1
|
Xu JX, Su YX, Chen YY, Huang YY, Chen ZS, Peng YC, Qi LN. Immune infiltration landscape and potential drug-targeted implications for hepatocellular carcinoma with 'progression/hyper-progression' recurrence. Ann Med 2025; 57:2456113. [PMID: 39865865 PMCID: PMC11774162 DOI: 10.1080/07853890.2025.2456113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 12/20/2024] [Accepted: 01/08/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND AND AIMS Hepatocellular carcinoma (HCC) recurrence was previously characterized into four types, and patients with progression/hyper-progression recurrence (type III-IV) have an extremely poor prognosis. However, the immune background of resectable HCC, particularly in patients who experience recurrence, remains underexplored. Therefore, this study aimed to describe the immune landscape of resectable HCC, especially postoperative type III-IV recurrent HCC, and explore potential immune-targeted anti-relapse strategies for treated populations. METHODS The differences in gene expression in patients with recurrent HCC (type I-II (solitary or multi-intrahepatic oligo recurrence) vs. type III-IV) were investigated using bulk sequencing. Multiple immune infiltration methods (single-sample gene set enrichment analysis (GSEA), Microenvironment Cell Populations-counter and ESTIMATE) were used, and patients were divided into four groups to identify four distinct immune subtypes: immune-enrichment/matrix-poor (IE1), immune-enrichment/matrix-rich (IE2), immune intermediate/matrix-rich (ITM) and immune desert/matrix-poor (ID). Co-expression and protein interaction analyses were used to identify characteristic genes in ITM closely associated with type III-IV recurrence, which was matched with drug targets for Huaier granules (HG) and lenvatinib. Virtual docking was used to identify potential therapeutic targets, and the results were verified using single-nuclei RNA sequencing and histological analysis. RESULTS ITM was closely related to type III-IV recurrence and exhibited immunotherapy potential. The potential efficacy of inhibiting CCNA2, VEGFA, CXCL8, PLK2, TIMP1, ITGB2, ALDOA, ANXA5 and CSK in ITM reversal was determined. Molecular docking demonstrated that the proteins of these genes could bind to HG or lenvatinib. The immunohistochemical findings demonstrated differential VEGFA (p < .01) and PLK2 (p < .001) expression in ITM type and ID in type III-IV recurrent HCC. CONCLUSIONS Three primary immunotypes of resectable HCC (IE2, ITM and ID) were identified, and HG and lenvatinib could potentially overcome immune checkpoint blockade (ICB) resistance in ITM patients with HCC, particularly those classified as type III-IV.
Collapse
Affiliation(s)
- Jing-Xuan Xu
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumour, Ministry of Education, Nanning, China
| | - Yue-Xiang Su
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumour, Ministry of Education, Nanning, China
| | - Yuan-Yuan Chen
- Department of Ultrasound, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yi-Yue Huang
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumour, Ministry of Education, Nanning, China
| | - Zu-Shun Chen
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yu-Chong Peng
- Department of General Surgery, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Lu-Nan Qi
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumour, Ministry of Education, Nanning, China
- Guangxi Liver Cancer Diagnosis and Treatment Engineering and Technology Research Center, Nanning, China
| |
Collapse
|
2
|
Liu C, Li M, Liu L, Xu Q, Zheng L, Wu C, Ren J, Zhang T, Wang H, Lin Z. TGF-β1 induces autophagy and mediates the effect on macrophages differentiation in primary liver cancer. Int Immunopharmacol 2025; 157:114799. [PMID: 40339499 DOI: 10.1016/j.intimp.2025.114799] [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: 02/04/2025] [Revised: 05/01/2025] [Accepted: 05/01/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND Tumor-associated macrophages (TAMs) are closely associated with tumor development and patient outcomes due to their plasticity and polarization capacity. Several distinct TAMs have been proposed, but a complete understanding of heterogeneity and differentiation spectrum of macrophage in human primary liver cancer remains elusive. METHODS Deep single-cell RNA sequencing (scRNA-seq) data from 19 primary liver cancer patients were used to profile the transcriptomes of TAMs in liver cancer. Ingenuity pathway analysis (IPA) and in vitro experiments were used to explore possible mechanisms responsible for related signaling pathways altered at the transcriptional level. Finally, we analyzed the relationship between the abundance of the TAMs and the survival outcomes of the 428 patients in the Cancer Genome Atlas (TCGA). RESULTS Transcriptional profiles allowed us to identify four distinct TAMs cell subsets based on molecular and functional properties and to reconstruct their developmental trajectory. Specifically, TAM_c4 was preferentially enriched and potentially expanded in the advanced-stage patients or those receiving immune checkpoint blockade therapy (ICT). Gene pathway analysis revealed aberrant TGFB1 activation in TAM_c4, which was experimentally confirmed to drive TAM phenotypic transitions via autophagy signaling. High abundance of TAM_c4 is found to be related to a short survival time and low abundance of CD8+ T cells in primary liver cancers. CONCLUSIONS This integrated transcriptome compendium and experimental validation offer both mechanistic insights and a resource for understanding TAM heterogeneity in primary liver cancers.
Collapse
Affiliation(s)
- Chao Liu
- Yichang Central People's Hospital, The First College of Clinical Medical Science, China Three Gorges University, Yichang, People's Republic of China
| | - Mingjie Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Lichao Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Qian Xu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Linlin Zheng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Cailing Wu
- Faculty of Medicine, JiuJiang University, Jiujiang, People's Republic of China
| | - Jinghua Ren
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, People's Republic of China
| | - Tao Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| | - Haihong Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| | - Zhenyu Lin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| |
Collapse
|
3
|
Wu Y, Tao Q, Xie J, Liu X, Zhou Y, Wei C, Zhang C, Wang J, Jin Y. Indole-3-carbinol inhibits PD-L1-mediated immune evasion in hepatocellular carcinoma via suppressing NF-κB p105 Ubiquitination. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156692. [PMID: 40215823 DOI: 10.1016/j.phymed.2025.156692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/10/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one of the most common malignant tumors worldwide, and immunotherapy has demonstrated significant therapeutic benefit in HCC. Indole-3-carbinol (I3C), a naturally occurring ingredient of cruciferous vegetables, significantly inhibits the growth of a wide range of tumors. However, its mechanism of action has not been fully elucidated. PURPOSE This study aims to verify and explore the immunomodulatory effect of I3C in HCC models, and to investigate the specific role and mechanism by which I3C affects PD-L1 expression through the ubiquitination of NF-κB p105. METHODS In vitro, I3C was treated with HepG2 cells and relevant indicators were analyzed. In vivo, the mouse HCC model was established and the effect of I3C on tumors and immune function was evaluated. Subsequently, the downstream target of I3C was found through target prediction, molecular docking, and molecular dynamics simulation. Finally, combined therapy was used to further investigate the effect of I3C on mouse HCC tumors. RESULTS We observed that I3C resulted in decreased programmed cell death ligand 1 (PD-L1) expression in HepG2 cells and increased CD8 T cell infiltration in tissues. Subsequently, target prediction and molecular docking demonstrated that I3C was able to efficiently bind to NF-κB p105. In addition, overexpression of NF-κB p105 upregulated PD-L1 expression and almost completely eliminated the inhibitory effect of I3C. Notably, the combination of I3C and PD-L1 monoclonal antibodies showed synergistic anti-tumor effects in the mouse HCC model. CONCLUSION This study demonstrated that I3C inhibits PD-L1-mediated immune evasion in HCC via suppressing NF-κB p105 ubiquitination. The role of I3C in tumors deserves further investigation and provides the foundation for the future development of novel immunotherapeutic drugs.
Collapse
Affiliation(s)
- Yongkang Wu
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 230032, Hefei, PR China; Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of National Education, Anhui Medical University, 230032, Hefei, PR China
| | - Qing Tao
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 230032, Hefei, PR China; Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of National Education, Anhui Medical University, 230032, Hefei, PR China
| | - Jing Xie
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 230032, Hefei, PR China; Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of National Education, Anhui Medical University, 230032, Hefei, PR China
| | - Xiao Liu
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 230032, Hefei, PR China; Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of National Education, Anhui Medical University, 230032, Hefei, PR China
| | - Yuanzhi Zhou
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 230032, Hefei, PR China; Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of National Education, Anhui Medical University, 230032, Hefei, PR China
| | - Chengyan Wei
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 230032, Hefei, PR China; Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of National Education, Anhui Medical University, 230032, Hefei, PR China
| | - Chunwei Zhang
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 230032, Hefei, PR China; Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of National Education, Anhui Medical University, 230032, Hefei, PR China
| | - Jingjing Wang
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 230032, Hefei, PR China; Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of National Education, Anhui Medical University, 230032, Hefei, PR China
| | - Yong Jin
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 230032, Hefei, PR China; Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of National Education, Anhui Medical University, 230032, Hefei, PR China.
| |
Collapse
|
4
|
Hu XY, Sun YK, Miao Y, Chen XL, Lu D, Zhou BY, Wang LF, Zhao CK, Yin HH, Li XL, Chen ZT, Zhang YQ, Zhu MR, Guan X, Wu EX, Han H, Sun LP, Lu Q, Xu HX. Preoperative identification of hepatocellular carcinoma from focal liver lesions ≤ 20 mm in high-risk patients using clinical and contrast-enhanced ultrasound features. Eur J Radiol 2025; 187:112076. [PMID: 40187198 DOI: 10.1016/j.ejrad.2025.112076] [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: 11/30/2024] [Revised: 03/04/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025]
Abstract
OBJECTIVE We aimed to develop and validate a prediction model to identify HCC in focal liver lesions (FLLs) ≤20 mm among patients at risk for HCC based on clinical and contrast-enhanced ultrasound (CEUS) features. METHODS Between January 2022 and July 2023, 386 patients (mean age 58 ± 11 years; 277 male) at risk for HCC with FLLs ≤20 mm and clinical and preoperative CEUS data from three centers were retrospectively enrolled. Three prediction models based on clinical data (Cli-M), CEUS features (CEUS-M), and combined clinical and CEUS features (Com-M) were constructed using the training cohort (187 patients). Their predictive performance was evaluated using the area under the receiver operating characteristic curve (AUC), calibration curve, and decision curve analysis (DCA) in the internal and external validation cohorts. All patients were reclassified using the American College of Radiology CEUS Liver Imaging Reporting and Data System (CEUS LI-RADS) and combined with the best-performing model (modified LI-RADS). RESULTS The AUCs of Com-M were 0.873-0.951 in the training, internal, and external validation cohorts, which were higher than those of Cli-M (0.749-0.795, all P < 0.05) and CEUS-M (0.848-0.899, all P < 0.05). The sensitivity of LR-5 of modified LI-RADS was significantly improved from 83.1 % to 88.9 % (p<0.001) in the training, internal and external validation cohort while there was no statistical different on its specificity (82.6 %-94.7 % vs 95.7 %-97.6 %., p = 0.162-0.650). CONCLUSIONS The model based on clinical and CEUS features can help identify HCC in FLLs ≤ 20 mm in high-risk patients.
Collapse
Affiliation(s)
- Xin-Yuan Hu
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yi-Kang Sun
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yao Miao
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiao-Ling Chen
- Department of Ultrasound, Zhongshan Hospital (Xiamen Branch), Fudan University, Xiamen 361015, China
| | - Dan Lu
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Bo-Yang Zhou
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Li-Fan Wang
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Chong-Ke Zhao
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Hao-Hao Yin
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiao-Long Li
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zi-Tong Chen
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ya-Qin Zhang
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ming-Rui Zhu
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xin Guan
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Er-Xuan Wu
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Hong Han
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Li-Ping Sun
- Department of Medical Ultrasound, Shanghai Tenth People's Hospital, Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, School of Medicine, Tongji University, Shanghai, China
| | - Qing Lu
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Hui-Xiong Xu
- Department of Ultrasound, Institute of Ultrasound in Medicine and Engineering, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
5
|
Liu Q, Liang Z, Wang J, Wang Y, Wang J, Wang S, Du Z, Zhao L, Wei Y, Huang D. Mannose-modified multifunctional iron-based nanozyme for hepatocellular carcinoma treatment by remodeling the tumor microenvironment. Colloids Surf B Biointerfaces 2025; 250:114548. [PMID: 39923382 DOI: 10.1016/j.colsurfb.2025.114548] [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: 10/19/2024] [Revised: 01/12/2025] [Accepted: 01/31/2025] [Indexed: 02/11/2025]
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related deaths worldwide, with conventional treatments often accompanied by severe side effects. Recently, nanozymes have been extensively employed in cancer therapy due to their enhanced enzymatic activities, stability compared to native enzymes. However, a standalone nanozyme exhibits insufficient targeting capability and fails to specifically localize to the pathological site. In this study, we successfully synthesized a multifunctional iron-based-nanozyme delivery system - Fe3O4-OA-DHCA-PEI-MAN@DSF modified with PEI and MAN by the thermal decomposition method. This mannose-modified nanozyme can specifically target HCC cells via an external magnetic field and mannose-mannose receptor (MRC2) binding. In addition, it exhibits good biocompatibility and pH-dependent drug release characteristics. Within the acidic tumor microenvironment, the iron-based nanozyme initiates intracellular fenton reactions, boosting reactive oxygen species (ROS) production, which ultimately induces apoptosis in HCC cells. Concurrently, the disulfiram small molecule released from the Fe3O4-OA-DHCA-PEI-MAN@DSF nanozyme binds to the FROUNT factor within monocyte-macrophages, thereby inhibiting their response to chemotactic signals emitted by liver cancer cells. This process ultimately suppresses the recruitment of macrophages by HCC cells, reshaping the tumor microenvironment and supporting effective liver cancer treatment. Moreover, this nanozyme system holds potential for MRI-guided targeted chemotherapy combined with chemodynamic therapy, aiming to refine the early diagnosis and precision treatment of hepatic carcinoma, and paving the way for the creation of sophisticated integrated nanoplatforms melding diagnostic and therapeutic functionalities.
Collapse
Affiliation(s)
- Qi Liu
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Artificial Intelligence, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan 030024, China
| | - Ziwei Liang
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Artificial Intelligence, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan 030024, China; NHC Key Laboratory of Glycoconjuates Research Department of Biochemistry and Molecular, Biology School of Basic Medical Sciences, Fudan University, Shanghai 200032, China; Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan 030032, China; Shanxi Provincial Key Laboratory for Functional Proteins, Shanxi Jinbo Bio-Pharmaceutical Co., Ltd, Taiyuan 030032, China.
| | - Jiapu Wang
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Artificial Intelligence, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan 030024, China
| | - Yuhui Wang
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Artificial Intelligence, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan 030024, China
| | - Jie Wang
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Artificial Intelligence, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan 030024, China
| | - Shaojie Wang
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Artificial Intelligence, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan 030024, China
| | - Zhi Du
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Artificial Intelligence, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan 030024, China; Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan 030032, China
| | - Liqin Zhao
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Artificial Intelligence, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan 030024, China; Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan 030032, China
| | - Yan Wei
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Artificial Intelligence, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan 030024, China; Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan 030032, China; Shanxi Provincial Key Laboratory for Functional Proteins, Shanxi Jinbo Bio-Pharmaceutical Co., Ltd, Taiyuan 030032, China.
| | - Di Huang
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Artificial Intelligence, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan 030024, China; Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan 030032, China; Shanxi Provincial Key Laboratory for Functional Proteins, Shanxi Jinbo Bio-Pharmaceutical Co., Ltd, Taiyuan 030032, China.
| |
Collapse
|
6
|
Liu X, Hao J, Yuan Y, Liu F. Synergic chemotherapeutic effects of docetaxel and carboplatin show cytotoxic and apoptotic effects in liver cancer nursing care: Role of oxidative stress and hemocompatibility. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04295-5. [PMID: 40434421 DOI: 10.1007/s00210-025-04295-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025]
Abstract
Recent findings have demonstrated that Docetaxel (DTX) and Carboplatin (CRP) exhibit significant anticancer and antiproliferative properties in cancer cells. This study aims to explore the effects of DTX on enhancing CRP-induced apoptosis in liver cancer cells and the associated molecular pathways. DTX and CRP cell viability against SMMC-7721 and Bel7402 cells by MTT test. The IC50 values of CRP, DTX, and DTX + CRP for 35 μM, 4.69 μM, and 3.12 μM for SMMC-7721 cells, respectively. The outcomes of the respective fluorescence staining assays displayed that DTX + CRP remarkably enhanced the reactive oxygen species, diminished MMP, and triggered apoptosis in SMMC-7721 cells. DTX + CRP diminished the levels of GSH, CAT, and SOD while enhancing MDA contents in SMMC-7721 cell lines. In SMMC-7721 cells subjected to DTX + CRP, the Bax, Bcl-2, CyC, caspase-3, -8, and -9 expressions were fourfold increased, while Bcl-2 expression was threefold reduced. DTX + CRP enhanced the anticancer efficacy of human liver cancer cells by causing cellular oxidative stress. The hemocompatibility of DTX, CRP, and DTX + CRP was measured at different concentrations. Overall, the results indicate that these DTX + CRP possess potential biomedical uses due to their reduced hemotoxicity, cytotoxicity, and enhanced physiological milieu stability.
