1
|
Chang TM, Fang WY, Hsu HP, Chu PY, Jiang SS, Huang KW, Hung WC, Lin HY, Tsai HJ. PCK2 promotes invasion and epithelial-to-mesenchymal transition in triple-negative breast cancer by promoting TGF-β/SMAD3 signaling through inhibiting TRIM67-mediated SMAD3 ubiquitination. Cancer Biol Ther 2025; 26:2478670. [PMID: 40081967 PMCID: PMC11913380 DOI: 10.1080/15384047.2025.2478670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/22/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025] Open
Abstract
PCK2, which encodes mitochondrial phosphoenolpyruvate carboxykinase (PEPCK-M), is upregulated in various cancers. We demonstrated high expression of PEPCK-M in approximately half of triple-negative breast cancers (TNBCs) previously. TNBC is associated with an aggressive phenotype and a high metastasis rate. In this study, we investigated the role of PCK2 in TNBC. PCK2 knockdown suppressed proliferation and mTOR signaling in TNBC cells. In addition, cell invasion/migration ability and the expression of epithelial-to-mesenchymal transition (EMT) markers were positively correlated with PCK2 expression in TNBC cells via regulation of transforming growth factor-β (TGF-β)/SMAD3 signaling. SMAD3 was positively regulated by PCK2 in TNBC cells. Knockdown of SMAD3 in PCK2-overexpressing TNBC cells reduced the expression levels of EMT markers, Snail and Slug, and suppressed cell invasion/migration. In addition, PCK2 knockdown attenuated the stimulatory effect of TGF-β on SMAD3 phosphorylation in TNBC cells. PEPCK-M promotes the protein and mRNA expression of SMAD3 via competitive binding to tripartite motif-containing 67 (TRIM67), an E3 ubiquitin ligase, to reduce SMAD3 ubiquitination, which leads to promoting nuclear translocation of SMAD3 and autoregulation of SMAD3 transcription. Moreover, high PCK2 mRNA expression was significantly associated with poor survival in TNBC patients. In conclusion, our study revealed for the first time that PCK2 activates TGF-β/SMAD3 signaling by regulating the expression and phosphorylation of SMAD3 by inhibiting TRIM67-mediated SMAD3 ubiquitination and promoting the stimulatory effect of TGF-β to promote TNBC invasion. The regulatory effect of PCK2 on mTOR and TGF-β/SMAD3 signaling suggests that PCK2 is a potential therapeutic target for suppressing TNBC progression.
Collapse
Affiliation(s)
- Tsung-Ming Chang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
- Department of Medical Laboratory Science, College of Medical Science and Technology, I-Shou University, Kaohsiung, Taiwan
| | - Wei-Yu Fang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Hui-Ping Hsu
- Department of Surgery, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Pei-Yi Chu
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
- Department of Pathology, Show Chwan Memorial Hospital, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Shih Sheng Jiang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Kuo-Wei Huang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Wen-Chun Hung
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Hui-You Lin
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Hui-Jen Tsai
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
2
|
Luo Y, Xu D, Yu C. Research progress on sepsis-associated encephalopathy by inhibiting pyroptosis. Gene 2025; 961:149560. [PMID: 40355013 DOI: 10.1016/j.gene.2025.149560] [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: 02/15/2025] [Revised: 04/28/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
Sepsis is a life-threatening condition characterized by multiple organ dysfunction syndrome resulted from dysregulated host responses to infection. Sepsis-associated encephalopathy (SAE) is one of the most common symptoms of acute-phase sepsis, with nearly 70 % of patients with sepsis ultimately developing SAE. Pyroptosis represents a type of cell death that is initiated by inflammation. This cell death type is associated with various infectious and noninfectious diseases. The gasdermin family proteins are crucial cell death executors and critical components in regulating the canonical pyroptosis pathway in microglia. In this review, we summarize the inhibitory effects of several drugs and genes on the pyroptosis pathway. Our findings suggest that several drugs (puerarin, VX765, HC067047, dexpramipexole, and Danhong injection), erbin gene, and TRIM45 knockdown improve SAE by suppressing the canonical pathway of NLRP3/caspase-1/gasdermin D-mediated pyroptosis. Therefore, they have significant importance in terms of brain protection. Moreover, we review the relevant literature published in recent years and summarize the research status and development prospects in this field to provide a basis for subsequent related research.
Collapse
Affiliation(s)
- Yanhua Luo
- Department of Yanbian University Hospital, Yanji, Jilin 133000, People's Republic of China
| | - Dahai Xu
- Department of Emergency Medicine, The First Hospital of Jilin University, Changchun Jilin 130000, People's Republic of China
| | - Chenglin Yu
- Department of Emergency Medicine, Yanbian University Hospital, Yanji, Jilin 133000, People's Republic of China.
| |
Collapse
|
3
|
Zhang Y, Shi H, Wang Y, Liu W, Li G, Li D, Wu W, Wu Y, Zhang Z, Ji Y, Zhu C, Bai W, Lei H, Xu H, Zhong H, Han B, Yang L, Liu L, Wang W, Zhao Y, Zhang Y, Wu Y. Noscapine derivative 428 suppresses ferroptosis through targeting GPX4. Redox Biol 2025; 83:103635. [PMID: 40305884 DOI: 10.1016/j.redox.2025.103635] [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: 02/01/2025] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 05/02/2025] Open
Abstract
Inhibiting ferroptosis represents a promising strategy to combat ferroptosis-related diseases. Here we show that 428, a selenide-containing noscapine derivative, effectively inhibits ferroptosis in various cell lines by enhancing the stability and activity of GPX4. TRIM41 was identified as a novel E3 ubiquitin ligase of GPX4 and 428 was demonstrated to bind to the selenocysteine residue Sec46 of GPX4 via the formation of a transient and reversible Se-Se bond, thereby blocking the interaction between GPX4 and TRIM41, stabilizing GPX4 and enhancing its activity. This unique dynamic covalent binding mode was preliminarily validated by structure-activity relationship analysis and molecular docking studies. Importantly, we demonstrated that 428 treatment alleviates bleomycin-induced pulmonary fibrosis in vivo by inhibiting ferroptosis. Overall, our studies identified a novel stabilizer and activator of GPX4, offering a potential therapeutic approach for the treatment of ferroptosis-related diseases and uncovering a new mechanism for regulating GPX4 degradation.
Collapse
Affiliation(s)
- Youping Zhang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Haoliang Shi
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, and School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yingying Wang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wanting Liu
- Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Guanyi Li
- School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Defeng Li
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, and School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Wenxuan Wu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yunzhao Wu
- Yusuf Hamied Department of Chemistry, University of Cambridge, CB2 1EW, Cambridge, UK
| | - Ziwei Zhang
- Yusuf Hamied Department of Chemistry, University of Cambridge, CB2 1EW, Cambridge, UK
| | - Yanjie Ji
- Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chujiao Zhu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wenhui Bai
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hu Lei
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hanzhang Xu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hua Zhong
- Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Baohui Han
- Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Li Yang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ligen Liu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wei Wang
- Department of Pharmacology and Toxicology and BIO5 Institute, University of Arizona, USA
| | - Yaxue Zhao
- School of Pharmaceutical Sciences, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China.
| | - Yongqiang Zhang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, and School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
| | - Yingli Wu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
4
|
Leach DA, Chatterjee N, Spahr K, de Almeida GS, Varela-Carver A, Shah TT, Winkler M, Ahmed HU, Bevan CL. Simultaneous inhibition of TRIM24 and TRIM28 sensitises prostate cancer cells to antiandrogen therapy, decreasing VEGF signalling and angiogenesis. Mol Oncol 2025. [PMID: 40411304 DOI: 10.1002/1878-0261.70065] [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: 08/06/2024] [Revised: 03/20/2025] [Accepted: 05/13/2025] [Indexed: 05/26/2025] Open
Abstract
Castrate-resistant prostate cancer (CRPC) is a likely outcome of hormone treatment for advanced prostate cancer. Although no longer dependent on androgen levels, CRPC remains driven by the androgen receptor (AR). One proposed progression mechanism is altered repertoires of coregulator proteins possessing the ability to alter AR activity. Increased expression of tripartite motif-containing 24 (TRIM24) and TRIM28-two members of a distinct bromodomain-containing subfamily of Tripartite motif (TRIM) coregulators-occurs in CRPC. Endogenous TRIM24 and TRIM28 interact with each other and AR, bind to chromatin and regulate genes such as the angiogenic factor vascular endothelial growth factor A (VEGFA) and oncogene MYC. Silencing of TRIM24 and TRIM28 simultaneously, but not either alone, sensitised CRPC model cell lines to the antiandrogen enzalutamide and bicalutamide. This re-sensitisation to antiandrogen therapeutics could then be reversed by addition of VEGF. Furthermore, both TRIM24 and TRIM28 expression associated with angiogenesis signatures in tumour samples, and conditioned media from TRIM24 and TRIM28-silenced cancer cells inhibited endothelial cell proliferation and formation of vascular tube structures. Our data suggest that TRIM24 and TRIM28 proteins interact, in gene-specific manners, to regulate AR activity, increase VEGF signalling and angiogenesis, and that targeting these coregulators may increase the effectiveness of antiandrogen therapy.
Collapse
Affiliation(s)
- Damien A Leach
- Division of Cancer, Imperial Centre for Translational & Experimental Medicine, Imperial College London, UK
| | - Nilesh Chatterjee
- Division of Cancer, Imperial Centre for Translational & Experimental Medicine, Imperial College London, UK
- St George's University of London, UK
| | - Kellie Spahr
- University of Michigan, Ann Arbor, MI, USA
- University of Pittsburgh, PA, USA
| | | | - Anabel Varela-Carver
- Division of Cancer, Imperial Centre for Translational & Experimental Medicine, Imperial College London, UK
| | - Taimur T Shah
- Imperial Urology, Division of Surgery, Imperial College Healthcare NHS Trust, London, UK
- Imperial Prostate, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, UK
| | - Mathias Winkler
- Imperial Urology, Division of Surgery, Imperial College Healthcare NHS Trust, London, UK
- Imperial Prostate, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, UK
| | - Hashim U Ahmed
- Imperial Urology, Division of Surgery, Imperial College Healthcare NHS Trust, London, UK
- Imperial Prostate, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, UK
| | - Charlotte L Bevan
- Division of Cancer, Imperial Centre for Translational & Experimental Medicine, Imperial College London, UK
| |
Collapse
|
5
|
Ly T, Pickard B, Pandey A, Yap M, Opara J, Arnold L, Martinez-Rivera N, Rosa-Molinar E, New J, Werner L, Farrokhian N, Gunewardena S, O'Neil M, Bur A, Anant S, Washburn MP, Barnaba C, Ding WX, Thomas SM. TRIM16 mediates secretory autophagy in head and neck cancer-associated fibroblasts. Autophagy 2025:1-24. [PMID: 40383937 DOI: 10.1080/15548627.2025.2508064] [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: 09/25/2024] [Revised: 05/06/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025] Open
Abstract
Improving treatment options for head and neck squamous cell carcinoma (HNSCC) requires a deeper understanding of the tumor microenvironment, particularly cancer-associated fibroblasts (CAFs). We previously reported that HNSCC-derived FGF2/bFGF (fibroblast growth factor 2) triggers cytokine release from CAFs via secretory autophagy. Here, using transmission electron microscopy, live-cell imaging, and immunofluorescence, we show that CAF autophagosomes transport cargo, including IL6, to the plasma membrane for secretion. Autophagy in CAFs is constitutive and independent of STAT3, MAPK1/ERK2-MAPK3/ERK1 and phosphoinositide 3-kinase (PI3K) signaling. Despite the significant role of secretory autophagy in CAFs, its molecular machinery has remained elusive. Using both a literature based, and an unbiased approach, we studied the molecular machinery involved in autophagosome trafficking in CAFs. We identified TRIM16, a protein previously reported to traffic to autophagosomes, upregulated in CAFs compared to normal oral fibroblasts. Immunohistochemistry of patient HNSCC stroma revealed co-expression of TRIM16 and LC3B, linking TRIM16 to autophagosome function. An unbiased proteomics profiling of immunoprecipitated LC3B+ vesicles in primary HNSCC CAFs revealed enrichment in trafficking proteins, focal adhesion, and mitochondrial proteins. We demonstrate that SEC22B, SNAP23, VAMP3, and STX4 colocalize with LC3B, IL6, and TRIM16 in CAFs. TRIM16 knockdown reduced autophagosomes at the plasma membrane and decreased IL6 secretion from CAFs. These findings uncover key molecular components involved in autophagy-mediated IL6 secretion in CAFs and suggest potential therapeutic targets for HNSCC.Abbreviations: ACTA2/αSMA: actin alpha 2, smooth muscle; CAF: cancer-associated fibroblasts; CM: conditioned media; CQ: chloroquine; DAPI: 4',6-diamidino-2-phenylindole; DMSO: dimethylsulfoxide; EGFP: enhanced green fluorescent protein; ELISA: enzyme-linked immunosorbent assay; ER: endoplasmic reticulum; FGF2/bFGF: fibroblast growth factor 2; FGFR: fibroblast growth factor receptor; GO: gene ontology; GORASP2/GRASP55: golgi reassembly stacking protein 2; HMGB1: high mobility group box 1; HNSCC: head and neck squamous cell carcinoma; HPV: human papillomavirus; IL6: interleukin 6; IP: immunoprecipitation; LC-MS/MS: liquid chromatography-mass spectrometry/mass spectrometry; LIR: LC3-interacting region; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MAPK1/ERK2: mitogen-activated protein kinase 1; MAPK3/ERK1: mitogen-activated protein kinase 3; NFs: normal oral fibroblasts; NSCLC: non-small cell lung cancer; PLA: proximity ligation assay; SQSTM1/p62: sequestosome 1; STAT3: signal transducer and activator of transcription 3; SNAP23: synaptosome associated protein 23; SNARE: soluble N-ethyl-maleimide-sensitive factor attachment protein receptor; STX4: syntaxin 4; TEM: transmission electron microscopy; TGFB1: transforming growth factor beta 1; TMA: tissue microarray; TRIM: tri-partite motif; VAMP: vesicle associated membrane protein; VC: vehicle control.
