1
|
Zhu Q, Yuan Z, Huo Q, Lu Q, Wu Q, Guo J, Fu W, Lu Y, Zhong L, Shang W, Cui D, Li S, Liu X, Tu K, Huang D, Xu Q, Hu X. YY1 induced USP13 transcriptional activation drives the malignant progression of hepatocellular carcinoma by deubiquitinating WWP1. Cell Mol Biol Lett 2025; 30:56. [PMID: 40319251 PMCID: PMC12049795 DOI: 10.1186/s11658-025-00733-7] [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: 08/21/2024] [Accepted: 04/16/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the sixth most prevalent cancer globally and the third leading cause of cancer-related mortality. Protein ubiquitination and deubiquitination play vital roles in human cancers. Ubiquitin-specific protease 13 (USP13) is a deubiquitinating enzyme (DUB) that is involved in many cellular processes. However, the mechanism by which USP13 regulates deubiquitination remains largely unknown. METHODS Clinical data were analyzed via online databases. USP13 expression in HCC cell lines and tissues was analyzed via western blotting and immunohistochemistry. A lentivirus was used to established stable USP13-knockdown and USP13-overexpression cells. Cell Counting Kit-8, colony formation, wound healing, Transwell, and sphere formation assays were used to detect the malignant behaviors of HCC cells in vitro. A subcutaneous mouse model was used to investigate the function of USP13 in vivo. Co-immunoprecipitation, chromatin immunoprecipitation and dual-luciferase reporter assays were conducted to explore the molecular regulation. RESULTS USP13 was upregulated in HCC cell lines and tissues, which predicted a poor prognosis in patients with HCC. Functional experiments in which USP13 was overexpressed or depleted revealed the oncogenic role of USP13 in driving HCC progression both in vitro and in vivo. Mechanistically, WW domain-containing ubiquitin E3 ligase 1 (WWP1) was identified as a binding protein of USP13. Furthermore, USP13 can interact with WWP1 and then remove the K29- and K48-linked polyubiquitination chains from WWP1 to stabilize the WWP1 protein via the ubiquitin-proteasome pathway. Moreover, Yin Yang 1 (YY1) was explored as a new transcription factor of USP13, and YY1 could also upregulate WWP1 expression through USP13. Moreover, YY1 and WWP1 were shown to participate in the oncogenic role of USP13. CONCLUSIONS Our findings revealed the functional YY1/USP13/WWP1 signaling axis in HCC, identifying a promising therapeutic target for anti-HCC treatment.
Collapse
Affiliation(s)
- Qingwei Zhu
- The Qingdao Medical College of Qingdao University, Qingdao, 266000, China
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Zibo Yuan
- The Qingdao Medical College of Qingdao University, Qingdao, 266000, China
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Qiang Huo
- Department of General Surgery, Zhoushan Dinghai Central Hospital (Dinghai District of Zhejiang Provincial People's Hospital), Zhoushan, 316000, China
| | - Qiliang Lu
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
- The Key Laboratory of Gastroenterology of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China
| | - Qingsong Wu
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
- Department of Hepatobiliary, Shandong Provincial Third Hospital, Shandong University, Jinan, 250031, China
| | - Junwei Guo
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
- The Second Clinical Medical College of Zhejiang, Chinese Medical University, Hangzhou, 310053, China
| | - Wen Fu
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
- Cancer Center, Department of Hematology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Ying Lu
- Department of Haematology, Affiliated People's Hospital of Ningbo University, Ningbo, 315000, China
| | - Lei Zhong
- Department of Laboratory Medicine, Tongxiang Traditional Chinese Medicine Hospital, Tongxiang, 314500, China
| | - Wenzhong Shang
- Department of Hematology, The first People's Hospital of Fuyang Hangzhou, Hangzhou, 311400, China
| | - Di Cui
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
- General Surgery, Cancer Center, Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Shuangshuang Li
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China
| | - Xin Liu
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China.
| | - Kangsheng Tu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Dongsheng Huang
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China.
| | - Qiuran Xu
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China.
| | - Xiaoge Hu
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China.
- General Surgery, Cancer Center, Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, 310014, China.
| |
Collapse
|
2
|
Hu H, Zhan X, Xiong Y, Yuan R, Du Y, Dong Q, Li S, Guo B, Li Z, Feng J, Xiong S, Xiong J, Li D, Fu B, Xu S, Guo J. Non-classic deubiquitinase USP13 inhibits bladder cancer metastasis through destabilizing cytoplasmic KDM3A. Oncogene 2025:10.1038/s41388-025-03410-3. [PMID: 40253486 DOI: 10.1038/s41388-025-03410-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/31/2025] [Accepted: 04/07/2025] [Indexed: 04/21/2025]
Abstract
Bladder cancer (BLCa) metastasis is a predominant cause of death for bladder cancer patients. Histone demethylase KDM3A specifically removes the repressive mono- or di-methyl marks from H3K9 and thus contributes to the activation of gene transcription. However, the underlying mechanisms of KDM3A in bladder cancer are poorly understood. Here, we report that high levels of KDM3A are associated with bladder cancer clinical progression. KDM3A silencing inhibits bladder cancer cell growth, cell migration and invasion in vitro and in vivo. Mechanistically, we identify that non-classic deubiquitinase USP13 interacts with KDM3A to promote its degradation in cytoplasm via the proteasome-specific pathway. USP13 was significantly down-regulated in bladder cancer tissues and negatively associated with KDM3A expression. Furthermore, we show in bladder injected-liver metastasis xenograft model that USP13 inhibits bladder cancer metastasis through destabilizing cytoplasmic KDM3A. Collectively, our findings identify KDM3A is an important regulator of bladder cancer cell growth and metastasis and targeting USP13/KDM3A complex could be a valuable strategy to ameliorate bladder cancer progression and metastasis.
Collapse
Affiliation(s)
- Hongji Hu
- Department of Urology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Urinary System Diseases, Department of Urology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xiangpeng Zhan
- Department of Urology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Urinary System Diseases, Department of Urology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yunqiang Xiong
- Department of Urology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Urinary System Diseases, Department of Urology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Ruize Yuan
- Department of Urology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Urinary System Diseases, Department of Urology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yuanzhuo Du
- Department of Urology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Urinary System Diseases, Department of Urology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Qianxi Dong
- Department of Urology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Urinary System Diseases, Department of Urology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Sheng Li
- Department of Urology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Urinary System Diseases, Department of Urology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Biao Guo
- Department of Urology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Urinary System Diseases, Department of Urology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Zhongqi Li
- Department of Urology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Urinary System Diseases, Department of Urology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jianhua Feng
- Department of Radiation Oncology, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Situ Xiong
- Department of Urology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Urinary System Diseases, Department of Urology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jing Xiong
- Department of Urology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Urinary System Diseases, Department of Urology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Dongshui Li
- Department of Andrology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Bin Fu
- Department of Urology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
- Jiangxi Provincial Key Laboratory of Urinary System Diseases, Department of Urology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| | - Songhui Xu
- Department of Urology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
- Jiangxi Provincial Key Laboratory of Urinary System Diseases, Department of Urology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| | - Ju Guo
- Department of Urology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
- Jiangxi Provincial Key Laboratory of Urinary System Diseases, Department of Urology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
3
|
Yi L, Shahatiaili A, Zhang L, He H, Chen L, Zhang Z, Gao F, Shao F, Gao Y, He J. USP13: A therapeutic target for combating tumorigenesis and antitumor therapy resistance. Int J Biol Macromol 2025; 304:140608. [PMID: 39900156 DOI: 10.1016/j.ijbiomac.2025.140608] [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: 11/01/2024] [Revised: 01/29/2025] [Accepted: 01/31/2025] [Indexed: 02/05/2025]
Abstract
Ubiquitin-specific peptidase 13 (USP13) has emerged as a key regulator of proteins critical to the hallmarks of cancer, playing an essential role in cellular regulation. This deubiquitinating enzyme, often overexpressed in malignancies, wields its molecular scissors precisely, snipping ubiquitin tags to rescue oncoproteins from degradation. Our review highlights the dual role of USP13 in cancer biology: while it predominantly fuels tumor growth and metastasis, USP13 occasionally functions as a tumor suppressor. USP13 is as a formidable factor in cancer therapy, fortifying tumors against an arsenal of treatments. It bolsters DNA repair mechanisms, ignites prosurvival autophagy, and even reprograms cell lineages to evade targeted therapies. However, USP13 is also a promising target in the treatment of cancer. We highlight burgeoning strategies to neutralize USP13, from small molecule inhibitors to innovative protein degraders, which may disarm cancer resistance mechanisms. We also offer suggestions for future USP13 research, emphasizing the need for structural insights and more potent inhibitors. This review highlights the critical role of USP13 in cancer and underscores its potential as a therapeutic target for advancing cancer treatment.
