1
|
Zhou Y, Wang F, Feng S, Li M, Zhu M. USP39 promote post-translational modifiers to stimulate the progress of cancer. Discov Oncol 2025; 16:749. [PMID: 40358671 PMCID: PMC12075731 DOI: 10.1007/s12672-025-02573-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 05/05/2025] [Indexed: 05/15/2025] Open
Abstract
Deubiquitinating enzymes (DUBs) are a class of crucial peptidyl hydrolases within the ubiquitin system, playing a significant role in reversing and strictly regulating ubiquitination, which is essential for various biological processes such as protein stability and cellular signal transduction. Ubiquitin-specific protease 39 (USP39) is an important member of the DUBs family. Recent studies have revealed that USP39 is involved in the regulation of multiple cellular activities including cell proliferation, migration, invasion, apoptosis, and DNA damage repair. USP39 also plays a significant role in the development and progression of various cancers. It is believed that USP39 is a unique enzyme that controls the ubiquitin process and is closely associated with the occurrence and progression of many cancers, including hepatocellular, lung, gastric, breast, and ovarian cancer. This review summarizes the structural and functional aspects of USP39 and its research advancements in tumors, investigates the key molecular mechanisms related to USP39, and provides references for tumor diagnosis and treatment.
Collapse
Affiliation(s)
- Yuli Zhou
- Key Laboratory of Tropical Translational Medicine, Ministry of Education and Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, 3 Xueyuan Road, Longhua District, Haikou, 571199, Hainan, People's Republic of China
| | - Fang Wang
- Key Laboratory of Tropical Translational Medicine, Ministry of Education and Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, 3 Xueyuan Road, Longhua District, Haikou, 571199, Hainan, People's Republic of China
| | - Siren Feng
- Key Laboratory of Tropical Translational Medicine, Ministry of Education and Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, 3 Xueyuan Road, Longhua District, Haikou, 571199, Hainan, People's Republic of China
| | - Mengsen Li
- Key Laboratory of Tropical Translational Medicine, Ministry of Education and Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, 3 Xueyuan Road, Longhua District, Haikou, 571199, Hainan, People's Republic of China.
- Department of Medical Oncology, Second Affiliated Hospital, Hainan Medical University, Haikou, 570216, China.
| | - Mingyue Zhu
- Key Laboratory of Tropical Translational Medicine, Ministry of Education and Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, 3 Xueyuan Road, Longhua District, Haikou, 571199, Hainan, People's Republic of China.
| |
Collapse
|
2
|
Chen Y, Zhang J, Yang J, Zhao J, Guo X, Zhang J, Gan J, Zhao W, Chen S, Zhang X, Lin Y, Jin J. Exploring the cancerous nexus: the pivotal and diverse roles of USP39 in cancer development. Discov Oncol 2025; 16:715. [PMID: 40347416 PMCID: PMC12065690 DOI: 10.1007/s12672-025-02480-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 04/24/2025] [Indexed: 05/12/2025] Open
Abstract
The ubiquitin-proteasome system enables post-transcriptional protein modification and is a major pathway for the degradation of most of them in eukaryotic cells. Among these, the ubiquitin-specific protease (USP) family is the most extensively studied. As an important member of the USP family, ubiquitin-specific protease 39 (USP39) plays an essential role in RNA splicing and protein regulation. This review comprehensively summarizes the structural characteristics and molecular functions of USP39, emphasizing its pivotal role in the regulation of cellular processes. Dysregulation of USP39 is closely associated with the progression of various cancers through mechanisms such as immune evasion, modulation of oncogenic signaling pathways, and altered RNA splicing. These processes impact key aspects of cancer biology, including proliferation, metastasis, and therapy resistance, underscoring the broad implications of USP39 in tumor progression. Recent studies position USP39 as a promising target for cancer treatment. Future research should explore its upstream regulatory networks, develop small-molecule inhibitors, and evaluate its potential for precision oncology. This review integrates the latest insight into USP39, providing a foundation for its clinical application in cancer therapy.
Collapse
Affiliation(s)
- Yujing Chen
- School of Pharmacy, Guilin Medical University, Guangxi, 541199, Guilin, People's Republic of China
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi, China
| | - Jingyi Zhang
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi, China
- Institute of Integrated Traditional Chinese and Western Medicine, Jining Medical University, Jining, Shandong, China
| | - Jinfeng Yang
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi, China
| | - Jiawei Zhao
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi, China
- Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Guilin, Guangxi, China
| | - Xiaotong Guo
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi, China
- Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Guilin, Guangxi, China
| | - Juzheng Zhang
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi, China
- Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Guilin, Guangxi, China
| | - Jinfeng Gan
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi, China
- Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Guilin, Guangxi, China
| | - Weijia Zhao
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi, China
- Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Guilin, Guangxi, China
| | - Siqi Chen
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi, China
- Department of Oral Bioscience, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Xinwen Zhang
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi, China
| | - Yi Lin
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi, China.
- Department of Ultrasound, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China.
| | - Jiamin Jin
- School of Pharmacy, Guilin Medical University, Guangxi, 541199, Guilin, People's Republic of China.
