positively contribute to the regulation of autophagy.
Collapse
Affiliation(s)
- Hanjie Jiang
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Claire Y Chiang
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
| | - Zan Chen
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland, USA; Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA; Department of Biophysics and Biophysical Chemistry, The Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Sara Nathan
- Department of Biophysics and Biophysical Chemistry, The Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Gabriel D'Agostino
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Guang Song
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Heng Zhu
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland, USA; Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sandra B Gabelli
- Department of Biophysics and Biophysical Chemistry, The Johns Hopkins School of Medicine, Baltimore, Maryland, USA; Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Philip A Cole
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
39
|
Shao Y, Jiang Z, He D, Shen J. NEDD4 attenuates phosgene-induced acute lung injury through the inhibition of Notch1 activation. J Cell Mol Med 2022; 26:2831-2840. [PMID: 35355403 PMCID: PMC9097839 DOI: 10.1111/jcmm.17296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 02/21/2022] [Accepted: 03/04/2022] [Indexed: 11/28/2022] Open
Abstract
Phosgene gas leakage can cause life-threatening acute lung injury (ALI), which is characterized by inflammation, increased vascular permeability, pulmonary oedema and oxidative stress. Although the downregulation of neuronal precursor cell-expressed developmentally downregulated 4 (NEDD4) is known to be associated with inflammation and oxidative damage, its functions in phosgene-induced ALI remain unclear. In this study, rats with phosgene-induced ALI were intravenously injected with NEDD4-overexpressing lentiviruses to determine the functions of NEDD4 in this inflammatory condition. NEDD4 expression was decreased in the lung parenchyma of phosgene-exposed control rats, whereas its expression level was high in the NEDD4-overexpressing rats. Phosgene exposure increased the wet-to-dry lung weight ratio, but NEDD4 abrogated this effect. NEDD4 overexpression attenuated phosgene-induced lung inflammation, lowering the high lung injury score (based on total protein, inflammatory cells and inflammatory factors in bronchoalveolar lavage fluid) and also reduced phosgene-induced oxidative stress and cell apoptosis. Finally, NEDD4 was found to interact with Notch1, enhancing its ubiquitination and thereby its degradation, thus attenuating the inflammatory responses to ALI. Therefore, we demonstrated that NEDD4 plays a protective role in alleviating phosgene-induced ALI, suggesting that enhancing the effect of NEDD4 may be a new approach for treating phosgene-induced ALI.
Collapse
Affiliation(s)
- Yiru Shao
- Center of Emergency & Intensive Care UnitJinshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health CommissionShanghaiChina
- Medical Research Center for Chemical Injury, Emergency and Critical Care of Chemical InjuryJinshan HospitalFudan UniversityShanghaiChina
| | - Zhifeng Jiang
- Center of Emergency & Intensive Care UnitJinshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health CommissionShanghaiChina
- Medical Research Center for Chemical Injury, Emergency and Critical Care of Chemical InjuryJinshan HospitalFudan UniversityShanghaiChina
| | - Daikun He
- Center of Emergency & Intensive Care UnitJinshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health CommissionShanghaiChina
- Medical Research Center for Chemical Injury, Emergency and Critical Care of Chemical InjuryJinshan HospitalFudan UniversityShanghaiChina
| | - Jie Shen
- Center of Emergency & Intensive Care UnitJinshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health CommissionShanghaiChina
- Medical Research Center for Chemical Injury, Emergency and Critical Care of Chemical InjuryJinshan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
40
|
Lohraseb I, McCarthy P, Secker G, Marchant C, Wu J, Ali N, Kumar S, Daly RJ, Harvey NL, Kawabe H, Kleifeld O, Wiszniak S, Schwarz Q. Global ubiquitinome profiling identifies NEDD4 as a regulator of Profilin 1 and actin remodelling in neural crest cells. Nat Commun 2022; 13:2018. [PMID: 35440627 PMCID: PMC9018756 DOI: 10.1038/s41467-022-29660-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 03/24/2022] [Indexed: 01/02/2023] Open
Abstract
The ubiquitin ligase NEDD4 promotes neural crest cell (NCC) survival and stem-cell like properties to regulate craniofacial and peripheral nervous system development. However, how ubiquitination and NEDD4 control NCC development remains unknown. Here we combine quantitative analysis of the proteome, transcriptome and ubiquitinome to identify key developmental signalling pathways that are regulated by NEDD4. We report 276 NEDD4 targets in NCCs and show that loss of NEDD4 leads to a pronounced global reduction in specific ubiquitin lysine linkages. We further show that NEDD4 contributes to the regulation of the NCC actin cytoskeleton by controlling ubiquitination and turnover of Profilin 1 to modulate filamentous actin polymerization. Taken together, our data provide insights into how NEDD4-mediated ubiquitination coordinates key regulatory processes during NCC development. Here the authors combine multi-omics approaches to uncover a role for ubiquitination and the ubiquitin ligase NEDD4 in targeting the actin binding protein Profilin 1 to regulate actin polymerisation in neural crest cells.