Collapse
Affiliation(s)
- Xiaozhen Liu
- Department of Interventional Therapy for Tumors and Vascular Diseases, Shanxi Norman Bethune Hospital, No. 99, Longcheng Street, Xiaodian District, Taiyuan, 030032, China
| | - Jiao Hao
- Department of Emergency, Shanxi Norman Bethune Hospital, Taiyuan, 030032, China
| | - Youyuan Yuan
- Department of Interventional Therapy for Tumors and Vascular Diseases, Shanxi Norman Bethune Hospital, No. 99, Longcheng Street, Xiaodian District, Taiyuan, 030032, China
| | - Fang Liu
- Department of Interventional Therapy for Tumors and Vascular Diseases, Shanxi Norman Bethune Hospital, No. 99, Longcheng Street, Xiaodian District, Taiyuan, 030032, China.
| |
Collapse
|
7
|
Zou Y, Wan X, Zhou Q, Zhu G, Lin S, Tang Q, Yang X, Wang S. Mechanisms of drug resistance in hepatocellular carcinoma. Biol Proced Online 2025; 27:19. [PMID: 40437363 PMCID: PMC12117952 DOI: 10.1186/s12575-025-00281-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 05/12/2025] [Indexed: 06/01/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is the most prevalent form of primary liver cancer, associated with high morbidity and mortality worldwide. Despite advancements in diagnostic methods and systemic treatments, including tyrosine kinase inhibitors (TKIs) and immune checkpoint inhibitors (ICIs), the development of drug resistance remains a significant challenge in HCC management. Traditional treatments such as surgical resection and transarterial chemoembolization offer limited efficacy, especially in advanced stages. Although novel therapies like lenvatinib, sorafenib, regorafenib, and ICIs have shown promise, their effectiveness is often hindered by primary and acquired resistance, leading to poor long-term survival outcomes. This review focuses on the molecular mechanisms underlying resistance to targeted therapies and immunotherapies in HCC. Key factors contributing to resistance include alterations in the tumor microenvironment (TME), immune evasion, hypoxia, changes in cellular metabolism, and genetic mutations. Additionally, molecular players such as ferroptosis, autophagy, apoptosis, endoplasmic reticulum stress, ABC transporters, and non-coding RNAs(ncRNAs) are discussed as contributors to drug resistance. Understanding these mechanisms is critical for the development of novel therapeutic strategies aimed at overcoming resistance, improving patient outcomes, and ultimately enhancing survival rates in HCC patients.
Collapse
Affiliation(s)
- Yongchun Zou
- Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Department of Oncology, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Chinese Medicine Guangdong Laboratory, State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Xinliang Wan
- Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Department of Oncology, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Chinese Medicine Guangdong Laboratory, State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Qichun Zhou
- Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Department of Oncology, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Chinese Medicine Guangdong Laboratory, State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Gangxing Zhu
- Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Department of Oncology, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Chinese Medicine Guangdong Laboratory, State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Shanshan Lin
- Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Department of Oncology, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Chinese Medicine Guangdong Laboratory, State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Qing Tang
- Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Department of Oncology, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Chinese Medicine Guangdong Laboratory, State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong, 510120, China.
| | - Xiaobing Yang
- Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Department of Oncology, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Chinese Medicine Guangdong Laboratory, State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong, 510120, China.
| | - Sumei Wang
- Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Department of Oncology, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Chinese Medicine Guangdong Laboratory, State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong, 510120, China.
| |
Collapse
|
8
|
Chen W, Li Y, Zhou Q, Peng W, Cao M, Zhao Y, Yang Z, Xiong S, Huang H, Liu L, Bai S, Cheng B. The cancer-associated fibroblast facilitates YAP liquid-liquid phase separation to promote cancer cell stemness in HCC. Cell Commun Signal 2025; 23:238. [PMID: 40413530 PMCID: PMC12103779 DOI: 10.1186/s12964-025-02256-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025] Open
Abstract
Cancer stem cells (CSCs) are strongly associated with the refractory characteristics of Hepatocellular carcinoma (HCC). However, the complex interaction between CSCs and the tumor microenvironment remains incompletely understood. In this study, we identified a novel long non-coding RNA (lncRNA) NEAT1 in cancer-associated fibroblast (CAFs)-derived extracellular vesicles (EVs) that play a critical role in the induction of CSCs and HCC tumorigenesis. NEAT1 was significantly overexpressed in human HCC tissues. Furthermore, high expression of lncRNA NEAT1 in EVs was found to be associated with poor prognosis. Knockdown of NEAT1 in CAFs inhibited invasion, migration, and tumor growth. Mechanistically, NEAT1 promoted cancer cell stemness, including 3D spheroid formation, by facilitating the liquid-liquid phase separation (LLPS) of the transcription factor YAP. Specifically, NEAT1 is directly bound to the intrinsic disordered region in the YAP protein, promoting the formation of LLPS biomolecular condensates. Additionally, a positive correlation between NEAT1 and Nanog was observed in clinical HCC tissues. In conclusion, our findings reveal that NEAT1 promotes HCC carcinogenesis and CSC induction by facilitating the LLPS of the YAP protein.
Collapse
Affiliation(s)
- Wei Chen
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road, No.1095, Wuhan, 430030, China
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, 999077, China
| | - Yanling Li
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road, No.1095, Wuhan, 430030, China
| | - Qiaodan Zhou
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road, No.1095, Wuhan, 430030, China
| | - Wang Peng
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road, No.1095, Wuhan, 430030, China
| | - Mengdie Cao
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road, No.1095, Wuhan, 430030, China
| | - Yuchong Zhao
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road, No.1095, Wuhan, 430030, China
| | - Zihan Yang
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan, 430065, China
| | - Si Xiong
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road, No.1095, Wuhan, 430030, China
| | - Hai Huang
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road, No.1095, Wuhan, 430030, China
| | - Luyao Liu
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road, No.1095, Wuhan, 430030, China
| | - Shuya Bai
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road, No.1095, Wuhan, 430030, China.
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road, No.1095, Wuhan, 430022, Hubei, China.
| | - Bin Cheng
- Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road, No.1095, Wuhan, 430030, China.
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road, No.1095, Wuhan, 430022, Hubei, China.
| |
Collapse
|
9
|
Zhao J, Li Y, Zhu J, Li H, Jin X. Ubiquitination in hepatocellular carcinoma immunity. J Transl Med 2025; 23:574. [PMID: 40410880 PMCID: PMC12102898 DOI: 10.1186/s12967-025-06592-2] [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: 02/10/2025] [Accepted: 05/08/2025] [Indexed: 05/25/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is the sixth most prevalent malignancy worldwide, and represents a major global health challenge. While surgical resection at early stages offers favorable prognosis with 5-year survival rates exceeding 70%, the clinical reality in China reveals a contrasting scenario, where over 60% of patients present with advanced disease, resulting in a dramatic decline in 5-year survival to below 12.5%. The immunological landscape plays a pivotal role in HCC pathogenesis and progression, comprising two complementary arms: the innate immune system's rapid-response mechanism for immediate tumor surveillance and the adaptive immune system's antigen-specific targeting with immunological memory capabilities. Emerging evidence has highlighted ubiquitination, a sophisticated post-translational modification system, as a critical regulator of immune homeostasis in HCC pathogenesis. This molecular process exerts precise control through three primary mechanisms: (1) Modulation of immune cell activation thresholds via proteasomal degradation of signaling proteins, (2) Orchestrating immune cell differentiation through stability regulation of transcriptional factors, and (3) Maintenance of immune tolerance by dynamic modification of checkpoint regulators. Such multifaceted regulation affects both innate immune recognition pathways (e.g., NF-κB and STING signaling) and adaptive immune effectors (particularly T cell receptor signaling cascades). This comprehensive review establishes a threefold Objective: First, to elucidate the mechanistic interplay between ubiquitination networks and HCC-related immune dysregulation; Second, to systematically analyze how innate immune-associated ubiquitination events drive hepatocarcinogenesis through chronic inflammation modulation; and third, to critically evaluate recent clinical advances combining ubiquitination-targeted therapies (e.g., proteasome inhibitors and E3 ligase modulators) with immunotherapeutic regimens. Our synthesis revealed that strategic manipulation of ubiquitination pathways can potentiate PD-1/PD-L1 blockade efficacy while mitigating therapeutic resistance, particularly through modulation of tumor-associated macrophages and exhausted T cell populations. By integrating fundamental mechanistic insights with translational clinical data, this review provides a conceptual framework for the development of next-generation diagnostic biomarkers and rational therapeutic combinations. The proposed strategy of ubiquitination-immune axis modulation holds significant potential to transform current HCC management paradigms, offering new avenues for precision immunotherapy for this challenging malignancy.
Collapse
Affiliation(s)
- Jianan Zhao
- Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, 315040, P. R. China
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, P. R. China
| | - Yuxuan Li
- Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, 315040, P. R. China
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, P. R. China
| | - Jie Zhu
- Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, 315040, P. R. China
| | - Hong Li
- Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, 315040, P. R. China.
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, P. R. China.
| | - Xiaofeng Jin
- Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, 315040, P. R. China.
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, P. R. China.
| |
Collapse
|
10
|
Huang W, Yang C, Cheng S, Fu S, Chen X, Zhu Y, Hu H, Gao F, He S. A DNA-Mediated Lysosomal Degradation Strategy for Targeted Degradation of PD-L1 Protein. J Med Chem 2025. [PMID: 40403183 DOI: 10.1021/acs.jmedchem.5c00675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2025]
Abstract
The expression of programmed cell death ligand 1 (PD-L1) enables tumor cells to evade immune surveillance by T-cells. The level of PD-L1 on the cell surface plays a crucial role in the effectiveness of PD-L1-targeted immune checkpoint blockade therapy. Therefore, we utilized the unique trafficking capabilities of scavenger receptors (SRs) to direct PD-L1 to lysosomes for degradation. By employing click chemistry to conjugate the PD-L1 inhibitor BMS-202 with dendritic DNA scaffolds, we created a bifunctional compound, PBL1, which is capable of simultaneously targeting both SRs and PD-L1. PBL1 effectively induces PD-L1 degradation both in vitro and in vivo, significantly reducing the off-target toxicity commonly associated with traditional PD-L1 inhibitors. The efficacy and specificity of PBL1 have been validated in A549 cells and zebrafish models. The development of this SRs-mediated lysosomal degradation strategy offers a promising new approach for cancer immunotherapy, providing a safer and more targeted alternative to existing PD-L1 inhibitors.
Collapse
Affiliation(s)
- Wenjing Huang
- School of Medicine or Institute of Translational Medicine, Shanghai Engineering Research Center of Organ Repair, Shanghai University, 99 Shangda Road, Shanghai 200444, P. R. China
| | - Can Yang
- School of Medicine or Institute of Translational Medicine, Shanghai Engineering Research Center of Organ Repair, Shanghai University, 99 Shangda Road, Shanghai 200444, P. R. China
| | - Sizhu Cheng
- School of Medicine or Institute of Translational Medicine, Shanghai Engineering Research Center of Organ Repair, Shanghai University, 99 Shangda Road, Shanghai 200444, P. R. China
| | - Shuyue Fu
- School of Medicine or Institute of Translational Medicine, Shanghai Engineering Research Center of Organ Repair, Shanghai University, 99 Shangda Road, Shanghai 200444, P. R. China
| | - Xinyu Chen
- School of Medicine or Institute of Translational Medicine, Shanghai Engineering Research Center of Organ Repair, Shanghai University, 99 Shangda Road, Shanghai 200444, P. R. China
| | - Yaojin Zhu
- School of Medicine or Institute of Translational Medicine, Shanghai Engineering Research Center of Organ Repair, Shanghai University, 99 Shangda Road, Shanghai 200444, P. R. China
| | - Honggang Hu
- School of Medicine or Institute of Translational Medicine, Shanghai Engineering Research Center of Organ Repair, Shanghai University, 99 Shangda Road, Shanghai 200444, P. R. China
| | - Fei Gao
- School of Medicine or Institute of Translational Medicine, Shanghai Engineering Research Center of Organ Repair, Shanghai University, 99 Shangda Road, Shanghai 200444, P. R. China
| | - Shipeng He
- School of Medicine or Institute of Translational Medicine, Shanghai Engineering Research Center of Organ Repair, Shanghai University, 99 Shangda Road, Shanghai 200444, P. R. China
| |
Collapse
|
11
|
Chen L, Guillot A, Tacke F. Reviewing the function of macrophages in liver disease. Expert Rev Gastroenterol Hepatol 2025:1-17. [PMID: 40387555 DOI: 10.1080/17474124.2025.2508963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 04/10/2025] [Accepted: 05/08/2025] [Indexed: 05/20/2025]
Abstract
INTRODUCTION The liver is a central metabolic organ, but is also hosting a unique immune microenvironment to sustain homeostasis and proper defense measures against injury threats in healthy individuals. Liver macrophages, mostly represented by the tissue-resident Kupffer cells and bone marrow- or monocyte-derived macrophages, are intricately involved in various aspects of liver homeostasis and disease, including tissue injury, inflammation, fibrogenesis and repair mechanisms. AREAS COVERED We review recent findings on defining the liver macrophage landscape and their functions in liver diseases with the aim of highlighting potential targets for therapeutic interventions. A comprehensive literature search in PubMed and Google Scholar was conducted to identify relevant literature up to date. EXPERT OPINION Liver macrophages orchestrate key homeostatic and pathogenic processes in the liver. Thus, targeting liver macrophages represents an attractive strategy for drug development, e.g. to ameliorate liver inflammation, steatohepatitis or fibrosis. However, translation from fundamental research to therapies remains challenging due to the versatile nature of the liver macrophage compartment. Recent and major technical advances such as single-cell and spatially-resolved omics approaches deepened our understanding of macrophage biology at a molecular level. Yet, further studies are needed to identify suitable, etiology- and stage-dependent strategies for the treatment of liver diseases.
Collapse
Affiliation(s)
- Lanlan Chen
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Adrien Guillot
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
12
|
Li J, Bai Y, Xiong F, Liu X, Hu J, Zhang G, Liu J, Wu S, Zheng C, Kan X. Atezolizumab Plus Bevacizumab Combined with or without Transarterial Chemoembolization in the Treatment of Advanced Hepatocellular Carcinoma: A Single-Center Retrospective Study. J Hepatocell Carcinoma 2025; 12:973-984. [PMID: 40395491 PMCID: PMC12090845 DOI: 10.2147/jhc.s515453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 05/09/2025] [Indexed: 05/22/2025] Open
Abstract
Purpose This study aimed to compare the efficacy and safety of atezolizumab plus bevacizumab (T+A) in combination with transarterial chemoembolization (TACE) (T+A+TACE) and T+A for patients with advanced hepatocellular carcinoma (HCC). Patients and Methods From December 2020 to August 2024, 83 patients with advanced HCC who received T+A+TACE treatment or T+A treatment in our hospital were included, and these patients were categorized into TACE+T+A group (n=52) and T+A group (n=31). The clinical outcomes between the two groups were analyzed and compared, and the prognostic factors that affected the efficacy were analyzed. Results The median overall survival (OS) and median progression-free survival (PFS) in the T+A+TACE group were significantly longer than those of in the T+A group (OS: 22.8 vs 16.9 months, P = 0.015; PFS: 7.1 vs 4.9 months, P = 0.006). A significantly higher objective response rate (ORR) and disease control rate (DCR) that are based on the modified RECIST were achieved in the T+A+TACE group than those of in the T+A group (ORR: 51.9% vs 6.5%, P < 0.001; DCR: 88.5% vs 54.8%, P < 0.001). No significant differences in adverse events (AEs) were observed between the two groups (P > 0.05). The T+A+TACE treatment was identified as a protective factor for OS and PFS. Conclusion TACE further improved the efficacy of T+A treatment for patients with advanced HCC, and it did not increase the incidence of AEs. T+A+TACE treatment is a promising treatment option for patients with advanced HCC.