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi, China.
- Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Guilin, Guangxi, China.
- Department of Ultrasound, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China.
| |
Collapse
|
3
|
Shen S, Xue G, Zeng Z, Peng L, Nie W, Zeng X. Toosendanin promotes prostate cancer cell apoptosis, ferroptosis and M1 polarization via USP39-mediated PLK1 deubiquitination. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03916-3. [PMID: 40056202 DOI: 10.1007/s00210-025-03916-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 02/11/2025] [Indexed: 03/10/2025]
Abstract
Toosendanin (TSN) can inhibit the malignant process of many cancers, and has the potential to be developed as an anti-tumor drug. However, the role and mechanism of TSN in prostate cancer (PCa) progression remain unclear. PCa cells (DU145 and LNCaP) were treated with TSN. Cell viability was detected by cell counting kit 8 assay. Cell proliferation, apoptosis and metastasis were assessed by colony formation assay, flow cytometry and transwell assay. Cell ferroptosis was evaluated by examining Fe2+, MDA and lipid-ROS levels. M1 polarization markers were analyzed by flow cytometry. Immunohistochemical staining, quantitative real-time PCR and western blot were used to detect ubiquitin-specific protease 39 (USP39) and polo-like kinase 1 (PLK1) expression. Cycloheximide treatment, Co-IP assay and ubiquitination assay were performed to confirm the regulation of USP39 on PLK1. In vivo experiments were employed to determine the effect of TSN and USP39 on PCa tumor growth. TSN treatment suppressed PCa cell proliferation, cell cycle, migration, and invasion, while enhanced apoptosis, ferroptosis, and M1 polarization. USP39 was upregulated in PCa tissues and cells, and its protein expression was reduced by TSN. USP39 overexpression reversed the regulation of TSN on PCa cell functions. PLK1 had elevated expression in PCa, and USP39 stabilized its protein expression by deubiquitination. USP39 knockdown inhibited PCa cell behaviors, and its regulation was abolished by PLK1 overexpression. Meanwhile, TSN reduced PCa tumor growth by regulating USP39/PLK1. TSN played anti-tumor role in PCa, which promoted PCa cell apoptosis, ferroptosis, and M1 polarization by inhibiting USP39/PLK1 axis.
Collapse
Affiliation(s)
- Siyao Shen
- Department of Urology, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, No.90, Bayi Avenue, Xihu District, Nanchang City, 330003, Jiangxi Province, China
- Department of Urology, The Fourth Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang City, 330003, Jiangxi Province, China
- Institute of Urology, Jiangxi University of Traditional Chinese Medicine, Nanchang City, 330003, Jiangxi Province, China
| | - Guifeng Xue
- Institute of Urology, Jiangxi University of Traditional Chinese Medicine, Nanchang City, 330003, Jiangxi Province, China
| | - Zhigang Zeng
- Department of Urology, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, No.90, Bayi Avenue, Xihu District, Nanchang City, 330003, Jiangxi Province, China
- Department of Urology, The Fourth Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang City, 330003, Jiangxi Province, China
| | - Liang Peng
- Department of Urology, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, No.90, Bayi Avenue, Xihu District, Nanchang City, 330003, Jiangxi Province, China
- Department of Urology, The Fourth Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang City, 330003, Jiangxi Province, China
| | - Weidong Nie
- Department of Urology, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, No.90, Bayi Avenue, Xihu District, Nanchang City, 330003, Jiangxi Province, China
- Department of Urology, The Fourth Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang City, 330003, Jiangxi Province, China
| | - Xiaochun Zeng
- Department of Urology, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, No.90, Bayi Avenue, Xihu District, Nanchang City, 330003, Jiangxi Province, China.
- Department of Urology, The Fourth Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang City, 330003, Jiangxi Province, China.
- Institute of Urology, Jiangxi University of Traditional Chinese Medicine, Nanchang City, 330003, Jiangxi Province, China.
| |
Collapse
|
4
|
Han H, Shi Q, Zhang Y, Ding M, He X, Liu C, Zhao D, Wang Y, Du Y, Zhu Y, Yuan Y, Wang S, Guo H, Wang Q. RBM12 drives PD-L1-mediated immune evasion in hepatocellular carcinoma by increasing JAK1 mRNA translation. Oncogene 2024; 43:3062-3077. [PMID: 39187545 DOI: 10.1038/s41388-024-03140-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/15/2024] [Accepted: 08/20/2024] [Indexed: 08/28/2024]
Abstract
Immunosuppression characterizes the tumour microenvironment in HCC, and recent studies have implicated RNA-binding proteins (RBPs) in the development of HCC. Here, we conducted a screen and identified RBM12 as a key protein that increased the expression of PD-L1, thereby driving immune evasion in HCC. Furthermore, RBM12 was found to be significantly upregulated in HCC tissues and was associated with a poor prognosis for HCC patients. Through various molecular assays and high-throughput screening, we determined that RBM12 could directly bind to the JAK1 mRNA via its 4th-RRM (RNA recognition motif) domain and recruit EIF4A2 through its 2nd-RRM domain, enhancing the distribution of ribosomes on JAK1 mRNA, which promotes the translation of JAK1 and the subsequent upregulation of its expression. As a result, the activated JAK1/STAT1 pathway transcriptionally upregulates PD-L1 expression, facilitating immune evasion in HCC. In summary, our findings provide insights into the significant contribution of RBM12 to immune evasion in HCC, highlighting its potential as a therapeutic target in the future. This graphical abstract shows that elevated expression of RBM12 in HCC can augment PD-L1-mediated tumour immune evasion by increasing the efficiency of JAK1 mRNA translation.