Collapse
Affiliation(s)
- Iman Lohraseb
- Centre for Cancer Biology, University of South Australia and SA Pathology, GPO Box 2471, Adelaide, 5000, Australia
| | - Peter McCarthy
- Centre for Cancer Biology, University of South Australia and SA Pathology, GPO Box 2471, Adelaide, 5000, Australia
| | - Genevieve Secker
- Centre for Cancer Biology, University of South Australia and SA Pathology, GPO Box 2471, Adelaide, 5000, Australia
| | - Ceilidh Marchant
- Centre for Cancer Biology, University of South Australia and SA Pathology, GPO Box 2471, Adelaide, 5000, Australia
| | - Jianmin Wu
- Kinghorn Cancer Centre & Cancer Division, Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, NSW, 2010, Australia
| | - Naveid Ali
- Bone Therapeutics Group, Bone Biology Division, Garvan Institute of Medical Research, Sydney, 2010, Australia
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia and SA Pathology, GPO Box 2471, Adelaide, 5000, Australia
| | - Roger J Daly
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Victoria, 3800, Australia
| | - Natasha L Harvey
- Centre for Cancer Biology, University of South Australia and SA Pathology, GPO Box 2471, Adelaide, 5000, Australia
| | - Hiroshi Kawabe
- Department of Molecular Neurobiology, Max Planck Institute for Experimental Medicine, Goettingen, 37075, Germany.,Department of Pharmacology, Gunma University Graduate School of Medicine, Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Oded Kleifeld
- Faculty of Biology, Technion-Israel Institute of Technology, Technion City, Haifa, 3200003, Israel
| | - Sophie Wiszniak
- Centre for Cancer Biology, University of South Australia and SA Pathology, GPO Box 2471, Adelaide, 5000, Australia
| | - Quenten Schwarz
- Centre for Cancer Biology, University of South Australia and SA Pathology, GPO Box 2471, Adelaide, 5000, Australia.
| |
Collapse
|
41
|
Aisenberg WH, McCray BA, Sullivan JM, Diehl E, DeVine LR, Alevy J, Bagnell AM, Carr P, Donohue JK, Goretzki B, Cole RN, Hellmich UA, Sumner CJ. Multiubiquitination of TRPV4 reduces channel activity independent of surface localization. J Biol Chem 2022; 298:101826. [PMID: 35300980 PMCID: PMC9010760 DOI: 10.1016/j.jbc.2022.101826] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 02/19/2022] [Accepted: 02/21/2022] [Indexed: 02/06/2023] Open
Abstract
Ubiquitin (Ub)-mediated regulation of plasmalemmal ion channel activity canonically occurs via stimulation of endocytosis. Whether ubiquitination can modulate channel activity by alternative mechanisms remains unknown. Here, we show that the transient receptor potential vanilloid 4 (TRPV4) cation channel is multiubiquitinated within its cytosolic N-terminal and C-terminal intrinsically disordered regions (IDRs). Mutagenizing select lysine residues to block ubiquitination of the N-terminal but not C-terminal IDR resulted in a marked elevation of TRPV4-mediated intracellular calcium influx, without increasing cell surface expression levels. Conversely, enhancing TRPV4 ubiquitination via expression of an E3 Ub ligase reduced TRPV4 channel activity but did not decrease plasma membrane abundance. These results demonstrate Ub-dependent regulation of TRPV4 channel function independent of effects on plasma membrane localization. Consistent with ubiquitination playing a key negative modulatory role of the channel, gain-of-function neuropathy-causing mutations in the TRPV4 gene led to reduced channel ubiquitination in both cellular and Drosophila models of TRPV4 neuropathy, whereas increasing mutant TRPV4 ubiquitination partially suppressed channel overactivity. Together, these data reveal a novel mechanism via which ubiquitination of an intracellular flexible IDR domain modulates ion channel function independently of endocytic trafficking and identify a contributory role for this pathway in the dysregulation of TRPV4 channel activity by neuropathy-causing mutations.
