1
|
Farrokhi Yekta R, Farahani M, Koushki M, Amiri-Dashatan N. Deciphering the potential role of post-translational modifications of histones in gastrointestinal cancers: a proteomics-based review with therapeutic challenges and opportunities. Front Oncol 2024; 14:1481426. [PMID: 39497715 PMCID: PMC11532047 DOI: 10.3389/fonc.2024.1481426] [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: 08/15/2024] [Accepted: 09/30/2024] [Indexed: 11/07/2024] Open
Abstract
Oncogenesis is a complex and multi-step process, controlled by several factors including epigenetic modifications. It is considered that histone modifications are critical components in the regulation of gene expression, protein functions, and molecular interactions. Dysregulated post-translationally modified histones and the related enzymatic systems are key players in the control of cell proliferation and differentiation, which are associated with the onset and progression of cancers. The most of traditional investigations on cancer have focused on mutations of oncogenes and tumor suppressor genes. However, increasing evidence indicates that epigenetics, especially histone post-translational modifications (PTMs) play important roles in various cancer types. Mass spectrometry-based proteomic approaches have demonstrated tremendous potential in PTMs profiling and quantitation in different biological systems. In this paper, we have made a proteomics-based review on the role of histone modifications involved in gastrointestinal cancers (GCs) tumorigenesis processes. These alterations function not only as diagnostic or prognostic biomarkers for GCs, but a deeper comprehension of the epigenetic regulation of GCs could facilitate the treatment of this prevalent malignancy through the creation of more effective targeted therapies.
Collapse
Affiliation(s)
- Reyhaneh Farrokhi Yekta
- Proteomics Research Center, System Biology Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoumeh Farahani
- Proteomics Research Center, System Biology Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Koushki
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
- Department of Clinical Biochemistry, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Nasrin Amiri-Dashatan
- Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
2
|
Zhao FY, Chen X, Wang JM, Yuan Y, Li C, Sun J, Wang HQ. O-GlcNAcylation of TRIM29 and OGT translation forms a feedback loop to promote adaptive response of PDAC cells to glucose deficiency. Cell Oncol (Dordr) 2024; 47:1025-1041. [PMID: 38345749 DOI: 10.1007/s13402-023-00915-5] [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] [Accepted: 12/29/2023] [Indexed: 07/04/2024] Open
Abstract
PURPOSE Glucose not only provides energy for tumor cells, but also provides various biomolecules that are essential for their survival, proliferation and invasion. Therefore, it is of great clinical significance to understand the mechanism of how tumor cells adapt to metabolic stress and maintain their survival. The aim of this research was to study the critical role of OGT and TRIM29 O-GlcNAc modification driven adaptability of PDAC cells to low glucose stress, which might have important medical implications for PDAC therapy. METHODS Western blotting, mass spectrometry and WGA-immunoprecipitation were used to examined the levels of OGT and O-GlcNAc glycosylated proteins in BxPC3 and SW1990 cells in normal culture and under glucose deprivation conditions. Crystal violet assay, flow cytometry, RIP, RT-qPCR, protein stability assay, biotin pull down were used to investigate the mechanism of OGT and TRIM29-mediated adaptive response to glucose deficiency in PDAC cells. RESULTS The current study found that under the condition of low glucose culture, the levels of OGT and O-GlcNAc glycosylation in PDAC cells were significantly higher than those in normal culture. Moreover, the high expression of OGT has a protective effect on PDAC cells under low glucose stress. This study confirmed that there was no significant change in mRNA level and protein degradation of OGT under low glucose stress, which was mainly reflected in the increase of protein synthesis. In addition, O-GlcNAc modification at T120 site plays a critical role in the metabolic adaptive responses mediated by TRIM29. CONCLUSIONS Taken together, our study indicated that O-GlcNAcylation of TRIM29 at T120 site and OGT translation forms a loop feedback to facilitate survival of PDAC under glucose deficiency.