Collapse
Affiliation(s)
- Thuc Ly
- Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
- Otolaryngology-Head and Neck Surgery, University of Kansas Medical Center, Kansas City, KS, USA
| | - Bailey Pickard
- Otolaryngology-Head and Neck Surgery, University of Kansas Medical Center, Kansas City, KS, USA
| | - Avisha Pandey
- Otolaryngology-Head and Neck Surgery, University of Kansas Medical Center, Kansas City, KS, USA
| | - Marrion Yap
- Otolaryngology-Head and Neck Surgery, University of Kansas Medical Center, Kansas City, KS, USA
| | - Julia Opara
- Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Levi Arnold
- Otolaryngology-Head and Neck Surgery, University of Kansas Medical Center, Kansas City, KS, USA
| | - Noraida Martinez-Rivera
- Microscopy and Analytical Imaging Shared Resource Laboratory, University of Kansas, Lawrence, KS, USA
| | - Eduardo Rosa-Molinar
- Microscopy and Analytical Imaging Shared Resource Laboratory, University of Kansas, Lawrence, KS, USA
- Pharmacology and Toxicology, and Neuroscience Graduate Program, University of Kansas, Lawrence, KS, USA
| | - Jacob New
- Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Lauryn Werner
- Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Nathan Farrokhian
- Otolaryngology-Head and Neck Surgery, University of Kansas Medical Center, Kansas City, KS, USA
| | - Sumedha Gunewardena
- Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Maura O'Neil
- Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Andres Bur
- Otolaryngology-Head and Neck Surgery, University of Kansas Medical Center, Kansas City, KS, USA
| | - Shrikant Anant
- Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Michael P Washburn
- Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Carlo Barnaba
- Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, USA
| | - Wen-Xing Ding
- Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Sufi Mary Thomas
- Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA
- Otolaryngology-Head and Neck Surgery, University of Kansas Medical Center, Kansas City, KS, USA
- Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
6
|
Zhou LP, Kang L, Zhang ZG, Jia CY, Zhao CH, Zhang XL, Zhang HQ, Zhang RJ, Shen CL. RBX1 mitigates ferroptosis by inhibiting NCOA4-mediated ferritinophagy and contributes to the attenuation of intervertebral disc degeneration. J Transl Med 2025; 23:514. [PMID: 40335979 PMCID: PMC12060535 DOI: 10.1186/s12967-025-06412-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 03/23/2025] [Indexed: 05/09/2025] Open
Abstract
Loss of nucleus pulposus (NP) cells is as one of the primary factors initiating intervertebral disc (IVD) degeneration (IVDD); however, the intrinsic physiological mechanisms of endogenous NP-derived stem cell (NPSC)-based therapy in IVDD remain poorly understood. Disturbed iron homeostasis is commonly observed in degenerative diseases, and an acidic microenvironment has been considered a crucial factor in IVDD. The molecular mechanism of ferroptosis in acidic microenvironments during IVDD has not been reported. Herein, we intended to investigate whether acidic conditions can induce ferroptosis in NPSCs and explore the mechanism of IVDD progression through NCOA4-mediated ferritinophagy, which is a type of selective autophagy mediating ferroptosis. The role of ring-box 1 (RBX1) in NCOA4-mediated ferritinophagy in NPSC ferroptosis and IVDD pathogenesis was also explored. First, clinical epidemiology research revealed that a reduction in serum ferritin level was an independent risk factor for IVDD. We then demonstrated that ferroptosis progressively increased in human NP tissues as IVDD advanced and the acidic conditions induced ferroptosis-associated decline in cell viability, reactive oxygen species accumulation, and extracellular matrix degradation in human NPSCs. In an acidic microenvironment, ferroptosis is promoted due to enhanced NCOA4-mediated ferritinophagy in NPSCs. A mechanistic study demonstrated that RBX1-mediated ubiquitination modulated NCOA4 expression and the inhibition of RBX1 promoted ferroptosis through NCOA4-mediated ferritinophagy in the human NPSCs. Our in vivo study further illustrated that RBX1 overexpression ameliorated ferroptotic effects on IVDD progression by suppressing NCOA4-mediated ferritinophagy. Results demonstrated the modulating role of RBX1 in NCOA4-mediated ferritinophagy and NPSC ferroptosis, providing valuable insights into the potential application of endogenous stem cell-based IVD self-repair and self-regeneration for IVDD treatment.
Collapse
Affiliation(s)
- Lu-Ping Zhou
- Department of Orthopedics and Spine Surgery, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
- Laboratory of Spinal and Spinal Cord Injury Regeneration and Repair, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
- Anhui Province Research Center for the Clinical Application of Digital Medical Technology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Liang Kang
- Department of Orthopedics and Spine Surgery, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
- Laboratory of Spinal and Spinal Cord Injury Regeneration and Repair, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
- Anhui Province Research Center for the Clinical Application of Digital Medical Technology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Zhi-Gang Zhang
- Department of Orthopedics and Spine Surgery, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
- Laboratory of Spinal and Spinal Cord Injury Regeneration and Repair, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
- Anhui Province Research Center for the Clinical Application of Digital Medical Technology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Chong-Yu Jia
- Department of Orthopedics and Spine Surgery, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
- Laboratory of Spinal and Spinal Cord Injury Regeneration and Repair, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
- Anhui Province Research Center for the Clinical Application of Digital Medical Technology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Chen-Hao Zhao
- Department of Orthopedics and Spine Surgery, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
- Laboratory of Spinal and Spinal Cord Injury Regeneration and Repair, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
- Anhui Province Research Center for the Clinical Application of Digital Medical Technology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Xian-Liang Zhang
- Department of Orthopedics and Spine Surgery, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
- Laboratory of Spinal and Spinal Cord Injury Regeneration and Repair, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
- Anhui Province Research Center for the Clinical Application of Digital Medical Technology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Hua-Qing Zhang
- Department of Orthopedics and Spine Surgery, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China
- Laboratory of Spinal and Spinal Cord Injury Regeneration and Repair, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
- Anhui Province Research Center for the Clinical Application of Digital Medical Technology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Ren-Jie Zhang
- Department of Orthopedics and Spine Surgery, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China.
- Laboratory of Spinal and Spinal Cord Injury Regeneration and Repair, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China.
- Anhui Province Research Center for the Clinical Application of Digital Medical Technology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China.
| | - Cai-Liang Shen
- Department of Orthopedics and Spine Surgery, The First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei, 230022, Anhui, China.
- Laboratory of Spinal and Spinal Cord Injury Regeneration and Repair, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China.
- Anhui Province Research Center for the Clinical Application of Digital Medical Technology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China.
| |
Collapse
|
7
|
Zhou P, Zhang Q, Yang Y, Liu D, Wu W, Jongkaewwattana A, Jin H, Zhou H, Luo R. TRIM14 restricts tembusu virus infection through degrading viral NS1 protein and activating type I interferon signaling. PLoS Pathog 2025; 21:e1013200. [PMID: 40435148 PMCID: PMC12118852 DOI: 10.1371/journal.ppat.1013200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 05/09/2025] [Indexed: 06/01/2025] Open
Abstract
Tembusu virus (TMUV), an emerging avian orthoflavivirus, causes severe economic losses due to egg-drop syndrome and fatal encephalitis in domestic waterfowl. To combat this threat, the host immune system plays a crucial role in controlling and eliminating TMUV infection. Understanding the mechanisms of this immune response is thus vital for developing effective strategies against the virus. In this study, we investigated the antiviral activities of duck TRIM family proteins (duTRIM) against TMUV, focusing particularly on duTRIM14 as a potent host restriction factor. We showed that overexpression of duTRIM14 significantly inhibits TMUV replication, while its deficiency leads to increased viral titers. We elucidate a novel mechanism by which duTRIM14 interacts with the TMUV NS1 protein, facilitating its K27/K29-linked polyubiquitination and subsequent proteasomal degradation. The Lys141 residue on NS1 was identified as critical for this process, with its removal significantly enhancing TMUV replication both in vitro and in vivo. Furthermore, we showed that duTRIM14 interacts with duck TBK1 (duTBK1), promoting its K63-linked polyubiquitination on Lys30 and Lys401, which substantially augments IFN-β production during TMUV infection. Taken together, these results provide a novel dual-action antiviral mechanism in which duTRIM14 suppresses TMUV replication by simultaneously promoting proteasomal degradation of NS1 and enhancing the host antiviral response by modulating duTBK1 activity.
Collapse
Affiliation(s)
- Peng Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Wuhan, China
| | - Qingxiang Zhang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Wuhan, China
| | - Yueshan Yang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Wuhan, China
| | - Dan Liu
- China Institute of Veterinary Drug Control, Beijing, PR China
| | - Wanrong Wu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Wuhan, China
| | - Anan Jongkaewwattana
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Klong Nueng, Pathum Thani, Thailand
| | - Hui Jin
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Wuhan, China
| | - Hongbo Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Wuhan, China
| | - Rui Luo
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affairs of the People’s Republic of China, Wuhan, China
| |
Collapse
|
8
|
Chabot E, Durantel D, Lucifora J. TRIM proteins: A 'swiss army knife' of antiviral immunity. PLoS Pathog 2025; 21:e1013147. [PMID: 40354393 PMCID: PMC12068639 DOI: 10.1371/journal.ppat.1013147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025] Open
Abstract
With their modular structure and E3 ubiquitin ligase activity, Tripartite motif (TRIM) proteins interact with a wide range of cellular and viral substrates. This review summarizes how they have emerged as key players in the antiviral response. Shortly, TRIM proteins were shown (i) to enhance pro-inflammatory cytokines production by interacting with pattern recognition receptors and downstream components of immune signaling pathways, (ii) to interfere with viral trafficking by interacting with the cytoskeleton, and (iii) to exhibit direct antiviral effects by targeting viral proteins for proteasomal degradation or inducing autophagy. This combination of actions underscores TRIMs as a potent innate defense system, but also makes them vulnerable to viral evasion strategies.
Collapse
Affiliation(s)
- Elise Chabot
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
- Master de Biologie, École Normale Supérieure de Lyon, Lyon Cedex, France
| | - David Durantel
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Julie Lucifora
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| |
Collapse
|
9
|
Cao Y, Kong L, Zhai Y, Hou W, Wang J, Liu Y, Wang C, Zhao W, Ji H, He P. Comprehensive analysis of TRIM56's prognostic value and immune infiltration in Pan-Cancer. Sci Rep 2025; 15:13673. [PMID: 40258986 PMCID: PMC12012147 DOI: 10.1038/s41598-025-97856-w] [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: 10/24/2024] [Accepted: 04/08/2025] [Indexed: 04/23/2025] Open
Abstract
TRIM56 plays a role in tumor development through the ubiquitination of several key substrate molecules. However, its relationship with tumor prognosis and immune infiltration remains unclear. The expression and localization of TRIM56 were analyzed from TCGA_GTEx, TCGA and HPA database. The effects of TRIM56 on the proliferation and migration of lung cancer cells A549 were evaluated by CCK-8 and wound healing assays. Correlations between TRIM56 expression and survival in patients were analyzed using the Kaplan-Meier Plotter and a nomogram model. Additionally, the relationship between TRIM56 and immune cell infiltration in tumors was explored via TIMER 2.0. Functional interactions and associated proteins of TRIM56 were examined using GEPIA 2.0 and the STING database. The signaling pathways influenced by TRIM56 were identified through GO and KEGG analyses. TRIM56 expression showed significant variation across 11 different tumor types when compared to normal tissues, with some tumors displaying high expression and others showing the opposite. TRIM56 inhibited the proliferation and migration of A549 cells. High TRIM56 expression was associated with shorter overall survival (OS) in patients with COAD, GBM, and LGG, but with longer OS in BLCA, KIRC, MESO, and SKCM. In BLCA and KIRC, high TRIM56 expression was closely linked to B cells, macrophages, and CD4(+) and CD8(+) T cell infiltration, contributing to a favorable prognosis. TRIM56 appears to affect tumor development through transcriptional regulatory complexes, transcriptional co-regulatory factor activity, and immune-related pathways.TRIM56 may play a critical role in tumor immunity and influence tumor prognosis. It holds potential as both a target for immunotherapy and a prognostic marker.
Collapse
Affiliation(s)
- Yunpeng Cao
- College of Basic Medicine, Chengde Medical University, Chengde, China
| | - Lingwei Kong
- Department of Orthopaedics, The Afliated Hospital of Chengde Medical University, Chengde, China
| | - Yulu Zhai
- College of Basic Medicine, Chengde Medical University, Chengde, China
| | - Weiyan Hou
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jiayuan Wang
- College of Basic Medicine, Chengde Medical University, Chengde, China
| | - Yuxi Liu
- College of Basic Medicine, Chengde Medical University, Chengde, China
| | - Chengru Wang
- College of Basic Medicine, Chengde Medical University, Chengde, China
| | - Wei Zhao
- Graduate School, Chengde Medical University, Chengde, China
| | - Hairu Ji
- Department of Pathology, Chengde Medical University, Chengde, China.
| | - Peiyuan He
- Department of Gastroenterology, The Afliated Hospital of Chengde Medical University, Chengde, China
| |
Collapse
|
10
|
Kim K, Kim JH, Kim I, Seong S, Kook H, Koh JT, Kim N. Tripartite motif-containing 27 negatively regulates NF-κB activation in bone remodeling. Mol Med 2025; 31:141. [PMID: 40251491 PMCID: PMC12008848 DOI: 10.1186/s10020-025-01204-7] [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: 11/29/2024] [Accepted: 04/09/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Tripartite motif-containing 27 (TRIM27) is highly expressed in the mouse thymus, spleen, and hematopoietic compartment cells and regulates cell proliferation, apoptosis, and innate immune responses. However, the role of TRIM27 in bone remodeling remains unknown. This study aimed to investigate the role of TRIM27 in the differentiation of osteoclasts and osteoblasts. METHODS We measured the effects of overexpression or knockdown of TRIM27 in osteoclasts and osteoblasts using real-time PCR and Western blot analysis to quantify the mRNA and protein levels of marker genes. Additionally, we performed an in vivo analysis of TRIM27 knockout mice through bone mineral density analysis and histological analysis. RESULTS TRIM27 deficiency decreased bone mineral density by enhancing osteoclast differentiation and inhibiting osteoblast differentiation. Overexpression of TRIM27 in osteoclast precursors suppressed osteoclast formation and resorption activity, and ectopic expression of TRIM27 in osteoblast precursors induced osteoblast differentiation and mineralization. Additionally, we found that TRIM27 attenuated NF-κB activation in both osteoclasts and osteoblasts by interacting with TAB2 and promoting TAB2 degradation through lysosomal-dependent pathways, thereby inhibiting NF-κB signaling. CONCLUSIONS Our results identify TRIM27 as a novel negative regulator of NF-κB in bone remodeling, suggesting that regulating TRIM27 may be useful in developing treatments for musculoskeletal diseases, such as osteoporosis.
Collapse
Affiliation(s)
- Kabsun Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Jung Ha Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Inyoung Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Semun Seong
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Hyun Kook
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Jeong-Tae Koh
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Nacksung Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea.
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
11
|
Liu R, Xiao Y, Zhang G, Han P, Lin Z, Song H. TRIM8 enhances chondrocyte ferroptosis by inhibiting YTHDF2-m6A mediated SREBF2 mRNA degradation to promote OA progression. Int Immunopharmacol 2025; 152:114441. [PMID: 40073810 DOI: 10.1016/j.intimp.2025.114441] [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: 11/19/2024] [Revised: 03/03/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025]
Abstract
Chondrocyte ferroptosis plays a crucial role in osteoarthritis (OA) progression. Our previous study demonstrated that TRIM8 knockdown alleviated IL-1β-induced chondrocyte injury. However, the involvement of TRIM8 in regulating OA progression through ferroptosis remains unclear. In this study, human OA and normal cartilage samples were collected to examine ferroptosis and TRIM8 expression. We found that both ferroptosis markers and TRIM8 protein levels were elevated in OA cartilage compared to controls. An OA cell model was established by stimulating chondrocytes with IL-1β. TRIM8 knockdown mitigated IL-1β-induced ferroptosis, extracellular matrix (ECM) degradation, and inflammation in chondrocytes. Mechanistically, TRIM8 facilitated the ubiquitylation of YTHDF2 via its RING domain, promoting YTHDF2 protein degradation. This inhibited YTHDF2-m6A-induced SREBF2 mRNA degradation, thereby upregulating SREBF2 expression and enhancing chondrocyte ferroptosis. As expected, SREBF2 overexpression reversed the protective effect of TRIM8 silencing on IL-1β-induced chondrocyte injury. An OA mouse model was established using destabilized medial meniscus surgery, and TRIM8 deficiency alleviated cartilage degradation and synovial inflammation. In conclusion, TRIM8 promotes chondrocyte ferroptosis by suppressing YTHDF2-m6A mediated SREBF2 mRNA degradation, thereby accelerating OA progression.