Collapse
Affiliation(s)
- William H Aisenberg
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Brett A McCray
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jeremy M Sullivan
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Erika Diehl
- Department of Chemistry, Biochemistry Section, Johannes Gutenberg-Universität Mainz, Mainz, Germany
| | - Lauren R DeVine
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jonathan Alevy
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Anna M Bagnell
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Patrice Carr
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jack K Donohue
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Benedikt Goretzki
- Institute of Organic Chemistry and Macromolecular Chemistry, Cluster of Excellence 'Balance of the Microverse', Friedrich-Schiller-Universität, Jena, Germany; Center for Biomolecular Magnetic Resonance (BMRZ), Goethe-Universität, Frankfurt am Main, Germany
| | - Robert N Cole
- Institute of Organic Chemistry and Macromolecular Chemistry, Cluster of Excellence 'Balance of the Microverse', Friedrich-Schiller-Universität, Jena, Germany
| | - Ute A Hellmich
- Institute of Organic Chemistry and Macromolecular Chemistry, Cluster of Excellence 'Balance of the Microverse', Friedrich-Schiller-Universität, Jena, Germany; Center for Biomolecular Magnetic Resonance (BMRZ), Goethe-Universität, Frankfurt am Main, Germany
| | - Charlotte J Sumner
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
42
|
Haouari S, Vourc’h P, Jeanne M, Marouillat S, Veyrat-Durebex C, Lanznaster D, Laumonnier F, Corcia P, Blasco H, Andres CR. The Roles of NEDD4 Subfamily of HECT E3 Ubiquitin Ligases in Neurodevelopment and Neurodegeneration. Int J Mol Sci 2022; 23:ijms23073882. [PMID: 35409239 PMCID: PMC8999422 DOI: 10.3390/ijms23073882] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 02/06/2023] Open
Abstract
The ubiquitin pathway regulates the function of many proteins and controls cellular protein homeostasis. In recent years, it has attracted great interest in neurodevelopmental and neurodegenerative diseases. Here, we have presented the first review on the roles of the 9 proteins of the HECT E3 ligase NEDD4 subfamily in the development and function of neurons in the central nervous system (CNS). We discussed their regulation and their direct or indirect involvement in neurodevelopmental diseases, such as intellectual disability, and neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease or Amyotrophic Lateral Sclerosis. Further studies on the roles of these proteins, their regulation and their targets in neurons will certainly contribute to a better understanding of neuronal function and dysfunction, and will also provide interesting information for the development of therapeutics targeting them.
Collapse
Affiliation(s)
- Shanez Haouari
- UMR 1253, iBrain, Université de Tours, Inserm, 37044 Tours, France; (S.H.); (M.J.); (S.M.); (C.V.-D.); (D.L.); (F.L.); (P.C.); (H.B.); (C.R.A.)
| | - Patrick Vourc’h
- UMR 1253, iBrain, Université de Tours, Inserm, 37044 Tours, France; (S.H.); (M.J.); (S.M.); (C.V.-D.); (D.L.); (F.L.); (P.C.); (H.B.); (C.R.A.)
- CHRU de Tours, Service de Biochimie et Biologie Moléculaire, 37044 Tours, France
- Correspondence: ; Tel.: +33-(0)2-34-37-89-10; Fax: +33-(0)2-47-36-61-85
| | - Médéric Jeanne
- UMR 1253, iBrain, Université de Tours, Inserm, 37044 Tours, France; (S.H.); (M.J.); (S.M.); (C.V.-D.); (D.L.); (F.L.); (P.C.); (H.B.); (C.R.A.)
- CHRU de Tours, Service de Génétique, 37044 Tours, France
| | - Sylviane Marouillat
- UMR 1253, iBrain, Université de Tours, Inserm, 37044 Tours, France; (S.H.); (M.J.); (S.M.); (C.V.-D.); (D.L.); (F.L.); (P.C.); (H.B.); (C.R.A.)
| | - Charlotte Veyrat-Durebex
- UMR 1253, iBrain, Université de Tours, Inserm, 37044 Tours, France; (S.H.); (M.J.); (S.M.); (C.V.-D.); (D.L.); (F.L.); (P.C.); (H.B.); (C.R.A.)
