1
|
Ma X, Chen B, Tang Y, Ju S, Jing R, Feng W, Zong W. Exploring the clinical application and biological function of novel tsRNA tsRNA-Asp-5-0002 in hepatocellular carcinoma. Gene 2025; 956:149459. [PMID: 40164240 DOI: 10.1016/j.gene.2025.149459] [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: 10/21/2024] [Revised: 03/05/2025] [Accepted: 03/28/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND Hepatocellular Carcinoma (HCC) is a highly fatal cancer, which is the most common malignant tumor globally. Recently, tRNA-derived small RNA (tsRNA) is a type of non-coding RNA that is closely related to various diseases. However, their precise existence and function in HCC remain unclear. We investigated the role of a novel tsRNA tsRNA-Asp-5-0002 in HCC. METHODS We screened novel tsRNAs in the TCGA database. There are significant differences in tsRNA-Asp-5-0002 expression in HCC compared with other digestive system tumors. qRT-PCR was applied to confirm the level of tsRNA-Asp-5-0002 in 150 HCC patients, 50 patients with benign liver lesions, and 120 healthy controls. tsRNA-Asp-5-0002 mimics was constructed to transfect HCC cells and explore the effect of tsRNA-Asp-5-0002 on malignant phenotype of HCC. Bioinformatics analysis was used to predict the potential regulatory pathways of tsRNA-Asp-5-0002 in HCC. RESULTS (1) tsRNA-Asp-5-0002 is down-regulated in HCC. (2) qRT-PCR detection tsRNA-Asp-5-0002 had high sensitivity and specificity. (3) Low-expressed tsRNA-Asp-5-0002 was related to TNM stage, differentiation, and LNM, and the ROC curve indicated that tsRNA-Asp-5-0002 had a better diagnostic performance for HCC. (4) tsRNA-Asp-5-0002 inhibits the proliferation and migration of HCC through the PLCB1/Wnt axis. CONCLUSIONS tsRNA-Asp-5-0002 can work as an auxiliary biomarker for HCC diagnosis. Over-expression of tsRNA-Asp-5-0002 restrains the malignant process of HCC, which may offer fresh tactics for the adjuvant treatment of HCC.
Collapse
Affiliation(s)
- Xinyue Ma
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Bairong Chen
- Medical School of Nantong University, Nantong University, Nantong, Jiangsu, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Yelan Tang
- Medical School of Nantong University, Nantong University, Nantong, Jiangsu, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Shaoqing Ju
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Rongrong Jing
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Wei Feng
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China; Medical School of Nantong University, Nantong University, Nantong, Jiangsu, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China.
| | - Wei Zong
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
2
|
Yao J, Yang H, Yuan M, Wang C, Liao H, Song R, Xu Z, Zeng X, Zhang Z. GINS4 silencing mediates hepatocellular cancer cell proliferation, cycle and ferroptosis through POLE2. Cell Signal 2025; 131:111742. [PMID: 40081544 DOI: 10.1016/j.cellsig.2025.111742] [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: 10/11/2024] [Revised: 02/10/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND GINS4 has been identified as a regulator associated with multiple types of cancers. However, the effects of GINS4 on hepatocellular carcinoma (HCC) have not been reported. METHODS GINS4 expression in HCC was evaluated utilizing UALCAN database. The relationship between the expression of GINS4 and the survival probability of HCC patients was analyzed using Kaplan-Meier Plotter. Cell viability was evaluated by CCK8 assay and EDU assay. qRT-PCR and western blot were performed to examine GINS4 expression. The level of cell cycle was measured by flow cytometry and western blot. Fe2+ level and ferroptosis-related proteins were measured by corresponding kits and western blot. Lipid peroxidation was explored by C11 BODIPY 581/591 probe. STRING database and HDOCK database were performed to predict the binding of GINS4 to POLE2. Immunofluorescence and western blotting was adopted for assessing cell autophagy and mTOR signaling pathway. Ki67 and GPX4 levels were measured by immunohistochemistry. The expression levels of POLE2/PI3K/AKT were assessed by western blot. RESULTS The data indicated that GINS4 expression was upregulated in HCC. Knockdown of GINS4 alleviated the proliferation and cycle and promoted ferroptosis of HuH7 cells. GINS4 was proved to bind to POLE2 and the silencing of GINS4 inhibited the expression of POLE2. GINS4 knockdown accelerated ferroptosis in HuH7 cells. POLE2 overexpression reversed the influences of GINS4 silencing on proliferation and cycle, and also ferroptosis. In addition, interference with GINS4 suppressed the activation of PI3K/AKT signaling via POLE2. In vivo experiments illustrated that GINS4 deletion suppressed HCC tumor growth, increased the GPX4 expression and restrained the Ki67 level, as well as reducing POLE2/PI3K/AKT signaling. CONCLUSION GINS4 silencing suppressed proliferation and cycle while promoted ferroptosis in HCC cells by regulating PI3K/AKT signaling via binding to POLE2.
Collapse
Affiliation(s)
- Jinni Yao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Anhui University of Science and Technology, Huainan 232007, China; Graduate School of Anhui University of Science and Technology, Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital of Anhui University of Science and Technology, Huainan 232007, China
| | - Huaicheng Yang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Anhui University of Science and Technology, Huainan 232007, China; First Clinical College of Anhui University of Science and Technology, Huainan 232007, China.
| | - Meng Yuan
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Anhui University of Science and Technology, Huainan 232007, China; Graduate School of Anhui University of Science and Technology, Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital of Anhui University of Science and Technology, Huainan 232007, China
| | - Congyu Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Anhui University of Science and Technology, Huainan 232007, China; First Clinical College of Anhui University of Science and Technology, Huainan 232007, China
| | - Heqiang Liao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Anhui University of Science and Technology, Huainan 232007, China; First Clinical College of Anhui University of Science and Technology, Huainan 232007, China
| | - Rui Song
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Anhui University of Science and Technology, Huainan 232007, China; Graduate School of Bengbu Medical University, Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital of Anhui University of Science and Technology, Huainan 232007, China
| | - Zhe Xu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Anhui University of Science and Technology, Huainan 232007, China; Graduate School of Bengbu Medical University, Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital of Anhui University of Science and Technology, Huainan 232007, China
| | - Xiangrui Zeng
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Anhui University of Science and Technology, Huainan 232007, China; Graduate School of Bengbu Medical University, Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital of Anhui University of Science and Technology, Huainan 232007, China
| | - Zheng Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Anhui University of Science and Technology, Huainan 232007, China; Graduate School of Bengbu Medical University, Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital of Anhui University of Science and Technology, Huainan 232007, China
| |
Collapse
|
3
|
Sun B, Zhao Y, Yang S, Li X, Li N, Wang Y, Han Q, Liu X, Tu Q, Zheng J, Zhang X. Celecoxib as a potential treatment for hepatocellular carcinoma in populations exposed to high PFAS levels. JOURNAL OF HAZARDOUS MATERIALS 2025; 489:137613. [PMID: 39955994 DOI: 10.1016/j.jhazmat.2025.137613] [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: 09/17/2024] [Revised: 02/12/2025] [Accepted: 02/12/2025] [Indexed: 02/18/2025]
Abstract
Per- and polyfluoroalkyl substances (PFAS), including perfluorooctane sulfonate and perfluorooctanoic acid, are associated with adverse human effects. However, few studies have assessed the effects of PFAS mixtures on hepatocellular carcinoma (HCC). In this study, we systematically investigated the effects and underlying mechanisms of PFAS mixtures on the proliferation, migration, and invasion of HCC cells (JHH-7 and Li-7) in vitro using a combination of biological techniques and high-coverage untargeted metabolomics. A six day exposure to a 5 μM PFAS mixture significantly enhanced the malignant progression of HCC in vitro. Metabolomic analysis identified the upregulation of prostaglandin E2 (PGE2) as a key factor associated with these effects. This hypothesis was further validated using celecoxib, a PGE2 inhibitor, which reduced PGE2 levels in HCC cells, consequently slowing their migration and invasion. Additionally, mice treated with celecoxib exhibited reduced tumor volumes compared with those treated with PFAS alone. These results suggest that PFAS exposure enhances HCC malignancy through the PI3K/AKT signaling pathway via increased PGE2 production. In conclusion, a 5 μM PFAS mixture accelerates HCC proliferation and invasion; moreover, celecoxib demonstrates potential as a therapeutic agent that inhibits these effects.
Collapse
Affiliation(s)
- Boshi Sun
- Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, No. 148 BaoJian-ro, Harbin, Heilongjiang Province 150086, China
| | - Yuqiao Zhao
- Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, No. 148 BaoJian-ro, Harbin, Heilongjiang Province 150086, China
| | - Shifeng Yang
- Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, No. 148 BaoJian-ro, Harbin, Heilongjiang Province 150086, China
| | - Xiaodong Li
- Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, No. 148 BaoJian-ro, Harbin, Heilongjiang Province 150086, China
| | - Nana Li
- Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, No. 148 BaoJian-ro, Harbin, Heilongjiang Province 150086, China
| | - Yujie Wang
- Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, No. 148 BaoJian-ro, Harbin, Heilongjiang Province 150086, China
| | - Qixiang Han
- Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, No. 148 BaoJian-ro, Harbin, Heilongjiang Province 150086, China
| | - Xuyun Liu
- Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, No. 148 BaoJian-ro, Harbin, Heilongjiang Province 150086, China
| | - Qiushi Tu
- Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, No. 148 BaoJian-ro, Harbin, Heilongjiang Province 150086, China
| | - Jie Zheng
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT 06510, United States.
| | - Xinyu Zhang
- Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, No. 148 BaoJian-ro, Harbin, Heilongjiang Province 150086, China.
| |
Collapse
|
4
|
Jiang S, Bao H. Exploring the mechanism of esculetin extracted from Chroogomphus rutilus in treating liver cancer based on network pharmacology, molecular docking, and in vivo experimental validation. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119837. [PMID: 40254108 DOI: 10.1016/j.jep.2025.119837] [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: 01/19/2025] [Revised: 04/08/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Chroogomphus rutilus (C. rutilus) is a traditional Chinese medicine recorded in the book Illustrations of Medicinal Fungi in China that possesses a long history of use for the treatment of various diseases, including cancer. Esculetin (ES), the primary pharmacologically active ingredient of C. rutilus, exerts significant therapeutic effects against liver cancer (LC). Nonetheless, the underlying therapeutic mechanisms of ES against LC remain poorly understood. AIM OF THE STUDY To investigate the mechanisms of ES in LC treatment. MATERIALS AND METHODS ES was isolated and identified from C. rutilus. Subsequently, related targets and mechanism of ES against LC were predicted through network pharmacology and molecular docking. The antitumor effect of ES was examined using H22 tumor-bearing mouse models. The antitumor mechanism of ES was elucidated and validated using TUNEL, enzyme-linked immunosorbent assay (ELISA), immunofluorescence analysis, Western blot (WB), and quantitative real-time polymerase chain reaction (qPCR). RESULTS The chemical structure was determined using NMR carbon and hydrogen spectra. Network pharmacology analysis indicated that ES exerted anti-LC effects via the PI3K/AKT signaling pathway and associated proteins. TUNEL and ELISA revealed that ES exhibited an obvious antitumor effect in vivo and that the levels of TNF-α, IFN-γ, IL-2, and IL-6 were significantly increased. Immunofluorescence, WB, and qPCR analyses showed that ES upregulated the protein expression of Bax, caspase-3, and caspase-9 and downregulated the protein expression of Bcl-2, VEGF, and p-AKT. CONCLUSION This study demonstrates that the mechanisms of ES in LC treatment include enhancing immunity, inhibiting angiogenesis, and promoting apoptosis of tumor cells.