Collapse
Affiliation(s)
- Fu-Ying Zhao
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang, 110122, China
| | - Xue Chen
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang, 110122, China
| | - Jia-Mei Wang
- Department of Laboratory Medicine, The 1st Affiliated Hospital, China Medical University, Shenyang, 110001, China
| | - Ye Yuan
- Central Laboratory, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, China
| | - Chao Li
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang, 110122, China
| | - Jia Sun
- Department of Biochemistry and Molecular Biology, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Hua-Qin Wang
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
3
|
Yang W, Wang S, Tong S, Zhang WD, Qin JJ. Expanding the ubiquitin code in pancreatic cancer. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166884. [PMID: 37704111 DOI: 10.1016/j.bbadis.2023.166884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/23/2023] [Accepted: 09/06/2023] [Indexed: 09/15/2023]
Abstract
The ubiquitin-proteasome system (UPS) is a fundamental regulatory mechanism in cells, vital for maintaining cellular homeostasis, compiling signaling transduction, and determining cell fates. These biological processes require the coordinated signal cascades of UPS members, including ubiquitin ligases, ubiquitin-conjugating enzymes, deubiquitinases, and proteasomes, to ubiquitination and de-ubiquitination on substrates. Recent studies indicate that ubiquitination code rewriting is particularly prominent in pancreatic cancer. High frequency mutation or aberrant hyperexpression of UPS members dysregulates ferroptosis, tumor microenvironment, and metabolic rewiring processes and contribute to tumor growth, metastasis, immune evasion, and acquired drug resistance. We conduct an in-depth overview of ubiquitination process in pancreatic cancer, highlighting the role of ubiquitin code in tumor-promoting and tumor-suppressor pathways. Furthermore, we review current UPS modulators and analyze the potential of UPS modulators as cancer therapy.
Collapse
Affiliation(s)
- Wenyan Yang
- College of Pharmaceutical Science, Zhejiang University of Technology, Huzhou 313200, China; Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Shiqun Wang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Shengqiang Tong
- College of Pharmaceutical Science, Zhejiang University of Technology, Huzhou 313200, China
| | - Wei-Dong Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Jiang-Jiang Qin
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China.
| |
Collapse
|
4
|
Wu M, Jin MM, Cao XH, Zhao L, Li YH. Silencing TRIM29 Sensitizes Non-small Cell Lung Cancer Cells to Anlotinib by Promoting Apoptosis via Binding RAD50. Curr Cancer Drug Targets 2024; 24:445-454. [PMID: 37644752 DOI: 10.2174/1568009623666230829143148] [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: 03/08/2023] [Revised: 06/13/2023] [Accepted: 07/25/2023] [Indexed: 08/31/2023]
Abstract
BACKGROUND Previous studies have proposed that the transcriptional regulatory factor tripartite motif containing 29 (TRIM29) is involved in carcinogenesis via binding with nucleic acid. TRIM29 is confirmed to be highly expressed when the cancer cells acquire therapy-resistant properties. We noticed that TRIM29 levels were significantly increased in anlotinib-resistant NCIH1975 (NCI-H1975/AR) cells via mining data information from gene expression omnibus (GEO) gene microarray (GSE142031; log2 fold change > 1, p < 0.05). OBJECTIVE Our study aimed to investigate the function of TRIM29 on the resistance to anlotinib in non-small cell lung cancer (NSCLC) cells, including NCI-H1975 and A549 cells. METHODS Real-time RT-PCR and western blot were used to detect TRIM29 expression in anlotinib- resistant NSCLC (NSCLC/AR) cells. Apoptosis were determined through flow cytometry, acridine orange/ethidium bromide staining as well as western blot. ELISA was used to measure the content of C-X3-C motif chemokine ligand 1. Co-Immunoprecipitation assay was performed to verify the interaction between TRIM29 and RAD50 double-strand break repair protein (RAD50). RESULTS TRIM29 expression was shown to be elevated in the cytoplasm and nucleus of NSCLC/ AR cells compared to normal NSCLC cells. Next, we demonstrated that TRIM29 knockdown facilitated apoptosis and enhanced the sensitivity to anlotinib in NSCLC/AR cells. Based on the refined results citing from the database BioGRID, it was proved that TRIM29 interacted with RAD50. Herein, RAD50 overexpression diminished the pro-apoptotic effect induced by silencing TRIM29 in anlotinib-resistant A549 (A549/AR) cells. CONCLUSION Finally, we concluded that the increased sensitivity to anlotinib in NSCLC/AR cells was achieved by knocking down TRIM29, besides, the positive effects of TRIM29 knockdown were attributed to the promotion of apoptosis via binding to RAD50 in NSCLC/AR cell nucleus. Therefore, TRIM29 might become a potential target for overcoming anlotinib resistance in NSCLC treatment.