- CHRU de Tours, Service de Biochimie et Biologie Moléculaire, 37044 Tours, France
| | - Débora Lanznaster
- UMR 1253, iBrain, Université de Tours, Inserm, 37044 Tours, France; (S.H.); (M.J.); (S.M.); (C.V.-D.); (D.L.); (F.L.); (P.C.); (H.B.); (C.R.A.)
| | - Frédéric Laumonnier
- UMR 1253, iBrain, Université de Tours, Inserm, 37044 Tours, France; (S.H.); (M.J.); (S.M.); (C.V.-D.); (D.L.); (F.L.); (P.C.); (H.B.); (C.R.A.)
| | - Philippe Corcia
- UMR 1253, iBrain, Université de Tours, Inserm, 37044 Tours, France; (S.H.); (M.J.); (S.M.); (C.V.-D.); (D.L.); (F.L.); (P.C.); (H.B.); (C.R.A.)
- CHRU de Tours, Service de Neurologie, 37044 Tours, France
| | - Hélène Blasco
- UMR 1253, iBrain, Université de Tours, Inserm, 37044 Tours, France; (S.H.); (M.J.); (S.M.); (C.V.-D.); (D.L.); (F.L.); (P.C.); (H.B.); (C.R.A.)
- CHRU de Tours, Service de Biochimie et Biologie Moléculaire, 37044 Tours, France
| | - Christian R. Andres
- UMR 1253, iBrain, Université de Tours, Inserm, 37044 Tours, France; (S.H.); (M.J.); (S.M.); (C.V.-D.); (D.L.); (F.L.); (P.C.); (H.B.); (C.R.A.)
- CHRU de Tours, Service de Biochimie et Biologie Moléculaire, 37044 Tours, France
| |
Collapse
|
43
|
Huang Z, Keramat S, Izadirad M, Chen ZS, Soukhtanloo M. The Potential Role of Exosomes in the Treatment of Brain Tumors, Recent Updates and Advances. Front Oncol 2022; 12:869929. [PMID: 35371984 PMCID: PMC8968044 DOI: 10.3389/fonc.2022.869929] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 02/21/2022] [Indexed: 12/05/2022] Open
Abstract
Exosomes are small endosomal derived membrane extracellular vesicles that contain cell-specific cargos such as lipid, protein, DNA, RNA, miRNA, long non-coding RNA, and some other cell components that are released into surrounding body fluids upon the fusion of multivesicular bodies (MVB) and the plasma membrane. Exosomes are a one-of-a-kind cell-to-cell communication mechanism that might pave the way for target therapy. The use of exosomes as a therapeutic potential in a variety of cancers has been and is still being investigated. One of the most important of these has been the use of exosomes in brain tumors therapy. Exosome contents play a crucial role in brain tumor progression by providing a favorable niche for tumor cell proliferation. Also, exosomes that are secreted from tumor cells, lead to the protection of tumor cells and their proliferation in the tumor environment by reducing the inflammatory response and suppression of the immune system. Although some treatment protocols such as surgery, chemotherapy, and radiotherapy are common in brain tumors, they do not result in complete remission in the treatment of some malignant and metastatic brain tumors. Identifying, targeting, and blocking exosomes involved in the progression of brain tumors could be a promising way to reduce brain tumor progression. On the other way, brain tumor therapy with effective therapeutic components such as siRNAs, mRNAs, proteins, could be developed. Finally, our research suggested that exosomes of nanoscale sizes might be a useful tool for crossing the blood-brain barrier and delivering effective content. However, further research is needed to fully comprehend the potential involvement of the exosome in brain tumor therapy protocols.