Collapse
Affiliation(s)
- Shuang Jiang
- Key Laboratory of Edible Fungi Resources and Utilization, Ministry of Agriculture and Rural Affairs, Jilin Agricultural University, No. 2888 Xincheng Street, Nanguan District, Changchun, Jilin, 130118, China; College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, 130118, China.
| | - Haiying Bao
- Key Laboratory of Edible Fungi Resources and Utilization, Ministry of Agriculture and Rural Affairs, Jilin Agricultural University, No. 2888 Xincheng Street, Nanguan District, Changchun, Jilin, 130118, China; College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, 130118, China.
| |
Collapse
|
5
|
Li R, Zhang G, Tao Q, Wu Z, Liu X, Wang R, Liu L, Niu Y, Du K, Wu R, Du F, Zheng X, Li Y, Shi X. Revealing the prognostic potential of natural killer cell-related genes in hepatocellular carcinoma: the key role of NRAS. Discov Oncol 2025; 16:807. [PMID: 40383831 DOI: 10.1007/s12672-025-02200-3] [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: 08/09/2024] [Accepted: 03/21/2025] [Indexed: 05/20/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a common malignancy associated with high morbidity and mortality rates worldwide. To improve the prognosis of HCC, early diagnosis is crucial. However, to date, little is known about the role of natural killer cell-related genes (NKCRGs) in predicting the prognosis of hepatocellular carcinoma patients. In this study, we identified 24 differentially expressed NKCRGs in HCC specimens from the TCGA dataset, including 22 upregulated genes and 2 downregulated genes. Functional enrichment analysis revealed that these genes were mainly involved in immune response pathways and various cancer-related pathways. Univariate analysis identified 21 prognostic NKCRGs, with eight genes (PAK1, MAP2K2, MAPK3, PLCG1, SHC1, HRAS, NRAS, and MICB) confirmed to be involved in HCC prognosis through Venn diagram analysis. A prognostic model was developed using LASSO-Cox regression, incorporating four genes (MAP2K2, SHC1, HRAS, and NRAS). The model's risk score was significantly associated with overall survival (OS) in both the TCGA and ICGC cohorts. Patients with high-risk scores had poorer OS, as demonstrated by Kaplan-Meier curves and ROC analyses. The risk score was not significantly correlated with gender or age but was higher in patients with advanced tumor grades and stages. Immune status analysis using ssGSEA showed higher enrichment scores for various immune cells and pathways in the high-risk group. Additionally, the risk score was positively correlated with the immune score, indicating its potential role in tumor microenvironment modulation. Expression analysis revealed that HRAS, SHC1, MAP2K2, and NRAS were upregulated in HCC tissues, with higher expressions of HRAS, MAP2K2, and NRAS associated with shorter OS. Knockdown experiments confirmed that silencing NRAS suppressed the proliferation of HCC cells, highlighting its potential as a therapeutic target. Overall, our findings suggest that the identified NKCRGs, particularly NRAS, play crucial roles in HCC progression and could serve as valuable prognostic markers and therapeutic targets.
Collapse
Affiliation(s)
- Ruixi Li
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Guangquan Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Qiang Tao
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Ziyun Wu
- The First Clinical Medical College of Nanchang University, Nanchang, 330031, China
| | - Xiaoping Liu
- The First Clinical Medical College of Nanchang University, Nanchang, 330031, China
| | - Rongrong Wang
- The First Clinical Medical College of Nanchang University, Nanchang, 330031, China
| | - Lei Liu
- Department of Clinical Pharmacy, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Yiran Niu
- The First Clinical Medical College of Nanchang University, Nanchang, 330031, China
| | - Kaile Du
- The First Clinical Medical College of Nanchang University, Nanchang, 330031, China
| | - Runpeng Wu
- The First Clinical Medical College of Nanchang University, Nanchang, 330031, China
| | - Fei Du
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Xiyan Zheng
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Yingliang Li
- Department of Breast Disease Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| | - Xianjie Shi
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China.
| |
Collapse
|
6
|
Zhang X, Sun P, Chu T, Feng Y, Zhang X. Comprehensive bioinformatics analysis of MEX3 family genes in hepatocellular carcinoma. Sci Rep 2025; 15:16971. [PMID: 40374855 PMCID: PMC12081770 DOI: 10.1038/s41598-025-02057-0] [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: 02/26/2025] [Accepted: 05/12/2025] [Indexed: 05/18/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a highly lethal malignancy associated with poor prognosis due to late-stage diagnosis and high recurrence rates. The MEX3 family genes has been implicated in various cancers; however, their roles in HCC remain largely unexplored. This study aims to systematically analyze the expression patterns, prognostic significance, and immune-related functions of MEX3A, MEX3B, MEX3C, and MEX3D in HCC using comprehensive bioinformatics approaches. We conducted a multi-level bioinformatics analysis to investigate the expression, prognostic significance, clinicopathological correlations, genetic alterations, immune associations, and functional mechanisms of MEX3 family members in HCC. Transcriptomic data from TCGA and GEO databases, along with experimental validation via qRT-PCR and Western blotting, were used to assess expression profiles. Kaplan-Meier, ROC curve, and Cox regression analyses were employed for prognostic evaluation. Co-expression, enrichment, and immune infiltration analyses further elucidated the functional and immunological relevance of MEX3 family genes. A prognostic model based on co-expressed genes was constructed and validated using LASSO and time-dependent ROC analyses. MEX3A, MEX3B, MEX3C, and MEX3D were significantly upregulated in HCC tissues compared to normal liver tissues (P < 0.05). ROC curve analysis demonstrated high diagnostic accuracy, particularly for MEX3A (AUC = 0.915). Kaplan-Meier survival analysis indicated that elevated MEX3A and MEX3C expression was associated with poorer overall survival (OS) and disease-specific survival (DSS) (P < 0.05). Mutation analysis revealed that MEX3A exhibited the highest alteration frequency (11%), primarily through gene amplifications. Immune infiltration analysis demonstrated significant correlations between MEX3 expression and multiple immune cell populations, including regulatory T cells (Tregs), cytotoxic T cells, and macrophages. Moreover, MEX3B, MEX3C, and MEX3D expression correlated with key immune checkpoint genes, including PDCD1, CD274, and CTLA4. Functional enrichment analysis revealed that MEX3 co-expressed genes were significantly involved in RNA metabolism, immune response regulation, and oncogenic signaling pathways. A 17-gene MEX3 co-expression-based prognostic model stratified patients into high- and low-risk groups with significantly different survival outcomes (AUC = 0.791 at 1 year). This study highlights the oncogenic potential of MEX3 family members in HCC and their associations with immune regulation. The findings suggest that MEX3 family genes could serve as potential biomarkers for HCC prognosis and immunotherapy responsiveness. Further experimental validation is warranted to elucidate the mechanistic roles of MEX3 family genes in HCC progression and immune evasion.
Collapse
Affiliation(s)
- Xuezhong Zhang
- Department of Laboratory Medicine, Zibo Central Hospital, Zibo, Shandong, China
| | - Peng Sun
- Department of Gastroenterology, Zibo Central Hospital, Zibo, Shandong, China
| | - Tingting Chu
- Department of Laboratory Medicine, Zibo Central Hospital, Zibo, Shandong, China
| | - Yuling Feng
- Department of Infection Disease and Hepatology Ward, Zibo Central Hospital, Zibo, Shandong, China.
| | - Xuebin Zhang
- Department of Anorectal Surgery, Dongying People's Hospital (Dongying Hospital of Shandong Provincial Hospital Group), Dongying, China.
| |
Collapse
|
7
|
Hong Y, Wang D, Liu Z, Chen Y, Wang Y, Li J. Decoding per- and polyfluoroalkyl substances (PFAS) in hepatocellular carcinoma: a multi-omics and computational toxicology approach. J Transl Med 2025; 23:504. [PMID: 40317014 PMCID: PMC12049027 DOI: 10.1186/s12967-025-06517-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 04/18/2025] [Indexed: 05/04/2025] Open
Abstract
BACKGROUND Per- and polyfluoroalkyl substances (PFAS), particularly perfluorooctanoic acid (PFOA) and perfluorooctane sulfonate (PFOS), are synthetic chemicals known for their widespread use and environmental persistence. These compounds have been increasingly linked to hepatotoxicity and the development of hepatocellular carcinoma (HCC). However, the molecular mechanisms by which PFAS contribute to HCC remain underexplored. METHODS This study employs a multi-omics approach that combines network toxicology, integrated machine learning, single-cell RNA sequencing, spatial transcriptomics, experimental validation, and molecular docking simulations to uncover the mechanisms through which PFAS exposure drives HCC. We analyzed publicly available transcriptomic data from several HCC cohorts and used differential gene expression analysis to identify targets associated with both PFAS exposure and HCC. We constructed a protein-protein interaction (PPI) network and a survival risk model, the PFAS-related HCC signature (PFASRHSig), based on integrated machine learning to identify prognostic biomarkers, with the goal of identifying core targets of PFAS in HCC progression and prognosis. RT-qPCR and immunohistochemical (IHC) staining were used to validate the expression levels of the targets in both tumor and normal tissues. Molecular docking simulations were conducted to assess the binding affinities between PFAS compounds and selected target proteins. RESULTS Functional enrichment studies revealed that PFAS targets were associated with metabolic signaling pathways, which are actively involved in lipid, glucose, drug metabolism, etc. Through integrated machine learning and PPI network analysis, we identified six genes, APOA1, ESR1, IGF1, PPARGC1A, SERPINE1, and PON1, that serve as core targets of PFAS in both HCC progression and prognosis. These targets were further validated via bulk RNA-seq, single-cell RNA-seq, and spatial transcriptomics, which revealed differential expression patterns across various cell types in the HCC tumor microenvironment. The results of RT-qPCR and IHC staining were consistent with the in silico findings. Molecular docking simulations revealed strong binding affinities between PFAS compounds and these core targets, supporting their potential roles in PFAS-induced hepatocarcinogenesis. CONCLUSIONS Our study highlights key molecular targets and pathways involved in PFAS-induced liver carcinogenesis and proposes a robust survival risk model (PFASRHSig) for HCC. These findings provide new insights into PFAS toxicity mechanisms and offer potential therapeutic targets for mitigating the health risks associated with PFAS exposure. Collectively, our findings help in advancing clinical applications by providing insights into disease mechanisms and potential therapeutic interventions.
Collapse
Affiliation(s)
- Yanggang Hong
- The Second School of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Deqi Wang
- The First School of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Zeyu Liu
- The Second School of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yuxin Chen
- The First School of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yi Wang
- The First School of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Jiajun Li
- The Second School of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| |
Collapse
|
8
|
Wei Y, Mao D, Liu T, Wu W, Yang Z, Liu X. The predictive value of PIV, PLR, LMR, NPR, and NLR for the prognosis of transarterial chemoembolization in patients with hepatocellular carcinoma combined with liver cirrhosis. BMC Gastroenterol 2025; 25:315. [PMID: 40301803 PMCID: PMC12042468 DOI: 10.1186/s12876-025-03815-0] [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: 12/10/2024] [Accepted: 03/24/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Transarterial chemoembolization (TACE) is a primary treatment for hepatocellular carcinoma (HCC) in patients with liver cirrhosis. Prognostic markers that reliably predict outcomes in these patients post-TACE remain insufficiently defined. Systemic inflammatory markers such as the Pan-Immunological Value (PIV), Platelet-Lymphocyte Ratio (PLR), Lymphocyte-Monocyte Ratio (LMR), Neutrophil-Platelet Ratio (NPR), and Neutrophil-Lymphocyte Ratio (NLR) offer potential prognostic insights for various disease. This study aims to evaluate this markers to ascertain their predictive value in determining prognosis post-TACE. METHODS This retrospective study involved 216 patients with HCC and cirrhosis treated with TACE at a single hospital from May 2017 to May 2023. Patients were stratified into good (n = 92) and poor prognosis groups (n = 124) based on one-year post-operative outcomes using the Response Evaluation Criteria in Solid Tumors (RECIST). We evaluated preoperative inflammatory markers, biochemical and imaging data, and utilized univariate and multivariate logistic regression analyses to determine predictive factors for prognosis. RESULTS Patients in the poor prognosis group exhibited significantly higher PIV, PLR, NLR, and NPR, and lower LMR (P < 0.05). Multivariate analysis identified PIV and NPR as the strongest independent predictors of poor prognosis (OR: 1.021, P < 0.001 and OR: 2.909, P < 0.001, respectively). ROC analysis demonstrated that PIV had the greatest predictive accuracy (AUC = 0.803). CONCLUSION PIV, PLR, LMR, NPR, and NLR serve as significant prognostic markers for patients with HCC undergoing TACE in the context of liver cirrhosis.