Collapse
Affiliation(s)
- Ruoxi Liu
- Department of Orthopaedics, the Second Affiliated Hospital of Xi'an JiaoTong University, Xi'an, China
| | - Yi Xiao
- Shaanxi Provincial Center for Disease Control and Prevention, Xi'an, China
| | - Gelin Zhang
- Department of Orthopaedics, the Second Affiliated Hospital of Xi'an JiaoTong University, Xi'an, China
| | - Pei Han
- Department of Orthopaedics, the Second Affiliated Hospital of Xi'an JiaoTong University, Xi'an, China
| | - Zhaoxing Lin
- Shaanxi Provincial Center for Disease Control and Prevention, Xi'an, China.
| | - Huanjin Song
- Department of Orthopaedics, the Second Affiliated Hospital of Xi'an JiaoTong University, Xi'an, China.
| |
Collapse
|
12
|
Hage A, Janes M, Best SM. A No-Brainer! The Therapeutic Potential of TRIM Proteins in Viral and Central Nervous System Diseases. Viruses 2025; 17:562. [PMID: 40285004 PMCID: PMC12031127 DOI: 10.3390/v17040562] [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: 03/22/2025] [Revised: 04/11/2025] [Accepted: 04/12/2025] [Indexed: 04/29/2025] Open
Abstract
Tripartite motif (TRIM) proteins comprise an important class of E3 ubiquitin ligases that regulate numerous biological processes including protein expression, cellular signaling pathways, and innate immunity. This ubiquitous participation in fundamental aspects of biology has made TRIM proteins a focus of study in many fields and has illuminated the negative impact they exert when functioning improperly. Disruption of TRIM function has been linked to the success of various pathogens and separately to the occurrence and development of several neurodegenerative diseases, making TRIM proteins an appealing candidate to study for novel therapeutic approaches. Here, we review the current findings on TRIM proteins that demonstrate their analogous properties in the distinct fields of viral infection and central nervous system (CNS) disorders. We also examine recent advancements in drug development and targeted protein degradation as potential strategies for TRIM-mediated therapeutic treatments and discuss the implications these technologies have on future research directions.
Collapse
Affiliation(s)
- Adam Hage
- Innate Immunity and Pathogenesis Section, Laboratory of Neurological Infections and Immunity, Division of Intramural Research, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA; (M.J.); (S.M.B.)
| | | | | |
Collapse
|
13
|
Zhang L, Liu S, Zhao Q, Liu X, Zhang Q, Liu M, Zhao W. The role of ubiquitination and deubiquitination in the pathogenesis of non-alcoholic fatty liver disease. Front Immunol 2025; 16:1535362. [PMID: 40292292 PMCID: PMC12021615 DOI: 10.3389/fimmu.2025.1535362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/19/2025] [Indexed: 04/30/2025] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is one of the most common chronic liver diseases and is closely associated with metabolic abnormalities. The causes of NAFLD are exceedingly complicated, and it is known that a variety of signaling pathways, endoplasmic reticulum stress, and mitochondrial dysfunction play a role in the pathogenesis of NAFLD. Recent studies have shown that ubiquitination and deubiquitination are involved in the regulation of the NAFLD pathophysiology. Protein ubiquitination is a dynamic and diverse post-translational alteration that affects various cellular biological processes. Numerous disorders, including NAFLD, exhibit imbalances in ubiquitination and deubiquitination. To highlight the significance of this post-translational modification in the pathogenesis of NAFLD and to aid in the development of new therapeutic approaches for the disease, we will discuss the role of enzymes involved in the processes of ubiquitination and deubiquitination, specifically E3 ubiquitin ligases and deubiquitinating enzymes that are important in the regulation of NAFLD.
Collapse
Affiliation(s)
- Lihui Zhang
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Zhengzhou, Henan, China
| | - Sutong Liu
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Zhengzhou, Henan, China
| | - Qing Zhao
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiaoyan Liu
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Qiang Zhang
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Minghao Liu
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Zhengzhou, Henan, China
| | - Wenxiao Zhao
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Zhengzhou, Henan, China
| |
Collapse
|
14
|
Wu J, Yang Z, Chen X, Hou S, Li N, Chang Y, Yin J, Xu J. TRIM36 Inhibits the Development of AOM/DSS-Induced Colitis-Associated Colorectal Cancer by Promoting the Ubiquitination and Degradation of GRB7. Mol Carcinog 2025; 64:668-679. [PMID: 39803720 DOI: 10.1002/mc.23871] [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: 08/09/2024] [Revised: 10/30/2024] [Accepted: 12/05/2024] [Indexed: 03/10/2025]
Abstract
Colorectal cancer (CRC) is among the most common cancer types for both sexes. Tripartite motif 36 (TRIM36) has been reported to be aberrantly expressed in several cancer types, suggesting its involvement in cancer progression. However, the role of TRIM36 in the colorectal carcinogenesis remain unknown. In our in vivo experiments, we investigated the role of TRIM36 in AOM/DSS-induced colitis-associated carcinogenesis using TRIM36-knockout (TRIM36 KO) mice. Subsequently, we overexpressed and knocked down TRIM36 expression in two CRC cell lines to further confirm the role of TRIM36 in vitro. The UALCAN database revealed a significant decrease in TRIM36 levels in CRC tissues, including colon adenocarcinoma and rectum adenocarcinoma. A significant correlation was observed between TRIM36 levels and the histological subtype, individual cancer stage, and nodal metastasis status. The downregulation of TRIM36 in CRC tissues was further confirmed using our own collected clinical specimens. Low expression of TRIM36 was found to be associated with unfavorable overall survival and recurrence-free survival in CRC. TRIM36 KO promoted inflammation, inhibited autophagy, and facilitated the development of AOM/DSS-induced CRC. TRIM36 overexpression inhibited proliferation, migration, and invasion, while activated autophagy in CRC cells. TRIM36 directly bound to and regulated the ubiquitination of GRB7 protein. The tumor-suppressive role of TRIM36 in CRC cells was mediated by GRB7. The TRIM36/GRB7 axis may represent a promising therapeutic target for the treatment of CRC.
Collapse
Affiliation(s)
- Ju Wu
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
- The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Dalian University, Dalian, China
| | - Zhengbo Yang
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
- The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Dalian University, Dalian, China
| | - Xi Chen
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
- The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Dalian University, Dalian, China
| | - Shuangshuang Hou
- Department of General Surgery, Fuyang Normal University Second Affiliated Hospital, Fuyang, China
| | - Nanbo Li
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Yaoyuan Chang
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
- The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Dalian University, Dalian, China
| | - Jiajun Yin
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
- The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Dalian University, Dalian, China
| | - Jian Xu
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
- The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Dalian University, Dalian, China
| |
Collapse
|
15
|
Zhang Y, Tan X, Wang L, Ji D, Zhang C, Peng W, Zhu R, Wang X, Zhou J, Feng Y, Sun Y. TRIM38 Suppresses the Progression of Colorectal Cancer via Enhancing CCT6A Ubiquitination to Inhibit the MYC Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411285. [PMID: 40047371 PMCID: PMC12021106 DOI: 10.1002/advs.202411285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 02/14/2025] [Indexed: 04/26/2025]
Abstract
Emerging evidence reveals the pivotal function of tripartite motif protein (TRIM) in colorectal cancer (CRC). However, the precise function of TRIM38 and its underlying mechanism in CRC remains to be elucidated, especially regarding its putative ubiquitination function. Here, it is identified that TRIM38 is downregulated in CRC tissues by DNA hypermethylation of its promoter. Further analysis demonstrates that decreased TRIM38 is correlated with unfavorable clinical features and poor prognosis. Moreover, TRIM38 functions as a tumor suppressor by inhibiting cell proliferation, metastasis, and AOM/DSS-induced tumorigenesis in CRC cells. Mechanistically, TRIM38 binds to the substrate protein CCT6A, leading to the degradation and K48-linked ubiquitination of CCT6A at the K127/K138 residues. The elevation of CCT6A protein level caused by TRIM38 downregulation diminishes the degradation of c-Myc protein, thereby activating the MYC pathway. The study elucidates a novel mechanism of TRIM38/CCT6A/c-Myc axis regulating CRC, potentially offering a new therapeutic target for its treatment.
Collapse
Affiliation(s)
- Yue Zhang
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu210029P. R. China
- The First School of Clinical Medicine Nanjing Medical UniversityNanjing210029P. R. China
- Colorectal Institute of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjing210029P. R. China
| | - Xinyu Tan
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu210029P. R. China
- The First School of Clinical Medicine Nanjing Medical UniversityNanjing210029P. R. China
- Colorectal Institute of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjing210029P. R. China
| | - Lu Wang
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu210029P. R. China
- Colorectal Institute of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjing210029P. R. China
| | - Dongjian Ji
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu210029P. R. China
- The First School of Clinical Medicine Nanjing Medical UniversityNanjing210029P. R. China
- Colorectal Institute of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjing210029P. R. China
| | - Chuan Zhang
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu210029P. R. China
- The First School of Clinical Medicine Nanjing Medical UniversityNanjing210029P. R. China
- Colorectal Institute of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjing210029P. R. China
| | - Wen Peng
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu210029P. R. China
- The First School of Clinical Medicine Nanjing Medical UniversityNanjing210029P. R. China
- Colorectal Institute of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjing210029P. R. China
| | - Renzhong Zhu
- Institute of Translational MedicineMedical CollegeYangzhou UniversityYangzhouJiangsu225000P. R. China
| | - Xiaowei Wang
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu210029P. R. China
- The First School of Clinical Medicine Nanjing Medical UniversityNanjing210029P. R. China
- Colorectal Institute of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjing210029P. R. China
| | - Jiahui Zhou
- The Affiliated Suzhou Hospital of Nanjing Medical UniversitySuzhou Municipal Hospital, Gusu SchoolNanjing Medical UniversitySuzhouJiangsu215000P. R. China
| | - Yifei Feng
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu210029P. R. China
- Colorectal Institute of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjing210029P. R. China
| | - Yueming Sun
- Department of General SurgeryThe First Affiliated Hospital of Nanjing Medical UniversityNanjingJiangsu210029P. R. China
- Colorectal Institute of Nanjing Medical UniversityNanjing210029P. R. China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational MedicineNanjing210029P. R. China
| |
Collapse
|
16
|
Zou J, Chen J, Deng L, Xu B, Yu T, Wang J, He C. Mechanistic insights into SENP1 and OCT4 interaction in promoting drug resistance and stem cell features in colon cancer. Am J Physiol Cell Physiol 2025; 328:C1260-C1278. [PMID: 40063360 DOI: 10.1152/ajpcell.00817.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 11/18/2024] [Accepted: 02/17/2025] [Indexed: 04/01/2025]
Abstract
This study explores the molecular mechanism by which sentrin/SUMO-specific protease 1 (SENP1) promotes cisplatin (Cis) resistance and tumor stem cell characteristics in colon adenocarcinoma (COAD) through deSUMOylation-mediated modification of octamer-binding transcription factor 4 (OCT4). By analyzing single-cell and transcriptome sequencing datasets, we identified key genes and regulatory pathways in both resistant and sensitive COAD cells. Malignant cells were isolated and evaluated for stemness using the infercnv package, and differential genes between Cis-resistant and -sensitive groups were identified. Machine learning algorithms highlighted essential genes, and databases predicted interaction sites between OCT4 and SENP1. In vitro experiments using enriched HCT116 stem cells revealed that SENP1 and OCT4 expression significantly elevated CD44 and CD133 levels, enhancing stemness. Functional assays showed that SENP1's deSUMOylation of OCT4 intensified Cis resistance, migration, and invasion in cisplatin-resistant cell line 116 (Cis-116) cells. In vivo, SENP1 knockdown reduced tumor growth and stem cell markers, whereas OCT4 overexpression escalated tumor metastasis and structural damage. These findings demonstrate that SENP1's modulation of OCT4 is central to COAD's resistance and stem cell properties, offering a novel target for COAD therapy.NEW & NOTEWORTHY This study uncovers the critical role of SENP1 in regulating OCT4 through deSUMOylation, driving Cis resistance and tumor stemness in COAD. Targeting this pathway may provide novel therapeutic strategies for COAD management.
Collapse
Affiliation(s)
- Jun Zou
- Department of Abdominal Oncology Surgery, Jiangxi Cancer Hospital, Nanchang, People's Republic of China
| | - Jing Chen
- Department of nursing, Nanchang Medical College, Nanchang, People's Republic of China
| | - Lei Deng
- Department of Medical Oncology, Jiangxi Cancer Hospital, Nanchang, People's Republic of China
| | - Bangran Xu
- Department of Abdominal Oncology Surgery, Jiangxi Cancer Hospital, Nanchang, People's Republic of China
| | - Tenghua Yu
- Department of Breast Surgery, Jiangxi Cancer Hospital, Nanchang, People's Republic of China
| | - Jun Wang
- General Surgery Department of the Trauma Center, Nanchang First Hospital, Nanchang, People's Republic of China
| | - Chongwu He
- Department of Breast Surgery, Jiangxi Cancer Hospital, Nanchang, People's Republic of China
| |
Collapse
|
17
|
Hu H, Li W, Ma P, Song J, Zhang X, Ruan L, Zhang J, Zheng Y. TRIM22 inhibits the metastasis of colorectal cancer through facilitating β-Catenin degradation. Exp Cell Res 2025; 446:114473. [PMID: 39978715 DOI: 10.1016/j.yexcr.2025.114473] [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: 10/02/2024] [Revised: 02/15/2025] [Accepted: 02/16/2025] [Indexed: 02/22/2025]
Abstract
Tripartite motif-containing 22 (TRIM22), a member of the tripartite motif protein family, has emerged as a putative tumor suppressor in various cancers. Nevertheless, its specific role and clinical significance in colorectal cancer (CRC) remain poorly characterized. Herein, we observed that TRIM22 expression was frequently downregulated in primary CRC tissues and was significantly correlated with better prognosis. Functional assays demonstrated that TRIM22 overexpression substantially attenuated the metastatic potential of CRC cells both in vitro and in vivo. Mechanistically, our results revealed that TRIM22 directly interacts with and ubiquitinates β-Catenin, a crucial transcription factor that drives CRC metastasis by modulating the epithelial-mesenchymal transition (EMT) process. Additionally, our data indicated that the anti-metastatic effect of TRIM22 relies on the degradation of β-catenin. In summary, this study is the first to deliberate the vital anti-tumor role of TRIM22 in CRC metastasis. We also provide new evidence suggesting that TRIM22 could be a prognostic marker and therapeutic target for inhibiting CRC progression.
Collapse
Affiliation(s)
- Haiyang Hu
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Wensheng Li
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Pengfei Ma
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Junxin Song
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Xiaobo Zhang
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Longhui Ruan
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Jing Zhang
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Youwei Zheng
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China.
| |
Collapse
|
18
|
Saxena T, Quan A, Chan E, Kozlova N, Matai L, Lee JD, Rupaimoole R, Beca F, Muranen T, Slack FJ. EGFR-induced lncRNA TRIDENT promotes drug resistance in non-small cell lung cancer via phospho-TRIM28-mediated DNA damage repair. Proc Natl Acad Sci U S A 2025; 122:e2415389122. [PMID: 40030013 PMCID: PMC11912419 DOI: 10.1073/pnas.2415389122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 01/06/2025] [Indexed: 03/19/2025] Open
Abstract
Long noncoding RNAs (lncRNAs) play numerous roles in cellular biology and alterations in lncRNA expression profiles have been implicated in a variety of cancers. Here, we identify and characterize a lncRNA, TRIM28 Interacting DNA damage repair Enhancing Noncoding Transcript (TRIDENT), whose expression is induced upon epithelial growth factor receptor (EGFR) activation, and which exerts pro-oncogenic functions in EGFR-driven non-small cell lung cancer. Knocking down TRIDENT leads to decreased tumor-cell proliferation in both in vitro and in vivo model systems and induces sensitization to chemotherapeutic drugs. Using ChIRP-MS analysis we identified TRIM28 as a protein interactor of TRIDENT. TRIDENT promotes phosphorylation of TRIM28 and knocking down TRIDENT leads to accumulation of DNA damage in cancer cells via decreased TRIM28 phosphorylation. Altogether, our results reveal a molecular pathway in which TRIDENT regulates TRIM28 phosphorylation to promote tumor cell growth and drug resistance. Our findings suggest that TRIDENT can be developed as a biomarker or therapeutic target for EGFR mutant non-small cell lung cancer.