Collapse
Affiliation(s)
- Lina Yi
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China; Central Laboratory & Shenzhen Key Laboratory of Epigenetics and Precision Medicine for Cancers, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China; Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Akezhouli Shahatiaili
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Haihua He
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China; Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Leifeng Chen
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhen Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fushan Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fei Shao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Laboratory of Translational Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yibo Gao
- Central Laboratory & Shenzhen Key Laboratory of Epigenetics and Precision Medicine for Cancers, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China; Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Laboratory of Translational Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Institute of Cancer Research, Henan Academy of Innovations in Medical Science, Zhengzhou, China; Department of Gastroenterology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancers Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China.
| | - Jie He
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China; Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Laboratory of Translational Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; State Key Laboratory of Molecular Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Institute of Cancer Research, Henan Academy of Innovations in Medical Science, Zhengzhou, China.
| |
Collapse
|
4
|
Xiao D, Chu X, Wang W, Peng M, Lv Q, Xu C, Duan H, Yang X. Inhibition of Cyclin D1 by Novel Biguanide Derivative YB-004 Increases the Sensitivity of Bladder Cancer to Olaparib via Causing G0 / G1 Arrest. Int J Biol Sci 2025; 21:1984-1998. [PMID: 40083696 PMCID: PMC11900825 DOI: 10.7150/ijbs.105072] [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: 10/12/2024] [Accepted: 01/12/2025] [Indexed: 03/16/2025] Open
Abstract
Bladder cancer (BC) is the 10th most common type of tumor worldwide, and recently approved immunotherapies and FGFR inhibitors have been shown to improve the prognosis of only a very limited subset of BC patients. Thus, the quest for more effective drugs remains an urgent priority for improving the quality of life of more BC patients. Previously, we demonstrated that a novel biguanide YB-004 has potent antitumor activity. In this study, we found that the novel biguanide YB-004 interrupts the cell cycle by reducing the expression of cyclin D1, causing G0/G1 phase arrest, and suppresses homologous recombination (HR) by inhibiting Rad51, thereby increasing DNA damage and blocking BC cell proliferation. Interestingly, our results further revealed that cell accumulation in the S and G2/M phases is the main reason why HR-proficient BC cells are not sensitive to olaparib, as these phases are conducive to HR activation and DNA repair. Thus, YB-004 increased the sensitivity of BC cells to olaparib by reversing the cell cycle changes and HR activation caused by olaparib. Taken together, these findings suggest that the combination of YB-004 with olaparib has great potential for the clinical treatment of HR-proficient BC.
Collapse
Affiliation(s)
- Di Xiao
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Oncology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, The Research Center of Reproduction and Translational Medicine of Hunan Province, Key Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of Education, School of Pharmaceutical Sciences, Health Science Center, Hunan Normal University, Changsha 410013, Hunan, China
- TCM and Ethnomedicine Innovation and Development International Laboratory, Innovative Material Medical Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Xuetong Chu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Oncology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, The Research Center of Reproduction and Translational Medicine of Hunan Province, Key Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of Education, School of Pharmaceutical Sciences, Health Science Center, Hunan Normal University, Changsha 410013, Hunan, China
| | - Weifan Wang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Oncology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, The Research Center of Reproduction and Translational Medicine of Hunan Province, Key Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of Education, School of Pharmaceutical Sciences, Health Science Center, Hunan Normal University, Changsha 410013, Hunan, China
| | - Mei Peng
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Oncology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, The Research Center of Reproduction and Translational Medicine of Hunan Province, Key Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of Education, School of Pharmaceutical Sciences, Health Science Center, Hunan Normal University, Changsha 410013, Hunan, China
| | - Qi Lv
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Oncology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, The Research Center of Reproduction and Translational Medicine of Hunan Province, Key Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of Education, School of Pharmaceutical Sciences, Health Science Center, Hunan Normal University, Changsha 410013, Hunan, China
| | - Cangcang Xu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Oncology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, The Research Center of Reproduction and Translational Medicine of Hunan Province, Key Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of Education, School of Pharmaceutical Sciences, Health Science Center, Hunan Normal University, Changsha 410013, Hunan, China
| | - Huaxin Duan
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Oncology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, The Research Center of Reproduction and Translational Medicine of Hunan Province, Key Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of Education, School of Pharmaceutical Sciences, Health Science Center, Hunan Normal University, Changsha 410013, Hunan, China
| | - Xiaoping Yang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Oncology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, The Research Center of Reproduction and Translational Medicine of Hunan Province, Key Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of Education, School of Pharmaceutical Sciences, Health Science Center, Hunan Normal University, Changsha 410013, Hunan, China
- FuRong Laboratory, Changsha 410078, Hunan, China
| |
Collapse
|
5
|
Bakkar M, Khalil S, Bhayekar K, Kushwaha ND, Samarbakhsh A, Dorandish S, Edwards H, Dou QP, Ge Y, Gavande NS. Ubiquitin-Specific Protease Inhibitors for Cancer Therapy: Recent Advances and Future Prospects. Biomolecules 2025; 15:240. [PMID: 40001543 PMCID: PMC11853158 DOI: 10.3390/biom15020240] [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: 01/20/2025] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Cancer management has traditionally depended on chemotherapy as the mainstay of treatment; however, recent advancements in targeted therapies and immunotherapies have offered new options. Ubiquitin-specific proteases (USPs) have emerged as promising therapeutic targets in cancer treatment due to their crucial roles in regulating protein homeostasis and various essential cellular processes. This review covers the following: (1) the structural and functional characteristics of USPs, highlighting their involvement in key cancer-related pathways, and (2) the discovery, chemical structures, mechanisms of action, and potential clinical implications of USP inhibitors in cancer therapy. Particular attention is given to the role of USP inhibitors in enhancing cancer immunotherapy, e.g., modulation of the tumor microenvironment, effect on regulatory T cell function, and influence on immune checkpoint pathways. Furthermore, this review summarizes the current progress and challenges of clinical trials involving USP inhibitors as cancer therapy. We also discuss the complexities of achieving target selectivity, the ongoing efforts to develop more specific and potent USP inhibitors, and the potential of USP inhibitors to overcome drug resistance and synergize with existing cancer treatments. We finally provide a perspective on future directions in targeting USPs, including the potential for personalized medicine based on specific gene mutations, underscoring their significant potential for enhancing cancer treatment. By elucidating their mechanisms of action, clinical progress, and potential future applications, we hope that this review could serve as a useful resource for both basic scientists and clinicians in the field of cancer therapeutics.
Collapse
Affiliation(s)
- Mohamad Bakkar
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences (EACPHS), Wayne State University, Detroit, MI 48201, USA; (M.B.); (K.B.); (N.D.K.); (A.S.); (S.D.)