Collapse
Affiliation(s)
- Min Wu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, No. 100, Huaihai Avenue, Hefei, Anhui, People's Republic of China
- Department of Respiratory and Critical Care Medicine, Anhui Public Health Clinical Center, No. 100, Huaihai Avenue, Hefei, Anhui, People's Republic of China
| | - Meng-Meng Jin
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, No. 100, Huaihai Avenue, Hefei, Anhui, People's Republic of China
- Department of Respiratory and Critical Care Medicine, Anhui Public Health Clinical Center, No. 100, Huaihai Avenue, Hefei, Anhui, People's Republic of China
| | - Xiao-Hui Cao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, No. 100, Huaihai Avenue, Hefei, Anhui, People's Republic of China
- Department of Respiratory and Critical Care Medicine, Anhui Public Health Clinical Center, No. 100, Huaihai Avenue, Hefei, Anhui, People's Republic of China
| | - Lei Zhao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, No. 100, Huaihai Avenue, Hefei, Anhui, People's Republic of China
- Department of Respiratory and Critical Care Medicine, Anhui Public Health Clinical Center, No. 100, Huaihai Avenue, Hefei, Anhui, People's Republic of China
| | - Yong-Huai Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, No. 100, Huaihai Avenue, Hefei, Anhui, People's Republic of China
- Department of Respiratory and Critical Care Medicine, Anhui Public Health Clinical Center, No. 100, Huaihai Avenue, Hefei, Anhui, People's Republic of China
| |
Collapse
|
5
|
Lv S, Zhang J, Peng X, Liu H, Liu Y, Wei F. Ubiquitin signaling in pancreatic ductal adenocarcinoma. Front Mol Biosci 2023; 10:1304639. [PMID: 38174069 PMCID: PMC10761520 DOI: 10.3389/fmolb.2023.1304639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignant tumor of the digestive system, characterized by rapid progression and being prone to metastasis. Few effective treatment options are available for PDAC, and its 5-year survival rate is less than 9%. Many cell biological and signaling events are involved in the development of PDAC, among which protein post-translational modifications (PTMs), such as ubiquitination, play crucial roles. Catalyzed mostly by a three-enzyme cascade, ubiquitination induces changes in protein activity mainly by altering their stability in PDAC. Due to their role in substrate recognition, E3 ubiquitin ligases (E3s) dictate the outcome of the modification. Ubiquitination can be reversed by deubiquitylases (DUBs), which, in return, modified proteins to their native form. Dysregulation of E3s or DUBs that disrupt protein homeostasis is involved in PDAC. Moreover, the ubiquitination system has been exploited to develop therapeutic strategies, such as proteolysis-targeting chimeras (PROTACs). In this review, we summarize recent progress in our understanding of the role of ubiquitination in the development of PDAC and offer perspectives in the design of new therapies against this highly challenging disease.
Collapse
Affiliation(s)
- Shengnan Lv
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jian Zhang
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xinyu Peng
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Huan Liu
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yan Liu
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Feng Wei
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
6
|
Gu J, Chen J, Xiang S, Zhou X, Li J. Intricate confrontation: Research progress and application potential of TRIM family proteins in tumor immune escape. J Adv Res 2023; 54:147-179. [PMID: 36736694 DOI: 10.1016/j.jare.2023.01.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 01/06/2023] [Accepted: 01/12/2023] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Tripartite motif (TRIM) family proteins have more than 80 members and are widely found in various eukaryotic cells. Most TRIM family proteins participate in the ubiquitin-proteasome degradation system as E3-ubiquitin ligases; therefore, they play pivotal regulatory roles in the occurrence and development of tumors, including tumor immune escape. Due to the diversity of functional domains of TRIM family proteins, they can extensively participate in multiple signaling pathways of tumor immune escape through different substrates. In current research and clinical contexts, immune escape has become an urgent problem. The extensive participation of TRIM family proteins in curing tumors or preventing postoperative recurrence and metastasis makes them promising targets. AIM OF REVIEW The aim of the review is to make up for the gap in the current research on TRIM family proteins and tumor immune escape and propose future development directions according to the current progress and problems. KEY SCIENTIFIC CONCEPTS OF REVIEW This up-to-date review summarizes the characteristics and biological functions of TRIM family proteins, discusses the mechanisms of TRIM family proteins involved in tumor immune escape, and highlights the specific mechanism from the level of structure-function-molecule-pathway-phenotype, including mechanisms at the level of protein domains and functions, at the level of molecules and signaling pathways, and at the level of cells and microenvironments. We also discuss the application potential of TRIM family proteins in tumor immunotherapy, such as possible treatment strategies for combination targeting TRIM family protein drugs and checkpoint inhibitors for improving cancer treatment.