Collapse
Affiliation(s)
- Tanvi Saxena
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Anan Quan
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Erica Chan
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Nina Kozlova
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Latika Matai
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Jonathan D. Lee
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Rajesha Rupaimoole
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Francisco Beca
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Taru Muranen
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Frank J. Slack
- Harvard Medical School Initiative for RNA Medicine, Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| |
Collapse
|
19
|
Ge S, Cen J, Liu X, Hong Y, Tang Y, Yu Y, Li H, Xie T, Wang C, Cai M, Qiu Y, Zeng X, Peng T, Li Q, Li Q, Wu X, Song XL, Zhao SC. TGFβ-activated Asporin interacts with STMN1 to promote prostate cancer docetaxel chemoresistance and metastasis by upregulating the Wnt/β-catenin signaling pathway. Drug Resist Updat 2025; 81:101227. [PMID: 40073743 DOI: 10.1016/j.drup.2025.101227] [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: 12/12/2024] [Revised: 02/22/2025] [Accepted: 03/02/2025] [Indexed: 03/14/2025]
Abstract
AIMS Prostate cancer (PCa) remains a significant challenge in oncology due to high rates of drug resistance following standard treatment with docetaxel-based chemotherapy. Asporin (ASPN) has been regarded as an oncogene and its upregulation is closely associated with malignant behavior and poor prognosis in multiple cancers. Studies indicated that abnormal activation of the Wnt/β-catenin signaling pathway is prevalent in PCa. This study investigated the important role of ASPN in regulating Wnt/β-catenin signaling pathway in docetaxel resistance and metastasis of PCa. METHODS The impacts of ASPN on the docetaxel chemoresistance and metastasis of PCa cells were investigated in vitro and in vivo assays. Lastly, the underlying mechanism of ASPN was revealed by Western blot, protein immunocoprecipitation, Immunofluorescence, Immunohistochemical staining, liquid chromatography-mass spectrometry, and rescue experiments. RESULTS In present study, we reported that ASPN is highly expressed in PCa cells and tissues. Functional and molecular analyses showed that ASPN is activated by TGFβ and interacts with STMN1. ASPN increases the expression of β-catenin and promotes its nuclear accumulation by mediating the activation of the Wnt/β-catenin signaling pathway, thereby enhancing the stemness and epithelial-mesenchymal transition (EMT) of PCa cells, ultimately facilitating the docetaxel resistance and metastasis of PCa cells. CONCLUSIONS Our findings identify ASPN as a novel upstream regulatory factor of Wnt/β-catenin signaling pathway, suggesting that targeting the ASPN/STMN1/β-catenin axis could be a promising strategy for PCa intervention.
Collapse
Affiliation(s)
- Shengdong Ge
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China; Department of Urology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou 510900, China
| | - Jinpeng Cen
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Xiaofeng Liu
- Department of Urology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou 510900, China
| | - Yaying Hong
- Department of Urology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361101, PR China
| | - Yuting Tang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, PR China
| | - Yuzhong Yu
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Haolin Li
- Department of Urology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, PR China
| | - Tao Xie
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Chong Wang
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Maoping Cai
- Department of Urology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai Urological Cancer Institute, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, PR China
| | - Yang Qiu
- Department of Urology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou 510900, China
| | - Xianzi Zeng
- Department of Urology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou 510900, China
| | - Tianming Peng
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China
| | - Qu Li
- Department of Urology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou 510900, China
| | - Qianyi Li
- First Clinical Medical College, Southern Medical University, Guangzhou, China Contact Information, PR China
| | - Xingcheng Wu
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100720, PR China.
| | - Xian-Lu Song
- Department of Radiotherapy, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510095, PR China.
| | - Shan-Chao Zhao
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, PR China; Department of Urology, The Fifth Affiliated Hospital, Southern Medical University, Guangzhou 510900, China.
| |
Collapse
|
20
|
Kong E, Polacek N. TRIM21 modulates stability of pro-survival non-coding RNA vtRNA1-1 in human hepatocellular carcinoma cells. PLoS Genet 2025; 21:e1011614. [PMID: 40096176 PMCID: PMC11940608 DOI: 10.1371/journal.pgen.1011614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 03/26/2025] [Accepted: 02/10/2025] [Indexed: 03/19/2025] Open
Abstract
Recent studies expanded our knowledge of diverse pro-survival functions of short non-coding vault RNAs. One of the human vault RNA paralogs, vtRNA1-1, modulates several intracellular processes, including proliferation, apoptosis, autophagy, and drug resistance in various types of human cancer cells. However, protein interaction partners and mechanisms by which vtRNA1-1 levels are controlled within the cells remained elusive. Here, we describe a regulatory process for vtRNA1-1 stabilization mediated by the newly identified interacting proteins, TRIM21 and TRIM25, in human hepatocellular carcinoma (HCC) cells. Depleting TRIM21 or TRIM25 reduced the stability of vtRNA1-1 both in vivo and in vitro. We also identified the responsible sequence of vtRNA1-1 for the stability regulation by TRIM21 and TRIM25 and revealed another critical factor for vtRNA1-1 stability, an NSUN2-mediated methylation at C69 of vtRNA1-1. Consequently, our findings demonstrated that the TRIM proteins govern the stability of vtRNA1-1 depending on its methylation status in HCC cells. Since vtRNA1-1 is crucial for pro-survival characteristics in HCC cells, insight into vtRNA1-1 protein binding partners and the regulation of its stability can impact the development of new anticancer strategies.
Collapse
Affiliation(s)
- EunBin Kong
- Department for Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Norbert Polacek
- Department for Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| |
Collapse
|
21
|
Zhang J, Zou B, Geng Y, Yin H, Qin B, Gao W, Lin X, Sun N. TRIM36 serves as a prognostic indicator linked to immune infiltration in KIRC. Heliyon 2025; 11:e42540. [PMID: 40028597 PMCID: PMC11870256 DOI: 10.1016/j.heliyon.2025.e42540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 11/26/2024] [Accepted: 02/06/2025] [Indexed: 03/05/2025] Open
Abstract
Renal cell carcinoma (RCC) represents approximately 85 % of all renal malignant tumors, with kidney renal clear cell carcinoma (KIRC) being the most typical subtype. The tripartite motif (TRIM) family is involved in cancer initiation, progression, and therapy resistance. While TRIM36 has exhibited anti-tumor effects in various cancers, its relationship with KIRC remains unclear. In our research, we studied the relationship between TRIM36 and KIRC. Through a combination of bioinformatic analyses and validation experiments, we noted a rise in TRIM36 expression in KIRC, and the upregulation of TRIM36 expression is associated with a poorer prognosis in KIRC. Also, our findings from wound healing assays and transwell migration assays showed that TRIM36 promotes the proliferation and migration of KIRC cells. To understand the underlying mechanisms, we screened relevant genes and conducted enrichment analysis. We identified that TRIM36 may interact with 5 hub genes and involve in the cell cycle and cell division processes in KIRC. Additionally, through immune infiltration analysis, we found that TRIM36 may interact with 6 tumor-infiltrating lymphocytes (TILs) and 6 immune inhibitors. In summary, our research identifies TRIM36 as a promising biomarker and comprehensively explores its promoting effect on the proliferation of KIRC.
Collapse
Affiliation(s)
- Jikai Zhang
- Xuzhou Medical University, Xuzhou, China
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, China
| | - Botao Zou
- Xuzhou Medical University, Xuzhou, China
| | | | - Hang Yin
- Xuzhou Medical University, Xuzhou, China
| | | | - Wanjun Gao
- Xuzhou Medical University, Xuzhou, China
| | - Xiaoman Lin
- Department of Obstetrics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Nan Sun
- Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
22
|
Qi H, Wang J, Cao L. TRIM44 facilitates aggressive behaviors in multiple myeloma through promoting ZEB1 deubiquitination. Discov Oncol 2025; 16:248. [PMID: 40014271 PMCID: PMC11867989 DOI: 10.1007/s12672-025-01933-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 02/04/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Tripartite motif-containing 44 (TRIM44) involves in various tumor development. This study investigated role of TRIM44 in multiple myeloma (MM). MATERIALS AND METHODS TRIM44 levels in bone marrow tissues and MM cell lines was detected by quantitative reverse transcription PCR (RT-qPCR). Cell viability, migration, and invasion of MM cells were evaluated under the interference of TRIM44 expression. The role of TRIM44 on regulating tumor growth in vivo was also investigated in subcutaneous tumor xenograft models. The protein interact between TRIM44 and Zinc Finger E-Box Binding Homeobox 1 (ZEB1) was also studied according IP followed by western blotting assay. RESULTS TRIM44 was all highly expressed in collected bone marrow tissues and MM cell lines. Cell viability, migration, and invasion of MM cells with low expression of TRIM44 was significantly inhibited. Over-expression of TRIM44 can down-regulate the ZEB1 ubiquitination to enhance the protein stability. CONCLUSIONS TRIM44 exerts as an oncogenic factor to induce the oncogenesis of MM by stabilizing ZEB1.
Collapse
Affiliation(s)
- Hui Qi
- Department of Hematology, Affiliated Hospital of Inner Mongolia Medical University, 1 Tongdao North Road, Huimin District, Hohhot, 010050, China
| | - Jing Wang
- Department of Rheumatology and Immunology, Affiliated Hospital of Inner Mongolia Medical University, 1 Tongdao North Road, Huimin District, Hohhot, 010050, China
| | - Lixia Cao
- Department of Hematology, Affiliated Hospital of Inner Mongolia Medical University, 1 Tongdao North Road, Huimin District, Hohhot, 010050, China.
| |
Collapse
|
23
|
Liu J, Qiu L, Chen J, Zeng T. Lycorine hydrochloride Suppresses the Proliferation and Invasion of Esophageal Cancer by Targeting TRIM22 and Inhibiting the JAK2/STAT3 and Erk Pathways. Cancers (Basel) 2025; 17:718. [PMID: 40075566 PMCID: PMC11898953 DOI: 10.3390/cancers17050718] [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: 11/05/2024] [Revised: 02/03/2025] [Accepted: 02/07/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Tumor metastasis and poor drug efficacy are two of the most common causes of therapeutic failure in cancer patients. The underlying molecular mechanism requires further exploration, and novel effective curative strategies are urgently needed. Nature is a rich source of novel drugs, and Lycorine hydrochloride (Lyc.HCL) is a natural alkaloid with tremendous therapeutic potential. However, the molecular mechanisms of its antitumor activity are still unknown. In the current study, we investigated the effects and mechanisms of Lyc.HCL against esophageal squamous cell carcinomas (ESCCs), which pose serious threats to human life. METHODS An MTS assay and a clone formation assay were used to assess the viability of ESCC cell lines after Lyc.HCL treatment in vitro. Apoptosis and cell cycle regulation were analyzed using flow cytometry. Wound healing and Transwell assays were used to analyze cell migration, while invasion was analyzed using the Matrigel Transwell assay. We detected the expression of tripartite motif-containing 22 (TRIM22) through immunohistochemistry and Western blotting. A docking experiment was performed to explore the targets of Lyc.HCL. The expression levels of Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) and phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/extracellular signal-regulated kinase (Erk) pathway components were detected through Western blotting. A rescue experiment was performed to determine the potential role of TRIM22. In addition, we explored the in vivo anti-ESCC effects and mechanism of Lyc.HCL by using it to treat tumor-bearing mice. RESULTS The Lyc.HCL treatment was found to inhibit esophageal squamous cell carcinoma cell proliferation both in vitro and in vivo by blocking the cell cycle at the G2 phase, inhibiting cell migration and invasion. We found that the TRIM22 protein was highly expressed in ESCCs but not in normal esophageal tissue. Lyc.HCL directly targeted TRIM22, decreasing the expression of TRIM22 and the JAK2/STAT3 and Erk signaling pathways, both in vitro and in vivo. Using animal experiments, we observed that the depletion of TRIM22 delayed tumor growth, but this effect was significantly reversed upon TRIM22 overexpression. CONCLUSIONS Taken together, these findings demonstrate that Lyc.HCL can effectively suppress ESCC both in vitro and in vivo by targeting TRIM22 and regulating the JAK2/STAT3 and Erk pathways. These results suggest that Lyc.HCL may serve as a potential novel therapeutic for ESCC, with TRIM22 emerging as a promising target for treatment.
Collapse
Affiliation(s)
- Jingyan Liu
- Department of Medical Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524023, China
- Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Liangxian Qiu
- Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Jialing Chen
- Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Tao Zeng
- Department of Medical Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524023, China
| |
Collapse
|
24
|
Wang YY, Choi MJ, Kim JH, Choi JH. Enhanced Expression of TRIM46 in Ovarian Cancer Cells Induced by Tumor-Associated Macrophages Promotes Invasion via the Wnt/β-Catenin Pathway. Cells 2025; 14:214. [PMID: 39937005 PMCID: PMC11817100 DOI: 10.3390/cells14030214] [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: 12/22/2024] [Revised: 01/19/2025] [Accepted: 01/28/2025] [Indexed: 02/13/2025] Open
Abstract
Metastasis presents significant challenges in ovarian cancer treatment. Tumor-associated macrophages (TAMs) within the tumor microenvironment (TME) facilitate metastasis through epithelial-mesenchymal transition, yet the molecular underlying mechanisms are not fully understood. Here, we identified that tripartite motif-containing 46 (TRIM46) is significantly upregulated in ovarian cancer cells treated with a conditioned medium derived from macrophages stimulated by ovarian cancer cells (OC-MQs). Furthermore, TRIM46 was highly expressed in late-stage ovarian cancer patients and was associated with poor prognosis. Silencing of TRIM46 suppressed cancer cell invasion stimulated by OC-MQ and mesenchymal marker expression without affecting cell viability. Gene set enrichment analysis showed that the Wnt/β-catenin pathway is enriched in the high-TRIM46 expression group. Importantly, the inhibition of TRIM46-mediated β-catenin nuclear translocation and ovarian cancer cell invasion was reversed by CHIR99021, a Wnt/β-catenin activator. Additionally, C-X-C motif chemokine ligand 8 (CXCL8) was identified as being highly expressed in peritoneal MQs from the ascites of ovarian cancer patients and was positively correlated with C-X-C chemokine receptor 1/2 (CXCR1/2) expression in tumor cells. Notably, pre-treatment with reparixin, a CXCR1/2 inhibitor, blocked OC-MQ-induced TRIM46 expression and cell invasion. These results suggest that CXCL8 derived from TAMs promotes human ovarian cancer cell invasion via the Wnt/β-catenin pathway by upregulating TRIM46.