Collapse
Affiliation(s)
- Yanan Wei
- Department of Oncology, The First Clinical Medical College, Three Gorges University/Yichang City Central People's Hospital, Yichang, 443000, China
| | - Dongting Mao
- Department of Clinical Laboratory, The First Clinical Medical College, Three Gorges University, Yichang City Central People's Hospital, Yichang, 443000, China
| | - Tiantian Liu
- Department of Oncology, The First Clinical Medical College, Three Gorges University/Yichang City Central People's Hospital, Yichang, 443000, China
| | - Wanyan Wu
- Department of Oncology, The First Clinical Medical College, Three Gorges University/Yichang City Central People's Hospital, Yichang, 443000, China
| | - Ziwei Yang
- Department of Oncology, The First Clinical Medical College, Three Gorges University/Yichang City Central People's Hospital, Yichang, 443000, China
| | - Xiuli Liu
- Department of Oncology, The First Clinical Medical College, Three Gorges University/Yichang City Central People's Hospital, Yichang, 443000, China.
| |
Collapse
|
9
|
Zhi Y, Guo Y, Li S, He X, Wei H, Laster K, Wu Q, Zhao D, Xie J, Ruan S, Lemoine NR, Li H, Dong Z, Liu K. FBL promotes hepatocellular carcinoma tumorigenesis and progression by recruiting YY1 to enhance CAD gene expression. Cell Death Dis 2025; 16:348. [PMID: 40289107 PMCID: PMC12034760 DOI: 10.1038/s41419-025-07684-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 04/13/2025] [Accepted: 04/17/2025] [Indexed: 04/30/2025]
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related death worldwide. Accumulating evidence suggests that epigenetic dysregulation contributes to the initiation and progression of HCC. We aimed to investigate key epigenetic regulators that contribute to tumorigenesis and progression, providing a theoretical basis for targeted therapy for HCC. We performed a comprehensive epigenetic analysis of differentially expressed genes in LIHC from the TCGA database. We identified fibrillarin (FBL), an rRNA 2'-O-methyltransferase, as an essential contributor to HCC. A series of in vitro and in vivo biological experiments were performed to investigate the potential mechanisms of FBL. FBL knockdown suppressed the proliferation of HCC cells. In vivo studies using cell-derived xenograft (CDX), patient-derived xenograft (PDX), and diethylnitrosamine (DEN)-induced HCC models in Fbl liver-specific knockout mice demonstrated the critical role of FBL in HCC carcinogenesis and progression. Mechanistically, FBL regulates the expression of CAD in HCC cells by recruiting YY1 to the CAD promoter region. We also revealed that fludarabine phosphate is a novel inhibitor of FBL and can inhibit HCC growth in vitro and in vivo. The antitumor activity of lenvatinib has been shown to be synergistically enhanced by fludarabine phosphate. Our study highlights the cancer-promoting role of the FBL-YY1-CAD axis in HCC and identifies fludarabine phosphate as a novel inhibitor of FBL. A schematic diagram depicting the FBL-YY1-CAD signaling pathway and its regulatory role in HCC progression.
Collapse
Affiliation(s)
- Yafei Zhi
- State Key Laboratory of Metabolic Dysregulation & Prevention and Treatment of Esophageal Cancer; The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou, China
- Innovation Center of Basic Research for Metabolic-Associated Fatty Liver Disease, Ministry of Education of China, Zhengzhou, China
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, China
- Cancer Chemistry International Collaboration Laboratory, Zhengzhou, China
| | - Yan Guo
- State Key Laboratory of Metabolic Dysregulation & Prevention and Treatment of Esophageal Cancer; The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| | - Shiliang Li
- Innovation Center for AI and Drug Discovery, East China Normal University, Shanghai, China
| | - Xinyu He
- State Key Laboratory of Metabolic Dysregulation & Prevention and Treatment of Esophageal Cancer; The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| | - Huifang Wei
- State Key Laboratory of Metabolic Dysregulation & Prevention and Treatment of Esophageal Cancer; The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| | - Kyle Laster
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| | - Qiong Wu
- State Key Laboratory of Metabolic Dysregulation & Prevention and Treatment of Esophageal Cancer; The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| | - Dengyun Zhao
- State Key Laboratory of Metabolic Dysregulation & Prevention and Treatment of Esophageal Cancer; The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| | - Jinxin Xie
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Shanshan Ruan
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Nicholas R Lemoine
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy; The School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China.
- Center for Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, UK.
| | - Honglin Li
- Innovation Center for AI and Drug Discovery, East China Normal University, Shanghai, China.
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China.
- Lingang Laboratory, Shanghai, China.
| | - Zigang Dong
- State Key Laboratory of Metabolic Dysregulation & Prevention and Treatment of Esophageal Cancer; The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China.
- Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou, China.
- Innovation Center of Basic Research for Metabolic-Associated Fatty Liver Disease, Ministry of Education of China, Zhengzhou, China.
- Cancer Chemistry International Collaboration Laboratory, Zhengzhou, China.
| | - Kangdong Liu
- State Key Laboratory of Metabolic Dysregulation & Prevention and Treatment of Esophageal Cancer; The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China.
- Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou, China.
- Innovation Center of Basic Research for Metabolic-Associated Fatty Liver Disease, Ministry of Education of China, Zhengzhou, China.
- Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, China.
- Cancer Chemistry International Collaboration Laboratory, Zhengzhou, China.
| |
Collapse
|
10
|
Ji K, Chen G, Wang Y, Li Y, Chen J, Feng M. YEATS2: a novel cancer epigenetic reader and potential therapeutic target. Cancer Cell Int 2025; 25:162. [PMID: 40287757 PMCID: PMC12034173 DOI: 10.1186/s12935-025-03797-9] [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: 09/28/2024] [Accepted: 04/21/2025] [Indexed: 04/29/2025] Open
Abstract
YEATS2, an evolutionarily conserved reader of histone acylation marks (H3K27ac, H3K27cr, H3K27bz), functions as a central oncogenic driver in diverse cancers, including non-small cell lung cancer (NSCLC), pancreatic ductal adenocarcinoma (PDAC), and hepatocellular carcinoma (HCC). Its structurally plastic YEATS domain bridges acyl-CoA metabolism to chromatin remodeling, amplifying transcription of survival genes such as MYC, BCL2, and PD-L1. YEATS2 orchestrates malignancy-specific programs-sustaining ribosome biogenesis in NSCLC through ATAC complex recruitment, enhancing NF-κB-dependent immune evasion in PDAC, and activating PI3K/AKT-driven metabolic rewiring in HCC. Structural studies demonstrate a unique aromatic cage architecture that selectively engages diverse acylated histones. Although pyrazolopyridine-based inhibitors targeting the YEATS domain show preclinical efficacy, developing isoform-selective agents remains challenging. Clinically, YEATS2 overexpression correlates with therapy resistance and may synergize with immune checkpoint blockade. This review integrates mechanistic insights into the role of YEATS2 in epigenetic regulation, evaluates its therapeutic potential, and proposes future directions: elucidating full-length complex topologies, mapping synthetic lethal interactors, and optimizing selective inhibitors. Disrupting YEATS2-mediated epigenetic adaptation presents novel opportunities for precision cancer therapy.
Collapse
Affiliation(s)
- Kangkang Ji
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
- Department of Clinical Medical Research, Binhai County People's Hospital, Clinical Medical College of Yangzhou University, Yancheng, 224500, Jiangsu, China
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Guoping Chen
- Department of Clinical Medical Research, Binhai County People's Hospital, Clinical Medical College of Yangzhou University, Yancheng, 224500, Jiangsu, China
| | - Yan Wang
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Yunyi Li
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Jian Chen
- Department of Head and Neck Surgery, Tongji Medical College, Hubei Cancer Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, 430070, China.
| | - Mingqian Feng
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, Hubei, 430070, China.
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China.
| |
Collapse
|
11
|
Cao L, Bi W. METTL16/IGF2BP2 axis enhances malignant progression and DDP resistance through up-regulating COL4A1 by mediating the m6A methylation modification of LAMA4 in hepatocellular carcinoma. Cell Div 2025; 20:9. [PMID: 40251670 PMCID: PMC12008873 DOI: 10.1186/s13008-025-00152-2] [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: 12/31/2024] [Accepted: 04/08/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the third most common malignant tumor after gastric cancer and esophageal cancer, which is a serious threat to human health. Methyltransferase-like protein 16 (METTL16) regulates the occurrence and development of various cancers, but its molecular mechanism in HCC has not been fully investigated. METHODS A series of databases were used to predict gene expression, methylation sites, correlation analysis, and protein interaction analysis. Gene expression levels were detected by quantitative real-time polymerase chain reaction (qRT-PCR), western blot, and immunohistochemistry (IHC). What's more, drug-resistant cell lines were established for drug resistance analysis. Cell proliferation was measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and 5-ethynyl-2'-deoxyuridine (EdU) staining. Flow cytometry, transwell and wound healing assays were used for apoptosis, invasion and migration, respectively. In addition, the regulatory mechanism of METTL16 in HCC was investigated by methylated RNA immunoprecipitation (MeRIP), RNA immunoprecipitation (RIP) and co-immunoprecipitation (Co-IP). Finally, constructing subcutaneous transplanted tumor in nude mice confirmed the effect of METTL16 in vivo. RESULTS METTL16 was up-regulated in HCC drug-resistant tissues and cells. Knockdown of METTL16 inhibited Cisplatin (DDP) resistance, proliferation, invasion and migration of HCC cells, but promoted apoptosis. Besides, laminin subunit alpha 4 (LAMA4), which was overexpressed in HCC drug-resistant tissues and cells, was selected as the target of METTL16. Mechanistically, METTL16 and m6A reader insulin like growth factor 2 mRNA binding protein 2 (IGF2BP2) co-regulated the m6A modification and mRNA stability of LAMA4, and LAMA4 weakened the effects of METTL16 knockdown on HCC drug-resistance. Meanwhile, LAMA4 bound to collagen type IV alpha 1 chain (COL4A1) and facilitated DDP resistance and HCC progression via COL4A1. Similarly, in vivo, METTL16 induced tumor growth, as well as LAMA4 and COL4A1 expression, and increased DDP resistance. CONCLUSION METTL16 and IGF2BP2 jointly mediated the m6A methylation modification of LAMA4, thereby promoting DDP resistance and malignant progression of HCC through regulation of COL4A1.
Collapse
Affiliation(s)
- Liming Cao
- Department of General Surgery, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang, 050000, China
| | - Wei Bi
- Department of General Surgery, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang, 050000, China.
| |
Collapse
|
12
|
Deng H, Wu D, He Y, Yu X, Liu J, Zhang Y, Leng B, Yuan X, Xiao L. E2F1-driven EXOSC10 transcription promotes hepatocellular carcinoma growth and stemness: a potential therapeutic target. Hereditas 2025; 162:60. [PMID: 40221814 PMCID: PMC11992873 DOI: 10.1186/s41065-025-00430-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND E2F Transcription Factor 1 (E2F1) is a transcription factor that plays a crucial role in the growth of many cancers, including hepatocellular carcinoma (HCC). Herein, this study probed the functions and underlying mechanisms of E2F1 in HCC tumorigenesis. METHODS The expression profiles of E2F1 and Exosome Component 10 (EXOSC10) were detected using qRT-PCR and western blotting. Functional experiments were carried out using 5-ethynyl-2'-deoxyuridine (EdU), flow cytometry, tube formation, and sphere formation assays in vitro, as well as xenograft experiments in vivo, respectively. Stemness-related proteins were assayed using western blotting. The interaction between E2F1 and EXOSC10 was verified using bioinformatics analysis and dual-luciferase reporter assay. RESULTS E2F1 was highly expressed in HCC tissues and cells, and was associated with advanced TNM stage, distant metastasis, and short survival rate. Functionally, knockdown of E2F1 suppressed HCC cell proliferation, angiogenesis, and stemness, and induced cell apoptosis. Mechanistically, E2F1 directly bound to the promoter region of EXOSC10 to up-regulate its expression. EXOSC10 silencing impaired HCC cell proliferation, angiogenesis, and stemness. Moreover, the anticancer effects of E2F1 knockdown were reversed by EXOSC10 elevation. In vivo assay, E2F1 deficiency suppressed HCC tumor growth and eliminated cancer stemness, while these effects were abolished by EXOSC10 up-regulation. CONCLUSION E2F1 promotes EXOSC10 transcription and then facilitates HCC growth and cancer stemness, revealing a potential target for HCC therapy.