Collapse
Affiliation(s)
- Jing Li
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, 430022, People’s Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, People’s Republic of China
| | - Yaowei Bai
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, 430022, People’s Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, People’s Republic of China
| | - Fu Xiong
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, 430022, People’s Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, People’s Republic of China
| | - Xiaocui Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, 430022, People’s Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, People’s Republic of China
| | - Junwen Hu
- Department of Oncology, The Third People’s Hospital of Yibin, Sichuan, 644000, People’s Republic of China
| | - Guilin Zhang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, 430022, People’s Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, People’s Republic of China
| | - Jiayun Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, 430022, People’s Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, People’s Republic of China
| | - Suyue Wu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, 430022, People’s Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, People’s Republic of China
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, 430022, People’s Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, People’s Republic of China
| | - Xuefeng Kan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, 430022, People’s Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, People’s Republic of China
| |
Collapse
|
13
|
Majeed NS, Mohammed MH, Hatem ZA, El-Sehrawy AAMA, Ganesan S, Singh A, Akoul MA, Sudan P, Singh R, Hamad HA. Interplay between NETosis and the lncRNA-microRNA regulatory axis in the immunopathogenesis of cancer. J Physiol Biochem 2025:10.1007/s13105-025-01082-x. [PMID: 40358898 DOI: 10.1007/s13105-025-01082-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 04/14/2025] [Indexed: 05/15/2025]
Abstract
Neutrophil extracellular traps (NETs), web-like complex structures secreted by neutrophils, have emerged as key players in the modulation of immune responses and the immunopathogenesis of immune disorders. Initially described for their antimicrobial function, NETs now play a part in the fundamental processes of cancer biology, including cancer initiation, metastatic dissemination, and immune evasion strategies. NETs hijack anti-tumor immunity by entrapping circulating cancer cells, fostering the growth of tumors, and reorganizing the tumor microenvironment such that it is pro-malignancy. Emerging evidence emphasizes the role of NETosis coupled with non-coding RNAs-long non-coding RNAs (lncRNAs) and microRNAs (miRNAs)-as key regulators of gene expression and controllers of processes vital for cancer growth, such as immune response and programmed cell death processes like apoptosis, necroptosis, pyroptosis, and ferroptosis. Aberrantly expressed non-coding RNAs have been attributed to immune dysregulation and excessive NET production, promoting tumor growth. NETs are also associated with a myriad of pathological conditions, such as autoimmune disorders, cystic fibrosis, sepsis, and thrombotic disorders. New therapeutic approaches-such as DNase therapy and PAD4 inhibitors-target NET production and their degradation to modify immune function and the efficiency of immunotherapies. Further clarification of the intricate interactions of NETosis, lncRNAs, and miRNAs has the potential to establish new strategies for the suppression of the growth of tumors and preventing immune evasion. This review seeks to elucidate the interactions between NETosis and the regulatory networks involving non-coding RNAs that significantly contribute to the immunopathogenesis of cancer.
Collapse
Affiliation(s)
| | - Mohammed Hashim Mohammed
- Medical Laboratory Techniques department, College of Health and medical technology, Al-Maarif University, Anbar, Iraq.
| | - Zainab Amer Hatem
- College of Science, Biotechnology Department, Diyala University, Diyala, Iraq
| | | | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Abhayveer Singh
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, 140401, India
| | - Marwa Azeez Akoul
- Biotechnology Department, College of Applied Science, Fallujah University, Anbar, Iraq
| | - Puneet Sudan
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, 140307, India
| | - Roshni Singh
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Hamad Ali Hamad
- Department of Pathological Analysis, Collage of Applied Sciences, University of Fallujah, Fallujah, Iraq
| |
Collapse
|
14
|
Yan L, Lv J, Xu M, Jia H, Li S. High Midkine Expression Correlates with Poor Prognosis and Immune Cell Infiltration in Hepatocellular Carcinoma. Int J Gen Med 2025; 18:2567-2579. [PMID: 40386763 PMCID: PMC12085142 DOI: 10.2147/ijgm.s490409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 03/26/2025] [Indexed: 05/20/2025] Open
Abstract
Objective This study investigated the role of MDK (Midkine) in hepatocellular carcinoma (HCC) through bioinformatics analysis and experimental validation, focusing on its relationship with tumor immune microenvironment and patient prognosis. Methods We employed the GEPIA database to analyze MDK expression patterns across cancer types and specifically in HCC versus normal tissues. MDK expression was validated through immunohistochemistry (IHC) in 100 paired HCC and adjacent tissue samples. Survival analyses were conducted using Kaplan-Meier and Cox regression methods. The relationship between MDK expression and immune cell infiltration was investigated using TIMER 2.0 database and verified through IHC staining of immune cell markers. Results MDK expression was significantly elevated in HCC tissues compared to adjacent normal tissues. High MDK expression strongly correlated with tumor number, vascular invasion, advanced clinical stage and poor prognosis, serving as an independent prognostic factor. Notably, elevated MDK expression predicted poor outcomes in patients receiving immunotherapy. Database analysis and IHC analysis revealed that MDK expression positively correlated with regulatory T (Treg) cell infiltration while negatively correlating with natural killer (NK) cell presence, suggesting its role in shaping the tumor immune microenvironment. Conclusion High MDK expression in HCC correlates with unfavorable patient outcomes and impacts immune cell infiltration. MDK may serve as a novel prognostic biomarker and potential therapeutic target in HCC treatment.
Collapse
Affiliation(s)
- Lili Yan
- Department of Gastroenterology, First Hospital of Qinhuangdao, Qinhuangdao, 066005, People’s Republic of China
| | - Ji Lv
- Department of Breast Surgery, First Hospital of Qinhuangdao, Qinhuangdao, 066005, People’s Republic of China
| | - Meimei Xu
- Department of Gastroenterology, First Hospital of Qinhuangdao, Qinhuangdao, 066005, People’s Republic of China
| | - Hongyu Jia
- Department of Gastroenterology, First Hospital of Qinhuangdao, Qinhuangdao, 066005, People’s Republic of China
| | - Shanshan Li
- Department of Gastroenterology, First Hospital of Qinhuangdao, Qinhuangdao, 066005, People’s Republic of China
| |
Collapse
|
15
|
Tu J, Liu X, Li K, Liu H, Li J, Zhu J, Xia N, Wang Q. A novel polysaccharide from Citrus aurantium L.: Structural properties and antitumor activities in vitro and invivo. JOURNAL OF ETHNOPHARMACOLOGY 2025; 347:119725. [PMID: 40216044 DOI: 10.1016/j.jep.2025.119725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 03/21/2025] [Accepted: 03/30/2025] [Indexed: 04/15/2025]
Abstract
A novel cold-water-soluble polysaccharide (CALP-1-1) was isolated and purified from Citrus aurantium L. Besides determining its in vitro and in vivo anti-tumor activities, its structure was characterised. The results reveal that CALP-1-1 mainly consists of Rha, Ara, Gal, GalA, and GlcA (molar ratio, 1:14.56:19.27:2.27:1.29) with three main linkages. Its average molecular weight was 2.04 × 103 kDa. Moreover, the triple helix structure of CALP-1-1 was proved by Congo-red and circular dichroism (CD). The in vitro experimental results demonstrate that CALP-1-1 significantly inhibited the proliferation of HepG2 cells with typical apoptotic features by inducing cell cycle arrest in the S phase. Furthermore, in vivo anti-tumor experiments suggest that CALP-1-1 could induce H22 solid tumor cells apoptosis and exhibit anti-tumor effects by protecting immune organs and intensifying the secretion of immune cells (macrophages, lymphocytes and NK cells). In conclusion, CALP-1-1 might be a promising component for cancer treatment.
Collapse
Affiliation(s)
- Jianqiu Tu
- Xinyang Agriculture and Forestry University, Research Center for Comprehensive Utilization of Food Resources of Ta-pieh Mountains, Comprehensive Utilization and Development Key Laboratory in Characteristic Food Resources, Xinyang, Xinyang City, Henan Province, 464000, PR China
| | - Xiaoyuan Liu
- Xinyang Agriculture and Forestry University, Research Center for Comprehensive Utilization of Food Resources of Ta-pieh Mountains, Comprehensive Utilization and Development Key Laboratory in Characteristic Food Resources, Xinyang, Xinyang City, Henan Province, 464000, PR China
| | - Kun Li
- Xinyang Agriculture and Forestry University, Research Center for Comprehensive Utilization of Food Resources of Ta-pieh Mountains, Comprehensive Utilization and Development Key Laboratory in Characteristic Food Resources, Xinyang, Xinyang City, Henan Province, 464000, PR China.
| | - Huiping Liu
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Food Nutrition and Safety, Ministry of Education of China, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, PR China.
| | - Jianfang Li
- Xinyang Agriculture and Forestry University, Research Center for Comprehensive Utilization of Food Resources of Ta-pieh Mountains, Comprehensive Utilization and Development Key Laboratory in Characteristic Food Resources, Xinyang, Xinyang City, Henan Province, 464000, PR China
| | - Jing Zhu
- Xinyang Agriculture and Forestry University, Research Center for Comprehensive Utilization of Food Resources of Ta-pieh Mountains, Comprehensive Utilization and Development Key Laboratory in Characteristic Food Resources, Xinyang, Xinyang City, Henan Province, 464000, PR China
| | - Nan Xia
- Xinyang Agriculture and Forestry University, Research Center for Comprehensive Utilization of Food Resources of Ta-pieh Mountains, Comprehensive Utilization and Development Key Laboratory in Characteristic Food Resources, Xinyang, Xinyang City, Henan Province, 464000, PR China
| | - Qing Wang
- Xinyang Agriculture and Forestry University, Research Center for Comprehensive Utilization of Food Resources of Ta-pieh Mountains, Comprehensive Utilization and Development Key Laboratory in Characteristic Food Resources, Xinyang, Xinyang City, Henan Province, 464000, PR China
| |
Collapse
|
16
|
Su R, Du Y, Tian P, Ma W, Hui Y, Yang S. Single-cell and spatial transcriptomics reveal correlation between RNA methylation-related miRNA risk model and immune infiltration in hepatocellular carcinoma. Front Oncol 2025; 15:1553239. [PMID: 40416872 PMCID: PMC12098086 DOI: 10.3389/fonc.2025.1553239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 04/09/2025] [Indexed: 05/27/2025] Open
Abstract
Introduction Increasing evidence highlights the pivotal role of RNA methylation and miRNAs in hepatocellular carcinoma (HCC). However, the risk associated with RNA methylation-related miRNAs (RMRMs) in the HCC immune microenvironment remains largely unknown. Here, we predicted the correlation between RMRM risk and immune cell infiltration in HCC using machine learning. Methods MiRNA sequencing data was used to identify RMRMs. A risk score model of HCC was developed utilizing four RMRMs, including miR-551a, miR-4739, miR-326, and miR-210-3p. Results Patients with high-risk scores exhibited poorer prognoses. Single-cell RNA sequencing (scRNA-seq) analysis revealed the high-risk group exhibited increased infiltration levels of several immune cell subtypes, including myeloid-derived suppressor cell (MDSC), macrophage, and T cells. The data integration of scRNA-seq and bulk RNA-seq showed the decreased TIDE score in the high-risk patients and the elevated levels of Macro-secreted phosphoprotein 1 (SPP1), MDSC-meiotic nuclear divisions 1 (MND1), γδ T cells, and Macro-complement C1q C chain (C1QC) predicted adverse prognosis. ScRNA-seq and spatial transcriptomics data integration unveiled the spatial distribution of RMRMs risk scores and their correlation with immune cell subtype localization. Risk model-based clustering of HCC samples revealed that cluster 2, characterized by a higher risk score, correlated with a poorer prognosis and reduced immune and stromal scores. In vitro, the overexpression of miR-4739 in Huh-7 cells significantly induced SPP1+ macrophages, and the culture medium derived from SPP1+ macrophages further promoted the proliferation and migration of Huh-7 cells. Furthermore, miR-4739 reduced m1A methylation by inhibiting tRNA methyltransferase 61A (TRMT61A) expression. Discussion Our study reveals that the RMRM risk model could effectively predict the prognosis of HCC, and SPP1+ macrophages regulated by miR-4739-RNA methylation promote the proliferation and migration of HCC cells. These results highlight the potential of RMRMs in predicting the prognosis of HCC.
Collapse
Affiliation(s)
- Rong Su
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yong Du
- Department of Anesthesiology, People’s Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, China
| | - Pan Tian
- Department of Neurology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Weifang Ma
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yongfeng Hui
- Department of Hepatobiliary Surgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Shaoqi Yang
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| |
Collapse
|
17
|
Lei W, Zhou K, Lei Y, Li Q, Zhu H. Pathogenesis and Systemic Treatment of Hepatocellular Carcinoma: Current Status and Prospects. Mol Cancer Ther 2025; 24:692-708. [PMID: 39417575 DOI: 10.1158/1535-7163.mct-24-0403] [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: 05/30/2024] [Revised: 08/14/2024] [Accepted: 10/08/2024] [Indexed: 10/19/2024]
Abstract
Hepatocellular carcinoma (HCC) remains one of the major threats to human health worldwide. The emergence of systemic therapeutic options has greatly improved the prognosis of patients with HCC, particularly those with advanced stages of the disease. In this review, we discussed the pathogenesis of HCC, genetic alterations associated with the development of HCC, and alterations in the tumor immune microenvironment. Then, important indicators and emerging technologies related to the diagnosis of HCC are summarized. Also, we reviewed the major advances in treatments for HCC, offering insights into future prospects for next-generation managements.
Collapse
Affiliation(s)
- Wanting Lei
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Kexun Zhou
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ye Lei
- College of Liberal Arts, Neijiang Normal University, Neijiang, China
| | - Qiu Li
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Zhu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Tong J, Tan Y, Ouyang W, Chang H. Targeting immune checkpoints in hepatocellular carcinoma therapy: toward combination strategies with curative potential. Exp Hematol Oncol 2025; 14:65. [PMID: 40317077 PMCID: PMC12046748 DOI: 10.1186/s40164-025-00636-5] [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: 01/20/2025] [Accepted: 03/07/2025] [Indexed: 05/04/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a primary liver cancer characterized by poor immune cell infiltration and a strongly immunosuppressive microenvironment. Traditional treatments have often yielded unsatisfactory outcomes due to the insidious onset of the disease. Encouragingly, the introduction of immune checkpoint inhibitors (ICIs) has significantly transformed the approach to HCC treatment. Moreover, combining ICIs with other therapies or novel materials is considered the most promising opportunity in HCC, with some of these combinations already being evaluated in large-scale clinical trials. Unfortunately, most clinical trials fail to meet their endpoints, and the few successful ones also face challenges. This indicates that the potential of ICIs in HCC treatment remains underutilized, prompting a reevaluation of this promising therapy. Therefore, this article provides a review of the role of immune checkpoints in cancer treatment, the research progress of ICIs and their combination application in the treatment of HCC, aiming to open up avenues for the development of safer and more efficient immune checkpoint-related strategies for HCC treatment.
Collapse
Affiliation(s)
- Jing Tong
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510631, China
| | - Yongci Tan
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510631, China
| | - Wenwen Ouyang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510631, China
| | - Haocai Chang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510631, China.
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, School of Optoelectronic Science and Engineering, South China Normal University, Guangzhou, 510631, China.
| |
Collapse
|
19
|
Guo X, Wu L, Lai J, Wu Y, Chen D. Causal Associations Between Lipids, NPC1L1, and Liver Cancer Risk: Insights From Mendelian Randomization and Bioinformatics. J Gastroenterol Hepatol 2025. [PMID: 40312834 DOI: 10.1111/jgh.16897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/12/2025] [Accepted: 01/20/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND AND AIM The study aims to investigate the potential causal effects of lipids on liver cancer risk and to analyze the possible impact of lipid-lowering drug targets on liver cancer. METHODS Genetic variants linked to lipid traits and drug targets were obtained from the Global Lipids Genetics Consortium and DrugBank. Liver cancer data were sourced from FinnGen. Mendelian randomization (MR) was used to assess causal relationships between lipid traits and liver cancer. Functional analyses included protein-protein interaction (PPI), KEGG pathway enrichment, transcription factor (TF) network analysis, and survival analysis. NPC1L1 expression, DNA methylation, and immune infiltration were analyzed using UALCAN, TCGA-LIHC, and TIMER, respectively. RESULTS MR analysis showed higher genetically predicted LDL-C levels reduced liver cancer risk (OR = 0.5981, p = 0.034). Drug target MR indicated that NPC1L1 inhibition (OR = 1.0638, p = 0.0311) and elevated PPARɑ levels (OR = 1.1339, p < 0.01) increased liver cancer risk. Functional analysis revealed NPC1L1 was highly expressed in liver cancer tissues due to hypomethylation and linked to immune cell infiltration, indicating its role in immune evasion and tumor progression. CONCLUSION The study demonstrates that elevated LDL-C levels are associated with a reduced risk of liver cancer and NPC1L1 plays a key role in regulating lipid metabolism and influencing immune evasion.