- Division of Pediatric Hematology and Oncology, Children’s Hospital of Michigan, Detroit, MI 48201, USA
| | - Sara Khalil
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.K.); (Q.P.D.)
| | - Komal Bhayekar
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences (EACPHS), Wayne State University, Detroit, MI 48201, USA; (M.B.); (K.B.); (N.D.K.); (A.S.); (S.D.)
| | - Narva Deshwar Kushwaha
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences (EACPHS), Wayne State University, Detroit, MI 48201, USA; (M.B.); (K.B.); (N.D.K.); (A.S.); (S.D.)
| | - Amirreza Samarbakhsh
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences (EACPHS), Wayne State University, Detroit, MI 48201, USA; (M.B.); (K.B.); (N.D.K.); (A.S.); (S.D.)
| | - Sadaf Dorandish
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences (EACPHS), Wayne State University, Detroit, MI 48201, USA; (M.B.); (K.B.); (N.D.K.); (A.S.); (S.D.)
| | - Holly Edwards
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA;
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute (KCI), Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Q. Ping Dou
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.K.); (Q.P.D.)
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA;
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute (KCI), Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Yubin Ge
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.K.); (Q.P.D.)
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA;
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute (KCI), Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Navnath S. Gavande
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences (EACPHS), Wayne State University, Detroit, MI 48201, USA; (M.B.); (K.B.); (N.D.K.); (A.S.); (S.D.)
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute (KCI), Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
6
|
Liu F, Wang Y, Xia L, Sun C, Li Y, Xia Y. Immunological characterization and prognostic of colon cancer evaluated by angiogenesis-related features: a computational analysis and in vitro experiments. Discov Oncol 2025; 16:101. [PMID: 39881026 PMCID: PMC11780071 DOI: 10.1007/s12672-025-01835-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 01/20/2025] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND Diseases are often caused by multiple factors, angiogenesis-related genes (ARGs) have been shown to be associated with cancer, however, their role in colon cancer had not been fully explored. This study investigated potential biomarkers based on ARGs to improve prognosis and treatment effect in colon cancer. METHODS ARGs associated with colon cancer prognosis were identified using Cox regression analysis and LASSO analysis. Furthermore, a prognostic model was constructed in colon cancer based on the 3 ARGs, and its biological function were analyzed. We evaluated the differences in tumor immune microenvironment based on prognostic signature. Finally, cell experiments confirmed the function of genes in colon cancer. RESULTS The prognostic value of ARGs in colon cancer patients has been comprehensively analyzed for the first time and identified 3 ARGs with prognostic values. A prognosis risk model was constructed based on 3 ARGs and its prognostic value was validated on an independent external colon cancer dataset. In colon cancer patients, this prognostic feature was an independent risk factor and was significantly correlated with clinical feature information of colon cancer patients. This feature was also related to the immune microenvironment of colon cancer. Cell experiments showed that high expression of TNF Receptor Superfamily Member 1B (TNFRSF1B) significantly promoted apoptosis and inhibited proliferation of colon cancer cells. Therefore, TNFRSF1B may become an important regulatory factor in the progression of colon cancer by participating in intracellular functional regulation. CONCLUSIONS This study constructed a prognostic risk model based on three ARGs and for the first time discovered that TNFRSF1B may become an important regulatory factor in cancer progression by participating in intracellular functional regulation.
Collapse
Affiliation(s)
- Fei Liu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Oncology, Anhui Public Health Clinical Center, Hefei, China
| | - Yi Wang
- Department of Oncology, The Third Affiliated Hospital of Anhui Medical University, Anhui, China
| | - Leiming Xia
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Hematology, Anhui Public Health Clinical Center, Hefei, China
| | - Chen Sun
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Oncology, Anhui Public Health Clinical Center, Hefei, China
| | - Yun Li
- School of Biology and Pharmaceutical Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Yunhong Xia
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
- Department of Oncology, Anhui Public Health Clinical Center, Hefei, China.
| |
Collapse
|
7
|
Yuan Y, Xue M, Zhou F, Gu L. USP13 promotes acute myeloid leukemia cell proliferation and autophagy by promoting ATG5. Tissue Cell 2024; 91:102494. [PMID: 39216303 DOI: 10.1016/j.tice.2024.102494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVE To elucidate the role of USP13 in acute myeloid leukemia (AML) by investigating its effects on cell growth, apoptosis and autophagy, and to explore the underlying mechanisms. METHODS The expression of USP13 in AML cells was assessed using quantitative PCR (qPCR) and immunoblotting. Cell Counting Kit-8 (CCK-8) and Edu staining were employed to evaluate the impact of USP13 on AML cell growth. Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and immunostaining assays were conducted to examine the effects of USP13 on apoptosis and autophagy in AML cells, and immunoblot assays were performed to determine the potential underlying mechanistic pathway. RESULTS USP13 expression was significantly elevated in AML cells, correlating with enhanced cell proliferation and resistance to apoptosis. Moreover, USP13 promoted autophagy in AML cells. Mechanistically, USP13 was found to be associated with upregulating ATG5 expression, which promoted AML progression. CONCLUSION USP13 promotes AML cell growth and autophagy by upregulating ATG5.
Collapse
Affiliation(s)
- Yuchu Yuan
- Inspection Division, Changshu blood bank, Changshu, Jiangsu 215500, China
| | - Meizhu Xue
- Department of Paediatrics, Changshu NO.1 People's Hospital, Changshu, Jiangsu 215500, China.
| | - Feng Zhou
- Inspection Division, Changshu Medicine Examination Institute, Changshu, Jiangsu 215500, China
| | - Lifang Gu
- Inspection Division, Changshu blood bank, Changshu, Jiangsu 215500, China
| |
Collapse
|
8
|
Guo F, Ji X, Xiong C, Sun H, Liang Z, Yan-Do R, Gai B, Gao F, Huang L, Li Z, Kuang BY, Shi P. Single-cell encoded gene silencing for high-throughput combinatorial siRNA screening. Nat Commun 2024; 15:9985. [PMID: 39562763 DOI: 10.1038/s41467-024-53419-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 10/09/2024] [Indexed: 11/21/2024] Open
Abstract
The use of combinatorial siRNAs shows great promise for drug discovery, but the identification of safe and effective siRNA combinations remains challenging. Here, we develop a massively multiplexed technology for systematic screening of siRNA-based cocktail therapeutics. We employ composite micro-carriers that are responsive to near infrared light and magnetic field to achieve photoporation-facilitated siRNA transfection to individual cells. Thus, randomized gene silencing by different siRNA formulations can be performed with high-throughput single-cell-based analyses. For screening anti-cancer siRNA cocktails, we test more than 1300 siRNA combinations for knocking down multiple genes related to tumor growth, discovering effective 3-siRNA formulations with an emphasis on the critical role of inhibiting Cyclin D1 and survivin, along with their complementary targets for synergic efficacy. This approach enables orders of magnitude reduction in time and cost associated with largescale siRNA screening, and resolves key insights to siRNA pharmacology that are not permissive to existing methods.
Collapse
Affiliation(s)
- Feng Guo
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, 999077, China
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering, Hong Kong Science Park, Hong Kong SAR, 999077, China
| | - Xianglin Ji
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, 999077, China
| | - Chuxiao Xiong
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, 999077, China
| | - Hailiang Sun
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, 999077, China
| | - Zhenghua Liang
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, 999077, China
| | - Richard Yan-Do
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, 999077, China
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering, Hong Kong Science Park, Hong Kong SAR, 999077, China
| | - Baowen Gai
- Department of Colorectal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Feng Gao
- Department of Colorectal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
| | - Linfeng Huang
- Wang-Cai Biochemistry Lab, Division of Natural and Applied Sciences & Global Health Research Center, Duke Kunshan University, Kunshan, Jiangsu, China
| | - Zhongping Li
- Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| | - Becki Yi Kuang
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, 999077, China
| | - Peng Shi
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR, 999077, China.