Collapse
Affiliation(s)
- Junjie Gu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jingyi Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shuaixi Xiang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xikun Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| | - Jing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
7
|
Chen Y, Ma J, Zhang M. TRIM29 promotes the progression of colorectal cancer by suppressing EZH2 degradation. Exp Biol Med (Maywood) 2023; 248:1527-1536. [PMID: 37837384 PMCID: PMC10676129 DOI: 10.1177/15353702231199070] [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: 02/01/2023] [Accepted: 07/08/2023] [Indexed: 10/16/2023] Open
Abstract
Colorectal cancer (CRC) is commonly diagnosed at the advanced stage and has a high mortality rate. Tripartite Motif Containing 29 (TRIM29) is an oncogene in numerous malignancies including CRC. However, the molecular mechanism of TRIM29 is largely unknown. In this study, we investigated the biological functions of TRIM29 and the underlying mechanisms. The expression of TRIM29 and Enhancer of Zeste Homolog 2 (EZH2) was predicted using the bioinformatic analysis and measured using a quantitative real-time polymerase chain reaction (PCR) and immunohistochemical assay. The biological functions of TRIM29 were analyzed using a cell counting kit-8, EdU and transwell assays, scratch test, and flow cytometry. The interaction between TRIM29 and EZH2 was assessed using protein immunoprecipitation. The stability of EZH2 was evaluated by treating it with cycloheximide. Our results indicated that TRIM29 levels were upregulated in CRC. Overexpression of TRIM29 promoted CRC cell proliferation and migration and suppressed apoptosis. The opposite result was obtained when TRIM29 was silenced. TRIM29 interacted with EZH2 mechanically and enhanced the protein stability of EZH2. Depletion of EZH2 reversed the effects of TRIM29, regarding its biological behaviors. Moreover, downregulation of TRIM29 inhibited tumor growth and improved the histopathological prognosis. In conclusion, EZH2 interacted with silenced TRIM29 to suppress its stability, thereby inhibiting cell proliferation, migration, and tumor growth, and promoting apoptosis in CRC. Our findings suggested that TRIM29 is a promising target for CRC therapy.
Collapse
Affiliation(s)
| | | | - Mingming Zhang
- Department of Gastrointestinal Surgery, Liuzhou People’s Hospital, Liuzhou 545006, Guangxi, China
| |
Collapse
|
8
|
Sun L, Wang D, Chen Z, Zhu X. TRIM29 knockdown prevented the colon cancer progression through decreasing the ubiquitination levels of KRT5. Open Life Sci 2023; 18:20220711. [PMID: 37671092 PMCID: PMC10476480 DOI: 10.1515/biol-2022-0711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 08/01/2023] [Accepted: 08/06/2023] [Indexed: 09/07/2023] Open
Abstract
To investigate the specific role of TRIM29 in colon cancer progression, bioinformatic analysis was performed on TRIM29. Colon cancer tissues were collected and colon cancer cells were cultured for further experiments. Cell viability and proliferation were determined using CCK-8, colony formation, and EDU staining assays. The mRNA and protein levels of TRIM29 and KRT5 were determined using quantitative real-time PCR and western blotting, respectively. The interaction between TRIM29 and KRT5 was detected using a co-immunoprecipitation (CO-IP) assay. Cycloheximide treatment was performed to analyse the stability of KRT5. TRIM29 was upregulated in colon cancer tissues and cells. TRIM29 knockdown decreased the cell viability and proliferation and ubiquitination levels of KRT5 and enhanced the protein stability and expression of KRT5. The CO-IP assay confirmed that TRIM29 and KRT5 binded to each other. KRT5 knockdown neutralises the inhibitory effect of sh-TRIM29 on colon cancer cell growth and TRIM29 knockdown prevented the proliferation of colon cancer cells by decreasing ubiquitination of KRT5, which enhanced the protein stability and expression of KRT5 in cancer cells. Thus, targeting TRIM29-mediated ubiquitination levels of KRT5 might be a new direction for colon cancer therapy.