Collapse
Affiliation(s)
- Yi-Yue Wang
- Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D, State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine & School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 561113, China;
- Department of Biomedical and Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; (M.-J.C.); (J.-H.K.)
| | - Min-Jun Choi
- Department of Biomedical and Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; (M.-J.C.); (J.-H.K.)
| | - Jin-Hyung Kim
- Department of Biomedical and Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; (M.-J.C.); (J.-H.K.)
| | - Jung-Hye Choi
- Department of Biomedical and Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; (M.-J.C.); (J.-H.K.)
| |
Collapse
|
25
|
Fu Q, Chen P, Wang Z, Liu B, Zhou Q, Anwar I, Wang Y. The function of TRIML2 on the temozolomide resistance in glioblastoma. Ann Med Surg (Lond) 2025; 87:506-514. [PMID: 40110305 PMCID: PMC11918695 DOI: 10.1097/ms9.0000000000002888] [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: 09/03/2024] [Accepted: 12/09/2024] [Indexed: 03/22/2025] Open
Abstract
Background Acquired resistance to temozolomide is a major challenge for the effective treatment of glioblastoma (GBM). TRIML2, a member of the TRIM family, plays an important role in cancer genesis, progression, and treatment resistance. However, its mechanism of action in GBM resistance to temozolomide remains unclear. Methods RNA bulk sequencing data from temozolomide-resistant U87 cells and wild-type U87 cells were downloaded from the NCBI public database (GEO: GSE193957) and analyzed. A temozolomide-resistant cell line (U87-TR) was induced with temozolomide, and the expression of TRIML2 in temozolomide-resistant and wild-type cell lines (U87-WT) was verified by cell activity assays, wound-healing assays, and western blotting. The alteration of resistance to temozolomide was assessed following the overexpression of TRIML2 in the resistant cell line by lentiviral transfection. The differences in TRIML2 expression between primary GBM and recurrent GBM after temozolomide chemotherapy were verified by immunofluorescence, immunohistochemistry, and western blotting. Results The expression of TRIML2 was significantly lower in U87-TR cells than in U87-WT cells. After the TRIML2 overexpressed in U87-TR cells, their resistance to temozolomide was significantly decreased and became sensitive to temozolomide treatment. TRIML2 expression was significantly decreased in the temozolimide-resistant GBM tumors; in contrast, TRIML2 was relatively high expressed in the temozolimide-sensitive GBM tumors. Conclusions TRIML2 inhibits temozolomide resistance in GBM and thus may serve as a novel therapeutic target for overcoming GBM resistance to temozolomide.
Collapse
Affiliation(s)
- Qiang Fu
- Department of Neurosurgery, First Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Peipei Chen
- Department of Clinical Nutrition, First Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Zening Wang
- Department of Neurosurgery, First Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Bo Liu
- Department of Neurosurgery, First Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Qingjiu Zhou
- Department of Neurosurgery, First Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Ilhamjan Anwar
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yongxin Wang
- Department of Neurosurgery, First Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| |
Collapse
|
26
|
Yang Z, Cai J, Li J, Liu X, Liu W, Cui K, Bai Z, Dong Y, Peng D, Duan Q, Shahzad A, Zhang Q. The Mechanism of TRIM21 Inhibiting the Invasion and Migration of ccRCC by Stabilizing ASS1. Mol Carcinog 2025; 64:260-278. [PMID: 39513657 DOI: 10.1002/mc.23840] [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: 08/30/2024] [Revised: 10/13/2024] [Accepted: 10/22/2024] [Indexed: 11/15/2024]
Abstract
Clear cell renal cell carcinoma (ccRCC) is characterized by its aggressive invasion and metastasis, presenting significant clinical challenges. Gaining insights into the molecular mechanisms underlying its progression is crucial for the development of effective therapeutic strategies. Addressing a critical knowledge gap in understanding ccRCC tumorigenesis, this study aims to elucidate the expression patterns of TRIM21 in ccRCC, unravel its impact on ccRCC patient prognosis, and investigate the regulatory role of TRIM21 in ASS1 expression and urea cycle dysregulation within the context of ccRCC. The results demonstrate that TRIM21 is downregulated in ccRCC, and low expression of TRIM21 predicts an unfavorable prognosis for ccRCC patients. Furthermore, the upregulation of TRIM21 can inhibit the migration and invasion of ccRCC cells by regulating the ubiquitination modification of ASS1. This not only expands the functional role of TRIM21 in ccRCC tumorigenesis but also demonstrates its ability to reverse urea cycle dysregulation through stabilizing ASS1 expression. Specifically, abnormal downregulation of TRIM21 in ccRCC reduces K63 ubiquitination modification of ASS1, leading to decreased stability of the ASS1 protein, aggravated urea cycle dysregulation, and facilitated migration and invasion of ccRCC cells. Additionally, reduction in ASS1 reverses the depressed migration and invasion caused by overexpression of TRIM21 in ccRCC cells. In summary, our findings contribute to a deeper understanding of the functional role played by TRIM21 in ccRCC progression, pinpoint a unique and novel regulatory mechanism involving ectopic downregulation-mediated ASS1 ubiquitination modification and urea cycle dysfunction during ccRCC progression, and provide fresh insights for further investigation into the pathogenesis and metabolic reprogramming associated with ccRCC.
Collapse
Affiliation(s)
- Zhe Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, China
- Departments of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Jihao Cai
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, China
| | - Jingjing Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, China
| | - Xiangjie Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, China
| | - Wenjing Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, China
| | - Kun Cui
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, China
| | - Ziyuan Bai
- Departments of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yurong Dong
- Departments of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Dongmei Peng
- Departments of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Qiuxin Duan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, China
| | - Asif Shahzad
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, China
| | - Qiao Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
27
|
Qin S, Chang F, Sun X, Li Z, Wang Y, Lei D. TRIM47 promotes hypopharyngeal and laryngeal cancers progression through promoting K63-linked ubiquitination of vimentin. Cancer Sci 2025; 116:367-380. [PMID: 39584529 PMCID: PMC11786321 DOI: 10.1111/cas.16397] [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: 04/29/2024] [Revised: 10/23/2024] [Accepted: 11/01/2024] [Indexed: 11/26/2024] Open
Abstract
Hypopharyngeal and laryngeal cancers which belong to head and neck squamous cell carcinoma (HNSCC) are the two most malignant types of head and neck cancer, characterized by a low 5-year survival rate, high recurrence and metastasis rate. It is vital to explore strategies to suppress metastasis and improve prognosis for patients with these cancers. In this research, we analyzed the clinical data and found that E3 ubiquitin ligase TRIM47 was upregulated in cancer tissues of hypopharyngeal cancer and was closely associated with poor survival outcomes. In terms of mechanism, we performed tandem affinity chromatography and denatured Ni-NTA Agarose pulldown. As a result, TRIM47 was found to interact with vimentin and control vimentin stabilization through ubiquitination, specifically in the form of K63 chains. Importantly, through experiments of cancer cell viability and migration, we found that TRIM47 could enhance the proliferation and metastasis abilities of cancer cells in a vimentin-dependent manner, thus promoting the advancement of hypopharyngeal and laryngeal cancers. TRIM47 was verified to regulate cancer cells metastasis in vivo using metastasis models. All these results imply that TRIM47 emerges as a potential biomarker for early diagnosis and metastasis prediction of hypopharyngeal and laryngeal cancers and represents a promising therapeutic target.
Collapse
Affiliation(s)
- Shichao Qin
- Department of OtorhinolaryngologyQilu Hospital of Shandong UniversityJinanShandongChina
- NHC Key Laboratory of Otorhinolaryngology (Shandong University)JinanShandongChina
| | - Fen Chang
- Department of OtorhinolaryngologyQilu Hospital of Shandong UniversityJinanShandongChina
- NHC Key Laboratory of Otorhinolaryngology (Shandong University)JinanShandongChina
| | - Xiangkai Sun
- Department of OtorhinolaryngologyQilu Hospital of Shandong UniversityJinanShandongChina
- NHC Key Laboratory of Otorhinolaryngology (Shandong University)JinanShandongChina
| | - Zinan Li
- Department of OtorhinolaryngologyQilu Hospital of Shandong UniversityJinanShandongChina
- NHC Key Laboratory of Otorhinolaryngology (Shandong University)JinanShandongChina
| | - Yin Wang
- Department of OtorhinolaryngologyQilu Hospital of Shandong UniversityJinanShandongChina
- NHC Key Laboratory of Otorhinolaryngology (Shandong University)JinanShandongChina
| | - Dapeng Lei
- Department of OtorhinolaryngologyQilu Hospital of Shandong UniversityJinanShandongChina
- NHC Key Laboratory of Otorhinolaryngology (Shandong University)JinanShandongChina
| |
Collapse
|
28
|
Che X, Guan X, Ruan Y, Shen L, Shen Y, Liu H, Zhu C, Zhou T, Wang Y, Feng W. TRIM4 modulates the ubiquitin-mediated degradation of hnRNPDL and weakens sensitivity to CDK4/6 inhibitor in ovarian cancer. Front Med 2025; 19:121-133. [PMID: 39643799 DOI: 10.1007/s11684-024-1103-5] [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] [Received: 03/26/2024] [Accepted: 07/26/2024] [Indexed: 12/09/2024]
Abstract
Ovarian cancer is the most lethal malignancy affecting the female reproductive system. Pharmacological inhibitors targeting CDK4/6 have demonstrated promising efficacy across various cancer types. However, their clinical benefits in ovarian cancer patients fall short of expectations, with only a subset of patients experiencing these advantageous effects. This study aims to provide further clinical and biological evidence for antineoplastic effects of a CDK4/6 inhibitor (TQB4616) in ovarian cancer and explore underlying mechanisms involved. Patient-derived ovarian cancer organoid models were established to evaluate the effectiveness of TQB3616. Potential key genes related to TQB3616 sensitivity were identified through RNA-seq analysis, and TRIM4 was selected as a candidate gene for further investigation. Subsequently, co-immunoprecipitation and GST pull-down assays confirmed that TRIM4 binds to hnRNPDL and promotes its ubiquitination through RING and B-box domains. RIP assay demonstrated that hnRNPDL binded to CDKN2C isoform 2 and suppressed its expression by alternative splicing. Finally, in vivo studies confirmed that the addition of siTRIM4 significantly improved the effectiveness of TQB3616. Overall, our findings suggest that TRIM4 modulates ubiquitin-mediated degradation of hnRNPDL and weakens sensitivity to CDK4/6 inhibitors in ovarian cancer treatment. TRIM4 may serve as a valuable biomarker for predicting sensitivity to CDK4/6 inhibitors in ovarian cancer.
Collapse
Affiliation(s)
- Xiaoxia Che
- Department of Gynecology and Obstetrics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xin Guan
- Department of Gynecology and Obstetrics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yiyin Ruan
- Department of Gynecology and Obstetrics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lifei Shen
- Department of Gynecology and Obstetrics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yuhong Shen
- Department of Gynecology and Obstetrics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hua Liu
- Department of Gynecology and Obstetrics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chongying Zhu
- Department of Gynecology and Obstetrics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Tianyu Zhou
- Department of Gynecology and Obstetrics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yiwei Wang
- Department of Gynecology and Obstetrics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Weiwei Feng
- Department of Gynecology and Obstetrics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
29
|
Yu C, Zhang C, Zhang Q, Zhang C, Han J, Li J. TRIM47 promotes head and neck squamous cell carcinoma malignant progression by degrading XAF1 through ubiquitination. iScience 2025; 28:111590. [PMID: 39834862 PMCID: PMC11743097 DOI: 10.1016/j.isci.2024.111590] [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: 03/06/2024] [Revised: 06/06/2024] [Accepted: 12/10/2024] [Indexed: 01/22/2025] Open
Abstract
Tripartite motif-containing 47 (TRIM47) is a member of the TRIM family, which has E3 ligase activity and has been demonstrated to be involved in tumor development. In this work, we found that TRIM47 is highly expressed in head and neck squamous cell carcinomas (HNSCC) tissues. TRIM47 overexpression promoted HNSCC cell proliferation. Downregulation of TRIM47 suppressed HNSCC cell proliferation and promoted apoptosis and autophagy. TRIM47 suppression caused cell proliferation inhibition and apoptosis promotion could be reversed by 3-MA, an autophagy inhibitor. In mechanism, TRIM47 interacted with XIAP-associated factor 1 (XAF1), promoting its ubiquitination and degradation. XAF1 promoted HNSCC cell apoptosis and autophagy. TRIM47 overexpression caused cell proliferation promotion and autophagy inhibition could be reversed by XAF1 overexpression. Animal experiments confirmed that the knockdown of TRIM47 inhibits tumor growth in vivo. Ultimately, TRIM47 promotes the ubiquitination and degradation of XAF1 and suppresses apoptosis and autophagy to promote the progression of HNSCC.
Collapse
Affiliation(s)
- Changyun Yu
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Chen Zhang
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Qianqian Zhang
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Cai Zhang
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jingjie Han
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jinying Li
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Department of Otolaryngology Head and Neck Surgery, Children’s Hospital Affiliated to Zhengzhou University, Zhengzhou 450018, China
| |
Collapse
|
30
|
Miao C, Huang Y, Zhang C, Wang X, Wang B, Zhou X, Song Y, Wu P, Chen ZS, Feng Y. Post-translational modifications in drug resistance. Drug Resist Updat 2025; 78:101173. [PMID: 39612546 DOI: 10.1016/j.drup.2024.101173] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/24/2024] [Accepted: 11/16/2024] [Indexed: 12/01/2024]
Abstract
Resistance to antitumor drugs, antimicrobial drugs, and antiviral drugs severely limits treatment effectiveness and cure rate of diseases. Protein post-translational modifications (PTMs) represented by glycosylation, ubiquitination, SUMOylation, acetylation, phosphorylation, palmitoylation, and lactylation are closely related to drug resistance. PTMs are typically achieved by adding sugar chains (glycosylation), small proteins (ubiquitination), lipids (palmitoylation), or functional groups (lactylation) to amino acid residues. These covalent additions are usually the results of signaling cascades and could be reversible, with the triggering mechanisms depending on the type of modifications. PTMs are involved in antitumor drug resistance, not only as inducers of drug resistance but also as targets for reversing drug resistance. Bacteria exhibit multiple PTMs-mediated antimicrobial drug resistance. PTMs allow viral proteins and host cell proteins to form complex interaction networks, inducing complex antiviral drug resistance. This review summarizes the important roles of PTMs in drug resistance, providing new ideas for exploring drug resistance mechanisms, developing new drug targets, and guiding treatment plans.
Collapse
Affiliation(s)
- Chenggui Miao
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong; Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Anhui University of Chinese Medicine, Hefei 230012, China; Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Yurong Huang
- Department of Respiratory Medicine, Center of Infectious Diseases and Pathogen Biology, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, The First Hospital, Jilin University, Changchun 130021, China
| | - Cheng Zhang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong
| | - Xiao Wang
- Department of Clinical Nursing, School of Nursing, Anhui University of Chinese Medicine, Hefei, China
| | - Bing Wang
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Xinyue Zhou
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Yingqiu Song
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Peng Wu
- Department of Anatomy, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Zhe-Sheng Chen
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA.
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 999077, Hong Kong.
| |
Collapse
|
31
|
Xu M, Deng C, Man Z, Zhu H. TRIM47 is a prognostic biomarker for gallbladder cancer and promotes tumor progression through regulating K63-linked ubiquitination of PARP1. Transl Oncol 2025; 51:102164. [PMID: 39489093 PMCID: PMC11567951 DOI: 10.1016/j.tranon.2024.102164] [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: 05/27/2024] [Revised: 09/08/2024] [Accepted: 10/27/2024] [Indexed: 11/05/2024] Open
Abstract
BACKGROUND Gallbladder cancer (GBC) is one of the most lethal malignancies worldwide with an extremely poor prognosis. Previous studies have suggested that tripartite motif containing 47 (TRIM47) is involved in the progression of numerous cancers. However, the molecular mechanism and function of TRIM47 in GBC remain unclear. METHODS The clinical significance of TRIM47 was evaluated using immunohistochemistry. Functional assays were performed in vitro and in vivo to determine the role of TRIM47 in GBC. Mass spectrometric analysis, western blotting, and immunoprecipitation assays were performed to investigate the molecular mechanisms involved. RESULTS In this study, TRIM47 was upregulated in GBC tissues and associated with shorter overall survival rates and TRIM47 was involved in GBC cell proliferation, migration, and apoptosis. Mechanistically, TRIM47 interacts with PARP1 and mediates the K63-linked polyubiquitination of PARP1, thereby stabilizing its expression. Furthermore, TRIM47 activated the AKT signaling pathway via PARP1. CONCLUSION The present study revealed that TRIM47 contributes to the progression of GBC and is therefore an important biomarker for predicting the prognosis of GBC and for therapeutic intervention.