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering, Hong Kong Science Park, Hong Kong SAR, 999077, China.
- Center of Super-Diamond and Advanced Films (COSDAF), City University of Hong Kong, Kowloon, Hong Kong SAR, 999077, China.
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, 518000, China.
| |
Collapse
|
9
|
Xu Z, Lei Z, Peng S, Fu X, Xu Y, Pan G. Dysregulation of deubiquitinases in gastric cancer progression. Front Oncol 2024; 14:1456710. [PMID: 39605891 PMCID: PMC11598704 DOI: 10.3389/fonc.2024.1456710] [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: 06/29/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024] Open
Abstract
Gastric cancer (GC), characterized by a high incidence rate, poses significant clinical challenges owing to its poor prognosis despite advancements in diagnostic and therapeutic approaches. Therefore, a comprehensive understanding of the molecular mechanisms driving GC progression is crucial for identifying predictive markers and defining treatment targets. Deubiquitinating enzymes (DUBs), also called deubiquitinases, function as reverse transcriptases within the ubiquitin-proteasome system to counteract protein degradation. Recent findings suggest that DUB dysregulation could be a crucial factor in GC pathogenesis. In this review, we examined recent research findings on DUBs in the context of GC, elucidating their molecular characteristics, categorizations, and roles while also exploring the potential mechanisms underlying their dysregulation in GC. Furthermore, we assessed the therapeutic efficacy of DUB inhibitors in treating malignancies and evaluated the prevalence of aberrant DUB expression in GC.
Collapse
Affiliation(s)
| | | | | | | | | | - Guoqing Pan
- First Affiliated Hospital of Kunming Medical University, Department of Pathology, Kunming, China
| |
Collapse
|
10
|
Chen J, Shan W, Jia Q, Chen Y, Jiang W, Tian Y, Huang X, Li X, Wang Z, Xia B. USP33 facilitates the ovarian cancer progression via deubiquitinating and stabilizing CBX2. Oncogene 2024; 43:3170-3183. [PMID: 39256572 DOI: 10.1038/s41388-024-03151-9] [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: 05/08/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 09/12/2024]
Abstract
Post-translational modifications of proteins play a pivotal role in both the initiation and progression of ovarian cancer. Despite the recognition of USP33 as a significant factor in various cancers, its specific function and underlying mechanisms in ovarian cancer remain elusive. Proteomics and ubiquitinomics approaches were coupled to screen novel substrate proteins directly regulated by USP33. Our findings unveil that USP33 was observed to eliminate K27- and K48-linked ubiquitin chains from CBX2 at the K277 position. Notably, acetylation of CBX2 at K199, catalyzed by lysine acetyltransferase GCN5, was found to enhance its interaction with USP33, subsequently promoting further deubiquitination and stabilization. Functionally, our experiments demonstrate that USP33 significantly enhances ovarian cancer proliferation and metastasis in a CBX2-dependent manner. Furthermore, analysis revealed a direct positive correlation between the expression levels of USP33 and CBX2 proteins in human specimens, with elevated levels being associated with reduced survival rates in ovarian cancer patients. These findings elucidate the mechanism by which USP33 augments ovarian cancer progression through the stabilization of CBX2, underscoring the USP33-CBX2 axis as a promising therapeutic target in ovarian cancer management.
Collapse
Affiliation(s)
- Jiming Chen
- Department of Gynecology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Wulin Shan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Department of Obstetrics and Gynecology, Anhui Provincial Cancer Hospital, Hefei, Anhui, China
| | - Qiucheng Jia
- Department of Gynecology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Yao Chen
- Department of Obstetrics and Gynecology, Anhui Provincial Cancer Hospital, Hefei, Anhui, China
- Bengbu Medical University, Bengbu, Anhui, China
| | - Wenjing Jiang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Department of Obstetrics and Gynecology, Anhui Provincial Cancer Hospital, Hefei, Anhui, China
| | - Yuan Tian
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Department of Obstetrics and Gynecology, Anhui Provincial Cancer Hospital, Hefei, Anhui, China
| | - Xu Huang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Department of Obstetrics and Gynecology, Anhui Provincial Cancer Hospital, Hefei, Anhui, China
| | - Xiaoyu Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Department of Obstetrics and Gynecology, Anhui Provincial Cancer Hospital, Hefei, Anhui, China
| | - Zengying Wang
- Department of Obstetrics and Gynecology, Anhui Provincial Cancer Hospital, Hefei, Anhui, China
- Bengbu Medical University, Bengbu, Anhui, China
| | - Bairong Xia
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
- Department of Obstetrics and Gynecology, Anhui Provincial Cancer Hospital, Hefei, Anhui, China.
- Bengbu Medical University, Bengbu, Anhui, China.
| |
Collapse
|
11
|
Chen C, Kang D, Chen Z, Shi P, Li Y, Qian S. DLD is a potential therapeutic target for COVID-19 infection in diffuse large B-cell lymphoma patients. Apoptosis 2024; 29:1696-1708. [PMID: 38581529 PMCID: PMC11416400 DOI: 10.1007/s10495-024-01959-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2024] [Indexed: 04/08/2024]
Abstract
Since the discovery of copper induces cell death(cuprotosis) in 2022, it has been one of the biggest research hotspots. cuprotosis related genes (CRGs) has been demonstrated to be a potential therapeutic target for cancer, however, the molecular mechanism of CRGs in coronavirus disease 2019 (COVID-19) infected in DLBCL patients has not been reported yet. Therefore, our research objective is first to elucidate the mechanism and role of CRGs in COVID-19. Secondly, we conducted univariate and multivariate analysis and machine learning to screen for CRGs with common expression differences in COVID-19 and DLBCL. Finally, the functional role and immune mechanism of genes in DLBCL were confirmed through cell experiments and immune analysis. The research results show that CRGs play an important role in the occurrence and development of COVID-19. Univariate analysis and machine learning confirm that dihydrolipoamide dehydrogenase (DLD) is the common key gene of COVID-19 and DLBCL. Inhibiting the expression of DLD can significantly inhibit the cycle progression and promote cell apoptosis of DLBCL cells and can target positive regulation of Lysine-specific demethylase 1 (LSD1, also known as KDM1A) to inhibit the proliferation of DLBCL cells and promote cell apoptosis. The immune analysis results show that high-expression of DLD may reduce T cell-mediated anti-tumor immunity by regulating immune infiltration of CD8 + T cells and positively regulating immune checkpoints LAG3 and CD276. Reducing the expression of DLD can effectively enhance T cell-mediated anti-tumor immunity, thereby clearing cancer cells and preventing cancer growth. In conclusion, DLD may be a potential therapeutic target for COVID-19 infection in DLBCL patients. Our research provides a theoretical basis for improving the clinical treatment of COVID-19 infection in DLBCL.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/virology
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- COVID-19/immunology
- COVID-19/genetics
- COVID-19/virology
- Apoptosis
- SARS-CoV-2
- Cell Line, Tumor
- Gene Expression Regulation, Neoplastic
- Machine Learning
Collapse
Affiliation(s)
- Can Chen
- Department of Hematology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Dandan Kang
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhenzhen Chen
- Department of Hematology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Pengfei Shi
- Department of Hematology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Yun Li
- Team of neonatal & infant development, health and nutrition, NDHN. School of Biology and Pharmaceutical Engineering, Wuhan Polytechnic University, Wuhan, China.