Collapse
Affiliation(s)
- Xiaoyan Guo
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Lili Wu
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jing Lai
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuankai Wu
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Dianke Chen
- Department of Medical Oncology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
20
|
Takehara Y, Waki Y, Morine Y, Saito Y, Teraoku H, Wada Y, Yamada S, Ikemoto T, Shimada M. Vascular endothelial growth factor released from lenvatinib-resistant hepatocellular carcinoma promotes malignant behavior and drug resistance through tumor-associated macrophages. Hepatol Res 2025; 55:752-762. [PMID: 40317612 DOI: 10.1111/hepr.14173] [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: 07/09/2024] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 05/07/2025]
Abstract
AIM Lenvatinib is one of the molecularly targeted drugs used worldwide in hepatocellular carcinoma (HCC) treatment, but the acquisition of drug resistance is a major problem. The mechanisms that make HCC lenvatinib-resistant (LR) in the tumor microenvironment are largely unknown. Here, we examined the impact of LR HCC cells on tumor-associated macrophages (TAMs) regarding tumor progression and drug resistance, focusing on vascular endothelial growth factor (VEGF) secretion by LR HCC cells. METHODS Hepatocellular carcinoma cells were cultured with lenvatinib to obtain cells with approximately 4-fold lenvatinib resistance. Monocyte-derived macrophages were cultured in the conditioned medium (CM) of LR HCC to induce LR TAMs. Secretion of VEGF by naïve HCC cells, LR HCC cells, TAMs, and LR TAMs was assessed. The macrophage phenotype and VEGFR2 expression in both TAMs were evaluated. Additionally, the effects of inhibiting VEGF secretion in LR HCC cells on the TAM malignant potential were investigated. Tumor-associated macrophages induced by VEGF-inhibited LR HCC were defined as modified LR TAMs. RESULTS Secretion of VEGF was elevated in LR HCC. Lenvatinib-resistant TAMs had increased expression of M2-like macrophage markers and increased expression of VEGFR2 compared with naïve HCC-derived TAMs. Lenvatinib-resistant TAM-CM promoted malignant behaviors and lenvatinib resistance in naïve HCC cells and LR HCC cells. However, the modified LR TAMs did not increase M2 polarization markers or decreased VEGFR2 expression. The elevated VEGF secretion in LR TAMs was not seen in modified LR TAMs. CONCLUSIONS Increased VEGF secretion from LR HCC promoted malignant behaviors and lenvatinib resistance through LR TAMs in the tumor microenvironment.
Collapse
Affiliation(s)
- Yukako Takehara
- Department of Digestive and Transplant Surgery, Tokushima University, Tokushima, Japan
| | - Yuhei Waki
- Department of Digestive and Transplant Surgery, Tokushima University, Tokushima, Japan
| | - Yuji Morine
- Department of Digestive and Transplant Surgery, Tokushima University, Tokushima, Japan
| | - Yu Saito
- Department of Digestive and Transplant Surgery, Tokushima University, Tokushima, Japan
| | - Hiroki Teraoku
- Department of Digestive and Transplant Surgery, Tokushima University, Tokushima, Japan
| | - Yuma Wada
- Department of Digestive and Transplant Surgery, Tokushima University, Tokushima, Japan
| | - Shinichiro Yamada
- Department of Digestive and Transplant Surgery, Tokushima University, Tokushima, Japan
| | - Tetsuya Ikemoto
- Department of Digestive and Transplant Surgery, Tokushima University, Tokushima, Japan
| | - Mitsuo Shimada
- Department of Digestive and Transplant Surgery, Tokushima University, Tokushima, Japan
| |
Collapse
|
21
|
Lai G, Xie B, Zhang C, Zhong X, Deng J, Li K, Liu H, Zhang Y, Liu A, Liu Y, Fan J, Zhou T, Wang W, Huang A. Comprehensive analysis of immune subtype characterization on identification of potential cells and drugs to predict response to immune checkpoint inhibitors for hepatocellular carcinoma. Genes Dis 2025; 12:101471. [PMID: 40092490 PMCID: PMC11907441 DOI: 10.1016/j.gendis.2024.101471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 04/12/2024] [Accepted: 11/02/2024] [Indexed: 03/19/2025] Open
Abstract
Immunosubtyping enables the segregation of immune responders from non-responders. However, numerous studies failed to focus on the integration of cellular heterogeneity and immunophenotyping in the prediction of hepatocellular carcinoma (HCC) patients' response to immune checkpoint inhibitors (ICIs). We categorized HCC patients into various immune subtypes based on feature scores linked to ICI response. Single-cell sequencing technology was to investigate the cellular heterogeneity of different immune subtypes and acquire significant ICI response-associated cells. Candidate drugs were identified using a blend of various drug databases and network approaches. HCC patients were divided into two distinct immune subtypes based on characterization scores of 151 immune-related gene sets. Patients in both subtypes showed varying overall survival, immunity levels, biological activities, and TP53 mutation rates. Subtype 1-related natural killer cells showed a positive correlation with immune-promoting scores but a negative correlation with immune-suppressing scores. Notably, docetaxel sensitivity in HCC patients rose as the levels of subtype 1-related natural killer cells increased. Our study demonstrated that immune subtypes have cellular heterogeneity in predicting response to ICIs. A combination of subtype 1-associated natural killer cells and docetaxel may offer new hope for ICI treatment in HCC.
Collapse
Affiliation(s)
- Guichuan Lai
- Department of Health Statistics, School of Public Health, Chongqing Medical University, Chongqing 401331, China
| | - Biao Xie
- Department of Health Statistics, School of Public Health, Chongqing Medical University, Chongqing 401331, China
| | - Cong Zhang
- Department of Health Statistics, School of Public Health, Chongqing Medical University, Chongqing 401331, China
| | - Xiaoni Zhong
- Department of Health Statistics, School of Public Health, Chongqing Medical University, Chongqing 401331, China
| | - Jielian Deng
- Department of Health Statistics, School of Public Health, Chongqing Medical University, Chongqing 401331, China
| | - Kangjie Li
- Department of Health Statistics, School of Public Health, Chongqing Medical University, Chongqing 401331, China
| | - Hui Liu
- Department of Health Statistics, School of Public Health, Chongqing Medical University, Chongqing 401331, China
| | - Yuan Zhang
- Department of Health Statistics, School of Public Health, Chongqing Medical University, Chongqing 401331, China
| | - Anbin Liu
- Department of Applied Statistics, School of Public Health, Chongqing Medical University, Chongqing 401331, China
| | - Yi Liu
- Department of Applied Statistics, School of Public Health, Chongqing Medical University, Chongqing 401331, China
| | - Jie Fan
- Department of Epidemiology, School of Public Health, Chongqing Medical University, Chongqing 401331, China
| | - Tianyi Zhou
- Department of Health Statistics, School of Public Health, Chongqing Medical University, Chongqing 401331, China
| | - Wei Wang
- Department of Applied Statistics, School of Public Health, Chongqing Medical University, Chongqing 401331, China
| | - Ailong Huang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
22
|
Chen H, Xiao Z, Lu Z, Xu N, Wei Q, Xu X. Targeted activation of junctional adhesion molecule-like protein + CD8 + T cells enhances immunotherapy in hepatocellular carcinoma. Chin J Cancer Res 2025; 37:212-226. [PMID: 40353078 PMCID: PMC12062980 DOI: 10.21147/j.issn.1000-9604.2025.02.08] [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: 09/11/2024] [Accepted: 03/24/2025] [Indexed: 05/14/2025] Open
Abstract
Objective Cytotoxic T lymphocytes (CTLs) play a crucial role in the therapeutic approach to hepatocellular carcinoma (HCC). Recent research has indicated that junctional adhesion molecule-like protein (JAML) enhances the antitumor activity of CD8+ T cells. Our study investigates the role of JAML+ CD8+ T cells in HCC. Methods We utilized time-of-flight mass cytometry and an orthotopic mouse model of HCC to examine histone modifications in tumor-infiltrating immune cells undergoing immunotherapy. Flow cytometry was used to assess CD4+ T cells differentiation and JAML expression in CD8+ T cells infiltrating HCC. Correlation analysis revealed a strong positive correlation between lactate dehydrogenase A+ (LDHA+) CD4+ T cells and JAML+ CD8+ T cells. Subsequently, we evaluated the therapeutic effects of an agonistic anti-JAML antibody, both alone and combined with immunotherapy. Finally, RNA sequencing was conducted to identify potential regulatory mechanisms. Results Immunotherapy significantly increased the percentage of CD8+ T cells infiltrating HCC and induced histone modifications, such as H3K18 lactylation (H3K18la) in CD4+ T cells. Flow cytometry analysis revealed that lactate promotes the differentiation of CD4+ T cells into Th1 cells. LDHA, an enzyme that converts pyruvate to lactate, plays a key role in this process. Correlation analysis revealed a strong positive relationship between LDHA+ CD4+ T cells and JAML+ CD8+ T cells in patients who responded to immunotherapy. Moreover, high JAML expression in CD8+ T cells was associated with a more favorable prognosis. In vivo experiments demonstrated that agonistic anti-JAML antibody therapy reduced tumor volume and significantly prolonged the survival of tumor-bearing mice, independent of the effects of anti-programmed cell death protein ligand-1 antibody (αPD-L1)-mediated immunotherapy. Pathway enrichment analysis further revealed that JAML enhances CTL responses through the oxidative phosphorylation pathway. Conclusions Activation of JAML enhances CTL responses in HCC treatment, independent of αPD-L1-mediated immunotherapy, providing a promising strategy for advanced HCC.
Collapse
Affiliation(s)
- Huan Chen
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Zhaofeng Xiao
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Zhengyang Lu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Nan Xu
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Qiang Wei
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou 310053, China
| | - Xiao Xu
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou 310053, China
| |
Collapse
|
23
|
Gao B, Lu Y, Lai X, Xu X, Gou S, Yang Z, Gong Y, Yang H. Metabolic reprogramming in hepatocellular carcinoma: mechanisms of immune evasion and therapeutic implications. Front Immunol 2025; 16:1592837. [PMID: 40370433 PMCID: PMC12075234 DOI: 10.3389/fimmu.2025.1592837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Accepted: 04/07/2025] [Indexed: 05/16/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related deaths worldwide, with limited treatment options for advanced stages. Metabolic reprogramming is a hallmark of cancer, enabling tumor cells to adapt to the harsh tumor microenvironment (TME) and evade immune surveillance. This review involves the role of metabolic reprogramming in HCC, focusing on the dysregulation of glucose, lipid, and amino acid metabolism, and its impact on immune evasion. Key metabolic pathways, such as the Warburg effect, fatty acid synthesis, and glutaminolysis, are discussed, along with their influence on tumor-associated macrophages (TAMs) and immune cell function. Targeting these metabolic alterations presents a promising therapeutic approach to enhance immunotherapy efficacy and improve HCC patient outcomes.
Collapse
Affiliation(s)
- Bocheng Gao
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yan Lu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xingyue Lai
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xi Xu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shuhua Gou
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhida Yang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yanju Gong
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hong Yang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
24
|
Liang Z, Li S, Wang Z, Zhou J, Huang Z, Li J, Bao H, Yam JWP, Xu Y. Unraveling the Role of the Wnt Pathway in Hepatocellular Carcinoma: From Molecular Mechanisms to Therapeutic Implications. J Clin Transl Hepatol 2025; 13:315-326. [PMID: 40206274 PMCID: PMC11976435 DOI: 10.14218/jcth.2024.00401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/13/2024] [Accepted: 12/23/2024] [Indexed: 04/11/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the deadliest malignant tumors in the world, and its incidence and mortality have increased year by year. HCC research has increasingly focused on understanding its pathogenesis and developing treatments.The Wnt signaling pathway, a complex and evolutionarily conserved signal transduction system, has been extensively studied in the genesis and treatment of several malignant tumors. Recent investigations suggest that the pathogenesis of HCC may be significantly influenced by dysregulated Wnt/β-catenin signaling. This article aimed to examine the pathway that controls Wnt signaling in HCC and its mechanisms. In addition, we highlighted the role of this pathway in HCC etiology and targeted treatment.
Collapse
Affiliation(s)
- Zixin Liang
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Shanshan Li
- School of Pharmacy, Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu Medical University, Bengbu, Anhui, China
| | - Zhiyu Wang
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Junting Zhou
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Ziyue Huang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Jiehan Li
- Department of Urology, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Haolin Bao
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Judy Wai Ping Yam
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yi Xu
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- School of Pharmacy, Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu Medical University, Bengbu, Anhui, China
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fuzhou, Fujian, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, China
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| |
Collapse
|
25
|
Zhao S, Chen F, Hu L, Li X, Gao Z, Chen M, Wang X, Song Z. Long non-coding rnas as key modulators of the immune microenvironment in hepatocellular carcinoma: implications for Immunotherapy. Front Immunol 2025; 16:1523190. [PMID: 40352941 PMCID: PMC12061944 DOI: 10.3389/fimmu.2025.1523190] [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: 11/05/2024] [Accepted: 04/02/2025] [Indexed: 05/14/2025] Open
Abstract
Hepatocellular carcinoma (HCC) represents a major global health challenge, characterized by its complex immune microenvironment that plays a pivotal role in tumor progression and therapeutic response. Long non-coding RNAs (lncRNAs) have emerged as critical regulators of various biological processes, including gene expression and immune cell function. This review explores the multifaceted roles of lncRNAs in modulating the immune microenvironment of HCC. We discuss how lncRNAs influence the infiltration and activation of immune cells, shape cytokine profiles, and regulate immune checkpoint molecules, thereby affecting the tumor's immunogenicity and response to immunotherapy. Furthermore, we highlight specific lncRNAs implicated in immune evasion mechanisms and their potential as biomarkers and therapeutic targets. By elucidating the intricate interplay between lncRNAs and the immune landscape in HCC, this review aims to provide insights into novel strategies for enhancing immunotherapeutic efficacy and improving patient outcomes.
Collapse
Affiliation(s)
| | | | | | | | | | - Minjie Chen
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Xiaoguang Wang
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Zhengwei Song
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| |
Collapse
|
26
|
Wang J, Liu ZX, Huang ZH, Wen J, Rao ZZ. Long non-coding RNA in the regulation of cell death in hepatocellular carcinoma. World J Clin Oncol 2025; 16:104061. [PMID: 40290684 PMCID: PMC12019274 DOI: 10.5306/wjco.v16.i4.104061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/02/2025] [Accepted: 02/25/2025] [Indexed: 03/26/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is the predominant form of primary liver cancer, accounting for 90% of all cases. Currently, early diagnosis of HCC can be achieved through serum alpha-fetoprotein detection, B-ultrasound, and computed tomography scanning; however, their specificity and sensitivity are suboptimal. Despite significant advancements in HCC biomarker detection, the prognosis for patients with HCC remains unfavorable due to tumor heterogeneity and limited understanding of its pathogenesis. Therefore, it is crucial to explore more sensitive HCC biomarkers for improved diagnosis, monitoring, and management of the disease. Long non-coding RNA (lncRNA) serves as an auxiliary carrier of genetic information and also plays diverse intricate regulatory roles that greatly contribute to genome complexity. Moreover, investigating gene expression regulation networks from the perspective of lncRNA may provide insights into the diagnosis and prognosis of HCC. We searched the PubMed database for literature, comprehensively classified regulated cell death mechanisms and systematically reviewed research progress on lncRNA-mediated cell death pathways in HCC cells. Furthermore, we prospectively summarize its potential implications in diagnosing and treating HCC.