Collapse
Affiliation(s)
- Ming Xu
- Department of Hepatic-Biliary-Pancreatic Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Chuanmin Deng
- Department of Hepatic-Biliary-Pancreatic Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Zhongran Man
- Department of Hepatic-Biliary-Pancreatic Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Hongyi Zhu
- Department of Hepatic-Biliary-Pancreatic Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China; Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
32
|
Chen S, He Z, Cai K, Zhang Y, Zhu H, Pang C, Zhang J, Wang D, Xu X. TRIM59/RBPJ positive feedback circuit confers gemcitabine resistance in pancreatic cancer by activating the Notch signaling pathway. Cell Death Dis 2024; 15:932. [PMID: 39725730 DOI: 10.1038/s41419-024-07324-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 12/07/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024]
Abstract
Pancreatic cancer (PC) is one of the most lethal malignant tumors that lacks effective treatment, and gemcitabine-based chemoresistance occurs frequently. Therefore, new therapeutic strategies for PC are urgently needed. Tripartite motif containing 59 (TRIM59) plays an important role in breast and lung cancer chemoresistance. However, the association between TRIM59 and gemcitabine resistance in PC remains unclear. We identified TRIM59 as an innovative E3 ubiquitin ligase that activated Notch signaling in PC. TRIM59 levels were increased in PC and positively correlated with poor prognosis and gemcitabine resistance in PC patients. TRIM59 facilitated gemcitabine resistance in PC cells in vitro and in vivo. TRIM59 interacted with recombination signal binding protein for immunoglobulin kappa J region (RBPJ) and stabilized it by promoting K63-linked ubiquitination. RBPJ transcriptionally upregulated TRIM59 expression, forming a positive feedback loop with TRIM59. We identified a novel TRIM59 inhibitor, catechin, and confirmed that it sensitized PC cells to gemcitabine. TRIM59 conferred gemcitabine resistance in PC by promoting RBPJ K63-linked ubiquitination, followed by activating Notch signaling. Therefore, our study provides a promising target for gemcitabine sensitization in PC treatment.
Collapse
Affiliation(s)
- Shiyu Chen
- Department of Hepatobiliary Pancreatic Surgery, South China Hospital, Medical School, Shenzhen University, Shenzhen, 518116, P. R. China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Zhiwei He
- Department of Hepatobiliary Surgery, Shenzhen University General Hospital & Shenzhen University Clinical Medical Academy Center, Shenzhen University, Shenzhen, 518000, China
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, 550001, China
| | - Kun Cai
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
- Department of Gastrointestinal Surgery, South China Hospital, Medical School, Shenzhen University, Shenzhen, 518116, P. R. China
| | - Yan Zhang
- Department of Hepatobiliary Pancreatic Surgery, South China Hospital, Medical School, Shenzhen University, Shenzhen, 518116, P. R. China
| | - Hongyan Zhu
- Department of Hepatobiliary Pancreatic Surgery, South China Hospital, Medical School, Shenzhen University, Shenzhen, 518116, P. R. China
| | - Chong Pang
- Department of Hepatobiliary Pancreatic Surgery, South China Hospital, Medical School, Shenzhen University, Shenzhen, 518116, P. R. China
| | - Jiaqi Zhang
- Department of Hepatobiliary Pancreatic Surgery, South China Hospital, Medical School, Shenzhen University, Shenzhen, 518116, P. R. China
| | - Dong Wang
- Department of Hepatobiliary Pancreatic Surgery, South China Hospital, Medical School, Shenzhen University, Shenzhen, 518116, P. R. China
| | - Xundi Xu
- Department of Hepatobiliary Pancreatic Surgery, South China Hospital, Medical School, Shenzhen University, Shenzhen, 518116, P. R. China.
| |
Collapse
|
33
|
Li X, Zhou F, Niu K, Wang Y, Shi Y, Li Y, Gao X, Zhao W, Chen T, Zhang Y. Emerging discoveries on the role of TRIM14: from diseases to immune regulation. Cell Death Discov 2024; 10:513. [PMID: 39719450 DOI: 10.1038/s41420-024-02276-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/04/2024] [Accepted: 12/16/2024] [Indexed: 12/26/2024] Open
Abstract
TRIM14 is an important member of the TRIM family and is widely expressed in a variety of tissues. Like other members of the TRIM family, TRIM14 is also involved in ubiquitination modifications. TRIM14 was initially reported as an interferon-stimulated gene (ISG). In recent years, many studies have focused on the regulatory role of TRIM14 in signaling pathways such as the PI3K/Akt, NF-κB, and cGAS/STING pathways and revealed its mechanism of action in a variety of pathophysiological processes, and the regulation of TRIM14 has attracted the interest of many researchers as a new direction for the treatment of various diseases. However, there are no reviews on the role of TRIM14 in diseases. In this paper, we will describe the structure of TRIM14, review its role in cancer, cardiovascular disease, cervical spondylosis, inflammation and antiviral immunity, and provide an outlook on future research directions.
Collapse
Affiliation(s)
- Xinhao Li
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Feilong Zhou
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Kaiyi Niu
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yizhu Wang
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yanlong Shi
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yunxin Li
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xin Gao
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Weijie Zhao
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Tianyi Chen
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yewei Zhang
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
34
|
Wang M, Qin Y, Ai X, Liu X. RBM15-dependent m6A modification mediates progression of non-small cell lung cancer cells. Mol Med 2024; 30:267. [PMID: 39716068 DOI: 10.1186/s10020-024-01018-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 11/28/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is the predominant form of lung cancer, contributing significantly to global health and economic challenges. This study elucidated the role of RBM15 in NSCLC progression through its involvement in m6A modifications. METHODS RBM15 levels in NSCLC tissues and cells were assessed via RT-qPCR and Western blotting. The impact of RBM15 knockdown on NSCLC proliferation, invasion, and migration was evaluated using CCK-8, colony formation, and Transwell assays. Expression levels of KLF1, TRIM13, and ANXA8 were determined by RT-qPCR and Western blot. m6A methylation levels were analyzed, while RIP and MeRIP assays were employed to explore the interaction between YTHDF1/YTHDF2/m6A and KLF1/TRIM13, as well as KLF1 binding to the ANXA8 promoter. The ubiquitination of ANXA8 was examined through ubiquitination assays. Xenograft and metastasis models were utilized to assess RBM15's role in vivo. RESULTS RBM15 was found to be overexpressed in NSCLC. Silencing RBM15 led to decreased cell proliferation, invasion, and migration of NSCLC cells. RBM15 upregulated KLF1 and downregulated TRIM13 via YTHDF1/YTHDF2, resulting in the promotion of ANXA8 expression. KLF1 overexpression or TRIM13 downregulation partially reversed the suppressive effects of RBM15 knockdown on NSCLC cell proliferation. ANXA8, upregulated in NSCLC, mitigated the inhibitory effects of RBM15 silencing on malignant behaviors. In vivo, RBM15 downregulation hindered NSCLC cell proliferation and metastasis by modulating the KLF1-TRIM13/ANXA8 axis. CONCLUSION RBM15-mediated m6A methylation enhances KLF1 expression and suppresses TRIM13 via YTHDF1/YTHDF2, thereby promoting ANXA8 and facilitating NSCLC progression. These findings provide novel insights and potential therapeutic targets for NSCLC treatment.
Collapse
Affiliation(s)
- Man Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Jilin University, 1 Xinmin Street, Changchun, 130021, Jilin, China
| | - Yujiao Qin
- Department of Respiratory Medicine, The First Affiliated Hospital of Jilin University, 1 Xinmin Street, Changchun, 130021, Jilin, China
| | - Xiaoqi Ai
- Department of Respiratory Medicine, The First Affiliated Hospital of Jilin University, 1 Xinmin Street, Changchun, 130021, Jilin, China
| | - Xiuhua Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Jilin University, 1 Xinmin Street, Changchun, 130021, Jilin, China.
| |
Collapse
|
35
|
Wu W, Yang J, Yu T, Zou Z, Huang X. The Role and Mechanism of TRIM Proteins in Gastric Cancer. Cells 2024; 13:2107. [PMID: 39768197 PMCID: PMC11674240 DOI: 10.3390/cells13242107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Tripartite motif (TRIM) family proteins, distinguished by their N-terminal region that includes a Really Interesting New Gene (RING) domain with E3 ligase activity, two B-box domains, and a coiled-coil region, have been recognized as significant contributors in carcinogenesis, primarily via the ubiquitin-proteasome system (UPS) for degrading proteins. Mechanistically, these proteins modulate a variety of signaling pathways, including Wnt/β-catenin, PI3K/AKT, and TGF-β/Smad, contributing to cellular regulation, and also impact cellular activities through non-signaling mechanisms, including modulation of gene transcription, protein degradation, and stability via protein-protein interactions. Currently, growing evidence indicates that TRIM proteins emerge as potential regulators in gastric cancer, exhibiting both tumor-suppressive and oncogenic roles. Given their critical involvement in cellular processes and the notable challenges of gastric cancer, exploring the specific contributions of TRIM proteins to this disease is necessary. Consequently, this review elucidates the roles and mechanisms of TRIM proteins in gastric cancer, emphasizing their potential as therapeutic targets and prognostic factors.
Collapse
Affiliation(s)
- Wangxi Wu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Jiangxi Provincial Key Laboratory of Bioengineering Drugs, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (W.W.); (T.Y.)
- The Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (J.Y.); (Z.Z.)
| | - Jinyu Yang
- The Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (J.Y.); (Z.Z.)
| | - Tian Yu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Jiangxi Provincial Key Laboratory of Bioengineering Drugs, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (W.W.); (T.Y.)
| | - Zhuoling Zou
- The Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (J.Y.); (Z.Z.)
| | - Xuan Huang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Jiangxi Provincial Key Laboratory of Bioengineering Drugs, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; (W.W.); (T.Y.)
- Chongqing Research Institute, Nanchang University, Chongqing 400010, China
| |
Collapse
|
36
|
Wu Q, Nandi D, Sharma D. TRIM-endous functional network of tripartite motif 29 (TRIM29) in cancer progression and beyond. Cancer Metastasis Rev 2024; 44:16. [PMID: 39644332 PMCID: PMC11625080 DOI: 10.1007/s10555-024-10226-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 10/16/2024] [Indexed: 12/09/2024]
Abstract
While most Tripartite motif (TRIM) family proteins are E3 ubiquitin ligases, some members have functions beyond the regulation of ubiquitination, impacting normal physiological processes and disease progression. TRIM29, an important member of the TRIM family, exerts a predominant influence on cancer growth, epithelial-to-mesenchymal transition, stemness and metastatic progression by directly potentiating multiple canonical oncogenic pathways. The cancer-promoting effect of TRIM29 is also evident in metabolic interventions and interference with the efficacy of cancer therapeutics. As expected for any key node in cancer, the expression of TRIM29 is tightly regulated by non-coding RNAs, epigenetic modulation, and post-translational regulation. A systematic discussion of how TRIM29 is regulated in cancer, its influences on cancer progression, and its impact on cancer therapeutics is presented in this review. We also explore the context-dependent alterations between TRIM29 function from oncogenic to tumor suppression. As TRIM29 is involved in multiple aspects of cancer progression, a better understanding of its biological impact in cancer may help improve prognosis and develop novel therapeutic combinations, leading to improved personalized cancer care.
Collapse
Affiliation(s)
- Qitong Wu
- Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Deeptashree Nandi
- Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Dipali Sharma
- Department of Oncology, Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans Street, CRB 1, Rm 145, Baltimore, MD, 21231, USA.
| |
Collapse
|
37
|
Weng C, Jin R, Jin X, Yang Z, He C, Zhang Q, Xu J, Lv B. Exploring the Mechanisms, Biomarkers, and Therapeutic Targets of TRIM Family in Gastrointestinal Cancer. Drug Des Devel Ther 2024; 18:5615-5639. [PMID: 39654601 PMCID: PMC11626976 DOI: 10.2147/dddt.s482340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 11/23/2024] [Indexed: 12/12/2024] Open
Abstract
Gastrointestinal region (GI) cancers are closely linked to the ubiquitination system, with the E3 ubiquitin ligase playing a crucial role by targeting various substrates. As E3 ubiquitin ligases, proteins of tripartite motif (TRIM) family play a role in cancer signaling, development, apoptosis, and formation. These proteins regulate diverse biological activities and signaling pathways. This study comprehensively outlines the functions of TRIM proteins in gastrointestinal physiology, contributing to our knowledge of the molecular pathways involved in gastrointestinal tumors. Gastrointestinal region (GI) cancers are closely linked to the ubiquitination system, with the E3 ubiquitin ligase playing a crucial role by targeting various substrates.
Collapse
Affiliation(s)
- Chunyan Weng
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, People’s Republic of China
| | - Rijuan Jin
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, People’s Republic of China
| | - Xiaoliang Jin
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, People’s Republic of China
| | - Zimei Yang
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, People’s Republic of China
| | - Chenghai He
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, People’s Republic of China
- Department of Gastroenterology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang Province, People’s Republic of China
| | - Qiuhua Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, People’s Republic of China
| | - Jingli Xu
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, People’s Republic of China
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang Province, People’s Republic of China
| | - Bin Lv
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, People’s Republic of China
| |
Collapse
|
38
|
Mafi A, Hedayati N, Kahkesh S, Khoshayand S, Alimohammadi M, Farahani N, Hushmandi K. The landscape of circRNAs in gliomas temozolomide resistance: Insights into molecular pathways. Noncoding RNA Res 2024; 9:1178-1189. [PMID: 39022676 PMCID: PMC11250881 DOI: 10.1016/j.ncrna.2024.05.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/01/2024] [Accepted: 05/20/2024] [Indexed: 07/20/2024] Open
Abstract
As the deadliest type of primary brain tumor, gliomas represent a significant worldwide health concern. Circular RNA (circRNA), a unique non-coding RNA molecule, seems to be one of the most alluring target molecules involved in the pathophysiology of many kinds of cancers. CircRNAs have been identified as prospective targets and biomarkers for the diagnosis and treatment of numerous disorders, particularly malignancies. Recent research has established a clinical link between temozolomide (TMZ) resistance and certain circRNA dysregulations in glioma tumors. CircRNAs may play a therapeutic role in controlling or overcoming TMZ resistance in gliomas and may provide guidance for a novel kind of individualized glioma therapy. To address the biological characteristics of circRNAs and their potential to induce resistance to TMZ, this review has highlighted and summarized the possible roles that circRNAs may play in molecular pathways of drug resistance, including the Ras/Raf/ERK PI3K/Akt signaling pathway and metabolic processes in gliomas.