- Kindstar Global Precision Medicine Institute, Wuhan, China.
| | - Shenxian Qian
- Department of Hematology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China.
- School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
12
|
Hu Z, Wu Y, Sun X, Tong Y, Qiu H, Zhuo E. ARMCX1 inhibits lung adenocarcinoma progression by recruiting FBXW7 for c-Myc degradation. Biol Direct 2024; 19:82. [PMID: 39285446 PMCID: PMC11406719 DOI: 10.1186/s13062-024-00532-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 09/09/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Armadillo Repeat Containing X-Linked 1 (ARMCX1), a member of the ARM Repeat X-linked protein family, exerts inhibitory function in various tumors. However, its biological role in lung adenocarcinoma (LUAD) and the underlying molecular mechanisms require further exploration. METHODS LUAD tissue microarrays and bioinformatic databases were used to evaluate the relationship between ARMCX1 and clinicopathological features. The influence of ARMCX1 on LUAD cell proliferation, migration, and invasion in vitro was determined by colony formation, CCK-8, EdU incorporation, cell cycle, wound healing, and Transwell assays. The impact of ARMCX1 on LUAD cell growth and metastasis in vivo was determined by subcutaneously transplanted tumor and pulmonary metastasis assays. Western blot, immunoprecipitation, immunofluorescence, cycloheximide, and proteasome inhibitor assays were finally conducted to explore the potential underlying molecular mechanisms. RESULTS ARMCX1 expression was downregulated in clinical LUAD samples due to which patient prognoses were poor. Functional experiments indicated that ARMCX1 overexpression inhibited the growth and metastasis of LUAD cells in vitro and in vivo. The molecular mechanism suggested that ARMCX1 recruits the E3 ubiquitin ligase FBXW7 for mediating ubiquitinated degradation of c-Myc, suppressing its nuclear accumulation, and ultimately inactivating cell cycle and epithelial-mesenchymal transition (EMT) signals. CONCLUSION ARMCX1 inhibits LUAD cell proliferation and metastasis by interacting with c-Myc and enhancing its ubiquitination and degradation. Consequently, it can act as a tumor suppressor in this disease. These results suggest that ARMCX1 is a potential target in the treatment of LUAD.
Collapse
Affiliation(s)
- Zhe Hu
- Department of Second Ward Oncology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
| | - Yilin Wu
- Department of Cardiology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
| | - Xiaoou Sun
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, China
| | - Yanli Tong
- Department of Pharmacy, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
| | - Houkuang Qiu
- Department of Laboratory Medicine, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
| | - Enqing Zhuo
- Department of Second Ward Oncology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China.
| |
Collapse
|
13
|
Lu P, Yang L, Lei Y, Zhao Y, Tang Z, Shang P, Zhou X, Wang P, Wang W, Feng F, Zhang Q. Low expression of Lnc-ENST00000535078 inhibits the migration, invasion, and enhances apoptosis of CTPE-induced malignantly transformed BEAS-2B cells. Toxicol Res (Camb) 2024; 13:tfae121. [PMID: 39175813 PMCID: PMC11336064 DOI: 10.1093/toxres/tfae121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 06/28/2024] [Accepted: 07/30/2024] [Indexed: 08/24/2024] Open
Abstract
Long non-coding RNA (LncRNA) plays an important role in malignant transformation of cells. This study aimed to explore the role of Lnc-ENST00000535078 in the malignant transformation of immortalized human bronchial epithelial cells (BEAS-2B) induced by coal tar pitch extract (CTPE). The malignant transformation model of BEAS-2B cells exposed to CTPE. Cell proliferation was examined by Cell counting kit-8 (CCK-8) assay. Colony formation assay was used to assess the colony of cells. Cell migration and invasion were detected by Transwell analysis. Cell cycle progression and apoptotic status were assessed by flow cytometry. Differentially expressed genes were screened by RNA sequencing. The results showed that Lnc-ENST00000535078 expression was highest in malignantly transformed BEAS-2B cells passaged to the 30th generation. Knockdown of Lnc-ENST00000535078 inhibited the migration, invasion and anti-apoptotic abilities of malignantly transformed BEAS-2B cells. Transcriptome analysis found 608 differential genes. CCND1 and FOS genes were screened out because of their levels were positive correlation with the expression of Lnc-ENST00000535078, which were consistent with the RNA sequencing results. In conclusion, Low expression of Lnc-ENST00000535078 inhibits the migration and invasion of malignant transformed BEAS-2B cells and promotes apoptosis in these cells. Lnc-ENST00000556926 might affect the malignant transformation of cells through the regulation of CCND1 and FOS. This study may provide a potential target for intervention in CTPE-induced lung cancer.
Collapse
Affiliation(s)
- Ping Lu
- Department of Toxicology, Zhengzhou University School of Public Health, Zhengzhou, Henan Province 450001, China
| | - Liu Yang
- Qinhuangdao Center for Disease Control and Prevention, Qinhuangdao, Hebei Province 066000, China
| | - Yanting Lei
- Department of Toxicology, Zhengzhou University School of Public Health, Zhengzhou, Henan Province 450001, China
| | - Yuezeng Zhao
- Department of Toxicology, Zhengzhou University School of Public Health, Zhengzhou, Henan Province 450001, China
| | - Zhihao Tang
- Department of Toxicology, Zhengzhou University School of Public Health, Zhengzhou, Henan Province 450001, China
| | - Pingping Shang
- Key Laboratory of Tobacco Chemistry, Zhengzhou Tobacco Research Institute, CNC, Hi-Tech Development Zone, Zhengzhou, Henan Province 450001, China
| | - Xiaolei Zhou
- Department of Pulmonary Medicine, Henan Provincial Chest Hospital, Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Pengpeng Wang
- Department of Occupational Health, Zhengzhou University School of Public Health, Hi-Tech Development Zone, Zhengzhou, Henan Province 450001, China
| | - Wei Wang
- Department of Occupational Health, Zhengzhou University School of Public Health, Hi-Tech Development Zone, Zhengzhou, Henan Province 450001, China
| | - Feifei Feng
- Department of Toxicology, Zhengzhou University School of Public Health, Zhengzhou, Henan Province 450001, China
| | - Qiao Zhang
- Department of Toxicology, Zhengzhou University School of Public Health, Zhengzhou, Henan Province 450001, China
| |
Collapse
|
14
|
Zang Y, Tian Z, Wang D, Li Y, Zhang W, Ma C, Liao Z, Gao W, Qian L, Xu X, Jia J, Liu Z. METTL3-mediated N 6-methyladenosine modification of STAT5A promotes gastric cancer progression by regulating KLF4. Oncogene 2024; 43:2338-2354. [PMID: 38879589 PMCID: PMC11271408 DOI: 10.1038/s41388-024-03085-2] [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: 02/19/2024] [Revised: 06/02/2024] [Accepted: 06/07/2024] [Indexed: 07/21/2024]
Abstract
N6-methyladenosine (m6A) is the predominant post-transcriptional RNA modification in eukaryotes and plays a pivotal regulatory role in various aspects of RNA fate determination, such as mRNA stability, alternative splicing, and translation. Dysregulation of the critical m6A methyltransferase METTL3 is implicated in tumorigenesis and development. Here, this work showed that METTL3 is upregulated in gastric cancer tissues and is associated with poor prognosis. METTL3 methylates the A2318 site within the coding sequence (CDS) region of STAT5A. IGF2BP2 recognizes and binds METTL3-mediated m6A modification of STAT5A through its GXXG motif in the KH3 and KH4 domains, leading to increased stability of STAT5A mRNA. In addition, both METTL3 and IGF2BP2 are positively correlated with STAT5A in human gastric cancer tissue samples. Helicobacter pylori infection increased the expression level of METTL3 in gastric cancer cells, thereby leading to the upregulation of STAT5A. Functional studies indicated that STAT5A overexpression markedly enhances the proliferation and migration of GC cells, whereas STAT5A knockdown has inhibitory effects. Further nude mouse experiments showed that STAT5A knockdown effectively inhibits the growth and metastasis of gastric cancer in vivo. Moreover, as a transcription factor, STAT5A represses KLF4 transcription by binding to its promoter region. The overexpression of KLF4 can counteract the oncogenic impact of STAT5A. Overall, this study highlights the crucial role of m6A in gastric cancer and provides potential therapeutic targets for gastric cancer.