Collapse
Affiliation(s)
- Haoyue Deng
- Department of Pathology, Suining Central Hospital, Suining, 629000, Sichuan, China
| | - Dingyong Wu
- Department of Oncology, Songshan General Hospital, Chongqing, 401120, China
| | - Yongpeng He
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Inaffiliationidualized Treatment, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, 400030, China
| | - Xiaolei Yu
- Department of Oncology, Fengning Manchu Autonomous County Hospital, No.737 Binhe Road, Chengde, 067000, Hebei, China
| | - Jifei Liu
- Department of Oncology, Fengning Manchu Autonomous County Hospital, No.737 Binhe Road, Chengde, 067000, Hebei, China
| | - Yanrui Zhang
- Department of Oncology, Fengning Manchu Autonomous County Hospital, No.737 Binhe Road, Chengde, 067000, Hebei, China
| | - Bing Leng
- Department of Oncology, Fengning Manchu Autonomous County Hospital, No.737 Binhe Road, Chengde, 067000, Hebei, China
| | - Xiaofeng Yuan
- Department of Oncology, Fengning Manchu Autonomous County Hospital, No.737 Binhe Road, Chengde, 067000, Hebei, China.
| | - Liguo Xiao
- Department of Oncology, Fengning Manchu Autonomous County Hospital, No.737 Binhe Road, Chengde, 067000, Hebei, China.
| |
Collapse
|
13
|
Sun T, Geng S, Ru Q, Zheng Y. METTL3 and HERC4: Elevated Expression and Impact on Hepatocellular Carcinoma Progression. Cancer Biother Radiopharm 2025; 40:173-184. [PMID: 39611657 DOI: 10.1089/cbr.2024.0126] [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] [Indexed: 11/30/2024] Open
Abstract
Background: Methyltransferase-like 3 (METTL3) and HECT and RLD domain containing E3 ubiquitin protein ligase 4 (HERC4) have been studied in the field of oncology; however, their roles and interaction in hepatocellular carcinoma (HCC) await elucidation. Methods: Initially, METTL3 and HERC4 expressions in normal and HCC samples were predicted employing the UALCAN database, and the targeting relationship between these two was explored via coimmunoprecipitation assay. Following the quantification on N6-methyladenosine (m6A) enrichment, the localization of METTL3 and HERC4 on HCC cells was visualized via immunofluorescence assay. The effects of METTL3 and HERC4 on HCC cells proliferation and migration were determined in vitro assays. METTL3 and HERC4 expressions were quantified via quantitative polymerase chain reaction, and those of metastasis-related proteins N-cadherin and vimentin were calculated with immunoblotting assay. Furthermore, the levels of angiogenic factors such as vascular endothelial growth factor and basic fibroblast growth factor were measured by enzyme-linked immunosorbent assay. Results: METTL3 and HERC4 expressed highly in HCC and their expressions were positively correlated with tumor grade. METTL3 overexpression enhanced the expression of HERC4 and promoted the proliferation and migration abilities of HCC cells. Specifically, METTL3 overexpression increased vimentin and N-cadherin expressions, while its silencing did conversely. Besides, HERC4 overexpression reversed the effects of METTL3 silencing on the proliferation and migration as well as the levels of angiogenic factors in HCC cells. Conclusion: This study reveals the upregulation of METTL3 and HERC4 expression in HCC and their role in HCC by enhancing the proliferation, migration, and angiogenesis potential of HCC cells.
Collapse
Affiliation(s)
- Tao Sun
- Department of Infectious Disease, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Shiyu Geng
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qingjing Ru
- Department of Infectious Disease, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yi Zheng
- Department of Infectious Disease, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
14
|
Huang J, Xie H, Li J, Huang X, Cai Y, Yang R, Yang D, Bao W, Zhou Y, Li T, Lu Q. Histone lactylation drives liver cancer metastasis by facilitating NSF1-mediated ferroptosis resistance after microwave ablation. Redox Biol 2025; 81:103553. [PMID: 39970777 PMCID: PMC11876915 DOI: 10.1016/j.redox.2025.103553] [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: 02/06/2025] [Accepted: 02/15/2025] [Indexed: 02/21/2025] Open
Abstract
Insufficient microwave ablation (IMWA) is linked to aggressive hepatocellular carcinoma (HCC) progression. An increase in lactate levels after sublethal heat stress (HS) has been confirmed in HCC. However, the role of lactate-related histone lactylation in the progression of HCC caused by sublethal HS remains unclear. Here, we found that the metastatic potential of HCC increased in a lactate-dependent manner after IMWA. Moreover, sublethal HS triggered an increase in H3K18la modification, as validated in a cell-derived xenograft mouse model and human HCC samples. By performing an integrated analysis of proteomic and transcriptomic profiles, we revealed that HCC cells exhibited increased intracellular iron ion homeostasis and developed resistance to platinum-based drugs after exposure to sublethal HS. We subsequently integrated proteomic and transcriptomic data with H3K18la-specific chromatin immunoprecipitation (ChIP) sequencing to identify candidate genes involved in sublethal heat treatment-induced HCC cell metastasis. Mechanically, an increase in H3K18la modification enhanced the transcriptional activity of NFS1 cysteine desulfurase (NFS1), a key player in iron‒sulfur cluster biosynthesis, thereby reducing the susceptibility of HCC to ferroptosis after IMWA. Knocking down NFS1 diminished the metastatic potential of sublethally heat-treated HCC cells. Additionally, NFS1 deficiency exhibited a synergistic effect with oxaliplatin, leading to the significant inhibition of the metastatic capability of HCC cells both in vitro and in vivo, regardless of sublethal HS treatment. In conclusion, our study revealed the oncogenic role of histone lactylation in HCC after IMVA. We also bridged histone lactylation with ferroptosis, providing novel therapeutic targets for HCC following microwave ablation, particularly when combined with oxaliplatin-based chemotherapy.
Collapse
Affiliation(s)
- Jiayan Huang
- Department of Ultrasound, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Huijing Xie
- Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Ju Li
- Laboratory of Ultrasound Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Xiaotong Huang
- Department of Ultrasound, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Yunshi Cai
- Liver Transplant Center, Transplant Center, West China Hospital, Sichuan University, Chengdu, 610041, China; Laboratory of Hepatic AI Translation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Rui Yang
- Department of Ultrasound, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Dongmei Yang
- Department of Ultrasound, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Wuyongga Bao
- Department of Ultrasound, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Yongjie Zhou
- Department of Liver Transplantation Center & Laboratory of Liver Transplantation, West China Hospital of Sichuan University, Chengdu, 641400, China
| | - Tao Li
- Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, 610041, China.
| | - Qiang Lu
- Department of Ultrasound, West China Hospital of Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
15
|
Yuan W, Lu G, Zhao Y, He X, Liao S, Wang Z, Lei X, Xie Z, Yang X, Tang S, Tang G, Deng X. Intranuclear TCA and mitochondrial overload: The nascent sprout of tumors metabolism. Cancer Lett 2025; 613:217527. [PMID: 39909232 DOI: 10.1016/j.canlet.2025.217527] [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: 11/27/2024] [Revised: 01/19/2025] [Accepted: 02/02/2025] [Indexed: 02/07/2025]
Abstract
Abnormal glucose metabolism in tumors is a well-known form of metabolic reprogramming in tumor cells, the most representative of which, the Warburg effect, has been widely studied and discussed since its discovery. However, contradictions in a large number of studies and suboptimal efficacy of drugs targeting glycolysis have prompted us to further deepen our understanding of glucose metabolism in tumors. Here, we review recent studies on mitochondrial overload, nuclear localization of metabolizing enzymes, and intranuclear TCA (nTCA) in the context of the anomalies produced by inhibition of the Warburg effect. We provide plausible explanations for many of the contradictory points in the existing studies, including the causes of the Warburg effect. Furthermore, we provide a detailed prospective discussion of these studies in the context of these new findings, providing new ideas for the use of nTCA and mitochondrial overload in tumor therapy.
Collapse
Affiliation(s)
- Weixi Yuan
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Guozhong Lu
- 922nd Hospital of Hengyang, 421001, Hunan, China
| | - Yin Zhao
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xiang He
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Senyi Liao
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Zhe Wang
- The Second Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Xiaoyong Lei
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; Department of Pharmacy, Xiangnan University, Chenzhou, 423000, China
| | - Zhizhong Xie
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xiaoyan Yang
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China; Department of Pharmacy, Xiangnan University, Chenzhou, 423000, China
| | - Shengsong Tang
- Hunan Province Key Laboratory for Antibody-based Drug and Intelligent Delivery Systems (2018TP1044), Hunan, 410007, China.
| | - Guotao Tang
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Xiangping Deng
- The First Affiliated Hospital, Department of Pharmacy, Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
16
|
Peng J, Liu W, Tian J, Shu Y, Zhao R, Wang Y. Non-coding RNAs as key regulators of epithelial-mesenchymal transition in breast cancer. Front Cell Dev Biol 2025; 13:1544310. [PMID: 40201201 PMCID: PMC11975958 DOI: 10.3389/fcell.2025.1544310] [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: 12/13/2024] [Accepted: 03/06/2025] [Indexed: 04/10/2025] Open
Abstract
This study examines the critical role of non-coding RNAs (ncRNAs) in regulating epithelial-mesenchymal transition (EMT) in breast cancer, a prevalent malignancy with significant metastatic potential. EMT, wherein cancer cells acquire mesenchymal traits, is fundamental to metastasis. ncRNAs-such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs)-modulate EMT by influencing gene expression and signaling pathways, affecting cancer cell migration and invasion. This review consolidates recent findings on ncRNA-mediated EMT regulation and explores their diagnostic and therapeutic potential. Specifically, miRNAs inhibit EMT-related transcription factors, while lncRNAs and circRNAs regulate gene expression through interactions with miRNAs, impacting EMT progression. Given the influence of ncRNAs on metastasis and therapeutic resistance, advancing ncRNA-based biomarkers and treatments holds promise for improving breast cancer outcomes.
Collapse
Affiliation(s)
- Jing Peng
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Wenhui Liu
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Jiaju Tian
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yuncong Shu
- School of life science, Lanzhou University, Lanzhou, China
| | - Rui Zhao
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yuping Wang
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
17
|
Ye Y, Zeng Y, Huang S, Zhu C, Wang Q. A Chemotherapy Response-Related Gene Signature and DNAJC8 as Key Mediators of Hepatocellular Carcinoma Progression and Drug Resistance. J Hepatocell Carcinoma 2025; 12:579-595. [PMID: 40130083 PMCID: PMC11932135 DOI: 10.2147/jhc.s506706] [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: 11/25/2024] [Accepted: 02/21/2025] [Indexed: 03/26/2025] Open
Abstract
Background Chemotherapy resistance in hepatocellular carcinoma presents a significant challenge to improved patient outcomes. Identifying genes associated with chemotherapy response can enhance treatment strategies and prognostic models. Methods We analyzed the expression of chemotherapy response-related gene in hepatocellular carcinoma using TCGA and GSE109211 cohorts. We constructed a prognostic model using Least Absolute Shrinkage and Selection Operator (LASSO) analysis and assessed its efficacy using Kaplan-Meier survival analysis. Additionally, we evaluated the immune landscape and gene mutation profiles between different chemotherapy response-related gene (CRRG) subtypes. DNAJC8's role in hepatocellular carcinoma cell functions and chemotherapy resistance was further explored through gene knockdown experiments in vitro and in vivo. Results Differential expression analysis identified 220 common genes associated with chemotherapy response. The prognostic model incorporating seven key genes efficiently distinguished responders from non-responders and indicated poorer overall survival for the CRRG-high subtype. The CRRG value correlated with tumor stage and grade, and mutation profiles showed distinct patterns between CRRG subtypes. The CRRG-high subtype exhibited an immune-suppressive phenotype with higher expression of PD-L1 and CTLA-4. High DNAJC8 expression was linked to poor prognosis in multiple cohorts. Knocking down DNAJC8 significantly inhibited hepatocellular carcinoma cell proliferation, migration, invasion, and reduced sorafenib IC50. Conclusion The seven-gene CRRG model, particularly DNAJC8, holds potential for predicting chemotherapy response and serves as a therapeutic target in hepatocellular carcinoma.