Collapse
Affiliation(s)
- Lihui Sun
- The Fifth Department of General Surgery, The Third Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Heping Road, Jinzhou, Liaoning 121000, China
| | - Dawei Wang
- The Second Department of General Surgery, Dalian Fifth People’s Hospital, Dalian, Liaoning 116081, China
| | - Zhenyu Chen
- The Fifth Department of General Surgery, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, China
| | - Xu Zhu
- The Fifth Department of General Surgery, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, China
| |
Collapse
|
9
|
Deng J, Yi X, Feng Z, Peng J, Li D, Li C, Deng B, Liu S, Sahu S, Hao L. Deubiquitinating enzyme USP10 promotes osteosarcoma metastasis and epithelial-mesenchymal transition by stabilizing YAP1. Cancer Med 2023. [PMID: 37184153 PMCID: PMC10358238 DOI: 10.1002/cam4.6074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 02/13/2023] [Accepted: 02/22/2023] [Indexed: 05/16/2023] Open
Abstract
BACKGROUND Osteosarcoma (OS) is a fatal adolescent tumor, which is susceptible to remote metastases at an early stage, and its treatment remains a major challenge. ubiquitin-specific protease 10 (USP10) is primarily located in the cytoplasm and can therefore deubiquitinate various cytoplasmic proteins. However, the expression and mechanism of USP10 in OS remain ambiguous. The aim of this study was to explore how USP10 affects Yes-associated protein1 (YAP1) to influence the metastasis and epithelial-mesenchymal transition (EMT). METHODS Western blotting, qRT-PCR, and immunohistochemical (IHC) analyses were performed to evaluate USP10 and YAP1 levels. Using wound healing and transwell tests, the roles and molecular pathways of USP10 and YAP1 ability to migrate and invade of OS were investigated, and cell morphological alterations were examined using phalloidin staining. RESULTS Our results indicated that USP10, a new type of deubiquitinating protease, is increased in OS tissues and cells contrasted with adjacent healthy tissues. Overexpression of USP10 correlated with tumor size, distant metastasis, and TNM stage, and was an independent factor of poor prognosis in OS patients. Also, USP10 expression is closely connected with the incident of OS metastasis and tumor size. Functional assays revealed that USP10 knockdown suppressed cell migrating and invading ability and inhibited the EMT of OS cells in vivo and in vitro. In addition, we showed that USP10 knockdown decreased the levels of YAP1, which is an important positive regulator of migration and invasion in many cancers. We also found a significant positive correlation between USP10 and YAP1 levels, further demonstrating that USP10-induced migration and EMT are based on YAP1 in OS cells. In a mechanistic way, USP10 stabilizes the expression of YAP1 by mediating its deubiquitination in OS cells. CONCLUSION Together, this study showed that USP10 can directly interact with YAP1 to reduce ubiquitinated YAP1, thereby stabilizing its protein levels and affecting EMT and distant metastasis in OS cells.
Collapse
Affiliation(s)
- Jianyong Deng
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xuan Yi
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zuxi Feng
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jie Peng
- Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Dan Li
- Department of Oncology, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Chen Li
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Binbin Deng
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shuaigang Liu
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Souradeep Sahu
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Liang Hao
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
10
|
Xu X, Qin Z, Zhang C, Mi X, Zhang C, Zhou F, Wang J, Zhang L, Hua F. TRIM29 promotes podocyte pyroptosis in diabetic nephropathy through the NF-kB/NLRP3 inflammasome pathway. Cell Biol Int 2023; 47:1126-1135. [PMID: 36841942 DOI: 10.1002/cbin.12006] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 09/09/2022] [Accepted: 02/20/2023] [Indexed: 02/27/2023]
Abstract
Diabetic nephropathy (DN) is one of the most common complications of diabetes. Gradual loss of podocytes is a sign of DN and pyroptosis mechanistically correlates with podocyte injury in DN; however, the mechanism(s) involved remain unknown. Here we reveal that TRIM29 is overexpressed in high glucose (HG)-treated murine podocytes cells and that TRIM29 silencing significantly inhibits podocyte damage due to HG treatment, as evidenced by lower desmin expression and greater nephrin expression. Additionally, flow cytometry analysis showed that TRIM29 silencing significantly inhibited HG treatment-induced pyroptosis, which was confirmed by immunoblotting for NLRP3, active Caspase-1, GSDMD-N, and phosphorylated NF-κB-p65. Conversely, overexpression of TRIM29 could trigger pyroptosis that was attenuated by NF-κB inhibition, indicating that TRIM29 promotes pyroptosis through the NF-κB pathway. Mechanistic studies revealed that TRIM29 interacts with IκBα to mediate its ubiquitination-dependent degradation, which in turn leads to NF-κB activation. Taken together, our data demonstrate that TRIM29 can promote podocyte pyroptosis by activating the NF-κB/NLRP3 pathway. Thus, TRIM29 represents a potentially novel therapeutic target that may also be clinically relevant in the management of DN.