Collapse
Affiliation(s)
- Yichen Zang
- Department of Biochemistry and Molecular Biology, Key Laboratory for Experimental Teratology of Chinese Ministry of Education, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhuangfei Tian
- Department of Biochemistry and Molecular Biology, Key Laboratory for Experimental Teratology of Chinese Ministry of Education, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Dandan Wang
- Department of Microbiology, Key Laboratory for Experimental Teratology of Chinese Ministry of Education, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yaxuan Li
- Department of Biochemistry and Molecular Biology, Key Laboratory for Experimental Teratology of Chinese Ministry of Education, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wenhui Zhang
- School of Clinical Medicine, Qingdao University, Qingdao, China
| | - Cunying Ma
- Department of Biochemistry and Molecular Biology, Key Laboratory for Experimental Teratology of Chinese Ministry of Education, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhenzhi Liao
- Department of Biochemistry and Molecular Biology, Key Laboratory for Experimental Teratology of Chinese Ministry of Education, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wenrong Gao
- Department of Microbiology, Key Laboratory for Experimental Teratology of Chinese Ministry of Education, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lilin Qian
- Department of Biochemistry and Molecular Biology, Key Laboratory for Experimental Teratology of Chinese Ministry of Education, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xia Xu
- Department of Biochemistry and Molecular Biology, Key Laboratory for Experimental Teratology of Chinese Ministry of Education, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jihui Jia
- Department of Microbiology, Key Laboratory for Experimental Teratology of Chinese Ministry of Education, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhifang Liu
- Department of Biochemistry and Molecular Biology, Key Laboratory for Experimental Teratology of Chinese Ministry of Education, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
15
|
Yang C, Zhang W, Xiang S, Chen L, Chun J, Chen H. Navel orange peel essential oil inhibits the growth and progression of triple negative breast cancer. BMC Complement Med Ther 2024; 24:233. [PMID: 38877505 PMCID: PMC11177363 DOI: 10.1186/s12906-024-04525-y] [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: 10/30/2023] [Accepted: 05/28/2024] [Indexed: 06/16/2024] Open
Abstract
BACKGROUND Triple Negative Breast Cancer (TNBC) is a particular type of breast cancer with the highest mortality rate. Essential oils are concerned more and more as potential anti-cancer drugs. METHODS TNBC cells were treated with different concentrations of navel orange peel essential oil (NOPEO), and then a variety of experiments were performed to investigate the changes in the growth and progression of TNBC cells. MTT assay was performed to detect the proliferation of TNBC cells. The changes of cell cycle and apoptosis were analyzed by FACS. In order to explored the migration of TNBC cells, scratch wound assay was carried out. Western blotting and qPCR were used to examine the expression of proteins and mRNA of related genes. Furthermore, RNA-seq was used to analyze the altered genes and explored the possible signal pathway. RESULTS NOPEO demonstrated dose- and time-dependent suppression of TNBC cell growth. TNBC cells showed an increased percentage of G2/M-phase cells and the protein levels of CyclinB1 and CyclinD1 were decreased after NOPEO treatment. The apoptotic cells were increased in the NOPEO treated TNBC cells. The migration mobility was significantly inhibited by NOPEO. In total, 1376 genes were found to be up-regulated and 1335 genes were down-regulated after NOPEO treatment. According to KEGG and GO pathways, the differentially expressed genes were related to MAPK, Jak/stat and FoxQ signaling pathways. CONCLUSION This investigation explored the bio-activity and molecular mechanisms of NOPEO against TNBC cells. These results indicated that NOPEO could suppress TNBC growth and migration perhaps via the MAPK and Jak/stat signaling pathways, which may provide theoretical reference for anticancer drug development. NOPEO may be a potential natural product for the chemotherapeutic of TNBC.
Collapse
Affiliation(s)
- Chao Yang
- Oncology Research Center, Jiangxi Provincial Key Laboratory of Traditional Chinese Medicine Diagnosis and Rehabilitation of Malignant Tumors, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
- College of Life Sciences, National Navel Orange Engineering Research Center, Gannan Normal University, Ganzhou, 341000, China
| | - Wenwen Zhang
- Oncology Research Center, Jiangxi Provincial Key Laboratory of Traditional Chinese Medicine Diagnosis and Rehabilitation of Malignant Tumors, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Shi Xiang
- Oncology Research Center, Jiangxi Provincial Key Laboratory of Traditional Chinese Medicine Diagnosis and Rehabilitation of Malignant Tumors, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Lai Chen
- Oncology Research Center, Jiangxi Provincial Key Laboratory of Traditional Chinese Medicine Diagnosis and Rehabilitation of Malignant Tumors, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Jiong Chun
- College of Life Sciences, National Navel Orange Engineering Research Center, Gannan Normal University, Ganzhou, 341000, China.
| | - Hui Chen
- Oncology Research Center, Jiangxi Provincial Key Laboratory of Traditional Chinese Medicine Diagnosis and Rehabilitation of Malignant Tumors, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
- College of Life Sciences, National Navel Orange Engineering Research Center, Gannan Normal University, Ganzhou, 341000, China.
| |
Collapse
|
16
|
Xu C, Cao G, Zhang H, Bai M, Yi X, Qu X. Avellanin A Has an Antiproliferative Effect on TP-Induced RWPE-1 Cells via the PI3K-Akt Signalling Pathway. Mar Drugs 2024; 22:275. [PMID: 38921586 PMCID: PMC11205091 DOI: 10.3390/md22060275] [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: 05/15/2024] [Revised: 06/10/2024] [Accepted: 06/10/2024] [Indexed: 06/27/2024] Open
Abstract
Cyclic pentapeptide compounds have garnered much attention as a drug discovery resource. This study focused on the characterization and anti-benign prostatic hyperplasia (BPH) properties of avellanin A from Aspergillus fumigatus fungus in marine sediment samples collected in the Beibu Gulf of Guangxi Province in China. The antiproliferative effect and molecular mechanism of avellanin A were explored in testosterone propionate (TP)-induced RWPE-1 cells. The transcriptome results showed that avellanin A significantly blocked the ECM-receptor interaction and suppressed the downstream PI3K-Akt signalling pathway. Molecular docking revealed that avellanin A has a good affinity for the cathepsin L protein, which is involved in the terminal degradation of extracellular matrix components. Subsequently, qRT-PCR analysis revealed that the expression of the genes COL1A1, COL1A2, COL5A2, COL6A3, MMP2, MMP9, ITGA2, and ITGB3 was significantly downregulated after avellanin A intervention. The Western blot results also confirmed that it not only reduced ITGB3 and FAK/p-FAK protein expression but also inhibited PI3K/p-PI3K and Akt/p-Akt protein expression in the PI3K-Akt signalling pathway. Furthermore, avellanin A downregulated Cyclin D1 protein expression and upregulated Bax, p21WAF1/Cip1, and p53 proapoptotic protein expression in TP-induced RWPE-1 cells, leading to cell cycle arrest and inhibition of cell proliferation. The results of this study support the use of avellanin A as a potential new drug for the treatment of BPH.