Collapse
Affiliation(s)
- Jiang Wang
- Children Medical Center, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410013, Hunan Province, China
| | - Zi-Xuan Liu
- Children Medical Center, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410013, Hunan Province, China
| | - Zhi-Hong Huang
- Children Medical Center, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410013, Hunan Province, China
| | - Jie Wen
- Department of Pediatric Orthopedics, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410013, Hunan Province, China
| | - Zhou-Zhou Rao
- Department of Physiology, Hunan Normal University School of Medicine, Changsha 410003, Hunan Province, China
| |
Collapse
|
27
|
Lu R, Abuduhailili X, Li Y, Wang S, Xia X, Feng Y. Integrated Analysis of PSMB8 Expression and Its Potential Roles in Hepatocellular Carcinoma. Dig Dis Sci 2025:10.1007/s10620-025-09040-9. [PMID: 40261568 DOI: 10.1007/s10620-025-09040-9] [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: 03/07/2025] [Accepted: 04/02/2025] [Indexed: 04/24/2025]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) represents a highly aggressive malignancy with significant global health implications. The proteasome subunit beta type-8 (PSMB8) gene, known for its association with hepatitis B virus susceptibility, has emerged as a potential regulator of tumor progression. However, its functional role and clinical significance in HCC remain poorly characterized. METHODS We conducted a comprehensive multi-omics analysis to elucidate the role of PSMB8 in HCC. PSMB8 expression profiles were derived from The Cancer Genome Atlas and validated using the GSE76427 dataset. Prognostic significance was assessed through Kaplan-Meier survival analysis. Then, we systematically evaluated the relationships between PSMB8 expression and clinicopathological features, somatic mutations, immune cell infiltration, immune regulatory genes, and immune checkpoint responses. Single-cell RNA sequencing data from the Tumor Immune Single-cell Hub database were analyzed to determine cell type-specific PSMB8 expression. Tissue-level validation was performed using multiplex immunofluorescence staining on HCC tissue microarrays. RESULTS PSMB8 demonstrated significant overexpression in HCC tissues and exhibited strong prognostic value. Single-cell analysis revealed predominant PSMB8 expression in T and B cell populations. Notably, PSMB8 expression showed significant positive correlations with immune checkpoint molecules PD-L1/CD274 and CD27. Functional enrichment analysis implicated PSMB8 in multiple oncogenic pathways, particularly proteasome-related processes. CONCLUSION Our findings position PSMB8 as a promising prognostic biomarker and potential therapeutic target in HCC. The observed associations with immune checkpoint molecules and proteasomal pathways suggest its potential role in modulating tumor immunity and protein homeostasis, warranting further investigation into its mechanistic contributions to HCC progression.
Collapse
Affiliation(s)
- Ruijiao Lu
- Department of Medical Laboratory Center, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, China
| | - Xieyidai Abuduhailili
- Department of Medical Laboratory Center, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, China
| | - Yuxia Li
- Department of Medical Laboratory Center, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, China
| | - Senyu Wang
- Good Clinical Research Practice, The First Huizhou Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Xigang Xia
- Department of Hepatobiliary Pancreatic Surgery, The First Huizhou Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Yangchun Feng
- Department of Medical Laboratory Center, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, China.
- Department of Medical Laboratory Center, The First Huizhou Affiliated Hospital of Guangdong Medical University, Guangdong, China.
| |
Collapse
|
28
|
Wang TC, Huang W, Li SX, Li JX, Zhou P, Wang LZ, Wei N, Cai WW, Hu JJ, Xiao YD. High Fibroblast Activation Protein Expression in Hepatocellular Carcinoma: CT Imaging Features and Histological Characteristics. Acad Radiol 2025:S1076-6332(25)00315-0. [PMID: 40254479 DOI: 10.1016/j.acra.2025.04.016] [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: 03/04/2025] [Revised: 04/06/2025] [Accepted: 04/06/2025] [Indexed: 04/22/2025]
Abstract
RATIONALE AND OBJECTIVES To clarify computed tomography (CT) imaging features of high fibroblast activation protein (FAP) expression in hepatocellular carcinoma (HCC) and to investigate their correlation to histological characteristics and prognosis. METHODS This retrospective multicenter study evaluated patients with HCC who underwent liver resection between August 2013 and June 2023. Histological staining for FAP was performed, and patients were classified into low and high-FAP expression groups. A predictive model was developed and validated to identify FAP expression levels among CT imaging features. Moreover, CT imaging-related histological characteristics were evaluated. With the predictive model, patients in training cohort were stratified into predicted low and high-FAP expression groups. Overall survival (OS) and recurrence-free survival (RFS) were compared accordingly. Besides, propensity score matching (PSM) and inverse probability of treatment weighting (IPTW) analysis were performed, and OS and RFS between the two groups were also compared. RESULTS In total, 1197 patients were included (high-FAP HCCs, n=267). Delayed central enhancement (OR: 15.196, 95% CI: 8.996-25.670, P<0.001) and dilated vasculature (OR: 7.455, 95% CI: 4.928-11.277, P<0.001) independently predicted high-FAP expression (AUCs: 0.779, training; 0.766, external validation). Based on these two CT imaging features, intratumoral fibrosis (ITF) and vessel-encapsulating tumor cluster (VETC) pattern were determined. Compared to low-FAP HCCs, high-FAP HCCs were more frequently with high ITF grade (54.7% vs. 22.4%; P<0.001) and VETC pattern (52.1% vs. 22.6%; P<0.001). Patients were divided into predicted low (n=564) and high (n=262) FAP expression groups. The predicted low-FAP group had significantly better or favorable trend toward improved OS and RFS than predicted high-FAP group before (P<0.001, OS and RFS), and after PSM (P=0.009, OS; P=0.005, RFS) and IPTW (P=0.019, OS; P=0.077, RFS). CONCLUSION Delayed central enhancement and dilated vasculature are independent factors of high-FAP expression in HCC. Noninvasive identifying FAP expression may offer potential prognostic and therapeutic insights.
Collapse
Affiliation(s)
- Tian-Cheng Wang
- Department of Radiology, the Second Xiangya Hospital of Central South University, No.139 Middle Renmin Road, Changsha 410011, China (T.C.W., W.H., S.X.L., J.J.H., Y.D.X.)
| | - Wei Huang
- Department of Radiology, the Second Xiangya Hospital of Central South University, No.139 Middle Renmin Road, Changsha 410011, China (T.C.W., W.H., S.X.L., J.J.H., Y.D.X.)
| | - Shu-Xian Li
- Department of Radiology, the Second Xiangya Hospital of Central South University, No.139 Middle Renmin Road, Changsha 410011, China (T.C.W., W.H., S.X.L., J.J.H., Y.D.X.)
| | - Jun-Xiang Li
- Department of Interventional Radiology, Guizhou Medical University Affiliated Cancer Hospital, Guiyang 550004, China (J.X.L.)
| | - Peng Zhou
- Department of Pathology, the Second Xiangya Hospital of Central South University, Changsha 410011, China (P.Z.)
| | - Li-Zhou Wang
- Department of Interventional Radiology, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China (L.Z.W.)
| | - Nan Wei
- Department of Diagnostic and Interventional Radiology, University Hospital Heidelberg, Heidelberg 69120, Germany (N.W.)
| | - Wen-Wu Cai
- Department of Liver Surgery, the Second Xiangya Hospital of Central South University, Changsha 410011, China (W.W.C.)
| | - Jun-Jiao Hu
- Department of Radiology, the Second Xiangya Hospital of Central South University, No.139 Middle Renmin Road, Changsha 410011, China (T.C.W., W.H., S.X.L., J.J.H., Y.D.X.)
| | - Yu-Dong Xiao
- Department of Radiology, the Second Xiangya Hospital of Central South University, No.139 Middle Renmin Road, Changsha 410011, China (T.C.W., W.H., S.X.L., J.J.H., Y.D.X.).
| |
Collapse
|
29
|
Tang K, Liu M, Zhang C. Construction of a prognostic model and identification of key genes in liver hepatocellular carcinoma based on multi-omics data. Sci Rep 2025; 15:13393. [PMID: 40251374 PMCID: PMC12008308 DOI: 10.1038/s41598-025-98038-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 04/09/2025] [Indexed: 04/20/2025] Open
Abstract
Liver hepatocellular carcinoma (LIHC) strongly contributes to global cancer mortality, highlighting the need for a deeper understanding of its molecular mechanisms to enhance patient prognosis and treatment approaches. We aimed to investigate the differential expression of immunogenic cell death-related genes (ICDRGs) and cellular senescence-related genes (CSRGs) in LIHC and their effects on patient prognosis. We combined the GSE25097, GSE46408, and GSE121248 datasets by eliminating batch effects and standardizing the data. After processing, 16 genes were identified as ICDR&CSR differentially expressed genes (ICDR&CSRDEGs), including UBE2T, HJURP, PTTG1, CENPA, and FOXM1. Gene set enrichment analysis indicated a strong enrichment of these genes in pre-Notch expression and processing. Gene set variation analysis revealed 20 pathways with significant differences between the LIHC and control groups. Mutation analysis identified TP53 as the most commonly mutated gene in LIHC samples. A prognostic risk model integrating 12 ICDR&CSRDEGs was developed, showing high precision at 1 year but diminished accuracy at 2 and 3 years. Our constructed prognostic risk model provides valuable insights for predicting patient outcomes and may guide future therapeutic interventions targeting these specific genes. Further research is needed to explore the mechanistic roles of these genes in LIHC progression and treatment response.
Collapse
Affiliation(s)
- Kun Tang
- Department of Hepatobiliary Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong Province, China
| | - Mingjiang Liu
- Department of Hepatobiliary Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong Province, China
| | - Cuisheng Zhang
- Department of Hepatobiliary Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong Province, China.
| |
Collapse
|
30
|
Su P, Han Y, Yi J, Hou Y, Xiao Y. Research status and frontiers in liver cancer immunotherapy: a bibliometric perspective on highly cited literature. Front Oncol 2025; 15:1587252. [PMID: 40276056 PMCID: PMC12018336 DOI: 10.3389/fonc.2025.1587252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 03/14/2025] [Indexed: 04/26/2025] Open
Abstract
Background Liver cancer is one of the major causes of cancer-related death in the world. As a breakthrough therapy, immunotherapy had significantly improved the prognosis of patients. However, the current research status and research hotspots in the field of liver cancer immunotherapy still lack systematic review. Based on the bibliometric analysis of highly cited papers, this study intended to reveal the current research status, research hotspots and future research trends in this field. Objective The purpose of this study was to analyze the national/regional contributions, authors and institutions cooperation network, keywords clustering and keywords burst analysis of highly cited papers on liver cancer immunotherapy through bibliometrics, so as to clarify the research frontier and development direction, and provide objective data support for future research direction and clinical practice. Methods The highly cited papers on liver cancer immunotherapy from the Web of Science core collection up to February 23, 2025 were retrieved, and 232 studies were included. CiteSpace was used to build a knowledge map, analyze the distribution of years, countries, authors, institutions and cooperation networks, and identify research hotspots and emerging trends through keyword clustering and burst detection. Results The number of highly cited papers continued to increase from 2014 and reached a peak in 2022. China and the United States had the highest number of publications and the centrality of cooperation networks. The author with the highest number of papers was Llovet, Josep M, whose research direction mainly focused on immune checkpoint inhibitor combination therapy and molecular typing. The author with the highest cooperation network centrality was Duda, Dan G, whose research team focused on tumor microenvironment regulation. Harvard University and the University of Barcelona played an important central role in the institutional collaboration. Keywords analysis showed that immune checkpoint inhibitors, tumor microenvironment and combination therapy were the core of liver cancer immunotherapy. Burst keywords such as cell lung cancer, pembrolizumab, advanced melanoma, blockade, lymphocytes, etc. had revealed the research frontier of liver cancer immunotherapy research. Conclusion The research on liver cancer immunotherapy had made multi-dimensional progress, with China and the United States leading the global cooperation. The main research directions were the combination strategy of immunization, the regulation of tumor microenvironment and the exploration of novel targets. In the future, it is necessary to optimize treatment resistance solutions, integrate interdisciplinary resources, and promote the development of precision and personalized treatment.
Collapse
Affiliation(s)
- Pan Su
- Teaching and Research Section of Clinical Nursing, Xiangya Hospital, Central South University, Changsha, China
| | - Yeqiong Han
- Teaching and Research Section of Clinical Nursing, Xiangya Hospital, Central South University, Changsha, China
| | - Jindong Yi
- Teaching and Research Section of Clinical Nursing, Xiangya Hospital, Central South University, Changsha, China
| | - Yu Hou
- Department of Pulmonology, Children’s Hospital, National Clinical Research Center For Child Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Yao Xiao
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
- International Joint Research Center of Minimally Invasive Endoscopic Technology Equipment & Standards, Xiangya Hospital, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
31
|
Liu T, Cui Y, Ouyang Y, Wang M, Yue S. Exosomal CCT3 as a biomarker for diagnosis and immune therapy response in patients diagnosed with hepatocellular carcinoma. Dig Liver Dis 2025:S1590-8658(25)00301-9. [PMID: 40221386 DOI: 10.1016/j.dld.2025.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 11/19/2024] [Accepted: 03/21/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the dominant type of liver cancer and is associated with a high mortality rate. However, HCC lacks biomarkers for diagnosis and immune therapy response. Tumor-derived exosomes (TDEs) carcinogen-specific molecules have been used for screening multiple biomarkers. This study aimed to identify new biomarkers for the diagnosis of HCC and response to immune checkpoint blockade (ICB) therapy. METHODS Analysis of differentially expressed genes (DEGs) in HCC and normal tissues was integrated using The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO), and ExoCarta datasets. The expression of CCT3 was validated in samples from patients with HCC using quantitative polymerase chain reaction (qPCR), Western blotting, and immunohistochemistry (IHC) techniques. RESULTS Exosomal CCT3 was identified as a potential biomarker with significant impact. The expression of CCT3 in different tumor stages and normal tissues adjacent to the tumors (NATs) was validated using qPCR, western blotting, and IHC. CCT3 expression significantly increased the number of activated natural killer cells in HCC, as confirmed by qPCR and IHC. CCT3 expression significantly increases the expression of immune checkpoints in HCC. HCC-derived exosomes significantly increase the enrichment of CCT3. CONCLUSION Exosomal CCT3 is a biomarker for diagnosis and ICB therapy of HCC via MYC pathway activation and immune infiltration.
Collapse
Affiliation(s)
- Tiange Liu
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China; Nankai University Affiliated Eye Hospital, Nankai University, Tianjin, China; Tianjin Eye Hospital, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, China
| | - Yanyan Cui
- The Affiliated Hospital of Chifeng University, Chifeng, Inner Mongolia, China
| | - Yiben Ouyang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Meilin Wang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China
| | - Shijing Yue
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 300071, China.
| |
Collapse
|
32
|
Liu GM, Guo R, Xu JW. A bibliometric and visual analysis based on immune checkpoint inhibitors for hepatocellular carcinoma: 2014 - 2024. Front Pharmacol 2025; 16:1520055. [PMID: 40260385 PMCID: PMC12009821 DOI: 10.3389/fphar.2025.1520055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 03/24/2025] [Indexed: 04/23/2025] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) have changed the treatment landscape of hepatocellular carcinoma (HCC), especially those with unresectable advanced stages. The field has progressed rapidly, and the research hotspots have significantly changed compared to previous years. The study aims to comprehensively review and analyze the development history, knowledge structure, current research focus, and emerging trends in ICIs for HCC. Materials and methods Reviews and articles published in English from The Web of Science Core Collection (WoSCC) database from 2014 to 2024 were systemically retrieved. Citespace, VOSviewer, and Bibliometrix R package were used for further bibliometric analysis and visualization for countries, institutions, authors, references, and keywords. Results 2,941 records were included for analysis. The literature on ICIs for HCC has continued to grow steadily over the past decade. Three major research centers have emerged: North America, Europe, and East Asia. The Chinese institution has the highest publication volume, but Kudo Masatoshi from Japan has the highest number of publications. At the same time, Richard S. Finn from the United States leads in citations and co-citations. The most prolific journal is "Cancers". The clustering and Timeline view of critical literature and keywords indicated that research on ICIs for HCC is rapidly advancing toward a more evidence-based, personalized, and multimodal approach. Immune evasion mechanisms, predictive biomarkers, and high-quality clinical trials focusing on Novel combination, conversion, and perioperative therapies, including ICIs, are emerging hotspots. Conclusion This study highlights the groundbreaking advancements of ICIs in treating HCC and shows a trend rapidly advancing towards a more evidence-based, personalized, and multimodal approach. The study updated the current understanding of ICIs in hepatocellular carcinoma and identified vital future directions for research, such as the exploration of mechanisms of immune evasion, developing predictive biomarkers, and combining therapy strategies.