Collapse
Affiliation(s)
- Yan Ye
- Ganzhou Key Laboratory of Molecular Medicine, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, People’s Republic of China
| | - Yanmei Zeng
- Ganzhou Key Laboratory of Molecular Medicine, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, People’s Republic of China
| | - Shenggang Huang
- Ganzhou Key Laboratory of Molecular Medicine, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, People’s Republic of China
- Department of Gastroenterology, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, People’s Republic of China
| | - Chunping Zhu
- Ganzhou Key Laboratory of Molecular Medicine, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, People’s Republic of China
- Department of Gastroenterology, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, People’s Republic of China
| | - Qingshui Wang
- College of Integrative Medicine, Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, People’s Republic of China
| |
Collapse
|
18
|
Chen J, Liu Y, Wang Z, Zhang B, Feng Y. Nanocarriers for the delivery of plant-extracted camptothecin derivatives and hepatocellular carcinoma treatment. Nat Prod Res 2025:1-12. [PMID: 40102036 DOI: 10.1080/14786419.2025.2479249] [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: 11/09/2024] [Revised: 02/21/2025] [Accepted: 03/09/2025] [Indexed: 03/20/2025]
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related deaths worldwide and a major cause of death in cirrhosis. The prognosis of HCC is poor, with a mortality rate close to the morbidity rate. Once HCC develops distant metastasis, the area affected by the cancer cells is significantly enlarged, and there is still no effective treatment for HCC. Therefore, the development of effective drugs is essential to improve the survival rate of HCC patients. We designed and optimised a delivery system for compound 1, derived from camptothecin extract, by developing a multifunctional fluorescent drug delivery platform composed of polylactic acid (PLA), chitosan (CS), and fluorescein isothiocyanate (FITC) (PLA-CS-FITC). This system successfully encapsulated CP1 and compound 1, forming a PLA-CS-FITC@CP1@1 composite nanocarrier with dual functions for drug delivery and real-time fluorescence monitoring. The effects of the system on HCC proliferation and its regulation were evaluated by treating HCC cells in vitro, which provided an experimental basis for the development of drugs for HCC.
Collapse
Affiliation(s)
- Jie Chen
- Hepatobiliary Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yanfeng Liu
- Hepatobiliary Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zelin Wang
- Hepatobiliary Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bingyuan Zhang
- Hepatobiliary Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yujie Feng
- Hepatobiliary Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
19
|
Liang X, Yang H, Hu P, Gan Z, Long S, Wang S, Yang X. Decoding the possible mechanism of action of Paeoniflorigenone in combating Aflatoxin B1-induced liver cancer: an investigation using network pharmacology and bioinformatics analysis. Toxicol Mech Methods 2025; 35:292-304. [PMID: 39350351 DOI: 10.1080/15376516.2024.2411621] [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: 04/10/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/11/2024]
Abstract
Moutan cortex has demonstrated antitumor properties attributed to its bioactive compound Paeoniflorigenone (PA). Nevertheless, there is limited research on the efficacy of PA in the prevention and treatment of hepatocellular carcinoma (HCC). We aimed to investigate the potential pharmacological mechanisms of PA in the treatment of Aflatoxin B1 (AFB1)-induced hepatocarcinogenesis using network pharmacology and bioinformatics analysis approaches. Through various databases and bioinformatics analysis approaches, 34 shared targets were identified as potential candidate genes for PA in fighting liver cancer caused by AFB1. Pathway analysis revealed involvement in cell cycle, HIF-1, and Rap1 pathways. A risk assessment model was developed using LASSO regression, showing an association between the identified genes and the tumor immune microenvironment. The genes within the risk model were found to be linked to the immune response in liver cancer. Molecular docking studies indicated that PA interacts with its targets through hydrogen bonding and hydrophobic interactions. This study provides insights into the possible mechanisms of PA in liver cancer treatment and offers a predictive model for assessing the risk level of individuals with liver cancer. These findings have significant implications for the therapeutic strategies in managing liver cancer patients.
Collapse
Affiliation(s)
- Xiaocong Liang
- Interventional Department, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
| | - Huiling Yang
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
| | - Pengrong Hu
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
| | - Ziyan Gan
- Oncology Department, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
| | - Shunqin Long
- Oncology Department, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
| | - Sumei Wang
- Oncology Department, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
| | - Xiaobing Yang
- Oncology Department, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
| |
Collapse
|
20
|
Yuan Y, Huang J, Wei G, Hu G, Yu H, Tao Y. Potential mechanism of circKIAA1429 accelerating the progression of hepatocellular carcinoma. Infect Agent Cancer 2025; 20:12. [PMID: 40025575 PMCID: PMC11872318 DOI: 10.1186/s13027-025-00645-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 02/14/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND This study investigates the underlying mechanism of circKIAA1429 (hsa_circ_0084922) in hepatocellular carcinoma (HCC) progression. METHODS circKIAA1429, SETD1A, NAP1L3, and GLIS2 expressions in HCC cells were detected by RT-qPCR or western blot. The stability of circKIAA1429 was tested after treatment with actinomycin D and Rnase R enzyme. circKIAA1429 expression was knocked down, followed by detection of cell proliferation, apoptosis, and migration/invasion using CCK-8, flow cytometry, and transwell. RIP and RNA pull-down were performed to validate the binding between circKIAA1429 and SETD1A, while ChIP analysis determined the enrichment of SETD1A and H3K4me3 or H3K27me3 on GLIS2 or NAP1L3 promoter. A nude mouse xenograft tumor model was establish to test the effect of circKIAA1429 on tumorigenicity. RESULTS circKIAA1429 and NAP1L3 were highly expressed in HCC cells, while GLIS2 was poorly expressed. Knockdown of circKIAA1429 repressed cell proliferation/invasion/migration and facilitated apoptosis. Mechanistically, circKIAA1429 directly interacted with SETD1A to reduce the enrichment of SETD1A and H3K4me3 or H3K27me3 on GLIS2 or NAP1L3 promoter, thus diminishing GLIS2 expression and elevating NAP1L3 expression. In vivo, circKIAA1429 promotes tumorigenesis via GLIS2/NAP1L3. CONCLUSION circKIAA1429 interacts with SETD1A to inhibit the enrichment of H3K4me3 and H3K27me3 on GLIS2 or NAP1L3 promoter, thus inhibiting/promoting the expression of GLIS2/NAP1L3 and accelerating the progression of HCC.
Collapse
Affiliation(s)
- Yiting Yuan
- Department of General Surgery, The First People's Hospital of Tongxiang, No. 1918, Xiaochang East Road, Wutong Street, Tongxiang City, Zhejiang Province, 314500, PR China
| | - Junwei Huang
- Department of General Surgery, The First People's Hospital of Tongxiang, No. 1918, Xiaochang East Road, Wutong Street, Tongxiang City, Zhejiang Province, 314500, PR China
| | - Guifen Wei
- Department of General Surgery, The First People's Hospital of Tongxiang, No. 1918, Xiaochang East Road, Wutong Street, Tongxiang City, Zhejiang Province, 314500, PR China
| | - Guang Hu
- Department of General Surgery, The First People's Hospital of Tongxiang, No. 1918, Xiaochang East Road, Wutong Street, Tongxiang City, Zhejiang Province, 314500, PR China
| | - Hongmei Yu
- Department of General Surgery, The First People's Hospital of Tongxiang, No. 1918, Xiaochang East Road, Wutong Street, Tongxiang City, Zhejiang Province, 314500, PR China
| | - Yiming Tao
- Department of General Surgery, The First People's Hospital of Tongxiang, No. 1918, Xiaochang East Road, Wutong Street, Tongxiang City, Zhejiang Province, 314500, PR China.
| |
Collapse
|
21
|
Haghir-Sharif-Zamini Y, Khosravi A, Hassan M, Zarrabi A, Vosough M. c-FLIP/Ku70 complex; A potential molecular target for apoptosis induction in hepatocellular carcinoma. Arch Biochem Biophys 2025; 765:110306. [PMID: 39818348 DOI: 10.1016/j.abb.2025.110306] [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: 11/18/2024] [Revised: 01/12/2025] [Accepted: 01/13/2025] [Indexed: 01/18/2025]
Abstract
Hepatocellular carcinoma (HCC) is one of the most lethal malignancies worldwide and the most common form of liver cancer. Despite global efforts toward early diagnosis and effective treatments, HCC is often diagnosed at advanced stages, where conventional therapies frequently lead to resistance and/or high recurrence rates. Therefore, novel biomarkers and promising medications are urgently required. Epi-drugs, or epigenetic-based medicines, have recently emerged as a promising therapeutic modality. Since the epigenome of the cancer cells is always dysregulated and this is followed by apoptosis-resistance, reprogramming the epigenome of cancer cells by epi-drugs (such as HDAC inhibitors (HDACis), and DNMT inhibitors (DNMTis)) could be an alternative approach to use in concert with established treatment protocols. C-FLIP, an anti-apoptotic protein, and Ku70, a member of the DNA repair system, bind together and make a cytoplasmic complex in certain cancers and induce resistance to apoptosis. Many epi-drugs, such as HDACis, can dissociate this complex through Ku70 acetylation and activate cellular apoptosis. The novel compounds for dissociating this complex could provide an innovative insight into molecular targeted HCC treatments. In this review, we address the innovative therapeutic potential of targeting c-FLIP/Ku70 complex by epi-drugs, particularly HDACis, to overcome apoptosis resistance of HCC cells. This review will cover the mechanisms by which the c-FLIP/Ku70 complex facilitates cancer cell survival, the impact of epigenetic alterations on the complex dissociation, and highlight HDACis potential in combination therapies, biomarker developments and mechanistic overviews. This review highlights c-FLIP ubiquitination and Ku70 acetylation levels as diagnostic and prognostic tools in HCC management.
Collapse
Affiliation(s)
- Yasamin Haghir-Sharif-Zamini
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Arezoo Khosravi
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Istanbul Okan University, Istanbul, 34959, Turkiye
| | - Moustapha Hassan
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul, 34396, Turkiye; Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan, 320315, Taiwan; Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai - 600 077, India.
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
22
|
Gao H, Fan L, Gai X, Fu R, Li G, Jing K, Xu J, Sun S. LINC01004/hsa-mir-125b-2-3p axis restrains ferroptosis in hepatocellular carcinoma by targeting HSPA4 via ceRNA mechanism. Technol Health Care 2025; 33:959-973. [PMID: 40105158 DOI: 10.1177/09287329241291430] [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] [Indexed: 03/20/2025]
Abstract
BackgroundHepatocellular carcinoma (HCC) is a primary cancer, accounting for 90% of primary liver cancer, mainly occurring in patients with cirrhosis and chronic liver disease.ObjectiveTo investigate the latent mechanisms of hepatocellular carcinoma (HCC) and find therapeutic targets.MethodsDifferentially expressed and overall survival related genes of HCC, and cell death related genes were intersected to obtain latent target genes. These genes were analyzed using ROC curve for diagnosing HCC. RT-qPCR and Western blot were performed to detect the expression level of genes. Wound healing tests were performed with or without si-HSPA4. Potential ceRNA axis was forecasted using TargetScan and miRanda and the dual luciferase reporter gene assay was used to verify the results. Finally, the images of H&E dye liquor-stained HCC tissue section, the CT images for patients in different tumor stage.ResultsLINC01004/hsa-miR-125b-2-3p/HSPA4 axis was forecasted and then was verified using dual-luciferase reporter assay. HSPA4 knockdown caused significant reduction of cell proliferation and ferroptosis. Si-HSPA4 related ferroptosis was generated through impairing iron transport via targeting restrain GPX4. For human subjects, the RT-qPCR analysis revealed the that the larger the tumor diameter, the higher the LINC01004, HSPA4, and GPX4 expression, and the lower the hsa-miR-125b-2-3p expression.ConclusionLINC01004/hsa-miR-125b-2-3p/HSPA4 regulatory axis involved in the ferroptosis of the progression of HCC via GPX4 dependent method, providing new therapeutic targets for HCC patients.