Collapse
Affiliation(s)
- Xiaohong Xu
- Department of Nephrology, Nanjing Drum Tower Hospital Group Suqian Hospital, Suqian, China.,Department of Nephrology, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, China
| | - Zihan Qin
- Department of Endocrinology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Ce Zhang
- Department of Nephrology, Nanjing Drum Tower Hospital Group Suqian Hospital, Suqian, China
| | - Xia Mi
- Department of Nephrology, Nanjing Drum Tower Hospital Group Suqian Hospital, Suqian, China
| | - Chi Zhang
- Department of Nephrology, Nanjing Drum Tower Hospital Group Suqian Hospital, Suqian, China
| | - Feihong Zhou
- Department of Nephrology, Nanjing Drum Tower Hospital Group Suqian Hospital, Suqian, China
| | - Junsheng Wang
- Department of Nephrology, Nanjing Drum Tower Hospital Group Suqian Hospital, Suqian, China
| | - Liexiang Zhang
- Department of Neurosurgery, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, China.,Department of Neurosurgery, Nanjing Drum Tower Hospital Group Suqian Hospital, Suqian, China
| | - Fei Hua
- Department of Endocrinology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
11
|
Ma J, Wu Y, Cheng S, Yang W, Zhong L, Li Q, Fang L. FBXO22 Accelerates Pancreatic Cancer Growth by Deactivation of the Hippo Pathway via Destabilizing LATS2. Dig Dis Sci 2022; 68:1913-1922. [PMID: 36515852 DOI: 10.1007/s10620-022-07780-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Dysregulation of ubiquitin ligases plays a crucial role in the development and progression of various human tumors. F-box only protein 22 (FBXO22), an F-box E3 ubiquitin ligase, has been reported to participate in diverse aspects of cancer progression. However, the clinical significance and biological function of FBXO22 in pancreatic cancer remain poorly understood. AIMS This study aimed to investigate the role of FBXO22 in promoting pancreatic cancer growth. METHODS FBXO22 expression was detected in pancreatic cancer and adjacent normal tissues using qRT-PCR, western blotting, and immunohistochemistry. Ectopic expression and knockdown of FBXO22 were performed to measure the impact on pancreatic cancer cells growth by CCK-8, colony formation, and tumorigenicity assay. Bioinformatics analysis uncovered the potential correlation between FBXO22 and various signaling pathways. Western blotting and immunoprecipitation were performed to identify FBXO22-interacting proteins. RESULTS We observed that FBXO22 was upregulated in samples obtained from patients with pancreatic cancer compared with its levels in the adjacent normal tissues, and an elevated FBXO22 level was obviously associated with poor prognosis among patients with pancreatic cancer. FBXO22 knockdown impaired pancreatic cancer cell growth both in vitro and in vivo, whereas FBXO22 overexpression accelerated pancreatic cancer cell growth. Furthermore, we found that FBXO22 contributed to pancreatic cancer cell growth by deactivating the Hippo pathway. Mechanistically, FBXO22 directly interacts with and destabilizes the large tumor suppressor 2 (LATS2), which is a critical regulator of the Hippo pathway. Blocking LATS2 leads to the loss of FBXO22-mediated oncogenic effect in pancreatic cancer. CONCLUSIONS These findings provide new insights into the upstream regulation of the Hippo pathway inactivation in pancreatic cancer growth and identify FBXO22 as a potential therapeutic target for this lethal malignant tumor.