Collapse
Affiliation(s)
- Gao-Min Liu
- Meizhou Clinical Medical College of Shantou University Medical College, Meizhou, China
- Department of Hepatobiliary Surgery, Meizhou People’s Hospital, Meizhou, China
| | - Rui Guo
- Department of Hepatobiliary Surgery, Meizhou People’s Hospital, Meizhou, China
| | - Ji-Wei Xu
- Meizhou Clinical Medical College of Shantou University Medical College, Meizhou, China
- Department of Hepatobiliary Surgery, Meizhou People’s Hospital, Meizhou, China
| |
Collapse
|
33
|
Li M, Wang Y, Li X, Xu J, Yan L, Tang S, Liu C, Shi M, Liu R, Zhao Y, Zhang Y, Yang L, Zhang Y, Wang G, Li Z, Guo Y, Feng Y, Liu P. Pharmacological targeting of the mitochondrial phosphatase PTPMT1 sensitizes hepatocellular carcinoma to ferroptosis. Cell Death Dis 2025; 16:257. [PMID: 40189563 PMCID: PMC11973169 DOI: 10.1038/s41419-025-07581-5] [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: 07/31/2024] [Revised: 03/07/2025] [Accepted: 03/21/2025] [Indexed: 04/09/2025]
Abstract
Protein tyrosine phosphatase mitochondrial 1 (PTPMT1), is a member of the protein tyrosine phosphatase superfamily localized on the mitochondrial inner membrane, and regulates the biosynthesis of cardiolipin. Given the important position of PTPMT1 in mitochondrial function and metabolism, pharmacological targeting of PTPMT1 is considered a promising manner in disease treatments. In this study, we mainly investigated the role of PTPMT1 in hepatocellular carcinoma (HCC) ferroptosis, a new type of cell death accompanied by significant iron accumulation and lipid peroxidation. Herein, the pharmacological inhibition of PTPMT1 was induced by alexidine dihydrochloride (AD, a dibiguanide compound). Human HCC cell lines with PTPMT1 knockout and PTPMT1 overexpression were established using CRISPR/Cas9 and lentiviral transduction methods, respectively. The position of PTPMT1 in regulating HCC ferroptosis was evaluated in vitro and in vivo. Our results indicated that pharmacological inhibition of PTPMT1, facilitated by AD treatment, heightens the susceptibility of HCC to cystine deprivation-ferroptosis, and AD treatment promoted the conversion from ferritin-bound Fe3+ to free Fe2+, which contributed to the labile iron pool in cytoplasm. Meanwhile, pharmacological inhibition of PTPMT1 also induced the formation of both swollen mitochondria and donut mitochondria, and enhanced the metabolism process form succinate to fumarate in mitochondrial tricarboxylic acid (TCA) cycle, which increased the sensitivity of HCC cells to cystine deprivation-induced ferroptosis. In total, our work reveals the close association of PTPMT1 with cysteine deprivation-induced ferroptosis, providing a novel insight into chemotherapy strategies against human HCC.
Collapse
Affiliation(s)
- Miaomiao Li
- Department of Critical Care Medicine, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Regenerative Medicine, School of Pharmaceutical Science, Jilin University, Changchun, China
| | - Yi Wang
- Department of Regenerative Medicine, School of Pharmaceutical Science, Jilin University, Changchun, China
| | - Xinyan Li
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jiayi Xu
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Liangwen Yan
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shenkang Tang
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Oncology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Chenyue Liu
- Department of Medical Image, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Mengjiao Shi
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Rongrong Liu
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yaping Zhao
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yi Zhang
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lan Yang
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yinggang Zhang
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Gang Wang
- Department of Critical Care Medicine, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Key Laboratory of Surgical Critical Care and Life Support, Xi'an Jiaotong University, Ministry of Education of China, Xi'an, China
| | - Zongfang Li
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of General Surgery, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ying Guo
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Yetong Feng
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Core Research Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Pengfei Liu
- International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Shaanxi Provincial Clinical Research Center for Hepatic & Splenic Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Key Laboratory of Environment and Genes Related To Diseases, Xi'an Jiaotong University, Ministry of Education of China, Xi'an, China.
| |
Collapse
|
34
|
Wang Y, Li Y, Lin Y, Cao C, Chen D, Huang X, Li C, Xu H, Lai H, Chen H, Zhou Y. Roles of the gut microbiota in hepatocellular carcinoma: from the gut dysbiosis to the intratumoral microbiota. Cell Death Discov 2025; 11:140. [PMID: 40185720 PMCID: PMC11971373 DOI: 10.1038/s41420-025-02413-z] [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: 08/08/2024] [Revised: 02/23/2025] [Accepted: 03/18/2025] [Indexed: 04/07/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is closely linked to alterations in the gut microbiota. This dysbiosis is characterized by significant changes in the microbial population, which correlate with the progression of HCC. Gut dysbiosis ultimately promotes HCC development in several ways: it damages the integrity of the gut-vascular barrier (GVB), alters the tumor microenvironment (TME), and even affects the intratumoral microbiota. Subsequently, intratumoral microbiota present a characteristic profile and play an essential role in HCC progression mainly by causing DNA damage, mediating tumor-related signaling pathways, altering the TME, promoting HCC metastasis, or through other mechanisms. Both gut microbiota and intratumoral microbiota have dual effects on HCC progression; a comprehensive understanding of their complex biological roles will provide a theoretical foundation for potential clinical applications in HCC treatment.
Collapse
Affiliation(s)
- Yiqin Wang
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yongqiang Li
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yong Lin
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Chuangyu Cao
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Dongcheng Chen
- Department of Gastroenterology and Hepatology, Baiyun Hospital of Guangzhou First People's Hospital (The Second People's Hospital of Baiyun District), Guangzhou, China
| | - Xianguang Huang
- Department of Gastroenterology and Hepatology, Baiyun Hospital of Guangzhou First People's Hospital (The Second People's Hospital of Baiyun District), Guangzhou, China
| | - Canhua Li
- Department of Gastroenterology and Hepatology, Baiyun Hospital of Guangzhou First People's Hospital (The Second People's Hospital of Baiyun District), Guangzhou, China
| | - Haoming Xu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Huasheng Lai
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Huiting Chen
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China.
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China.
| | - Yongjian Zhou
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China.
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China.
| |
Collapse
|
35
|
Zhu R, Zhao S, Cao J, Liu Y, Liang R. Comprehensive analysis of GPN1 in human cancer and its effects on the migration of hepatocellular carcinoma cells. BIOMOLECULES & BIOMEDICINE 2025; 25:1111-1125. [PMID: 39524004 PMCID: PMC11984376 DOI: 10.17305/bb.2024.11310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/04/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
To investigate the prognostic value of GPN1 in cancer and its role in the migration of hepatocellular carcinoma (HCC or LIHC) cells, we used several databases to assess GPN1 expression levels and effects in human tumors. Furthermore, experiments were conducted to verify changes in GPN1 expression in HCC cell lines and explore its biological function. We found that GPN1 gene and protein expression were significantly increased in several tumor tissues. Higher GPN1 expression was associated with unfavorable overall survival. Additionally, there was a strong association between GPN1 expression and several clinicopathological features, according to multivariate Cox regression analysis. Moreover, GPN1 gene mutation and methylation were present in some tumors. A relationship was also found between GPN1 expression and immune infiltration. Notably, immune checkpoint analysis showed that GPN1 expression was correlated with PD-1/PDL-1 and CTLA-4, suggesting it may serve as a biomarker for predicting immune subtypes and response to immunotherapy in HCC. Enrichment analysis in HCC indicated that GPN1 is primarily involved in RNA metabolism. Additionally, drug sensitivity analysis revealed that GPN1 appeared to be responsive to 16 drugs. Finally, GPN1 upregulation was confirmed to promote the migration of HCC cells. This study provides a comprehensive overview of GPN1 in human cancer and demonstrates that GPN1 contributes to the migration of HCC cells, potentially serving as a prognostic and immunotherapy biomarker.
Collapse
Affiliation(s)
- Rongtao Zhu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Senfeng Zhao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiahui Cao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yin Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruopeng Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
36
|
Sun P, Xu H, Guo C, Yang L, Zhang X, Lu B, Chen L, Huang J. TMEM115 as an Oncogenic and Immunological Biomarker in Hepatocellular Carcinoma. Liver Int 2025; 45:e70048. [PMID: 40052693 DOI: 10.1111/liv.70048] [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: 08/15/2024] [Revised: 02/11/2025] [Accepted: 02/18/2025] [Indexed: 05/13/2025]
Abstract
BACKGROUND Transmembrane (TMEM) proteins are involved in fundamental biological processes such as material transport and signal transduction. TMEM115 is a member of the TMEM protein family, but its significance in hepatocellular carcinoma (HCC) remains unclear. In this study, we investigate the clinical predictive significance and potential functions of TMEM115 in HCC. METHODS Bioinformatics was used to investigate TMEM115 mRNA expression and immune infiltration score. Through multiplex immunohistochemistry analysis, we assessed its protein expression and association with HCC patient clinical features, prognosis and immune cell infiltration in HCC. Through in vitro and in vivo experiments, we evaluated the biological functions of TMEM115 in HCC cells and its impact on the immune microenvironment. RESULTS TMEM115 mRNA and protein levels were significantly higher in HCC tissues compared to paracancerous liver tissues. Its protein expression correlated with clinical characteristics and overall survival in HCC patients. In HCC tissues, higher TMEM115 protein expression corresponded to lower proportions of CD66b+ neutrophils and CD8+ T cells and a higher proportion of CD4+ T cells. Furthermore, patients with low TMEM115 expression displayed higher programmed cell death ligand-1 and lower lymphocyte activation gene 3 protein expression. Functionally, TMEM115 knockdown inhibited the proliferation, migration and invasion of HCC cells. In orthotopic models, TMEM115 knockdown inhibited the growth of HCC and affected the infiltration of immune cells. CONCLUSIONS Our findings show TMEM115 as a promising prognostic indicator for HCC and hold promise in predicting responses to immune therapy, emphasising its potential clinical relevance and intricate involvement in the immune microenvironment of HCC.
Collapse
Affiliation(s)
- Pingping Sun
- Department of Clinical and Translational Research Center, Affiliated Hospital of Nantong University & Medical School of Nantong University, Nantong, China
| | - Haiyan Xu
- Department of Clinical and Translational Research Center, Affiliated Hospital of Nantong University & Medical School of Nantong University, Nantong, China
| | - Chengfeng Guo
- Department of Clinical and Translational Research Center, Affiliated Hospital of Nantong University & Medical School of Nantong University, Nantong, China
| | - Lei Yang
- Department of Clinical and Translational Research Center, Affiliated Hospital of Nantong University & Medical School of Nantong University, Nantong, China
| | - Xiaojing Zhang
- Department of Clinical and Translational Research Center, Affiliated Hospital of Nantong University & Medical School of Nantong University, Nantong, China
| | - Bing Lu
- Department of Clinical and Translational Research Center, Affiliated Hospital of Nantong University & Medical School of Nantong University, Nantong, China
| | - Lei Chen
- Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, People's Republic of China
| | - Jianfei Huang
- Department of Clinical and Translational Research Center, Affiliated Hospital of Nantong University & Medical School of Nantong University, Nantong, China
- Institute of Oncology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
37
|
Shen Z, Wang Y, Gao J, Gu W, Ren Z, Xu L, Qian R, Miao Q, Hu X, Wu Y, Liu W, Cai Y, Wan CC, Zhu Y, Sun L, Yan T. The EZH2/MCM Complex/hTERT axis facilitates hepatocellular carcinoma progression by inhibiting cellular senescence. Mech Ageing Dev 2025; 224:112040. [PMID: 39933657 DOI: 10.1016/j.mad.2025.112040] [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: 11/17/2024] [Revised: 02/07/2025] [Accepted: 02/07/2025] [Indexed: 02/13/2025]
Abstract
The complex pathogenesis of hepatocellular carcinoma (HCC) limits the effectiveness of current therapies. Through RNA sequencing of cancerous and adjacent non-cancerous tissues from six HCC patients, we identified a significant upregulation of MCM2-7 genes, which encode proteins that form the MCM complex, a DNA helicase involved in DNA replication and cell cycle progression. We focused on MCM2, MCM3, and MCM7, and observed that knockdown of these proteins inhibited HCC cell proliferation. Further analysis revealed a critical regulatory axis involving EZH2, the MCM complex, and hTERT. EZH2 was found to be highly correlated with MCM complex gene expression and directly bound to the MCM gene promoters, regulating their expression. This EZH2/MCM complex/hTERT axis may play a key role in suppressing cellular senescence, thereby promoting HCC progression. Knocking down MCM complex genes reduced hTERT expression, inducing HCC cell senescence and enhancing the therapeutic efficacy of sorafenib. These findings suggest that the EZH2/MCM complex/hTERT axis could serve as a novel therapeutic target for HCC.
Collapse
Affiliation(s)
- Ziyi Shen
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai 200444, China
| | - Yuanhui Wang
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai 200444, China
| | - Jie Gao
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai 200444, China
| | - Wei Gu
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai 200444, China
| | - Ziyi Ren
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai 200444, China
| | - Luanqi Xu
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai 200444, China
| | - Rui Qian
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai 200444, China
| | - Qinyi Miao
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai 200444, China
| | - Xiaomeng Hu
- University and College Key Lab of Natural Product Chemistry and Application in Xinjiang, School of Chemistry and Environmental Science, Yili Normal University, Yining 835000, China
| | - Yan Wu
- University and College Key Lab of Natural Product Chemistry and Application in Xinjiang, School of Chemistry and Environmental Science, Yili Normal University, Yining 835000, China
| | - Wei Liu
- University and College Key Lab of Natural Product Chemistry and Application in Xinjiang, School of Chemistry and Environmental Science, Yili Normal University, Yining 835000, China
| | - Yi Cai
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Chunpeng Craig Wan
- Jiangxi Key Laboratory for Postharvest Technology and Nondestructive Testing of Fruits and Vegetables, College of Agronomy, Jiangxi Agricultural University, Nanchang 330045, China
| | - Yansong Zhu
- Department of Medicine Yong Loo Lin School of Medicine, National University of Singapore, 117549, Singapore.
| | - Lei Sun
- Department of Pathology, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China.
| | - Tingdong Yan
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai 200444, China.
| |
Collapse
|
38
|
Yang S, Chen J, Xie K, Liu F. NPC1 promotes the progression of hepatocellular carcinoma by mediating the accumulation of neutrophils into the tumor microenvironment. FEBS Open Bio 2025; 15:661-673. [PMID: 39707615 PMCID: PMC11961396 DOI: 10.1002/2211-5463.13951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 11/20/2024] [Accepted: 12/02/2024] [Indexed: 12/23/2024] Open
Abstract
Hepatocellular carcinoma remains a significant threat to human health. Recent studies have found that the intake of cellular cholesterol contributes to the development and progression of hepatocellular carcinoma, although the exact mechanisms remain unclear. Our analysis of transcriptomic and proteomic databases has identified increased mRNA and protein expression levels of NPC1, a cholesterol intracellular transporter protein, in hepatocellular carcinoma tissues. This increase is significantly associated with a worse prognosis for patients. To corroborate these findings, we performed immunohistochemical staining of NPC1 on liver tissue samples from patients, revealing significantly higher expression levels of NPC1 in hepatocellular carcinoma tissues compared to normal tissues. Subsequent investigations have revealed that NPC1 expression does not significantly influence the proliferation of hepatocellular carcinoma cells in vitro. However, it has a substantial inhibitory effect on the progression of hepatocellular carcinoma tumors when observed in vivo. Utilizing flow cytometry to monitor cellular changes within the tumor microenvironment has led us to discover that NPC1 plays a crucial role in the regulation of neutrophil recruitment within the tumor. Using further neutrophil depletion experiments, we determined that the role of NPC1 in advancing hepatocellular carcinoma progression truly relies on neutrophils. These observations are further reinforced by a comprehensive analysis of clinical databases alongside immunohistochemistry findings. In conclusion, our research suggests that NPC1's overexpression could contribute to hepatocellular carcinoma progression by promoting neutrophil recruitment, positioning NPC1 as a promising new biomarker and therapeutic target for hepatocellular carcinoma.