Collapse
Affiliation(s)
- Han Gao
- Department of Biochemistry and Molecular Biology, Qiqihar Medical University, Qiqihar, China
| | - Li Fan
- Research Institute of Medicine and Pharmacy, Qiqihar Medical University, Qiqihar, China
| | - Xue Gai
- Department of Imaging, First Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Rong Fu
- Departmentof Oncology, First Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Guohua Li
- Department of Imaging, First Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Kexin Jing
- Department of Imaging, First Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Jingwei Xu
- General Surgery, First Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Shengjian Sun
- Department of Imaging, First Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| |
Collapse
|
23
|
Nagaraju GP, Sandhya T, Srilatha M, Ganji SP, Saddala MS, El-Rayes BF. Artificial intelligence in gastrointestinal cancers: Diagnostic, prognostic, and surgical strategies. Cancer Lett 2025; 612:217461. [PMID: 39809357 DOI: 10.1016/j.canlet.2025.217461] [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: 11/05/2024] [Revised: 12/12/2024] [Accepted: 01/11/2025] [Indexed: 01/16/2025]
Abstract
GI (Gastrointestinal) malignancies are one of the most common and lethal cancers globally. The dawn of precision medicine and developing technologies have reduced the mortality rates for GI malignancies, underscoring the main role of early detection methods for survival rate improvement. Artificial intelligence (AI) is a new technology that may improve GI cancer screening, treatment, and therapeutic efficiency for better patient care. AI could accelerate the development of targeted therapies by analyzing considerable data from the genome and identifying biomarkers connected with GI tumors. This opens up new avenues toward more tailored and personalized approaches, raising efficacy while reducing undesired side effects. For instance, AI may improve treatment outcomes by accurately predicting patient responses to therapeutic regimens, helping oncologists choose the most effective treatment options. This review will outline the transformative potential of AI in GI oncology by emphasizing the incorporation of AI-based technologies to enhance patient care.
Collapse
Affiliation(s)
- Ganji Purnachandra Nagaraju
- School of Medicine, Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Tatekalva Sandhya
- Department of Computer Science, Sri Venkateswara University, Tirupati, 517502, AP, India
| | - Mundla Srilatha
- Department of Biotechnology, Sri Venkateswara University, Tirupati, 517502, AP, India
| | - Swapna Priya Ganji
- School of Medicine, Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Madhu Sudhana Saddala
- Bioinformatics, Genomics and Proteomics, University of California, Irvine, Los Angeles, 92697, USA
| | - Bassel F El-Rayes
- School of Medicine, Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
| |
Collapse
|
24
|
Jian Y, Li Y, Zhou Y, Mu W. Pollutants in Microenvironmental Cellular Interactions During Liver Inflammation Cancer Transition and the Application of Multi-Omics Analysis. TOXICS 2025; 13:163. [PMID: 40137490 PMCID: PMC11945810 DOI: 10.3390/toxics13030163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/18/2025] [Accepted: 02/25/2025] [Indexed: 03/29/2025]
Abstract
This study categorizes pollutant-induced inflammation-cancer transition into three stages: non-alcoholic fatty liver disease (NAFLD), liver fibrosis, and hepatocellular carcinoma (HCC). It systematically reveals the temporal heterogeneity of pollutant-induced liver damage. The findings indicate that pollutants not only directly damage hepatocytes but also modulate key cells in the immune microenvironment, such as hepatic stellate cells (HSCs) and Kupffer cells, thereby amplifying inflammatory and fibrotic responses, ultimately accelerating the progression of HCC. Mechanistically, in the early stage (NAFLD), pollutants primarily cause hepatocyte injury through oxidative stress and lipid metabolism dysregulation. During the fibrosis stage, pollutants promote liver fibrosis by inducing extracellular matrix accumulation, while in the HCC stage, they drive tumorigenesis via activation of the Wnt/β-catenin pathway and p53 inactivation. Through multi-omics analyses, this study identifies critical pathogenic molecules and signaling pathways regulated by pollutants, providing new insights into their pathogenic mechanisms, potential biomarkers, and therapeutic targets. These findings offer valuable guidance for the development of diagnostic and therapeutic strategies for liver diseases and the formulation of environmental health risk prevention measures.
Collapse
Affiliation(s)
| | | | | | - Wei Mu
- School of Public Health, Center for Single-Cell Omics, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (Y.J.); (Y.L.); (Y.Z.)
| |
Collapse
|
25
|
Wei C, Chen J, Huang T, Zhou L, Xu Y, Lin Q, Qin Y, Tang Z, Yang W, Fang M. The microRNA landscape and regulatory network in Clonorchis sinensis-infected hepatocellular carcinoma: implications for tumor progression. Parasit Vectors 2025; 18:68. [PMID: 39985046 PMCID: PMC11846337 DOI: 10.1186/s13071-025-06689-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 01/27/2025] [Indexed: 02/23/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a leading cause of cancer mortality globally, and its progression is associated with various factors, including parasitic infections such as Clonorchis sinensis (C. sinensis). Although C. sinensis infection has been implicated in HCC, the molecular mechanisms, particularly the role of microRNAs (miRNAs), remain poorly understood. This study aims to fill this gap by investigating the miRNA expression profiles in C. sinensis+ and C. sinensis- HCC tissues. METHOD We performed miRNA sequencing on HCC tissues from C. sinensis+ and C. sinensis- patients, followed by bioinformatics analyses to identify differentially expressed miRNAs (DEMs) and their target genes. Gene Ontology (GO) enrichment analysis was conducted to explore relevant biological processes, while a competitive endogenous RNA (ceRNA) network was constructed to investigate the interactions among miRNAs, long noncoding RNAs (lncRNAs), and messenger RNAs (mRNAs). Additionally, we performed survival analysis using Gene Expression Profiling Interactive Analysis 2 (GEPIA2) based on the The Cancer Genome Atlas-Liver Hepatocellular Carcinoma (TCGA-LIHC) cohort and assessed the clinical relevance of DEMs. Key miRNAs identified from this analysis were further validated through quantitative real‑time polymerase chain reaction (qRT-PCR) assays to confirm their expression in MHCC97H. RESULTS Our research identified significant miRNA dysregulation in C. sinensis+ HCC tumors compared with C. sinensis- HCC tumors. Notably, miR-143-3p, miR-10a-5p, and miR-100-5p were upregulated in C. sinensis+ HCC, contributing to immune responses and tumor progression, while let-7 family members and miR-221-3p were downregulated, affecting metabolic pathways. GO enrichment analysis highlighted the involvement of developmental processes, immune system regulation, and metabolic reprogramming in C. sinensis+ HCC. The construction of a ceRNA network revealed key interactions between miRNAs, lncRNAs, and mRNAs in C. sinensis+ HCC, suggesting regulatory mechanisms that could be potential therapeutic targets. Additionally, validation through qRT-PCR confirmed these findings, highlighting miRNA dysregulation as a critical factor in C. sinensis+ HCC progression. CONCLUSIONS This study provides novel insights into the role of miRNAs in C. sinensis-infected HCC progression. The findings highlight the critical role of miRNA dysregulation in the progression of C. sinensis-associated HCC, emphasizing the potential for therapeutic interventions targeting these molecular alterations in affected patients.
Collapse
Affiliation(s)
- Caibiao Wei
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, 530021, China
| | - Junxian Chen
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, 530021, China
| | - Taijun Huang
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, 530021, China
| | - Lingling Zhou
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, 530021, China
| | - Yulong Xu
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, 530021, China
| | - Qiumei Lin
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, 530021, China
| | - Yuling Qin
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, 530021, China
| | - Zeli Tang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Guangxi Medical University, Nanning, 530021, China.
| | - Weilong Yang
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.
| | - Min Fang
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, 530021, China.
- Engineering Research Center for Tissue and Organ Injury and Repair Medicine, Guangxi Medical University Cancer Hospital, Nanning, 530021, China.
| |
Collapse
|
26
|
Xu K, Zhang H, Dai H, Mao W. Machine learning and multi-omics characterization of SLC2A1 as a prognostic factor in hepatocellular carcinoma: SLC2A1 is a prognostic factor in HCC. Gene 2025; 938:149178. [PMID: 39681148 DOI: 10.1016/j.gene.2024.149178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 12/18/2024]
Abstract
Hepatocellular carcinoma (HCC) is characterized by high incidence, significant mortality, and marked heterogeneity, making accurate molecular subtyping essential for effective treatment. Using multi-omics data from HCC patients, we applied diverse clustering algorithms to identify three HCC subtypes (HSs) with distinct prognostic characteristics. Among these, HS1 emerged as an immune-compromised subtype associated with the poorest prognosis. Additionally, we developed a novel, robust, and highly accurate machine learning-guided prognostic signature (MLPS) by integrating multiple machine learning algorithms and their combinations. Our study also identified SLC2A1, the core gene of MLPS, as being highly expressed during advanced stages of tumor progression. Knockdown experiments demonstrated that reducing SLC2A1 expression significantly suppressed the malignant behavior of HCC cells. Furthermore, SLC2A1 expression was linked to responsiveness to dasatinib and vincristine, suggesting potential therapeutic relevance. MLPS and SLC2A1 offer promising tools for individualized prognosis prediction and targeted therapy in HCC, providing new opportunities to improve patient outcomes.
Collapse
Affiliation(s)
- Kangjie Xu
- Zhongda Hospital, Southeast University, Jiangsu Province, Nanjing 210009, PR China; Binhai County People's Hospital, Jiangsu Province, Yancheng 224000, PR China
| | - Houliang Zhang
- Zhongda Hospital, Southeast University, Jiangsu Province, Nanjing 210009, PR China
| | - Hua Dai
- Yangzhou University Clinical Medical College, Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Jiangsu Province, Yangzhou 225009, PR China.
| | - Weipu Mao
- Zhongda Hospital, Southeast University, Jiangsu Province, Nanjing 210009, PR China; Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
27
|
Wang Z, Liu Z, Lv M, Luan Z, Li T, Hu J. Novel histone modifications and liver cancer: emerging frontiers in epigenetic regulation. Clin Epigenetics 2025; 17:30. [PMID: 39980025 PMCID: PMC11841274 DOI: 10.1186/s13148-025-01838-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 02/08/2025] [Indexed: 02/22/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related death worldwide, and its onset and progression are closely associated with epigenetic modifications, particularly post-translational modifications of histones (HPTMs). In recent years, advances in mass spectrometry (MS) have revealed a series of novel HPTMs, including succinylation (Ksuc), citrullination (Kcit), butyrylation (Kbhb), lactylation (Kla), crotonylation (Kcr), and 2-hydroxyisobutyrylation (Khib). These modifications not only expand the histone code but also play significant roles in key carcinogenic processes such as tumor proliferation, metastasis, and metabolic reprogramming in HCC. This review provides the first comprehensive analysis of the impact of novel HPTMs on gene expression, cellular metabolism, immune evasion, and the tumor microenvironment. It specifically focuses on their roles in promoting tumor stem cell characteristics, epithelial-mesenchymal transition (EMT), and therapeutic resistance. Additionally, the review highlights the dynamic regulation of these modifications by specific enzymes, including "writers," "readers," and "erasers."
Collapse
Affiliation(s)
- Zhonghua Wang
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Jinan, 250021, Shandong, People's Republic of China
| | - Ziwen Liu
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Jinan, 250021, Shandong, People's Republic of China
| | - Mengxin Lv
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Jinan, 250021, Shandong, People's Republic of China
| | - Zhou Luan
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Jinan, 250021, Shandong, People's Republic of China
| | - Tao Li
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Jinan, 250021, Shandong, People's Republic of China
| | - Jinhua Hu
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Jinan, 250021, Shandong, People's Republic of China.
| |
Collapse
|
28
|
Rong H, Jiang Y. METTL14 suppresses the migration and invasion of hepatocellular carcinoma cells by m6A methylation of RPLP2. Sci Rep 2025; 15:5660. [PMID: 39955344 PMCID: PMC11830075 DOI: 10.1038/s41598-025-87701-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 01/21/2025] [Indexed: 02/17/2025] Open
Abstract
Fluctuating N(6)-methyladenosine (m6A) levels affect the progression of hepatocellular carcinoma (HCC). METTL14, a m6A methyltransferase, acts as a tumor suppressor in HCC; however, its underlying mechanisms need further clarification. This study aimed to clarify the role of METTL14 in HCC and the underlying molecular mechanism. Cellular behaviors were evaluated using cell counting kit-8, EdU, and Transwell assays. The molecular mechanism was analyzed using methylated RNA binding protein immunoprecipitation, dual-luciferase reporter assay, and RNA stability determination. The results demonstrated that METTL14 expression was decreased in HCC tissues and cells, and its overexpression suppressed cellular proliferation, migration, and invasion. Moreover, RPLP2 was negatively correlated to METTL14, and it was highly expressed in HCC tissues and cells. METTL14 promoted the m6A modification of RPLP2 and reduced its stability, thereby inhibiting malignant behaviors. Besides, YTHDC2 decreased RPLP2 expression and reversed the stability induced by METTL14. In conclusion, METTL14 inhibits HCC progression by regulating the YTHDC2-m6A-RPLP2 axis.