Collapse
Affiliation(s)
- Alireza Mafi
- Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Neda Hedayati
- School of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Samaneh Kahkesh
- Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Sara Khoshayand
- School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Najma Farahani
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| |
Collapse
|
39
|
Tang Y, Meng X, Luo X, Yao W, Tian L, Zhang Z, Zhao Y, Xiao J, Zhu H, Hu J. Arginine methylation-dependent TRIM47 stability mediated by CARM1 promotes the metastasis of hepatocellular carcinoma. Cell Death Discov 2024; 10:477. [PMID: 39567506 PMCID: PMC11579460 DOI: 10.1038/s41420-024-02244-4] [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: 05/08/2024] [Revised: 11/06/2024] [Accepted: 11/11/2024] [Indexed: 11/22/2024] Open
Abstract
The tripartite motif (TRIM) protein family has been shown to play important roles in the occurrence and development of various tumors. However, the biological functions of TRIM47 and its regulatory mechanism in hepatocellular carcinoma (HCC) remain unexplored. Here, we showed that TRIM47 was upregulated in HCC tissues compared with adjacent normal tissues, especially at advanced stages, and associated with poor prognosis in HCC patients. Functional studies demonstrated that TRIM47 enhanced the migration and invasion ability of HCC cells in vitro and in vivo. Mechanistically, TRIM47 promotes HCC metastasis through interacting with SNAI1 and inhibiting its degradation by proteasome. Moreover, TRIM47 was di-methylated by CARM1 at its arginine 210 (R210) and arginine 582 (R582), which protected TRIM47 from the ubiquitination and degradation mediated by E3 ubiquitin ligase complex CRL4CRBN. Collectively, our study reveals a pro-metastasis role of TRIM47 in HCC, unveils a unique mechanism controlling TRIM47 stability by CARM1 mediated arginine methylation, and highlights the role of the CARM1-CRL4CRBN-TRIM47-SNAI1 axis in HCC metastasis. This work may provide potential therapeutic targets for metastatic HCC treatment.
Collapse
Affiliation(s)
- Yuzhe Tang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, China
| | - Xiang Meng
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, China
| | - Xia Luo
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wentao Yao
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, China
| | - Li Tian
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, China
| | - Zijian Zhang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, China
| | - Yuan Zhao
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, China
| | - Juan Xiao
- Institute of Neuroscience and Brain Diseases, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China.
| | - Haichuan Zhu
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, China.
| | - Jia Hu
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, China.
| |
Collapse
|
40
|
Wang J, Ye J, Liu R, Chen C, Wang W. TRIM47 drives gastric cancer cell proliferation and invasion by regulating CYLD protein stability. Biol Direct 2024; 19:106. [PMID: 39516831 PMCID: PMC11546413 DOI: 10.1186/s13062-024-00555-1] [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: 07/16/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
The expression of TRIM47, a member of the TRIM protein and E3 ubiquitin ligase families, is elevated in various cancers, such as non-small cell lung cancer and colorectal cancer, and is linked to poor prognosis. This study aimed to investigate the role of TRIM47 in gastric cancer development. Using The Cancer Genome Atlas-Stomach Adenocarcinoma (TCGA-STAD) dataset and analysis of 20 patient samples from our center, TRIM47 was found to be significantly up-regulated in gastric cancer tissues and associated with advanced N-stage and poor prognosis. We constructed stable TRIM47 knockdown and overexpressing gastric cancer cell lines. CCK8, EDU, colony formation, wound healing, and Transwell tests were used to evaluate the effects on cell proliferation, invasion, and migration. The results showed that TRIM47 knockdown inhibited the proliferation, migration and invasion of gastric cancer cells, while TRIM47 overexpression promoted these behaviors. These results were further confirmed in vivo. In the mechanism part, we found that TRIM47 interacts with CYLD protein. Moreover, TRIM47 promotes K48-linked ubiquitination, leading to the degradation of CYLD by the proteasome, thereby activating the NF-κB pathway and regulating the biological behavior of gastric cancer cells. Taken together, our study demonstrated that TRIM47 is involved in the proliferation and metastasis of gastric cancer through the CYLD/NF-κB pathway.
Collapse
Affiliation(s)
- Jianguo Wang
- Department of General Surgery, Renmin Hospital of Wuhan University, 99 Ziyang Road, Wuhan, Hubei, 430060, PR China
- Laboratory of General Surgery, Renmin Hospital of Wuhan University, 99 Ziyang Road, Wuhan, Hubei, 430060, PR China
- Department of Hepatobiliary Surgery, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| | - Jing Ye
- Department of General Surgery, Renmin Hospital of Wuhan University, 99 Ziyang Road, Wuhan, Hubei, 430060, PR China
- Laboratory of General Surgery, Renmin Hospital of Wuhan University, 99 Ziyang Road, Wuhan, Hubei, 430060, PR China
| | - Rongqiang Liu
- Department of General Surgery, Renmin Hospital of Wuhan University, 99 Ziyang Road, Wuhan, Hubei, 430060, PR China
- Laboratory of General Surgery, Renmin Hospital of Wuhan University, 99 Ziyang Road, Wuhan, Hubei, 430060, PR China
| | - Chen Chen
- Department of General Surgery, Renmin Hospital of Wuhan University, 99 Ziyang Road, Wuhan, Hubei, 430060, PR China.
- Laboratory of General Surgery, Renmin Hospital of Wuhan University, 99 Ziyang Road, Wuhan, Hubei, 430060, PR China.
| | - Weixing Wang
- Department of General Surgery, Renmin Hospital of Wuhan University, 99 Ziyang Road, Wuhan, Hubei, 430060, PR China.
- Laboratory of General Surgery, Renmin Hospital of Wuhan University, 99 Ziyang Road, Wuhan, Hubei, 430060, PR China.
| |
Collapse
|
41
|
Maghsoudloo M, Mokhtari K, Jamali B, Gholamzad A, Entezari M, Hashemi M, Fu J. Multifaceted role of TRIM28 in health and disease. MedComm (Beijing) 2024; 5:e790. [PMID: 39534556 PMCID: PMC11554878 DOI: 10.1002/mco2.790] [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: 06/02/2024] [Revised: 09/28/2024] [Accepted: 09/28/2024] [Indexed: 11/16/2024] Open
Abstract
The TRIM (tripartite motif) family, with TRIM28 as a key member, plays a vital role in regulating health and disease. TRIM28 contains various functional domains essential for transcriptional regulation, primarily through its interaction with KRAB-ZNF proteins, which influence chromatin remodeling and gene expression. Despite extensive research, the precise mechanisms by which TRIM28 impacts health and disease remain elusive. This review delves into TRIM28's multifaceted roles in maintaining health, contributing to a variety of diseases, and influencing cancer progression. In cancers, TRIM28 exhibits a dual nature, functioning as both a tumor promoter and suppressor depending on the cellular context and cancer type. The review also explores its critical involvement in processes such as DNA repair, cell cycle regulation, epithelial-to-mesenchymal transition, and the maintenance of stem cell properties. By uncovering TRIM28's complex roles across different cancers and other diseases, this review underscores its potential as a therapeutic target. The significance of TRIM28 as a versatile regulator opens the door to innovative therapeutic strategies, particularly in cancer treatment and the management of other diseases. Ongoing research into TRIM28 may yield key insights into disease progression and novel treatment options.
Collapse
Affiliation(s)
- Mazaher Maghsoudloo
- Key Laboratory of Epigenetics and Oncologythe Research Center for Preclinical MedicineSouthwest Medical UniversityLuzhouSichuanChina
| | - Khatere Mokhtari
- Department of Cellular and Molecular Biology and MicrobiologyFaculty of Biological Science and TechnologyUniversity of IsfahanIsfahanIran
| | - Behdokht Jamali
- Department of Microbiology and GeneticKherad Institute of Higher EducationBusheherIran
| | - Amir Gholamzad
- Farhikhtegan Medical Convergence Sciences Research CenterFarhikhtegan Hospital Tehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research CenterFarhikhtegan Hospital Tehran Medical SciencesIslamic Azad UniversityTehranIran
- Department of GeneticsFaculty of Advanced Science and TechnologyTehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research CenterFarhikhtegan Hospital Tehran Medical SciencesIslamic Azad UniversityTehranIran
- Department of GeneticsFaculty of Advanced Science and TechnologyTehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Junjiang Fu
- Key Laboratory of Epigenetics and Oncologythe Research Center for Preclinical MedicineSouthwest Medical UniversityLuzhouSichuanChina
| |
Collapse
|
42
|
Wei S, Huang X, Zhu Q, Chen T, Zhang Y, Tian J, Pan T, Zhang L, Xie T, Zhang Q, Kuang X, Lei E, Li Y. TRIM65 deficiency alleviates renal fibrosis through NUDT21-mediated alternative polyadenylation. Cell Death Differ 2024; 31:1422-1438. [PMID: 38951701 PMCID: PMC11519343 DOI: 10.1038/s41418-024-01336-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/19/2024] [Accepted: 06/25/2024] [Indexed: 07/03/2024] Open
Abstract
Chronic kidney disease (CKD) is a major global health concern and the third leading cause of premature death. Renal fibrosis is the primary process driving the progression of CKD, but the mechanisms behind it are not fully understood, making treatment options limited. Here, we find that the E3 ligase TRIM65 is a positive regulator of renal fibrosis. Deletion of TRIM65 results in a reduction of pathological lesions and renal fibrosis in mouse models of kidney fibrosis induced by unilateral ureteral obstruction (UUO)- and folic acid. Through screening with a yeast-hybrid system, we identify a new interactor of TRIM65, the mammalian cleavage factor I subunit CFIm25 (NUDT21), which plays a crucial role in fibrosis through alternative polyadenylation (APA). TRIM65 interacts with NUDT21 to induce K48-linked polyubiquitination of lysine 56 and proteasomal degradation, leading to the inhibition of TGF-β1-mediated SMAD and ERK1/2 signaling pathways. The degradation of NUDT21 subsequently altered the length and sequence content of the 3'UTR (3'UTR-APA) of several pro-fibrotic genes including Col1a1, Fn-1, Tgfbr1, Wnt5a, and Fzd2. Furthermore, reducing NUDT21 expression via hydrodynamic renal pelvis injection of adeno-associated virus 9 (AAV9) exacerbated UUO-induced renal fibrosis in the normal mouse kidneys and blocked the protective effect of TRIM65 deletion. These findings suggest that TRIM65 promotes renal fibrosis by regulating NUDT21-mediated APA and highlight TRIM65 as a potential target for reducing renal fibrosis in CKD patients.
Collapse
Affiliation(s)
- Sisi Wei
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Xuan Huang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies; Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Qing Zhu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies; Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Tao Chen
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Yan Zhang
- Department of Biological Sciences, College of Sciences and Arts, Michigan Technological University, Houghton, MI, 49931-1295, USA
| | - Juan Tian
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Tingyu Pan
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Lv Zhang
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Tao Xie
- The National Engineering Research Center for Bioengineering Drugs and the Technologies; Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Qi Zhang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies; Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Xian Kuang
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Enjun Lei
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Yong Li
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
43
|
Sreepangi S, Baha H, Opoku LA, Jones NX, Konadu M, Alem F, Barrera MD, Narayanan A. Host-Driven Ubiquitination Events in Vector-Transmitted RNA Virus Infections as Options for Broad-Spectrum Therapeutic Intervention Strategies. Viruses 2024; 16:1727. [PMID: 39599842 PMCID: PMC11599102 DOI: 10.3390/v16111727] [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: 06/11/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/29/2024] Open
Abstract
Many vector-borne viruses are re-emerging as public health threats, yet our understanding of the virus-host interactions critical for productive infection remains limited. The ubiquitination of proteins, including host- and pathogen-derived proteins is a highly prominent and consistent post-translational modification that regulates protein function through signaling and degradation. Viral proteins are documented to hijack the host ubiquitination machinery to modulate multiple host processes including antiviral defense mechanisms. The engagement of the host ubiquitination machinery in the post-translational modification of viral proteins to support aspects of the viral life cycle including assembly and egress is also well documented. Exploring the role ubiquitination plays in the life cycle of vector-transmitted viral pathogens will increase the knowledge base pertinent to the impact of host-enabled ubiquitination of viral and host proteins and the consequences on viral pathogenesis. In this review, we explore E3 ligase-regulated ubiquitination pathways functioning as proviral and viral restriction factors in the context of acutely infectious, vector-transmitted viral pathogens and the potential for therapeutically targeting them for countermeasures development.
Collapse
Affiliation(s)
- Sanskruthi Sreepangi
- School of Systems Biology, College of Science, George Mason University, Fairfax, VA 22030, USA; (S.S.); (H.B.); (L.A.O.); (N.X.J.); (M.K.); (M.D.B.)
| | - Haseebullah Baha
- School of Systems Biology, College of Science, George Mason University, Fairfax, VA 22030, USA; (S.S.); (H.B.); (L.A.O.); (N.X.J.); (M.K.); (M.D.B.)
| | - Lorreta Aboagyewa Opoku
- School of Systems Biology, College of Science, George Mason University, Fairfax, VA 22030, USA; (S.S.); (H.B.); (L.A.O.); (N.X.J.); (M.K.); (M.D.B.)
| | - Naomi X. Jones
- School of Systems Biology, College of Science, George Mason University, Fairfax, VA 22030, USA; (S.S.); (H.B.); (L.A.O.); (N.X.J.); (M.K.); (M.D.B.)
| | - Maame Konadu
- School of Systems Biology, College of Science, George Mason University, Fairfax, VA 22030, USA; (S.S.); (H.B.); (L.A.O.); (N.X.J.); (M.K.); (M.D.B.)
| | - Farhang Alem
- Institute of Biohealth Innovation, George Mason University, Fairfax, VA 22030, USA;
| | - Michael D. Barrera
- School of Systems Biology, College of Science, George Mason University, Fairfax, VA 22030, USA; (S.S.); (H.B.); (L.A.O.); (N.X.J.); (M.K.); (M.D.B.)
| | - Aarthi Narayanan
- Department of Biology, College of Science, George Mason University, Fairfax, VA 22030, USA
| |
Collapse
|
44
|
Shen J, Lai Y, Wu Y, Lin X, Zhang C, Liu H. Ubiquitination in osteosarcoma: unveiling the impact on cell biology and therapeutic strategies. Cancer Biol Med 2024; 21:j.issn.2095-3941.2024.0231. [PMID: 39475222 PMCID: PMC11523277 DOI: 10.20892/j.issn.2095-3941.2024.0231] [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: 07/01/2024] [Accepted: 08/30/2024] [Indexed: 11/02/2024] Open
Abstract
Ubiquitination, a multifaceted post-translational modification, regulates protein function, degradation, and gene expression. The pivotal role of ubiquitination in the pathogenesis and progression of cancer, including colorectal, breast, and liver cancer, is well-established. Osteosarcoma, an aggressive bone tumor predominantly affecting adolescents, also exhibits dysregulation of the ubiquitination system, encompassing both ubiquitination and deubiquitination processes. This dysregulation is now recognized as a key driver of osteosarcoma development, progression, and chemoresistance. This review highlights recent progress in elucidating how ubiquitination modulates tumor behavior across signaling pathways. We then focus on the mechanisms by which ubiquitination influences osteosarcoma cell function. Finally, we discuss the potential for targeting the ubiquitin-proteasome system in osteosarcoma therapy. By unraveling the impact of ubiquitination on osteosarcoma cell physiology, we aim to facilitate the development of novel strategies for prognosis, staging, treatment, and overcoming chemoresistance.