Collapse
Affiliation(s)
- Zoufang Huang
- Ganzhou Key Laboratory of Hematology, Department of Hematology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Shayan Keramat
- Department of Hematology and Blood Bank, Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran
| | - Mehrdad Izadirad
- Department of Hematology and Blood Bank, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, St John’s University, New York, NY, United States
| | - Mohammad Soukhtanloo
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
44
|
The Role of NEDD4 E3 Ubiquitin–Protein Ligases in Parkinson’s Disease. Genes (Basel) 2022; 13:genes13030513. [PMID: 35328067 PMCID: PMC8950476 DOI: 10.3390/genes13030513] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/03/2022] [Indexed: 01/25/2023] Open
Abstract
Parkinson’s disease (PD) is a debilitating neurodegenerative disease that causes a great clinical burden. However, its exact molecular pathologies are not fully understood. Whilst there are a number of avenues for research into slowing, halting, or reversing PD, one central idea is to enhance the clearance of the proposed aetiological protein, oligomeric α-synuclein. Oligomeric α-synuclein is the main constituent protein in Lewy bodies and neurites and is considered neurotoxic. Multiple E3 ubiquitin-protein ligases, including the NEDD4 (neural precursor cell expressed developmentally downregulated protein 4) family, parkin, SIAH (mammalian homologues of Drosophila seven in absentia), CHIP (carboxy-terminus of Hsc70 interacting protein), and SCFFXBL5 SCF ubiquitin ligase assembled by the S-phase kinase-associated protein (SKP1), cullin-1 (Cul1), a zinc-binding RING finger protein, and the F-box domain/Leucine-rich repeat protein 5-containing protein FBXL5), have been shown to be able to ubiquitinate α-synuclein, influencing its subsequent degradation via the proteasome or lysosome. Here, we explore the link between NEDD4 ligases and PD, which is not only via α-synuclein but further strengthened by several additional substrates and interaction partners. Some members of the NEDD4 family of ligases are thought to crosstalk even with PD-related genes and proteins found to be mutated in familial forms of PD. Mutations in NEDD4 family genes have not been observed in PD patients, most likely because of their essential survival function during development. Following further in vivo studies, it has been thought that NEDD4 ligases may be viable therapeutic targets in PD. NEDD4 family members could clear toxic proteins, enhancing cell survival and slowing disease progression, or might diminish beneficial proteins, reducing cell survival and accelerating disease progression. Here, we review studies to date on the expression and function of NEDD4 ubiquitin ligases in the brain and their possible impact on PD pathology.
Collapse
|
45
|
NEDD4L binds the proteasome and promotes autophagy and bortezomib sensitivity in multiple myeloma. Cell Death Dis 2022; 13:197. [PMID: 35236820 PMCID: PMC8891287 DOI: 10.1038/s41419-022-04629-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 01/20/2022] [Accepted: 02/07/2022] [Indexed: 11/30/2022]
Abstract
Multiple myeloma (MM) remains an incurable plasma cell cancer characterized by abnormal secretion of monoclonal immunoglobulins. The molecular mechanism that regulates the drug sensitivity of MM cells is being intensively studied. Here, we report an unexpected finding that the protein encoded by neural precursor cell-expressed developmentally downregulated gene 4L (NEDD4L), which is a HECT E3 ligase, binds the 19S proteasome, limiting its proteolytic function and enhancing autophagy. Suppression of NEDD4L expression reduced bortezomib (Bor) sensitivity in vitro and in vivo, mainly through autophagy inhibition mediated by low NEDD4L expression, which was rescued by an autophagy activator. Clinically, elevated expression of NEDD4L is associated with a considerably increased probability of responding to Bor, a prolonged response duration, and improved overall prognosis, supporting both the use of NEDD4L as a biomarker to identify patients most likely to benefit from Bor and the regulation of NEDD4L as a new approach in myeloma therapy.
Collapse
|
46
|
Mello e Souza T. Unraveling molecular and system processes for fear memory. Neuroscience 2022; 497:14-29. [DOI: 10.1016/j.neuroscience.2022.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 03/02/2022] [Accepted: 03/14/2022] [Indexed: 11/26/2022]
|
47
|
Wang T, Woodman P, Humphrey SJ, Petersen J. Environmental control of Pub1 (NEDD4 family E3 ligase) in Schizosaccharomyces pombe is regulated by TORC2 and Gsk3. Life Sci Alliance 2022; 5:5/5/e202101082. [PMID: 35121625 PMCID: PMC8817228 DOI: 10.26508/lsa.202101082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 11/24/2022] Open
Abstract
The NEDD4 family E3 ligase Pub1 is regulated by the nutrient environment, TORC2, and Gsk3 signalling pathway to control the level of amino acid transporters on the plasma membrane and thus nutrient uptake. Cells respond to changing nutrient environments by adjusting the abundance of surface nutrient transporters and receptors. This can be achieved by modulating ubiquitin-dependent endocytosis, which in part is regulated by the NEDD4 family of E3 ligases. Here we report novel regulation of Pub1, a fission yeast Schizosaccharomyces pombe member of the NEDD4-family of E3 ligases. We show that nitrogen stress inhibits Pub1 function, thereby increasing the abundance of the amino acid transporter Aat1 at the plasma membrane and enhancing sensitivity to the toxic arginine analogue canavanine. We show that TOR complex 2 (TORC2) signalling negatively regulates Pub1, thus TORC2 mutants under nutrient stress have decreased Aat1 at the plasma membrane and are resistant to canavanine. Inhibition of TORC2 signalling increases Pub1 phosphorylation, and this is dependent on Gsk3 activity. Addition of the Tor inhibitor Torin1 increases phosphorylation of Pub1 at serine 199 (S199) by 2.5-fold, and Pub1 protein levels in S199A phospho-ablated mutants are reduced. S199 is conserved in NEDD4 and is located immediately upstream of a WW domain required for protein interaction. Together, we describe how the major TORC2 nutrient-sensing signalling network regulates environmental control of Pub1 to modulate the abundance of nutrient transporters.