Collapse
Affiliation(s)
- Songhai Yang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
- Department of General SurgeryThe First People's Hospital of HefeiChina
| | - Jiangming Chen
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Kun Xie
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Fubao Liu
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| |
Collapse
|
39
|
Yang J, Hu B, Zhang G, Wu K, Zhang X, Ji M, Zhang B, Shi H, Li D. Protocadherin 17 weakens the lenvatinib resistance of liver cancer through inducing ferroptosis. Exp Cell Res 2025; 447:114495. [PMID: 40049312 DOI: 10.1016/j.yexcr.2025.114495] [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: 11/14/2024] [Revised: 01/23/2025] [Accepted: 03/04/2025] [Indexed: 03/09/2025]
Abstract
Lenvatinib has been employed in the treatment of advanced liver cancer; however, its clinical application is significantly impeded by frequent drug resistance. Recent studies have revealed that lenvatinib treatment triggers ferroptosis in liver cancer cells, providing a novel approach to addressing lenvatinib resistance. In this study, we initially validated the induction of ferroptosis by lenvatinib in liver cancer cells. Remarkably, protocadherin 17 (PCDH17), an adhesion-related protein, was found to be down-regulated in liver cancer, and overexpression of PCDH17 could induce ferroptosis in liver cancer cells. Importantly, silencing PCDH17 inhibited the impact of lenvatinib on liver cancer cell ferroptosis, while overexpression of PCDH17 had the opposite effect. These findings were further confirmed using a xenograft tumor model in BALB/c nude mice. Additionally, lenvatinib-resistant (LR) liver cancer cells were generated for additional validation purposes. It was observed that LR-liver cancer cells lost their susceptibility to ferroptosis induction by lenvatinib; however, overexpression of PCDH17 reactivated their sensitivity to ferroptosis. Corresponding results were also verified in BALB/c nude mice models. In conclusion, these results suggest that as a novel regulator of ferroptosis, PCDH17 can alleviate lenvatinib resistance and potentially enhance the therapeutic efficacy of lenvatinib in treating liver cancer.
Collapse
Affiliation(s)
- Jun Yang
- Department of General Surgery, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China; Research Center of Digestive Diseases, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Bin Hu
- Research Center of Digestive Diseases, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Department of General Surgery, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Guowei Zhang
- Research Center of Digestive Diseases, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Kai Wu
- Research Center of Digestive Diseases, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xue Zhang
- Research Center of Digestive Diseases, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Mengxuan Ji
- Research Center of Digestive Diseases, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Bin Zhang
- Research Center of Digestive Diseases, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Hengliang Shi
- Research Center of Digestive Diseases, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China; Institute of Digestive Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China; Central Laboratory, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Dechun Li
- Department of General Surgery, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China.
| |
Collapse
|
40
|
Parhira S, Zhu G, Wangteeraprasert A, Sawong S, Suknoppakit P, Somran J, Kaewpaeng N, Pansooksan K, Pekthong D, Srisawang P. Enhancement of apoptosis in HCT116 and HepG2 cells by Coix lacryma-jobi var. lacryma-jobi seed extract in combination with sorafenib. CHINESE HERBAL MEDICINES 2025; 17:322-339. [PMID: 40256710 PMCID: PMC12009101 DOI: 10.1016/j.chmed.2025.02.005] [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: 07/16/2024] [Revised: 09/24/2024] [Accepted: 02/19/2025] [Indexed: 04/22/2025] Open
Abstract
Objective Coix lacryma-jobi, a highly regarded Asian herb widely used in traditional Chinese medicine, is recognized for its dual benefits in promoting overall health and treating various diseases. While it exhibits moderate anticancer efficacy when used alone, this study investigated the enhanced anticancer potential of raw and cooked Coix lacryma-jobi var. lacryma-jobi (CL) seed extracts in combination with sorafenib against HCT116 and HepG2 cancer cell lines. The combination of sorafenib with other anticancer agents, including natural extracts, has garnered significant attention as a promising strategy for developing more effective cancer therapies. Methods Dry powders of raw (R) and cooked (C) CL seeds, obtained from a local commercial source in Thailand, were extracted and fractionated using ethanol (E), dichloromethane (D), ethyl acetate (A), and water (W) to produce eight fractions: CLRE, CLCE, CLRD, CLCD, CLRA, CLCA, CLRW, and CLCW. The coixol content in raw and cooked seed extracts was quantified and expressed as μg of coixol per gram of extract. The cytotoxic effects of these fractions were evaluated against HCT116 and HepG2 cells using the MTT assay. Fractions demonstrating the most significant cytotoxic responses were combined with sorafenib to evaluate their synergistic effects. Apoptosis induction and mitochondrial membrane potential (MMP) were assessed, and the underlying mechanism of apoptosis was explored by analyzing reactive oxygen species (ROS) generation and antioxidant protein expression levels. Additionally, the combination treatment's effect on the phosphatidylinositol-3 kinase (PI3K)/protein kinase B (AKT)/mechanistic target of rapamycin (mTOR) pathway was investigated. Results One gram of CLCE and CLCD extracts contained higher coixol levels (7.02 μg and 9.69 μg, respectively) compared to CLRE and CLRD (2.66 μg and 5.96 μg, respectively). Coixol content in CLRA, CLRW, and CLCW fractions was undetectable under the study conditions. All extract fractions exhibited IC50 values exceeding 1 mg/mL after 24- and 48-hour incubations with HCT116 and HepG2 cells, indicating limited cytotoxicity when used independently. CLRD and CLCD fractions were selected for combination studies at a concentration of 1 mg/mL, combined with sub-IC50 concentrations of sorafenib to minimize its side effects. This combination significantly increased cytotoxicity, inducing apoptosis in HCT116 and HepG2 cells by elevating ROS levels and reducing the expression of superoxide dismutase 2 and catalase. Furthermore, the combination treatment downregulated the PI3K/AKT/mTOR pathway, indicating a targeted anticancer mechanism. Conclusion The combination of CLCD with sorafenib demonstrates significant potential as a strategy for future anticancer therapies. This CL seed extract, cultivated and commercially available in Thailand, shows promise as a natural supplement to enhance the efficacy of chemotherapy in upcoming clinical anticancer applications.
Collapse
Affiliation(s)
- Supawadee Parhira
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok 65000, Thailand
- Center of Excellence for Innovation in Chemistry, Naresuan University, Phitsanulok 65000, Thailand
- Center of Excellence for Environmental Health and Toxicology, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok 65000, Thailand
| | - Guoyuan Zhu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa 999078, Macau
| | | | - Suphunwadee Sawong
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Pennapha Suknoppakit
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Julintorn Somran
- Department of Pathology, Faculty of Medicine, Naresuan University, Phitsanulok 65000, Thailand
| | - Naphat Kaewpaeng
- Center of Excellence for Innovation in Chemistry, Naresuan University, Phitsanulok 65000, Thailand
- Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok 65000, Thailand
| | - Khemmachat Pansooksan
- Center of Excellence for Innovation in Chemistry, Naresuan University, Phitsanulok 65000, Thailand
- Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok 65000, Thailand
| | - Dumrongsak Pekthong
- Center of Excellence for Innovation in Chemistry, Naresuan University, Phitsanulok 65000, Thailand
- Center of Excellence for Environmental Health and Toxicology, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok 65000, Thailand
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok 65000, Thailand
| | - Piyarat Srisawang
- Center of Excellence for Innovation in Chemistry, Naresuan University, Phitsanulok 65000, Thailand
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
- Center of Excellence in Medical Biotechnology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| |
Collapse
|
41
|
Chen L, Jiang XD, Liu XP, Lee YZ, Tham CL, Yusof R, Gao S, Lee MT. Mcl-1 is an important target protein for kaempferol from persimmon leaves in sensitizing ABT-199 to induce apoptosis in hepatoma cancer cells. Med Oncol 2025; 42:146. [PMID: 40169432 DOI: 10.1007/s12032-025-02696-3] [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: 12/17/2024] [Accepted: 03/19/2025] [Indexed: 04/03/2025]
Abstract
Overexpression of Mcl-1 causes hepatocellular carcinoma resistance to Bcl-2 inhibitors, but there are currently no direct Mcl-1 inhibitors available for clinical application. Our previous research demonstrated that kaempferol from persimmon leaves (KPL) can sensitize ABT-199 to inhibit liver cancer cell proliferation. This study further explored the effect of KPL sensitizing ABT-199 on liver cancer cell apoptosis and its potential mechanisms. The inhibitory effects of KPL and ABT-199, both individually and in combination, on the proliferation of HepG2, Huh7, and HCCLM3 cells were evaluated. Cell apoptosis and mitochondrial morphology were assessed with flow cytometry and confocal microscopy, respectively. Apoptosis and changes in Mcl-1 protein expression were evaluated after siMcl-1 knockdown. Molecular docking simulations were used to analyze the interactions of KPL and ABT-199, both individually and in combination, with Mcl-1 protein. The effect of KPL on Mcl-1 stability was investigated with proteasome inhibitor MG132. The results demonstrated that KPL showed a strong sensitizing effect on ABT-199 (CI value < 1), enhanced liver cancer cell proliferation inhibition and increased apoptosis rate. Combined treatment led to mitochondrial fragmentation and swelling, and significantly reduced Mcl-1 expression. siMcl-1 interference resulted in little difference in apoptosis rates and Mcl-1 expression between the combination treatment and untreated groups. Molecular docking revealed that KPL increased the affinity of ABT-199 for Mcl-1, whereas MG132 prevented KPL from downregulating Mcl-1 expression. These findings suggest that KPL enhances ABT-199-induced apoptosis in HCC cells by targeting Mcl-1 protein through increasing the affinity between ABT-199 and Mcl-1, while also promoting Mcl-1 degradation by affecting post-translational modifications.
Collapse
Affiliation(s)
- Li Chen
- Faculty of Pharmaceutical Sciences, UCSI University, 56000, Kuala Lumpur, Malaysia
- Department of Pharmacology, College of Medicine, Guangxi University of Science and Technology, Liuzhou, 545006, China
| | - Xu Dong Jiang
- Department of Pharmacology, College of Medicine, Guangxi University of Science and Technology, Liuzhou, 545006, China
| | - Xue Ping Liu
- Department of Pharmacology, College of Medicine, Guangxi University of Science and Technology, Liuzhou, 545006, China
| | - Yu Zhao Lee
- School of Healthy Aging, Aesthetic and Regenerative Medicine, Faculty of Medicine and Health Sciences, UCSI University, 56000, Kuala Lumpur, Malaysia
| | - Chau Ling Tham
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
- Natural Medicine and Product Research Laboratory (NaturMeds), Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Rohana Yusof
- Faculty of Applied Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Si Gao
- Department of Pharmacology, College of Medicine, Guangxi University of Science and Technology, Liuzhou, 545006, China.
| | - Ming Tatt Lee
- Faculty of Pharmaceutical Sciences, UCSI University, 56000, Kuala Lumpur, Malaysia.
- UCSI Wellbeing Research Centre, UCSI University, 56000, Kuala Lumpur, Malaysia.
| |
Collapse
|
42
|
Wu D, Liu Z, Sun Y, Gou C, Shang R, Lu M, Zhang R, Wei H, Li C, Shi Y, Zhang C, Wang Y, Wei D, Chen Z, Bian H. Integrated Analysis of the Anoikis-Related Signature Identifies Rac Family Small GTPase 3 as a Novel Tumor-Promoter Gene in Hepatocellular Carcinoma. MedComm (Beijing) 2025; 6:e70125. [PMID: 40123831 PMCID: PMC11928873 DOI: 10.1002/mco2.70125] [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: 08/29/2024] [Revised: 12/30/2024] [Accepted: 12/31/2024] [Indexed: 03/25/2025] Open
Abstract
Anoikis resistance in hepatocellular carcinoma (HCC) cells boosts survival and metastasis. This study aimed to establish an anoikis-related genes (ARGs)-based model for predicting HCC patients' outcomes and investigate the clinicopathological significance and function of crucial ARGs. The transcriptional expression patterns for HCC cohorts were compiled from TCGA, GEO and ICGC. Univariate and LASSO multivariate analyses were performed to screen for prognostic ARGs. Gain- and loss-of-function studies, RNA sequencing, and mass spectrometry were employed to elucidate the underlying mechanisms of ARGs in HCC. We established a five-gene ARGs risk model for HCC prognosis, with an AUC value of 0.812 for 1-year survival. Among the five genes, Rac family small GTPase 3 (RAC3) was upregulated in HCC relative to adjacent normal tissues and negatively correlated to overall survival and disease-free survival of patients with HCC. Silence of RAC3 in HCC cells resulted in an increased cell apoptosis and diminished cell proliferation and invasion. Mechanistically, we uncovered that RAC3 binding with SOX6 propelled the advancement of HCC cells through NNMT-mediated stimulation of the cAMP/MAPK/Rap1 signaling. In particular, EHop-016, a small molecule inhibitor targeting RAC3, significantly suppressed HCC progression.
Collapse
Affiliation(s)
- Dong Wu
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Ze‐Kun Liu
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Ying Sun
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Chu‐Heng Gou
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
- Department of Hepatobiliary SurgeryXijing HospitalFourth Military Medical UniversityXi'anChina
| | - Run‐Ze Shang
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Meng Lu
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Ren‐Yu Zhang
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Hao‐Lin Wei
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Can Li
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Ying Shi
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Cong Zhang
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Yu‐Tong Wang
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Ding Wei
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Zhi‐Nan Chen
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| | - Huijie Bian
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anChina
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesFourth Military Medical UniversityXi'anChina
| |
Collapse
|
43
|
Gu Y, Zhang Z, Huang H, Zhu W, Liu H, Zhang R, Weng N, Sun X. The dual role of CXCL9/SPP1 polarized tumor-associated macrophages in modulating anti-tumor immunity in hepatocellular carcinoma. Front Immunol 2025; 16:1528103. [PMID: 40230843 PMCID: PMC11994707 DOI: 10.3389/fimmu.2025.1528103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 03/13/2025] [Indexed: 04/16/2025] Open
Abstract
Introduction The main challenge for cancer therapy lies in immuno-suppressive tumor micro-environment. Reprogramming tumor-associated macrophages (TAMs) into an anti-tumor phenotype is a promising strategy. Methods A comprehensive analysis by combing multi-regional single-cell, bulk and spatial transcriptome profiling with radiomics characterization was conducted to dissect the heterogeneity of TAMs and resolve the landscape of the CXCL9:SPP1 (CS) macrophage polarity in HCC. Results TAMs were particularly increased in HCC. SPP1+ TAMs and CXCL9+ TAMs were identified as the dominant subtypes with different evolutionary trajectories. SPP1+ TAMs, located in the tumor core, co-localized with cancer-associated fibroblasts to promote tumor growth and further contributed to worse prognosis. In contrast, CXCL9+ TAMs, located in the peritumoral region, synergized with CD8+ T cells to create an immunostimulatory micro-environment. For the first time, we explored the applicability of CS polarity in HCC tumors and revealed several key transcription factors involved in shaping this polarity. Moreover, CS polarity could serve as a potential indicator of prognostic and micro-environmental status for HCC patients. Based on medical imaging data, we developed a radiomics tool, RCSP (Radiogenomics-based CXCL9/SPP1 Polarity), to assist in non-invasively predicting the CS polarity in HCC patients. Conclusion Our research sheds light on the regulatory roles of SPP1+ TAMs and CXCL9+ TAMs in the micro-environment and provides new therapeutic targets or insights for the reprogramming of targeted macrophages in HCC.
Collapse
Affiliation(s)
- Yu Gu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Zhihui Zhang
- College of Acupuncture-Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hao Huang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Wenyong Zhu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Hongjia Liu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Rongxin Zhang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Nan Weng
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Xiao Sun
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| |
Collapse
|
44
|
Li X, Li Y, Zhu J, Yang Y, Yang S. Bibliometric analysis of nanoparticle research for diagnostics and therapeutics in hepatocellular carcinoma. DISCOVER NANO 2025; 20:61. [PMID: 40159297 PMCID: PMC11955440 DOI: 10.1186/s11671-025-04226-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 02/25/2025] [Indexed: 04/02/2025]
Abstract
OBJECTIVES The aim of this study was to explore the bibliometric analysis of nanomaterials-based therapies for hepatocellular carcinoma as a means of assessing the current state of development and future trends in the field. MATERIALS AND METHODS Literature on hepatocellular carcinoma and nanomedicine interactions was searched from the core database of the Web of Science and bibliometric and visualisation analyses were performed using VOSviewer, CiteSpace and GraphPad Prism data analysis software. We focused on important keywords, countries, authors, affiliations, journals, and literature in the field of nanomaterials for HCC. RESULTS The search resulted in the finalization of 421 documents. The search resulted in the finalization of 421 documents. From 2008 to 2023, nanomedicine research in HCC has developed rapidly, and the number of published papers has steadily increased, increasing by about 2300%. There are currently 57 countries involved in research in this area. Among them, The USA had the strongest international cooperation network and cooperated most closely with China. Gene delivery and carbon nanotubes were early keywords, immunotherapy and nanocarriers are recent research hotspots. It is important that the selection of nanocarriers and drug delivery have become the core trends driving the development of hepatocellular carcinoma. CONCLUSION The combination of nanomaterials with traditional imaging techniques such as MRI can improve the early diagnosis rate of HCC. Nanomaterials can achieve precise targeting of cancer cells by encapsulating drugs, loading bioactive molecules or modifying specific targeting ligands, thus significantly improving drug efficacy and effectively reducing adverse reactions in therapy.