Collapse
Affiliation(s)
- Jianlin Shen
- Department of Orthopedics, Affiliated Hospital of Putian University, Putian 351100, China
- Central Laboratory, Affiliated Hospital of Putian University, Putian 351100, China
| | - Yue Lai
- Department of Orthopedics, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Yanjiao Wu
- Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde), Foshan 528000, China
| | - Xuan Lin
- Department of Environmental and Biological Engineering, Putian University, Putian 351100, China
| | - Cheng Zhang
- Department of Trauma Center, Zhongda Hospital, Southeast University, Nanjing 210000, China
| | - Huan Liu
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
45
|
Yang Y, Hao X, Zhang J, Gao T, Huo M, Liu W, Hu T, Ma T, Yuan B, Zhang M, Teng X, Yu H, Huang W, Wang Y. The E3 ligase TRIM22 functions as a tumor suppressor in breast cancer by targeting CCS for proteasomal degradation to inhibit STAT3 signaling. Cancer Lett 2024; 600:217157. [PMID: 39127340 DOI: 10.1016/j.canlet.2024.217157] [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: 01/02/2024] [Revised: 07/02/2024] [Accepted: 08/03/2024] [Indexed: 08/12/2024]
Abstract
Deregulation of E3 ubiquitin ligases drives the proliferation and metastasis of various cancers; however, the underlying mechanisms remain unknown. This study aimed to investigate the role of tripartite motif-containing 22 (TRIM22), a poorly investigated E3 ubiquitin ligase in the TRIM family, as a tumor suppressor in breast cancer. High expression of TRIM22 in breast cancer correlated with better prognosis. Functional experiments demonstrated that TRIM22 significantly inhibited the proliferation and invasion of breast cancer cells. Label-free proteomics and biochemical analyses revealed that the copper chaperone for superoxide dismutase (CCS), an oncoprotein that is upregulated in breast cancer and promotes the growth and invasion of breast cancer cells, was a target of TRIM22 for degradation via K27-linked ubiquitination. Notably, the ability of the coiled-coil domain-defective mutants of TRIM22 to induce CCS ubiquitination and degradation diminished, with lysine 76 of the CCS serving as the ubiquitination site. Moreover, the TRIM22-mediated inhibition of the proliferation and invasion of breast cancer cells was restored by ectopic CCS expression. RNA-sequencing experiments using Gene Set Enrichment Analysis demonstrated that TRIM22 is involved in the JAK-STAT signaling pathway. TRIM22 overexpression also improved reactive oxygen species levels in breast cancer cells and inhibited STAT3 phosphorylation, which was restored via CCS overexpression or N-acetyl-l-cysteine treatment. Chromatin immunoprecipitation-quantitative polymerase chain reaction results showed that TRIM22 overexpression decreased the enrichment of phosphorylated STAT3 in FN1, VIM and JARID2 promoters. Clinically, low TRIM22 expression correlated with high CCS expression and decreased survival rates in patients with breast cancer. Moreover, TRIM22 upregulation was associated with a better prognosis in patients with breast cancer who received classical therapy. TRIM22 expression was downregulated in many cancer types, including colon, kidney, lung, and prostate cancers. To the best of our knowledge, the E3 ubiquitin ligase TRIM22 was first reported as a tumor suppressor that inhibits the proliferation and invasion of breast cancer cells through CCS ubiquitination and degradation. TRIM22 is a potential prognostic biomarker in patients with breast cancer.
Collapse
Affiliation(s)
- Yunkai Yang
- Key Laboratory of Cancer and Microbiome, 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, 100021, China
| | - Xinhui Hao
- Key Laboratory of Cancer and Microbiome, 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, 100021, China; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Jingyao Zhang
- Key Laboratory of Cancer and Microbiome, 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, 100021, China
| | - Tianyang Gao
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Miaomiao Huo
- Key Laboratory of Cancer and Microbiome, 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, 100021, China
| | - Wei Liu
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Ting Hu
- Key Laboratory of Cancer and Microbiome, 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, 100021, China
| | - Tianyu Ma
- Key Laboratory of Cancer and Microbiome, 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, 100021, China
| | - Baowen Yuan
- Key Laboratory of Cancer and Microbiome, 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, 100021, China
| | - Min Zhang
- Key Laboratory of Cancer and Microbiome, 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, 100021, China
| | - Xu Teng
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Hefen Yu
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Wei Huang
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yan Wang
- Key Laboratory of Cancer and Microbiome, 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, 100021, China; Institute of Cancer Research, Henan Academy of Innovations in Medical Sciences, Zhengzhou, Henan Province, 450000, China; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China; Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
46
|
Chiang DC, Yap BK. TRIM25, TRIM28 and TRIM59 and Their Protein Partners in Cancer Signaling Crosstalk: Potential Novel Therapeutic Targets for Cancer. Curr Issues Mol Biol 2024; 46:10745-10761. [PMID: 39451518 PMCID: PMC11506413 DOI: 10.3390/cimb46100638] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/22/2024] [Accepted: 09/23/2024] [Indexed: 10/26/2024] Open
Abstract
Aberrant expression of TRIM proteins has been correlated with poor prognosis and metastasis in many cancers, with many TRIM proteins acting as key oncogenic factors. TRIM proteins are actively involved in many cancer signaling pathways, such as p53, Akt, NF-κB, MAPK, TGFβ, JAK/STAT, AMPK and Wnt/β-catenin. Therefore, this review attempts to summarize how three of the most studied TRIMs in recent years (i.e., TRIM25, TRIM28 and TRIM59) are involved directly and indirectly in the crosstalk between the signaling pathways. A brief overview of the key signaling pathways involved and their general cross talking is discussed. In addition, the direct interacting protein partners of these TRIM proteins are also highlighted in this review to give a picture of the potential protein-protein interaction that can be targeted for future discovery and for the development of novel therapeutics against cancer. This includes some examples of protein partners which have been proposed to be master switches to various cancer signaling pathways.
Collapse
Affiliation(s)
| | - Beow Keat Yap
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor, Penang 11800, Malaysia
| |
Collapse
|
47
|
Bansod S, Dodhiawala PB, Geng Y, Bulle A, Liu P, Li L, Townsend R, Grierson PM, Held JM, Adhikari H, Lim KH. The TRIM4 E3 ubiquitin ligase degrades TPL2 and is modulated by oncogenic KRAS. Cell Rep 2024; 43:114667. [PMID: 39178114 PMCID: PMC11472288 DOI: 10.1016/j.celrep.2024.114667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 03/24/2024] [Accepted: 08/06/2024] [Indexed: 08/25/2024] Open
Abstract
Loss-of-function mutations in the C terminus of TPL2 kinase promote oncogenesis by impeding its proteasomal degradation, leading to sustained protein expression. However, the degradation mechanism for TPL2 has remained elusive. Through proximity-dependent biotin identification (BioID), we uncovered tripartite motif-containing 4 (TRIM4) as the E3 ligase that binds and degrades TPL2 by polyubiquitination of lysines 415 and 439. The naturally occurring TPL2 mutants R442H and E188K exhibit impaired TRIM4 binding, enhancing their stability. We further discovered that TRIM4 itself is stabilized by another E3 ligase, TRIM21, which in turn is regulated by KRAS. Mutant KRAS recruits RNF185 to degrade TRIM21 and subsequently TRIM4, thereby stabilizing TPL2. In the presence of mutant KRAS, TPL2 phosphorylates and degrades GSK3β, resulting in β-catenin stabilization and activation of the Wnt pathway. These findings elucidate the physiological mechanisms regulating TPL2 and its exploitation by mutant KRAS, underscoring the need to develop TPL2 inhibitors for KRAS-mutant cancers.
Collapse
Affiliation(s)
- Sapana Bansod
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA; Division of Endocrinology, Metabolism & Lipid Research, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Paarth B Dodhiawala
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA; Division of Endocrinology, Metabolism & Lipid Research, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Yutong Geng
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA; Division of Endocrinology, Metabolism & Lipid Research, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Ashenafi Bulle
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA; Division of Endocrinology, Metabolism & Lipid Research, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Peng Liu
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA; Division of Endocrinology, Metabolism & Lipid Research, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA; Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Lin Li
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA; Division of Endocrinology, Metabolism & Lipid Research, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Reid Townsend
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA
| | - Patrick M Grierson
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA; Division of Endocrinology, Metabolism & Lipid Research, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Jason M Held
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA; Division of Endocrinology, Metabolism & Lipid Research, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Hema Adhikari
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA
| | - Kian-Huat Lim
- Division of Oncology, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA; Division of Endocrinology, Metabolism & Lipid Research, Department of Internal Medicine, Barnes-Jewish Hospital and The Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
48
|
Peretz CAC, Kennedy VE, Walia A, Delley CL, Koh A, Tran E, Clark IC, Hayford CE, D'Amato C, Xue Y, Fontanez KM, May-Zhang AA, Smithers T, Agam Y, Wang Q, Dai HP, Roy R, Logan AC, Perl AE, Abate A, Olshen A, Smith CC. Multiomic single cell sequencing identifies stemlike nature of mixed phenotype acute leukemia. Nat Commun 2024; 15:8191. [PMID: 39294124 PMCID: PMC11411136 DOI: 10.1038/s41467-024-52317-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 08/30/2024] [Indexed: 09/20/2024] Open
Abstract
Despite recent work linking mixed phenotype acute leukemia (MPAL) to certain genetic lesions, specific driver mutations remain undefined for a significant proportion of patients and no genetic subtype is predictive of clinical outcomes. Moreover, therapeutic strategy for MPAL remains unclear, and prognosis is overall poor. We performed multiomic single cell profiling of 14 newly diagnosed adult MPAL patients to characterize the inter- and intra-tumoral transcriptional, immunophenotypic, and genetic landscapes of MPAL. We show that neither genetic profile nor transcriptome reliably correlate with specific MPAL immunophenotypes. Despite this, we find that MPAL blasts express a shared stem cell-like transcriptional profile indicative of high differentiation potential. Patients with the highest differentiation potential demonstrate inferior survival in our dataset. A gene set score, MPAL95, derived from genes highly enriched in the most stem-like MPAL cells, is applicable to bulk RNA sequencing data and is predictive of survival in an independent patient cohort, suggesting a potential strategy for clinical risk stratification.
Collapse
Affiliation(s)
- Cheryl A C Peretz
- Division of Hematology and Oncology, Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Vanessa E Kennedy
- Division of Hematology and Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Anushka Walia
- Division of Hematology and Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Cyrille L Delley
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Andrew Koh
- Division of Hematology and Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Elaine Tran
- Division of Hematology and Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Iain C Clark
- Department of Bioengineering, University of California Berkeley, Berkeley, CA, USA
| | | | | | - Yi Xue
- Fluent Biosciences Inc., Watertown, MA, USA
| | | | | | | | - Yigal Agam
- Fluent Biosciences Inc., Watertown, MA, USA
| | - Qian Wang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, People's Republic of China
| | - Hai-Ping Dai
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, People's Republic of China
| | - Ritu Roy
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Aaron C Logan
- Division of Hematology and Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Alexander E Perl
- Department of Medicine, Division of Hematology-Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Adam Abate
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Adam Olshen
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Catherine C Smith
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA.
- Division of Hematology and Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
49
|
Yao Y, Zhou S, Yan Y, Fu K, Xiao S. The tripartite motif-containing 24 is a multifunctional player in human cancer. Cell Biosci 2024; 14:103. [PMID: 39160596 PMCID: PMC11334367 DOI: 10.1186/s13578-024-01289-3] [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: 04/09/2024] [Accepted: 08/15/2024] [Indexed: 08/21/2024] Open
Abstract
Tripartite motif-containing 24 (TRIM24), also known as transcriptional intermediary factor 1α (TIF1α), is the founding member of TIF1 family. Recent evidence indicates that aberrant expression of TRIM24, functions as an oncogene, is associated with poor prognosis across various cancer types. TRIM24 exhibits a multifaceted structure comprising an N-terminal TRIM region with a RING domain, B-box type 1 and type 2 domains, and a coiled-coil region, as well as a C-terminal plant-homeodomain (PHD)-bromodomain. The bromodomain serves as a 'reader' of epigenetic histone marks, regulating chromatin structure and gene expression by linking associated proteins to acetylated nucleosomal targets, thereby controlling transcription of genes. Notably, bromodomains have emerged as compelling targets for cancer therapeutic development. In addition, TRIM24 plays specialized roles as a signal transduction molecule, orchestrating various cellular signaling cascades in cancer cells. Herein, we review the recent advancements in understanding the functions of TRIM24, and demonstrate the research progress in utilizing TRIM24 as a target for cancer therapy.
Collapse
Affiliation(s)
- Yuanbing Yao
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Department of General Surgery, Xiangya Hospital, Central South University, 87# Xiangya Road, Changsha, 410008, Hunan, China
| | - Sheng Zhou
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Department of General Surgery, Xiangya Hospital, Central South University, 87# Xiangya Road, Changsha, 410008, Hunan, China
- Department of Ultrasound, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yue Yan
- Yanbian University Medical School, Yanji, Jilin, China
| | - Kai Fu
- Institute of Molecular Precision Medicine and Hunan Key Laboratory of Molecular Precision Medicine, Department of General Surgery, Xiangya Hospital, Central South University, 87# Xiangya Road, Changsha, 410008, Hunan, China.
- Hunan Key Laboratory of Animal Models for Human Diseases, Central South University, 87# Xiangya Road, Changsha, 410008, Hunan, China.
- Center MOE Key Lab of Rare Pediatric Diseases & Hunan Key Laboratory of Medical Genetics of the School of Life Sciences, Central South University, 87# Xiangya Road, Changsha, 410008, Hunan, China.
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, 87# Xiangya Road, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, 87# Xiangya Road, Changsha, 410008, Hunan, China.
| | - Shuai Xiao
- The First Affiliated Hospital, Department of Gastrointestinal Surgery, Hengyang Medical School, University of South China, 69# Chuanshan Road, Hengyang, 421001, Hunan, China.
| |
Collapse
|
50
|
Letafati A, Taghiabadi Z, Ardekani OS, Abbasi S, Najafabadi AQ, Jazi NN, Soheili R, Rodrigo R, Yavarian J, Saso L. Unveiling the intersection: ferroptosis in influenza virus infection. Virol J 2024; 21:185. [PMID: 39135112 PMCID: PMC11321227 DOI: 10.1186/s12985-024-02462-3] [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: 05/30/2024] [Accepted: 08/06/2024] [Indexed: 08/15/2024] Open
Abstract
The influenza virus (IFV) imposes a considerable health and economic burden globally, requiring a comprehensive understanding of its pathogenic mechanisms. Ferroptosis, an iron-dependent lipid peroxidation cell death pathway, holds unique implications for the antioxidant defense system, with possible contributions to inflammation. This exploration focuses on the dynamic interplay between ferroptosis and the host defense against viruses, emphasizing the influence of IFV infections on the activation of the ferroptosis pathway. IFV causes different types of cell death, including apoptosis, necrosis, and ferroptosis. IFV-induced ferroptotic cell death is mediated by alterations in iron homeostasis, intensifying the accumulation of reactive oxygen species and promoting lipid peroxidation. A comprehensive investigation into the mechanism of ferroptosis in viral infections, specifically IFV, has great potential to identify therapeutic strategies. This understanding may pave the way for the development of drugs using ferroptosis inhibitors, presenting an effective approach to suppress viral infections.
Collapse
Affiliation(s)
- Arash Letafati
- Department of Virology, Faculty of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Taghiabadi
- Department of Microbiology and Virology of Medicine, Mashhad University of Medical Science, Mashhad, Iran
| | - Omid Salahi Ardekani
- Department of Bacteriology & Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Simin Abbasi
- Department of Virology, Faculty of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Qaraee Najafabadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Negar Nayerain Jazi
- Department of Bacteriology & Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Roben Soheili
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Science, Islamic Azad University, Tehran, Iran
| | - Ramón Rodrigo
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Jila Yavarian
- Department of Virology, Faculty of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University, Rome, Italy.
| |
Collapse
|