Collapse
Affiliation(s)
- Tingting Wang
- Flinders Health and Medical Research Institute, Flinders Centre for Innovation in Cancer, Flinders University, Adelaide, Australia
| | - Philip Woodman
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Sean J Humphrey
- Charles Perkins Centre, School of Life and Environmental Sciences, The University of Sydney, Camperdown, Australia
| | - Janni Petersen
- Flinders Health and Medical Research Institute, Flinders Centre for Innovation in Cancer, Flinders University, Adelaide, Australia
| |
Collapse
|
48
|
Yuan H, Fang CL, Deng YP, Huang J, Niu RZ, Chen JL, Chen TB, Zhu ZQ, Chen L, Xiong LL, Wang TH. A2B5-positive oligodendrocyte precursor cell transplantation improves neurological deficits in rats following spinal cord contusion associated with changes in expression of factors involved in the Notch signaling pathway. Neurochirurgie 2022; 68:188-195. [PMID: 34543615 DOI: 10.1016/j.neuchi.2021.09.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 08/21/2021] [Accepted: 09/04/2021] [Indexed: 02/05/2023]
Abstract
BACKGROUND Oligodendrocyte precursor cells (OPCs) are myelinated glial cells of the central nervous system (CNS), able to regenerate oligodendrocytes and myelin. This study aimed to elucidate the effect of A2B5-positive (A2B5+) OPC transplantation in rats with spinal cord contusion (SCC) and to investigate changes in expression of various factors involved in the Notch signaling pathway after OPC transplantation. METHODS OPCs were obtained from induced pluripotent stem cells (iPSCs) originating from mouse embryo fibroblasts (MEFs). After identification of iPSCs and iPSC-derived OPCs, A2B5+ OPCs were transplanted into the injured site of rats with SCC one week after SCC insult. Behavioral tests evaluated motor and sensory function 7 days after OPC transplantation. Real-time quantitative polymerase chain reaction (RT-qPCR) determined the expression of various cytokines related to the Notch signaling pathway after OPC transplantation. RESULTS IPSC-derived OPCs were successfully generated from MEFs, as indicated by positive immunostaining of A2B5, PDGFα and NG2. Further differentiation of OPCs was identified by immunostaining of Olig2, Sox10, Nkx2.2, O4, MBP and GFAP. Importantly, myelin formation was significantly enhanced in the SCC+ OPC group and SCI-induced motor and sensory dysfunction was largely alleviated by A2B5+ OPC transplantation. Expression of factors involved in the Notch signaling pathway (Notch-1, Numb, SHARP1 and NEDD4) was significantly increased after OPC transplantation. CONCLUSIONS A2B5+ OPC transplantation attenuates motor and sensory dysfunction in SCC rats by promoting myelin formation, which may be associated with change in expression of factors involved in the Notch signaling pathway.