Collapse
Affiliation(s)
- Xiaoqing Li
- Central Laboratory, Yanbian University Hospital, Yanji, 133000, China
- Key Laboratory of Pathobiology (Yanbian University Hospital), State Ethnic Affairs Commission, Yanji, 133000, China
| | - Yue Li
- Central Laboratory, Yanbian University Hospital, Yanji, 133000, China
- Key Laboratory of Pathobiology (Yanbian University Hospital), State Ethnic Affairs Commission, Yanji, 133000, China
| | - Jingyan Zhu
- Central Laboratory, Yanbian University Hospital, Yanji, 133000, China
- Key Laboratory of Pathobiology (Yanbian University Hospital), State Ethnic Affairs Commission, Yanji, 133000, China
| | - Yang Yang
- Central Laboratory, Yanbian University Hospital, Yanji, 133000, China
- Key Laboratory of Pathobiology (Yanbian University Hospital), State Ethnic Affairs Commission, Yanji, 133000, China
| | - Shipeng Yang
- Central Laboratory, Yanbian University Hospital, Yanji, 133000, China.
- Key Laboratory of Pathobiology (Yanbian University Hospital), State Ethnic Affairs Commission, Yanji, 133000, China.
| |
Collapse
|
45
|
Liu X, Kang X, Kang H, Yan H. The immunosuppressive role of MDSCs in HCC: mechanisms and therapeutic opportunities. Cell Commun Signal 2025; 23:155. [PMID: 40148954 PMCID: PMC11951757 DOI: 10.1186/s12964-025-02170-7] [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: 12/30/2024] [Accepted: 03/23/2025] [Indexed: 03/29/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a prevalent malignancy with a significant global burden. Despite substantial advancements in HCC treatment in recent years, therapeutic efficacy remains constrained by immune evasion mechanisms within the tumor microenvironment (TME). Myeloid-derived suppressor cells (MDSCs), as critical immunosuppressive elements of the TME, have garnered increasing attention for their role in tumor progression. Recent studies emphasize their central involvement in promoting immune evasion, tolerance, and immunosuppression in HCC. This review examines the contributions of MDSCs to HCC pathogenesis, elucidates their underlying mechanisms, and discusses ongoing clinical trials, emphasizing their potential as therapeutic targets for improving clinical outcomes.
Collapse
Affiliation(s)
- Xiling Liu
- School of Public Health, Hebei Medical University, Shijiazhuang, 050017, China
- Hebei Key Laboratory of Immune Mechanism of Major Infectious Diseases and New Technology of Diagnosis and Treatment, The Fifth Hospital of Shijiazhuang, Shijiazhuang, 050021, China
| | - Xichun Kang
- Beijing Fangshan District Center for Disease Control and Prevention, Beijing, 102488, China
| | - Haiyan Kang
- Hebei Key Laboratory of Immune Mechanism of Major Infectious Diseases and New Technology of Diagnosis and Treatment, The Fifth Hospital of Shijiazhuang, Shijiazhuang, 050021, China
- Department of the Sixth Infection, The Fifth Hospital of Shijiazhuang, Shijiazhuang, 050021, China
| | - Huimin Yan
- School of Public Health, Hebei Medical University, Shijiazhuang, 050017, China.
- Hebei Key Laboratory of Immune Mechanism of Major Infectious Diseases and New Technology of Diagnosis and Treatment, The Fifth Hospital of Shijiazhuang, Shijiazhuang, 050021, China.
| |
Collapse
|
46
|
Li HS, Zhang XF, Fu J, Yuan B. Efficacy of microwave ablation vs laparoscopic hepatectomy for primary small liver cancer: A comparative study. World J Gastrointest Surg 2025; 17:101786. [PMID: 40162382 PMCID: PMC11948124 DOI: 10.4240/wjgs.v17.i3.101786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/13/2024] [Accepted: 01/07/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND In-depth comparative investigations in terms of clinical efficacies of liver tumor microwave ablation (MWA) and laparoscopic hepatectomy (LH), which are both important treatment modalities for liver neoplasms, have been limited in patients diagnosed with primary small liver cancer (PSLC). AIM To compare and analyze the clinical efficacy of liver tumor MWA and LH for PSLC. METHODS This study retrospectively analyzed the medical records of 123 patients with PSLC admitted to Xuzhou Central Hospital from January 2015 to November 2022 and categorized them based on treatment modalities into the LH and MWA groups. The LH group, consisting of 61 cases, received LH, and the MWA group, which included 62 cases, underwent liver tumor MWA. Basic data and various perioperative indicators were compared between the two groups, including changes in liver function indicators [alanine aminotransferase (ALT), glutamic aminotransferase (AST), and total bilirubin (TBIL)] pre- and post-treatment, and efficacy and postoperative complications were analyzed. RESULTS No statistically significant difference was observed between the two groups in terms of age, gender, tumor diameter, liver function Child-Pugh classification and number of tumors, body mass index, and educational status (P > 0.05). The overall effective rate was higher in the MWA group than in the LH group (98.39% vs 88.52%) (χ 2 = 4.918, P = 0.027). The MWA group exhibited less operation time, intraoperative bleeding, defecation time, and hospital stay than the LH group (P < 0.05). No difference was found in liver function indicators between the two groups pre-treatment (P > 0.05), and ALT, AST, and TBIL levels decreased in both groups post-treatment, with the MWA group demonstrating lower levels (P < 0.05). The MWA and LH groups exhibited postoperative complication rates of 4.84% and 19.67%, respectively, with statistically significant differences between the two groups (P = 0.012, χ 2 = 6.318). CONCLUSION MWA is more effective in treating PSLC, and it promotes faster postoperative recovery for patients, and more security improves liver function and reduces postoperative complications compared to LH.
Collapse
Affiliation(s)
- Huan-Song Li
- Department of Hepatobiliary Pancreatic Center, Xuzhou Central Hospital, Xuzhou 221009, Jiangsu Province, China
| | - Xuan-Feng Zhang
- Department of Hepatobiliary Pancreatic Center, Xuzhou Central Hospital, Xuzhou 221009, Jiangsu Province, China
| | - Jun Fu
- Department of Hepatobiliary Pancreatic Center, Xuzhou Central Hospital, Xuzhou 221009, Jiangsu Province, China
| | - Bo Yuan
- Department of Hepatobiliary Pancreatic Center, Xuzhou Central Hospital, Xuzhou 221009, Jiangsu Province, China
| |
Collapse
|
47
|
Yang Z, Feng X, Yu H, Lv L, Gao C, Liu W, Yi S, Jia C, Fu B. Identification of tumor immune infiltration-associated VPS72 and prognostic significance of VPS72 and CD8A in hepatocellular carcinoma. Discov Oncol 2025; 16:410. [PMID: 40146476 PMCID: PMC11950588 DOI: 10.1007/s12672-025-02017-0] [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: 09/29/2024] [Accepted: 02/25/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Copy Number Alterations (CNAs)-driven genes have gained attention as potential markers for predicting the response to immune checkpoint blockade in cancer treatment. Among them, VPS72 has emerged as a promising candidate in hepatocellular carcinoma (HCC). However, the relationship between VPS72 and immune infiltration remains unclear. METHODS TIMER analysis was performed to identify immune populations in bulk-RNAseq data. Then, we investigated the relationship between VPS72 and immune infiltration in HCC using diverse data sources, including the TCGA and GEO databases, clinical specimens, and animal models. RESULTS Our findings in the immunogenomic and TCGA-LIHC studies revealed significant enrichment of VPS72 among IRG in the altered group. Differential analysis and KEGG pathway analysis further highlighted the involvement of differentially expressed genes (DETs) in pathways related to the T cell receptor signaling pathway. Importantly, TIMER analysis suggested that low expression of VPS72 was associated with high infiltration of CD8 + T cells in multiple publicly available HCC datasets. To validate these findings, we conducted in vivo experiments and observed higher CD8A expression in VPS72-knockdown tumors. Additionally, in our patient cohort, individuals with low VPS72 expression exhibited higher CD8A expression. Furthermore, we identified a co-expression subtype characterized by low VPS72 and high CD8A levels, which showed a more favorable disease-free survival outcome in HCC. CONCLUSIONS The expression of VPS72 in tumors is associated with the tumor infiltration. VPS72 and CD8A coexpression are prognostic biomarkers in HCC.
Collapse
Affiliation(s)
- Zhou Yang
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-Sen University, Guangzhou, China
| | - Xiao Feng
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-Sen University, Guangzhou, China
| | - Haoyuan Yu
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-Sen University, Guangzhou, China
| | - Lei Lv
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-Sen University, Guangzhou, China
| | - Chengli Gao
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-Sen University, Guangzhou, China
| | - Wei Liu
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangdong Province Engineering Laboratory for Transplantation Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Shuhong Yi
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-Sen University, Guangzhou, China
| | - Changchang Jia
- Cell-Gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| | - Binsheng Fu
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
48
|
Zhang Z, Rao C, Hu M, Yan W, Du Z. Highly expressed GCN1 is associated with cancer progression and poor prognosis in hepatocellular carcinoma patients. Cancer Cell Int 2025; 25:107. [PMID: 40114124 PMCID: PMC11927180 DOI: 10.1186/s12935-025-03732-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 03/06/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND General control non-derepressible protein 1 (GCN1), a ribosome-binding protein, has been implicated in the development and progression of multiple cancers. However, the potential role of GCN1 in hepatocellular carcinoma (HCC) has not yet been investigated. METHODS The expression of GCN1 in HCC was analyzed using multiple databases. Bioinformatics analysis was employed to investigate the correlation of GCN1 expression with clinical significance and immune infiltration in HCC. Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, gene set enrichment analysis (GSEA), and in vitro experiments were conducted to study the function and potential mechanisms of GCN1 in HCC. RESULTS GCN1 was significantly upregulated in HCC, which was associated with worse clinicopathological features and poorer prognosis of the patients. GCN1 expression was closely associated with immune cell infiltration in HCC. GSEA analysis showed that GCN1 was involved in several tumor-related signaling pathways, including cell cycle, DNA replication, and Wnt signaling pathway. Knockdown of GCN1 inhibited the proliferation, invasion and migration of HCC cells, and also down-regulated the expression levels of cell cycle protein cyclin B1 (CCNB1), cyclin D1 (CCND1), and Wnt signaling pathway-related proteins Wnt3A and β-catenin. CONCLUSION GCN1 overexpression was associated with HCC progression and poor prognosis, and GCN1 knockdown could suppress the proliferation, migration and invasion ability of HCC cells by regulating Wnt signaling pathway, suggesting the potential of GCN1 as a prognostic and therapeutic target for HCC.
Collapse
Affiliation(s)
- Zhongchao Zhang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, Hubei, 430000, China
| | - Caijun Rao
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mingcun Hu
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Yan
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, Hubei, 430000, China.
| | - Zhipeng Du
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, Hubei, 430000, China.
| |
Collapse
|
49
|
Zhang J, Tian T, Tian S, Yao J, Zhang Y, Xie R, Yang T, Han B. Study on the Mechanism of QRICH1 Mediating PRMT1 to Regulate the Arginine Methylation Modification of cGAS to Promote Arsenics-Induced Pyroptosis in Hepatocellular Carcinoma Cells. J Hepatocell Carcinoma 2025; 12:597-614. [PMID: 40124968 PMCID: PMC11930257 DOI: 10.2147/jhc.s505266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 03/04/2025] [Indexed: 03/25/2025] Open
Abstract
Purpose This study aims to investigate the mechanism of action of arsenic-based agents against hepatocellular carcinoma (HCC) and to identify effective drug targets for HCC treatment. Methods Huh7 and HepG2 cells treated with NaAsO2 were assessed for cell viability, pyroptosis, migration, and invasion after undergoing lentiviral transfection. An orthotopic liver tumor model was established and divided into a model group and a treatment group. Proteins associated with QRICH1, PRMT1, cGAS-STING, and the classical pyroptosis pathway were quantified using Western blotting. The intracellular expression and localization of PRMT1 and NLRP3 in HCC were analyzed through cellular immunofluorescence. Co-immunoprecipitation (Co-IP) was performed to examine the protein interactions between PRMT1 and cGAS, as well as between STING and NLRP3. Chromatin immunoprecipitation (ChIP) was used to confirm QRICH1 enrichment in the PRMT1 promoter region. Results NaAsO2 treatment significantly inhibited the proliferation of Huh7 and HepG2 cells and effectively blocked their migration and invasion capabilities, while promoting cellular pyroptosis. Quantitative polymerase chain reaction(QRCR) and ChIP assays confirmed that NaAsO2 regulates PRMT1 expression by down-regulate QRICH1 binding in the PRMT1 promoter region. Additionally, NaAsO2 decreased the expression of the QRICH1-PRMT1 complex and upregulated the cGAS-STING signaling pathway, activating the downstream NLRP3-dependent classical pyroptosis pathway. Overexpression of QRICH1 reversed these effects. Conclusion NaAsO2 inhibits the expression of the QRICH1-PRMT1 axis, activates cGAS-STING signaling pathway transduction, and induces pyroptosis in HCC cells, thereby increasing the infiltration of immune cells in liver cancer tissues.
Collapse
Affiliation(s)
- Jiayuan Zhang
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
| | - Tian Tian
- Department of Eugenic Genetics, Guiyang Maternal and Child Health Care Hospital, Guiyang, Guizhou, 550003, People’s Republic of China
| | - Shanshan Tian
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
| | - Jinhai Yao
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
| | - Yingwan Zhang
- Qianxinan People’s Hospital, Qianxinan Affiliated Hospital of Zunyi Medical University, Xingyi, Guizhou, 562400, People’s Republic of China
| | - Rujia Xie
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
| | - Ting Yang
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
| | - Bing Han
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou, People’s Republic of China
| |
Collapse
|
50
|
Lu Z, Chai X, Li S. Machine learning-based identification of telomere-related gene signatures for prognosis and immunotherapy response in hepatocellular carcinoma. Mol Cytogenet 2025; 18:6. [PMID: 40102883 PMCID: PMC11921577 DOI: 10.1186/s13039-025-00705-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 02/06/2025] [Indexed: 03/20/2025] Open
Abstract
Telomere in cancers shows a main impact on maintaining chromosomal stability and unlimited proliferative capacity of tumor cells to promote cancer development and progression. So, we targeted to detect telomere-related genes(TRGs) in hepatocellular carcinoma (HCC) to develop a novel predictive maker and response to immunotherapy. We sourced clinical data and gene expression datasets of HCC patients from databases including TCGA and GEO database. The TelNet database was utilized to identify genes associated with telomeres. Genes with altered expression from TCGA and GSE14520 were intersected with TRGs, and Cox regression analysis was conducted to pinpoint genes strongly linked to survival prognosis. The risk model was developed using the Least Absolute Shrinkage and Selection Operator (LASSO) regression technique. Subsequently, evaluation of the risk model focused on immune cell infiltration, checkpoint genes, drug responsiveness, and immunotherapy outcomes across both high- and low-risk patient groups. We obtained 25 TRGs from the overlapping set of 34 genes using Cox regression analysis. Finally, six TRGs (CDC20, TRIP13, EZH2, AKR1B10, ESR1, and DNAJC6) were identified to formulate the risk score (RS) model, which independently predicted prognosis for HCC. The high-risk group demonstrated worse survival outcomes and showed elevated levels of infiltration by Macrophages M0 and Tregs. Furthermore, a notable correlation was observed between the genes in the risk model and immune checkpoint genes. The RS model, derived from TRGs, has been validated for its predictive value in immunotherapy outcomes. In conclusion, this model not only predicted the prognosis of HCC patients but also their immune responses, providing innovative strategies for cancer therapy.
Collapse
Affiliation(s)
- Zhengmei Lu
- Department of Infectious Diseases, Wenzhou Medical University Affiliated, Zhoushan Hospital, Zhoushan, 316000, China
| | - Xiaowei Chai
- Dermatology, Tongji University, Shanghai, 200040, China
| | - Shibo Li
- Department of Infectious Diseases, Wenzhou Medical University Affiliated, Zhoushan Hospital, Zhoushan, 316000, China.
| |
Collapse
|