Collapse
Affiliation(s)
- Chang Xu
- Faculty of Pharmacy/Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China; (C.X.); (G.C.); (H.Z.)
| | - Guangping Cao
- Faculty of Pharmacy/Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China; (C.X.); (G.C.); (H.Z.)
- Guangxi Key Laboratory of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Hong Zhang
- Faculty of Pharmacy/Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China; (C.X.); (G.C.); (H.Z.)
| | - Meng Bai
- Faculty of Pharmacy/Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China; (C.X.); (G.C.); (H.Z.)
- Guangxi Key Laboratory of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Xiangxi Yi
- Faculty of Pharmacy/Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China; (C.X.); (G.C.); (H.Z.)
- Guangxi Key Laboratory of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Xinjian Qu
- Faculty of Pharmacy/Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China; (C.X.); (G.C.); (H.Z.)
- Guangxi Key Laboratory of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China
| |
Collapse
|
17
|
Wang Y, Chen X, Li Y, Zhang Z, Xia L, Jiang J, Chai Y, Wang Z, Wan Y, Li T, Jin F, Li H. SLC27A2 is a potential immune biomarker for hematological tumors and significantly regulates the cell cycle progression of diffuse large B-cell lymphoma. BMC Med Genomics 2024; 17:105. [PMID: 38664735 PMCID: PMC11046844 DOI: 10.1186/s12920-024-01853-3] [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: 10/31/2023] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Research on the fatty acid metabolism related gene SLC27A2 is currently mainly focused on solid tumors, and its mechanism of action in hematological tumors has not been reported. METHOD This study aims to explore the pathological and immune mechanisms of the fatty acid metabolism related gene SLC27A2 in hematological tumors and verify its functional role in hematological tumors through cell experiments to improve treatment decisions and clinical outcomes of hematological tumors. RESULT This study identified the fatty acid metabolism related gene SLC27A2 as a common differentially expressed gene between DLBCL and AML. Immune microenvironment analysis showed that SLC27A2 was significantly positively correlated with T cell CD4 + , T cell CD8 + , endothelial cells, macrophages, and NK cells in DLBCL. In AML, there is a significant negative correlation between SLC27A2 and B cells, T cell CD8 + , and macrophages. SLC27A2 participates in the immune process of hematological tumors through T cell CD8 + and macrophages. The GESA results indicate that high expression of SLC27A2 is mainly involved in the fatty acid pathway, immune pathway, and cell cycle pathway of DLBCL. The low expression of SLC27A2 is mainly involved in the immune pathway of AML. Therefore, SLC27A2 is mainly involved in the pathological mechanisms of hematological tumors through immune pathways, and cell experiments have also confirmed that SLC27A2 is involved in the regulation of DLBCL cells. CONCLUSION In summary, our research results comprehensively report for the first time the mechanism of action of SLC27A2 in the immune microenvironment of DLBCL and AML, and for the first time verify the cycle and apoptotic effects of the fatty acid related gene SLC27A2 in DLBCL cells through cell experiments. Research can help improve the treatment of AML and DLBCL patients.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/pathology
- Cell Cycle
- Tumor Microenvironment/immunology
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Gene Expression Regulation, Neoplastic
- Hematologic Neoplasms/genetics
- Hematologic Neoplasms/immunology
- Hematologic Neoplasms/pathology
- Cell Line, Tumor
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/metabolism
- Fatty Acids/metabolism
Collapse
Affiliation(s)
- Yi Wang
- Department of Oncology, The Third Affiliated Hospital of Anhui Medical, Anhui, China
| | - Xue Chen
- Graduate School Internal Medicine, Bengbu Medical College, Anhui, China
| | - Yun Li
- Kindstar Global Precision Medicine Institute, Wuhan, China
- Department of Scientific Research Project, Wuhan Kindstar Medical Laboratory Co., Ltd, Wuhan, Hubei, China
| | - Zhixue Zhang
- Department of Hematology, The Ji'an Central Hospital, Jiangxi, China
| | - Leiming Xia
- Department of Hematology, The First Affiliated Hospital of Anhui Medical, Anhui, China
| | - Jiang Jiang
- Department of Ophthalmology, The First Affiliated Hospital of Anhui Medical, Hefei, Anhui, China
| | - Yuqin Chai
- Department of Oncology, The Third Affiliated Hospital of Anhui Medical, Anhui, China
| | - Ziming Wang
- Department of Oncology, The Third Affiliated Hospital of Anhui Medical, Anhui, China
| | - Yu Wan
- Department of Oncology, The Third Affiliated Hospital of Anhui Medical, Anhui, China
| | - Tongyu Li
- Ningbo Clinical Research Center for Hematologic Malignancies, the First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Fengbo Jin
- Department of Hematology, The First Affiliated Hospital of Anhui Medical, Anhui, China.
| | - Hongxia Li
- Department of Oncology, The Third Affiliated Hospital of Anhui Medical, Anhui, China.
- Graduate School Internal Medicine, Bengbu Medical College, Anhui, China.
| |
Collapse
|
18
|
Li KQ, Bai X, Ke AT, Ding SQ, Zhang CD, Dai DQ. Ubiquitin-specific proteases: From biological functions to potential therapeutic applications in gastric cancer. Biomed Pharmacother 2024; 173:116323. [PMID: 38401523 DOI: 10.1016/j.biopha.2024.116323] [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: 12/04/2023] [Revised: 02/17/2024] [Accepted: 02/19/2024] [Indexed: 02/26/2024] Open
Abstract
Deubiquitination, a post-translational modification regulated by deubiquitinases, is essential for cancer initiation and progression. Ubiquitin-specific proteases (USPs) are essential elements of the deubiquitinase family, and are overexpressed in gastric cancer (GC). Through the regulation of several signaling pathways, such as Wnt/β-Catenin and nuclear factor-κB signaling, and the promotion of the expression of deubiquitination- and stabilization-associated proteins, USPs promote the proliferation, metastasis, invasion, and epithelial-mesenchymal transition of GC. In addition, the expression of USPs is closely related to clinicopathological features, patient prognosis, and chemotherapy resistance. USPs therefore could be used as prognostic biomarkers. USP targeting small molecule inhibitors have demonstrated strong anticancer activity. However, they have not yet been tested in the clinic. This article provides an overview of the latest fundamental research on USPs in GC, aiming to enhance the understanding of how USPs contribute to GC progression, and identifying possible targets for GC treatment to improve patient survival.
Collapse
Affiliation(s)
- Kai-Qiang Li
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province 110032, China
| | - Xiao Bai
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province 110032, China
| | - Ang-Ting Ke
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province 110032, China
| | - Si-Qi Ding
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province 110032, China
| | - Chun-Dong Zhang
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province 110032, China
| | - Dong-Qiu Dai
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province 110032, China; Cancer Center, the Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province 110032, China.
| |
Collapse
|
19
|
Shou Z, Bai Z, Huo K, Zheng S, Shen Y, Zhou H, Huang X, Meng H, Xu C, Wu S, Li N, Chen C. Immobilizing c(RGDfc) on the surface of metal-phenolic networks by thiol-click reaction for accelerating osteointegration of implant. Mater Today Bio 2024; 25:101017. [PMID: 38495914 PMCID: PMC10940948 DOI: 10.1016/j.mtbio.2024.101017] [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: 12/05/2023] [Revised: 02/14/2024] [Accepted: 03/02/2024] [Indexed: 03/19/2024] Open
Abstract
The limited osteointegration often leads to the failure of implant, which can be improved by fixing bioactive molecules onto the surface, such as arginyl-glycyl-aspartic acid (RGD): a cell adhesion motif. Metal-Phenolic Networks (MPNs) have garnered increasing attention from different disciplines in recent years due to their simple and rapid process for depositing on various substrates or particles with different shapes. However, the lack of cellular binding sites on MPNs greatly blocks its application in tissue engineering. In this study, we present a facile and efficient approach for producing PC/Fe@c(RGDfc) composite coatings through the conjugation of c(RGDfc) peptides onto the surface of PC/Fe-MPNs utilizing thiol-click reaction. By combined various techniques (ellipsometry, X-ray photoelectron spectroscopy, Liquid Chromatography-Mass Spectrometry, water contact angle, scanning electronic microscopy, atomic force microscopy) the physicochemical properties (composition, coating mechanism and process, modulus and hydrophilicity) of PC/Fe@c(RGDfc) surface were characterized in detail. In addition, the PC/Fe@c(RGDfc) coating exhibits the remarkable ability to positively modulate cellular attachment, proliferation, migration and promoted bone-implant integration in vivo, maintaining the inherent features of MPNs: anti-inflammatory, anti-oxidative properties, as well as multiple substrate deposition. This work contributes to engineering MPNs-based coatings with bioactive molecules by a facile and efficient thiol-click reaction, as an innovative perspective for future development of surface modification of implant materials.