Collapse
Affiliation(s)
- Jingsheng Ma
- Department of Organ Transplantation, The Second Affiliated Hospital of Nanchang University, Nanchang, 330038, Jiangxi, China
| | - Yajun Wu
- School of Medical Laboratory, Nanchang Medical College, Nanchang, 330006, Jiangxi, China
| | - Shibao Cheng
- Surgery of Hepatobiliary and Pancreatic, The Third Hospital of Nanchang, Nanchang, 330008, Jiangxi, China
| | - Wentao Yang
- Department of Organ Transplantation, The Second Affiliated Hospital of Nanchang University, Nanchang, 330038, Jiangxi, China
| | - Lin Zhong
- Department of Organ Transplantation, The Second Affiliated Hospital of Nanchang University, Nanchang, 330038, Jiangxi, China
| | - Qigen Li
- Department of Organ Transplantation, The Second Affiliated Hospital of Nanchang University, Nanchang, 330038, Jiangxi, China
| | - Lu Fang
- Surgery of Hepatobiliary and Pancreatic, The Second Affiliated Hospital of Nanchang University, No. 1 Min De Road, Nanchang, 330006, Jiangxi Province, China.
| |
Collapse
|
12
|
Li WW, Yuan H, Kong S, Tian SB. E3 ubiquitin ligase TRIM55 promotes metastasis of gastric cancer cells by mediating epithelial-mesenchymal transition. World J Gastrointest Oncol 2022; 14:2183-2194. [PMID: 36438697 PMCID: PMC9694263 DOI: 10.4251/wjgo.v14.i11.2183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 09/28/2022] [Accepted: 10/27/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Gastric cancer (GC) is considered a major global health problem. The role of TRIM55, a member of the three-domain protein family, in GC is unknown.
AIM To determine the expression of TRIM55 in GC tissues and its relationship with clinicopathological characteristics, and to investigate the effects of TRIM55 on the malignant biological behavior of GC cells.
METHODS Differential expression of TRIM55 in GC and para-cancer tissues was detected by immunohistochemistry, and the relationship between TRIM55 level and clinicopathological characteristics and prognosis was analyzed. Gain-of-function, loss-of-function, cell counting kit-8 assay, colony formation, transwell assay, wound healing assay, and western blot analysis were used to assess the potential role of TRIM55 in the development of GC.
RESULTS TRIM55 expression was significantly increased in GC tissues compared with adjacent normal tissues. High expression of TRIM55 was associated with advanced pathological stage and poor prognosis. Overexpression of TRIM55 promoted invasion and metastasis of GC cells in vitro by regulating epithelial-mesenchymal transition (EMT), whereas knockdown of TRIM55 had the opposite effect. Our data showed that TRIM55 is highly expressed in GC tissues, and is associated with poor prognosis. TRIM55 plays the role of an oncogene in GC, and it promotes metastasis of GC through the regulation of EMT.
CONCLUSION TRIM55 may be a possible target for the diagnosis and prognosis of GC patients.
Collapse
Affiliation(s)
- Wei-Wei Li
- Department of Pulmonary and Critical Care Medicine, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250021, Shandong Province, China
- Department of Critical Care Medicine, The 960th Hospital of the People's Liberation Army Joint Logistics Support Force, Jinan 250031, Shandong Province, China
| | - Hao Yuan
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong Province, China
| | - Shuai Kong
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong Province, China
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250021, Shandong Province, China
| | - Shu-Bo Tian
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong Province, China
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250021, Shandong Province, China
| |
Collapse
|
13
|
Hsu CY, Yanagi T, Ujiie H. TRIM29 in Cutaneous Squamous Cell Carcinoma. Front Med (Lausanne) 2022; 8:804166. [PMID: 34988104 PMCID: PMC8720877 DOI: 10.3389/fmed.2021.804166] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/02/2021] [Indexed: 12/13/2022] Open
Abstract
Tripartite motif (TRIM) proteins play important roles in a wide range of cell physiological processes, such as signal transduction, transcriptional regulation, innate immunity, and programmed cell death. TRIM29 protein, encoded by the ATDC gene, belongs to the RING-less group of TRIM protein family members. It consists of four zinc finger motifs in a B-box domain and a coiled-coil domain, and makes use of the B-box domain as E3 ubiquitin ligase in place of the RING. TRIM29 was found to be involved in the formation of homodimers and heterodimers in relation to DNA binding; additional studies have also demonstrated its role in carcinogenesis, DNA damage signaling, and the suppression of radiosensitivity. Recently, we reported that TRIM29 interacts with keratins and FAM83H to regulate keratin distribution. Further, in cutaneous SCC, the expression of TRIM29 is silenced by DNA methylation, leading to the loss of TRIM29 and promotion of keratinocyte migration. This paper reviews the role of TRIM family proteins in malignant tumors, especially the role of TRIM29 in cutaneous SCC.
Collapse
Affiliation(s)
- Che-Yuan Hsu
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Teruki Yanagi
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hideyuki Ujiie
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|