Collapse
Affiliation(s)
- Hexu Han
- Department of Gastroenterology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu, 225300, China
| | - Qian Shi
- Huzhou Key Laboratory of Translational Medicine, The First Affliated Hospital of Huzhou University, Huzhou, Zhejiang, 313000, China
| | - Yue Zhang
- Clinical Medical Laboratory Center, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu, 225300, China
| | - Mingdong Ding
- Department of Infectious Diseases, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu, 225300, China
| | - Xianzhong He
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Innovative Institute of Tumor Immunity and Medicine (ITIM), Anhui Provincial Innovation Institute for Pharmaceutical Basic Research, Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, Anhui, 230000, China
| | - Cuixia Liu
- Department of Gastroenterology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu, 225300, China
| | - Dakun Zhao
- Department of Gastroenterology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu, 225300, China
| | - Yifan Wang
- Department of Gastroenterology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu, 225300, China
| | - Yanping Du
- Department of Gastroenterology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu, 225300, China
| | - Yichao Zhu
- Department of Hepatobiliary Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu, 225300, China
| | - Yin Yuan
- Department of Hepatobiliary Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu, 225300, China.
| | - Siliang Wang
- Department of pharmacy, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210000, China.
| | - Huimin Guo
- Department of Gastroenterology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210000, China.
| | - Qiang Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University, Innovative Institute of Tumor Immunity and Medicine (ITIM), Anhui Provincial Innovation Institute for Pharmaceutical Basic Research, Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, Hefei, Anhui, 230000, China.
| |
Collapse
|
5
|
Pan M, Luo M, Liu L, Chen Y, Cheng Z, Wang K, Huang L, Tang N, Qiu J, Huang A, Xia J. EGR1 suppresses HCC growth and aerobic glycolysis by transcriptionally downregulating PFKL. J Exp Clin Cancer Res 2024; 43:35. [PMID: 38287371 PMCID: PMC10823730 DOI: 10.1186/s13046-024-02957-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 01/14/2024] [Indexed: 01/31/2024] Open
Abstract
BACKGROUND Hepatocellular Carcinoma (HCC) is a matter of great global public health importance; however, its current therapeutic effectiveness is deemed inadequate, and the range of therapeutic targets is limited. The aim of this study was to identify early growth response 1 (EGR1) as a transcription factor target in HCC and to explore its role and assess the potential of gene therapy utilizing EGR1 for the management of HCC. METHODS In this study, both in vitro and in vivo assays were employed to examine the impact of EGR1 on the growth of HCC. The mouse HCC model and human organoid assay were utilized to assess the potential of EGR1 as a gene therapy for HCC. Additionally, the molecular mechanism underlying the regulation of gene expression and the suppression of HCC growth by EGR1 was investigated. RESULTS The results of our investigation revealed a notable decrease in the expression of EGR1 in HCC. The decrease in EGR1 expression promoted the multiplication of HCC cells and the growth of xenografted tumors. On the other hand, the excessive expression of EGR1 hindered the proliferation of HCC cells and repressed the development of xenografted tumors. Furthermore, the efficacy of EGR1 gene therapy was validated using in vivo mouse HCC models and in vitro human hepatoma organoid models, thereby providing additional substantiation for the anti-cancer role of EGR1 in HCC. The mechanistic analysis demonstrated that EGR1 interacted with the promoter region of phosphofructokinase-1, liver type (PFKL), leading to the repression of PFKL gene expression and consequent inhibition of PFKL-mediated aerobic glycolysis. Moreover, the sensitivity of HCC cells and xenografted tumors to sorafenib was found to be increased by EGR1. CONCLUSION Our findings suggest that EGR1 possesses therapeutic potential as a tumor suppressor gene in HCC, and that EGR1 gene therapy may offer benefits for HCC patients.
Collapse
Affiliation(s)
- Mingang Pan
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing, 400016, China
| | - Muyu Luo
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing, 400016, China
| | - Lele Liu
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing, 400016, China
| | - Yunmeng Chen
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing, 400016, China
| | - Ziyi Cheng
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing, 400016, China
| | - Kai Wang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing, 400016, China
| | - Luyi Huang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing, 400016, China
| | - Ni Tang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing, 400016, China
| | - Jianguo Qiu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Chongqing Medical University, Chongqing, 400016, China.
| | - Ailong Huang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing, 400016, China.
| | - Jie Xia
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|