Collapse
Affiliation(s)
- Haiyan Rong
- Laboratory Medicine Diagnostic Centre, The First Affiliated Hospital, Xinjiang Medical University, No.118, Liyushan Road, Xinshi District, Urumqi, 830011, Xinjiang, China
| | - Yan Jiang
- Laboratory Medicine Diagnostic Centre, The First Affiliated Hospital, Xinjiang Medical University, No.118, Liyushan Road, Xinshi District, Urumqi, 830011, Xinjiang, China.
| |
Collapse
|
29
|
Liu R, Ye J, Wang J, Ma W, Qiu Z, Yu J, Wang W. Single-cell landscape of dynamic changes in CD8 + T cells, CD4 + T cells and exhausted T cells in hepatocellular carcinoma. Sci Rep 2025; 15:4130. [PMID: 39900964 PMCID: PMC11791069 DOI: 10.1038/s41598-025-88377-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 01/28/2025] [Indexed: 02/05/2025] Open
Abstract
Hepatocellular carcinoma has a high incidence and poor prognosis. In this study, we investigated the value of T-cell-related genes in prognosis by single-cell sequencing data in hepatocellular carcinoma. Twelve cases of hepatocellular carcinoma single-cell sequencing were included in the study. The high dimensional weighted gene co-expression network analysis (hdWGCNA) was used to identify gene modules associated with CD4+ T cells, CD8+ T cells and exhausted T cells. Altered signaling pathway activity in exhausted T cells was uncovered by the AUCell algorithm. xCELL, TIMER, QUANTISEQ, CIBERSORT and CIBERSORT-abs were performed to explore immune cell infiltration. Immune checkpoint inhibitor genes and TIDE methods were used to predict immunotherapy response. Finally, immunohistochemistry and real-time PCR were used to verify gene expression. The hdWGCNA algorithm identified 40 genes strongly associated with CD4+ T cells, CD8+ T cells and exhausted T cells. Seven genes were finally selected for transcriptome data modeling. The results of the three independent datasets suggested that the model had strong prognostic value. Model genes were critical factors influencing CD4+ T cell and CD8+ T cell infiltration in patients. The efficacy of PD-1 immunotherapy was higher in patients belonging to the low-risk group. Alterations in signaling pathways' activity within exhausted T cells were crucial factors contributing to the decline in immune function. Differential expression of seven genes in CD8+ T cells, CD4+ T cells and exhausted T cells were key targets for improving immunotherapy response in HCC.
Collapse
Affiliation(s)
- Rongqiang Liu
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Jing Ye
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Jianguo Wang
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Wangbin Ma
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Zhendong Qiu
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Jia Yu
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| | - Weixing Wang
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| |
Collapse
|
30
|
Tan X, Xun L, Yin Q, Chen C, Zhang T, Shen T. Epigenetic Modifications in HBV-Related Hepatocellular Carcinoma. J Viral Hepat 2025; 32:e14044. [PMID: 39868653 DOI: 10.1111/jvh.14044] [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: 07/31/2024] [Revised: 11/13/2024] [Accepted: 11/30/2024] [Indexed: 01/28/2025]
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver cancer. Hepatitis B virus (HBV) is the main pathogen for HCC development. HBV covalently closed circular DNA (cccDNA) forms extra-host chromatin-like minichromosomes in the nucleus of hepatocytes with host histones, non-histones, HBV X protein (HBx) and HBV core protein (HBc). Epigenetic alterations are dynamic and reversible, which regulate gene expression without altering the DNA sequence and play a pivotal role in the regulation of HCC onset and progression. The aim of this review is to elucidate the deregulation of epigenetic mechanisms involved in the pathogenesis of HBV-related HCC (HBV-HCC), including post-translational histone and non-histone modifications, DNA hypermethylation and hypomethylation, non-coding RNA modification on HBV cccDNA minichromosomes and host factors, effecting the replication/transcription of HBV cccDNA and transcription/translation of host genes, and thus HBV-HCC progression. It is expected that the epigenetic regulation perspective provides new ways for more in-depth development of therapeutic control of HBV-HCC.
Collapse
Affiliation(s)
- Xiaoqing Tan
- Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
- Department of Pulmonary and Critical Care Medicine, Yunnan Provincial Key Laboratory for Clinical Virology, Institute of Basic and Clinical Medicine, The First People's Hospital of Yunnan Province, Kunming, Peoples republic of China, China
| | - Linting Xun
- Department of Gastroenterology, the First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, People's Republic of China
| | - Qi Yin
- Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, People's Republic of China, China
| | - Chaohui Chen
- Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
| | - Tao Zhang
- Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
- Department of Pulmonary and Critical Care Medicine, Yunnan Provincial Key Laboratory for Clinical Virology, Institute of Basic and Clinical Medicine, The First People's Hospital of Yunnan Province, Kunming, Peoples republic of China, China
| | - Tao Shen
- Medical School, Kunming University of Science and Technology, Kunming, People's Republic of China
- Department of Pulmonary and Critical Care Medicine, Yunnan Provincial Key Laboratory for Clinical Virology, Institute of Basic and Clinical Medicine, The First People's Hospital of Yunnan Province, Kunming, Peoples republic of China, China
| |
Collapse
|
31
|
Zhang Z, Gao Y, Qian Y, Wei B, Jiang K, Sun Z, Zhang F, Yang M, Baldi S, Yu X, Zuo Y, Ren S. The Lyn/RUVBL1 Complex Promotes Colorectal Cancer Liver Metastasis by Regulating Arachidonic Acid Metabolism Through Chromatin Remodeling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2406562. [PMID: 39665272 PMCID: PMC11792055 DOI: 10.1002/advs.202406562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 11/26/2024] [Indexed: 12/13/2024]
Abstract
Liver metastasis is a common cause of death in colorectal cancer (CRC) patients, but epigenetic remodeling and metabolic reprogramming for CRC liver metastasis remain unclear. The study revealed that the Lyn/RUVBL1 complex is highly expressed in CRC and is closely correlated with liver metastasis. On the one hand, ATAC-seq and HiCut suggested that Lyn/RUVBL1 regulates the expression of TRIB3 through the POL II-mediated chromatin conformation of TRIB3 and thus the expression of β-catenin. This promotes the proliferation and migration of CRC through β-catenin-mediated upregulation of MMP9 and VEGF. On the other hand, metabolomics revealed that Lyn/RUVBL1 regulates the expression of PGE2 through the enzyme COX2, thereby promoting arachidonic acid (AA) metabolism. CUT-Tag showed that Lyn/RUVBL1 silencing reduces the H3K27ac level in the COX2 promoter. Then, it is found that COX2 is regulated by the transcription factor FOXA1. Lyn/RUVBL1 modulates AA metabolism by regulating the chromatin accessibility of FOXA1. AA metabolism promotes the metastasis of CRC by affecting β-catenin nuclear translocation and upregulating MMP9 and VEGF. These findings suggest that the Lyn/RUVBL1 complex mediates epigenetic remodeling to regulate the metabolic reprogramming of AA, highlighting its role in promoting the metastasis of CRC.
Collapse
Affiliation(s)
- Zhenyu Zhang
- Department of General SurgeryThe Second Hospital of Dalian Medical UniversityDalian116023China
- Department of Clinical BiochemistryCollege of Laoratory Medicine, Dalian Medical UniversityDalian116044China
| | - Yina Gao
- Department of General SurgeryThe Second Hospital of Dalian Medical UniversityDalian116023China
- Department of Clinical BiochemistryCollege of Laoratory Medicine, Dalian Medical UniversityDalian116044China
| | - Yuanyuan Qian
- Department of Clinical BiochemistryCollege of Laoratory Medicine, Dalian Medical UniversityDalian116044China
| | - Bowen Wei
- Department of Clinical BiochemistryCollege of Laoratory Medicine, Dalian Medical UniversityDalian116044China
| | - Kexin Jiang
- Department of Clinical BiochemistryCollege of Laoratory Medicine, Dalian Medical UniversityDalian116044China
| | - Zhiwei Sun
- Department of General SurgeryThe Second Hospital of Dalian Medical UniversityDalian116023China
| | - Feifan Zhang
- Department of General SurgeryThe Second Hospital of Dalian Medical UniversityDalian116023China
| | - Mingming Yang
- Department of Clinical BiochemistryCollege of Laoratory Medicine, Dalian Medical UniversityDalian116044China
| | - Salem Baldi
- Department of Clinical BiochemistryCollege of Laoratory Medicine, Dalian Medical UniversityDalian116044China
| | - Xiaoqi Yu
- Department of Clinical BiochemistryCollege of Laoratory Medicine, Dalian Medical UniversityDalian116044China
| | - Yunfei Zuo
- Department of Clinical BiochemistryCollege of Laoratory Medicine, Dalian Medical UniversityDalian116044China
| | - Shuangyi Ren
- Department of General SurgeryThe Second Hospital of Dalian Medical UniversityDalian116023China
| |
Collapse
|
32
|
Liu Z, Ling ZQ. Golgi scaffold protein PAQR11 in pan-cancer landscape: A comprehensive bioinformatics exploration of expression patterns, prognostic significance, and potential immunological function. Heliyon 2025; 11:e41724. [PMID: 39906812 PMCID: PMC11791267 DOI: 10.1016/j.heliyon.2025.e41724] [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: 08/01/2024] [Revised: 09/28/2024] [Accepted: 01/03/2025] [Indexed: 02/06/2025] Open
Abstract
Background The progestin and adipoQ receptor family member, PAQR11, is recognized for its roles in vesicle trafficking, mitogenic signaling, and metastatic spread, positioning it as a crucial regulator in cancer biology. PAQR11 influences lipid metabolism and susceptibility to ferroptosis in cancer cells. This study aims to investigate the prognostic significance of PAQR11, its relevance to immune responses, and its association with drug sensitivity across various cancer types. By elucidating these aspects, the research seeks to assess PAQR11's potential as a biomarker and therapeutic target in oncology. Methods We conducted a comprehensive bioinformatics analysis using publicly available pan-cancer datasets from TCGA, GEO, UALCAN, TIMER, GEPIA2, KM plotter, and TISIDB. This analysis encompassed gene expression profiles across 33 cancer types, with a focus on PAQR11's expression patterns, prognostic significance, and immunological relevance. In addition, the study explored the correlation between PAQR11 expression and drug sensitivity, alongside its molecular and pathological characteristics in various tumors. Results Our findings demonstrate elevated PAQR11 expression levels across multiple cancer types, which significantly correlate with patient prognostic outcomes. The analysis further revealed PAQR11's involvement in immunological and epigenetic processes, underscoring its critical role in cancer progression and treatment response. Notably, a strong correlation between PAQR11 expression and drug sensitivity was identified, suggesting its potential influence on the initiation and progression of various cancers and highlighting its promise as a therapeutic target. Conclusions The comprehensive analysis of PAQR11 underscores its significance as a biomarker for cancer prognosis and its role in regulating immunological and epigenetic processes. These findings offer valuable insights that could inform early detection strategies and the development of novel therapeutic approaches. Further exploration and validation of PAQR11 are essential, highlighting the need for its integration into future oncological research and treatment strategy development. Trial registration Not applicable.