Collapse
Affiliation(s)
- H Yuan
- Institute of Neuroscience, Kunming Medical University, Kunming 650031, Yunnan, China; Department of Spine Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - C-L Fang
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China; Department of Anesthesiology, National Traditional Chinese Medicine Clinical Research Base and Western Medicine Translational Medicine Research Center, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Y-P Deng
- Department of Anesthesiology, National Traditional Chinese Medicine Clinical Research Base and Western Medicine Translational Medicine Research Center, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - J Huang
- Institute of Neuroscience, Kunming Medical University, Kunming 650031, Yunnan, China
| | - R-Z Niu
- Laboratory Animal Department, Kunming Medical University, Kunming 650031, Yunnan, China
| | - J-L Chen
- Laboratory Animal Department, Kunming Medical University, Kunming 650031, Yunnan, China
| | - T-B Chen
- Laboratory Animal Department, Kunming Medical University, Kunming 650031, Yunnan, China
| | - Z-Q Zhu
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - L Chen
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - L-L Xiong
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - T-H Wang
- Institute of Neuroscience, Kunming Medical University, Kunming 650031, Yunnan, China; Laboratory Animal Department, Kunming Medical University, Kunming 650031, Yunnan, China; Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
49
|
The Ubiquitin E3 Ligase Nedd4 Regulates the Expression and Amyloid-β Peptide Export Activity of P-Glycoprotein. Int J Mol Sci 2022; 23:ijms23031019. [PMID: 35162941 PMCID: PMC8834788 DOI: 10.3390/ijms23031019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/12/2022] [Accepted: 01/16/2022] [Indexed: 02/04/2023] Open
Abstract
The ATP-binding cassette transporter, P-glycoprotein (P-gp), has been demonstrated to facilitate the clearance of amyloid-beta (Aβ) peptides, exporting the neurotoxic entity out of neurons and out of the brain via the blood–brain barrier. However, its expression and function diminish with age and in Alzheimer’s disease. P-gp is known to undergo ubiquitination, a post-translational modification that results in internalisation and/or degradation of the protein. NEDD4-1 is a ubiquitin E3 ligase that has previously been shown to ubiquitinate P-gp and reduce its cell surface expression. However, whether this effect translates into altered P-gp activity remains to be determined. siRNA was used to knockdown the expression of Nedd4 in CHO-APP cells. Western blot analysis confirmed that absence of Nedd4 was associated with increased P-gp protein expression. This was accompanied by increased transport activity, as shown by export of the P-gp substrate calcein-AM, as well as enhanced secretion of Aβ peptides, as shown by ELISA. These results implicate Nedd4 in the regulation of P-gp, and highlight a potential approach for restoring or augmenting P-gp expression and function to facilitate Aβ clearance from the brain.
Collapse
|
50
|
Xie W, Jin S, Zhang C, Yang S, Wu Y, Zhao Y, Songyang Z, Cui J. Selective autophagy controls the stability of TBK1 via NEDD4 to balance host defense. Cell Death Differ 2022; 29:40-53. [PMID: 34257412 PMCID: PMC8738727 DOI: 10.1038/s41418-021-00833-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/04/2021] [Accepted: 07/06/2021] [Indexed: 01/03/2023] Open
Abstract
As a core kinase of antiviral immunity, the activity and stability of TANK-binding kinase 1 (TBK1) is tightly controlled by multiple post-translational modifications. Although it has been demonstrated that TBK1 stability can be regulated by ubiquitin-dependent proteasome pathway, it is unclear whether another important protein degradation pathway, autophagosome pathway, can specifically affect TBK1 degradation by cargo receptors. Here we report that E3 ubiquitin ligase NEDD4 functions as a negative regulator of type I interferon (IFN) signaling by targeting TBK1 for degradation at the late stage of viral infection, to prevent the host from excessive immune response. Mechanically NEDD4 catalyzes the K27-linked poly-ubiquitination of TBK1 at K344, which serves as a recognition signal for cargo receptor NDP52-mediated selective autophagic degradation. Taken together, our study reveals the regulatory role of NEDD4 in balancing TBK1-centered type I IFN activation and provides insights into the crosstalk between selective autophagy and antiviral signaling.
Collapse
Affiliation(s)
- Weihong Xie
- grid.12981.330000 0001 2360 039XGuangdong Province Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong P. R. China
| | - Shouheng Jin
- grid.12981.330000 0001 2360 039XGuangdong Province Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong P. R. China
| | - Chenqiu Zhang
- grid.12981.330000 0001 2360 039XGuangdong Province Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong P. R. China
| | - Shuai Yang
- grid.12981.330000 0001 2360 039XGuangdong Province Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong P. R. China
| | - Yaoxing Wu
- grid.12981.330000 0001 2360 039XGuangdong Province Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong P. R. China
| | - Yong Zhao
- grid.12981.330000 0001 2360 039XGuangdong Province Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong P. R. China
| | - Zhou Songyang
- grid.12981.330000 0001 2360 039XGuangdong Province Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong P. R. China
| | - Jun Cui
- grid.12981.330000 0001 2360 039XGuangdong Province Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong P. R. China
| |
Collapse
|