Collapse
Affiliation(s)
- Zeyu Shou
- Department of Orthopedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, People's Republic of China
| | - Zhibiao Bai
- Department of Orthopedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Kaiyuan Huo
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Shengwu Zheng
- Wenzhou Celecare Medical Instruments Co., Ltd, Wenzhou, 325000, People's Republic of China
| | - Yizhe Shen
- Department of Orthopedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Han Zhou
- Department of Orthopedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Xiaojing Huang
- Department of Orthopedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Hongming Meng
- Department of Orthopedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Chenwei Xu
- Department of Orthopedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Shaohao Wu
- Department of Orthopedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Na Li
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, People's Republic of China
| | - Chun Chen
- Department of Orthopedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
- Key Laboratory of Intelligent Treatment and Life Support for Critical Diseases of Zhejiang Province, Wenzhou, 325000, Zhejiang, People's Republic of China
- Zhejiang Engineering Research Center for Hospital Emergency and Process Digitization, Wenzhou, Zhejiang, 325000, People's Republic of China
| |
Collapse
|
20
|
Wang Z, Zhang Y, Shen Y, Zhou H, Gao Y, Zhu C, Qin X. Unlocking hepatocellular carcinoma aggression: STAMBPL1-mediated TRAF2 deubiquitination activates WNT/PI3K/NF-kb signaling pathway. Biol Direct 2024; 19:18. [PMID: 38419066 PMCID: PMC10903047 DOI: 10.1186/s13062-024-00460-7] [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: 02/06/2024] [Accepted: 02/22/2024] [Indexed: 03/02/2024] Open
Abstract
STAM Binding Protein Like 1 (STAMBPL1), functions as a deubiquitinase (DUB) and plays a significant role in various types of cancers. However, its effect as a DUB participating in the HCC tumorigenesis and progression still unknown. In the study, the upregulation and strong prognosis value of STAMBPL1 were identified in HCC patients. Functionally, STAMBPL1 significantly promoted HCC cells proliferation and metastasis, and it interacts with TRAF2 and stabilize it via the deubiquitination at the K63 residue. The TRAF2 upregulation stabilized by STAMBPL1 overexpression transfers of P65 protein into the nucleus and activates the WNT/PI3K/ NF-kb signaling pathway. The 251-436 sites of STAMBPL1 particularly interact with the 294-496 sites of TRAF2, thereby exerting the function of DUB and removing the ubiquitin molecules attached to TRAF2. Our research unveiled a new function of STAMBPL1 in mediating TRAF2 deubiquitination and stabilization, thereby activating the WNT/PI3K/NF-kb signaling pathway, suggesting its potential as a novel biomarker and therapeutic target for HCC.
Collapse
Affiliation(s)
- Zhihuai Wang
- Nanjing Medical University, Nanjing, 211166, China
- Department of General Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, 213000, China
| | - Yinjie Zhang
- Nanjing Medical University, Nanjing, 211166, China
- Department of General Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, 213000, China
| | - Yuhang Shen
- Nanjing Medical University, Nanjing, 211166, China
- Department of General Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, 213000, China
| | - Haiyang Zhou
- Nanjing Medical University, Nanjing, 211166, China
- Department of General Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, 213000, China
| | - Yuan Gao
- Department of General Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, 213000, China
| | - Chunfu Zhu
- Nanjing Medical University, Nanjing, 211166, China.
- Department of General Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, 213000, China.
| | - Xihu Qin
- Nanjing Medical University, Nanjing, 211166, China.
- Department of General Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, 213000, China.
| |
Collapse
|
21
|
Chen C, Li T, Li Y, Chen Z, Shi P, Li Y, Qian S. GPX4 is a potential diagnostic and therapeutic biomarker associated with diffuse large B lymphoma cell proliferation and B cell immune infiltration. Heliyon 2024; 10:e24857. [PMID: 38333875 PMCID: PMC10850411 DOI: 10.1016/j.heliyon.2024.e24857] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 01/08/2024] [Accepted: 01/16/2024] [Indexed: 02/10/2024] Open
Abstract
At present, GPX4's role in the occurrence and development of diffuse large B lymphoma (DLBCL) is rarely reported. This study's purpose is to explore GPX4's significance in the diagnosis, treatment, and pathological mechanisms of DLBCL. The TIMER 2.0, GEPIA, and GEO databases were used to analyze GPX4's expression levels in DLBCL tissue, peripheral blood, and single cells, and evaluate its potential performance as a therapeutic and diagnostic marker. Cell experiments validate GPX4's role in DLBCL cells. And revealed the potential mechanism of GPX4's action from three aspects: immunity, pathogenic gene expression, and protein interaction. The results indicate that GPX4 can be used as a biomarker for treatment and diagnosis (FC > 1.5, P < 0.05, AUC>0.8, KM-P value < 0.05). In single cell data, GPX4 also showed high expression in immune cells. Besides, cell experiments have confirmed that GPX4's high expression can inhibit DLBCL cells' proliferation. Meanwhile, we found a negative correlation between GPX4 and the 16 core DLBCL's pathogenic genes, and a significant negative correlation with immune B cell infiltration. In summary, GPX4 can serve as a potential therapeutic and diagnostic marker for DLBCL. GPX4's high expression can lead to a good prognosis in DLBCL patients, which may be related to its inhibition of cancer cell proliferation, high expression of key pathogenic genes, and infiltration of immune B cells.
Collapse
Affiliation(s)
- Can Chen
- Department of Hematology, Affiliated Hangzhou First People's Hospital, West Lake University, School of Medicine, Hangzhou, China
| | - TongYu Li
- Department of Hematology, Ningbo First Hospital, Ningbo, China
| | - Yiwei Li
- Department of Hematology, Affiliated Hangzhou First People's Hospital, West Lake University, School of Medicine, Hangzhou, China
| | - Zhenzhen Chen
- Department of Hematology, Affiliated Hangzhou First People's Hospital, West Lake University, School of Medicine, Hangzhou, China
| | - Pengfei Shi
- Department of Hematology, Affiliated Hangzhou First People's Hospital, West Lake University, School of Medicine, Hangzhou, China
| | - Yun Li
- Team of Neonatal & Infant Development, Health and Nutrition, NDHN, School of Biology and Pharmaceutical Engineering, Wuhan Polytechnic University, Wuhan, China
- Kindstar Global Precision Medicine Institute, Wuhan, China
- Department of Scientific Research Project, Wuhan Kindstar Medical Laboratory Co., Ltd., Wuhan, China
| | - Shenxian Qian
- Department of Hematology, Affiliated Hangzhou First People's Hospital, West Lake University, School of Medicine, Hangzhou, China
| |
Collapse
|