Collapse
Affiliation(s)
- Zhu Liu
- Zhejiang Cancer Institute, Zhejiang Cancer Hospital, No.1 Banshan East Rd., Gongshu District, Hangzhou, 310022, Zhejiang, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310018, China
| | - Zhi-Qiang Ling
- Zhejiang Cancer Institute, Zhejiang Cancer Hospital, No.1 Banshan East Rd., Gongshu District, Hangzhou, 310022, Zhejiang, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310018, China
| |
Collapse
|
33
|
Shi J, Zhu X, Yang JB. Advances and challenges in molecular understanding, early detection, and targeted treatment of liver cancer. World J Hepatol 2025; 17:102273. [PMID: 39871899 PMCID: PMC11736488 DOI: 10.4254/wjh.v17.i1.102273] [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: 10/14/2024] [Revised: 11/12/2024] [Accepted: 11/27/2024] [Indexed: 01/06/2025] Open
Abstract
In this review, we explore the application of next-generation sequencing in liver cancer research, highlighting its potential in modern oncology. Liver cancer, particularly hepatocellular carcinoma, is driven by a complex interplay of genetic, epigenetic, and environmental factors. Key genetic alterations, such as mutations in TERT, TP53, and CTNNB1, alongside epigenetic modifications such as DNA methylation and histone remodeling, disrupt regulatory pathways and promote tumorigenesis. Environmental factors, including viral infections, alcohol consumption, and metabolic disorders such as nonalcoholic fatty liver disease, enhance hepatocarcinogenesis. The tumor microenvironment plays a pivotal role in liver cancer progression and therapy resistance, with immune cell infiltration, fibrosis, and angiogenesis supporting cancer cell survival. Advances in immune checkpoint inhibitors and chimeric antigen receptor T-cell therapies have shown potential, but the unique immunosuppressive milieu in liver cancer presents challenges. Dysregulation in pathways such as Wnt/β-catenin underscores the need for targeted therapeutic strategies. Next-generation sequencing is accelerating the identification of genetic and epigenetic alterations, enabling more precise diagnosis and personalized treatment plans. A deeper understanding of these molecular mechanisms is essential for advancing early detection and developing effective therapies against liver cancer.
Collapse
Affiliation(s)
- Ji Shi
- Department of Research and Development, Ruibiotech Company Limited, Beijing 100101, China
| | - Xu Zhu
- Department of Research and Development, Ruibiotech Company Limited, Beijing 100101, China
| | - Jun-Bo Yang
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, Guangdong Province, China.
| |
Collapse
|
34
|
Wu X, Zhang X, Yu X, Liang H, Tang S, Wang Y. Exploring the association between air pollution and the incidence of liver cancers. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 290:117437. [PMID: 39671760 DOI: 10.1016/j.ecoenv.2024.117437] [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: 10/03/2024] [Revised: 11/20/2024] [Accepted: 11/26/2024] [Indexed: 12/15/2024]
Abstract
Liver cancer, namely hepatocellular carcinoma (HCC), is a major global health concern deeply influenced by environmental factors. Air pollutants emerged as significant contributors to its incidence. This review explores the association between air pollution-specifically particulate matter (PM2.5), industrial chemicals like vinyl chloride, and benzene-and the increased risk of liver cancer. Mechanistically, air pollutants may cause liver damage by inducing oxidative stress, inflammation, and genetic mutations, contributing to cancer development. Epidemiological evidence from cohort and geographic studies highlights a positive correlation between long-term exposure to air pollutants and elevated incidence and mortality of liver cancer. Furthermore, air pollution has been shown to worsen survival outcomes in liver cancer patients, particularly those diagnosed at early stages. The review emphasizes the need for stricter air quality regulations and relevant research for underlying mechanisms exposed to air pollution. Addressing air pollution exposure could be crucial for reducing liver cancer risks and improving public health outcomes.
Collapse
Affiliation(s)
- Xin Wu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shengyang, China
| | - Xin Zhang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shengyang, China
| | - Xiaopeng Yu
- Oncology Department, Shengjing Hospital of China Medical University, Shengyang, China
| | - Hongyuan Liang
- Department of Radiology, Shengjing Hospital of China Medical University, Shengyang, China.
| | - Shaoshan Tang
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shengyang, China.
| | - Yao Wang
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shengyang, China.
| |
Collapse
|
35
|
Luan S. The role of histone lactylation genes in hepatocellular carcinoma prognostic models and their immune cell infiltration features: a comprehensive analysis of single-cell, spatial transcriptome, Mendelian randomization and experiment. Discov Oncol 2025; 16:29. [PMID: 39789404 PMCID: PMC11717769 DOI: 10.1007/s12672-025-01775-1] [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: 09/30/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025] Open
Abstract
INTRODUCTION With the increasing impact of hepatocellular carcinoma (HCC) on society, there is an urgent need to propose new HCC diagnostic biomarkers and identification models. Histone lysine lactylation (Kla) affects the prognosis of cancer patients and is an emerging target in cancer treatment. However, the potential of Kla-related genes in HCC is poorly understood. METHODS A variety of machine learning methods were used to construct and validate a model of differentially expressed Kla genes with comprehensive evaluations included ROC, Kaplan‒Meier curve, Cox regression, decision curve. Immune infiltration gathered with spatial transcriptome was performed using integrated data from multiple databases. Furthermore, single-cell analysis was used to discover the cell-cell communication and Mendelian randomization was used to study the causal relationships between immune cell and HCC. Lastly, qRT-PCR was used to verify the expression of Kla genes. RESULTS We established a model consisting of 12 genes that had well prognostic performance and were identified as independent prognostic factors. Single-cell analysis showed that CD8 T+ cells and conventional dendritic cells were enriched in HCC patients. Spatial transcriptomics analysis indicated that the Kla genes influenced the immune characteristics of HCC. Mendelian randomization results showed that TBNK and monocytes were the main risk factors. qRT-PCR validation results indicated that the expression of multiple genes in Huh7 cells was significantly higher than in LO2 cells. CONCLUSION Overall, a Kla-related model was established, which may provide new strategies and insights for the treatment and diagnosis of HCC.
Collapse
Affiliation(s)
- Shanjie Luan
- Shandong University School of Medicine, 44 Wenhua Xi Road, Jinan, 250012, Shandong, China.
| |
Collapse
|
36
|
Cherradi S, Roux S, Dupuy M, Tabone-Eglinger S, Tuaillon E, Ziol M, Assenat E, Duong HT. Modelling hepatocellular carcinoma microenvironment phenotype to evaluate drug efficacy. Sci Rep 2025; 15:1179. [PMID: 39774014 PMCID: PMC11706984 DOI: 10.1038/s41598-024-84304-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is the third most common cause of cancer-related death worldwide. Treating HCC is challenging because of the poor drug effectiveness and the lack of tools to predict patient responses. To resolve these issues, we established a patient-centric spheroid model using HepG2, TWNT-1, and THP-1 co-culture, that mimics HCC phenotype. We developed a target-independent cell killing (TICK) exclusion strategy to monitor the therapeutic response. We demonstrated that our model reproduced the Barcelona Clinic Liver Cancer (BCLC) molecular classification, displayed known alterations of epigenetic players, and responded to tyrosine kinase inhibitors (TKIs) such as sorafenib, cabozantinib, and lenvatinib in a patient-dependent manner. Importantly, we reported for the first time that our model correctly predicted 34 clinical outcomes to TKIs out of 37 case studies on 32 HCC patients confirming that patient-centric spheroids, combined with our TICK exclusion strategy, are valuable models for drug discovery and opening a near perspective to personalized care.
Collapse
Affiliation(s)
- Sara Cherradi
- PredictCan Biotechnologies SAS, Biopôle Euromédecine, Grabels, France
| | - Salomé Roux
- PredictCan Biotechnologies SAS, Biopôle Euromédecine, Grabels, France
| | - Marie Dupuy
- Service d'Oncologie Médicale, Hôpital Saint Eloi, Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Séverine Tabone-Eglinger
- Plateforme de Gestion des Echantillons Biologiques, Centre Léon Bérard, Cancer Research Center of Lyon, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Lyon, France
| | - Edouard Tuaillon
- Centre de Ressources Biologiques (CRB), Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Marianne Ziol
- Centre de Ressources Biologiques du Groupe hospitalier Paris Seine Saint-Denis, Paris, France
| | - Eric Assenat
- Service d'Oncologie Médicale, Hôpital Saint Eloi, Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Hong Tuan Duong
- PredictCan Biotechnologies SAS, Biopôle Euromédecine, Grabels, France.
| |
Collapse
|
37
|
Kong X, Niu Z, Wang H, Liu M, Ma C, Lu J, Zhou X, Zhu H. Left-lateral decubitus jackknife position for laparoscopic resection of right posterior liver tumors: A safe and effective approach. Langenbecks Arch Surg 2025; 410:25. [PMID: 39755910 DOI: 10.1007/s00423-024-03595-3] [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: 07/25/2024] [Accepted: 12/22/2024] [Indexed: 01/06/2025]
Abstract
PURPOSE To compare outcomes of LLR in VI/VII of the liver in Left-lateral Decubitus Jackknife Position (LDJP) and traditional Supine Position (SP). We used propensity score matching (PSM) to analyze clinical outcomes. PATIENTS & METHODS This study retrospectively analyzed patients undergoing LLR for liver tumors in segments VI and/or VII at Shandong Provincial Hospital from 2018 to 2023. A total of 218 cases were included (LDJP, n = 94; SP, n = 124). Matched 1:1 PSM groups were created and clinical indicators compared between groups. RESULTS 218 LLR patients, 94 LDJP and 124 supine. After 1:1 PSM, each group had 62 patients. No significant differences in clinical or laboratory parameters. All surgeries were successful, 1 LDJP conversion to open resection and 4 SP conversions (P = 0.375). LDJP average surgery duration: 220.6 ± 29.9 min, supine position: 262.6 ± 35.6 min (P < 0.001). LDJP perioperative blood loss: 169.0 ± 74.4 mL, supine position: 231.6 ± 84.6 mL (P < 0.001). Four LDJP patients required intraoperative blood transfusion compared to 16 supine position patients (P = 0.012). All cases had negative margins postoperatively. No significant differences in postoperative complications (8 LDJP vs 9 supine, P = 0.675) or length of hospital stay (25 LDJP vs 26 supine, ≥ 7 days) (P = 1.000). CONCLUSION Laparoscopic partial hepatectomy in LDJP for hepatic VI/VII tumor safe and feasible. Reduces operative time, blood loss, transfusion requirement, improving outcomes.
Collapse
Affiliation(s)
- Xiaohan Kong
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital, Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, 250021, China
| | - Zheyu Niu
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital, Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, 250021, China
- Department of Clinical Research, Qilu Synva Pharmaceutical Co. Ltd, Dezhou, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Heng Wang
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, China
| | - Meng Liu
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital, Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, 250021, China
| | - Chaoqun Ma
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital, Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, 250021, China
| | - Jun Lu
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital, Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, 250021, China
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, China
| | - Xu Zhou
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital, Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, 250021, China
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, China
| | - Huaqiang Zhu
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital, Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, 250021, China.
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, China.
| |
Collapse
|
38
|
Chen M, Yang Y, Hu G, Peng Z, Wen W. The promoting effect of the POU3F2/METTL16/PFKM cascade on glycolysis and tumorigenesis of hepatocellular carcinoma. Ann Hepatol 2025; 30:101776. [PMID: 39756795 DOI: 10.1016/j.aohep.2025.101776] [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: 10/22/2024] [Revised: 12/02/2024] [Accepted: 12/17/2024] [Indexed: 01/07/2025]
Abstract
INTRODUCTION AND OBJECTIVES Deregulation of m6A methylation, the most prevailing RNA modification, participates in cancer pathogenesis. METTL16, an atypical methyltransferase, functions as a pro-tumorigenic factor in hepatocellular carcinoma (HCC). Here, we explored the action of METTL16 on HCC glycolysis and the associated mechanism. MATERIALS AND METHODS Expression analysis was done by quantitative PCR, immunoblotting, or immunohistochemistry. Cell sphere formation, invasiveness, apoptosis, proliferation and viability were detected by sphere formation, transwell, flow cytometry, EdU and CCK-8 assays, respectively. Xenograft studies were performed to analyze the role in vivo. Methylated RNA immunoprecipitation (MeRIP) and RIP assays were used to verify the METTL16/PFKM relationship. PFKM mRNA stability was tested by actinomycin D treatment. Chromatin immunoprecipitation (ChIP) and luciferase assays were performed to analyze the POU3F2/METTL16 relationship. RESULTS In HCC, METTL16 expression was elevated, and increased levels of METTL16 transcript predicted poor HCC prognosis. METTL16 deficiency resulted in suppressed HCC cell growth, invasiveness and sphere formation. Moreover, METTL16 depletion diminished HCC cell glycolysis. Mechanistically, PFKM expression was positively associated with METTL16 expression. METTL16 mediated m6A methylation to stabilize PFKM mRNA via an IGF2BP3-dependent manner. Restored PFKM expression exerted a counteracting effect on METTL16 deficiency-mediated in vitro cell phenotype alterations and in vivo xenograft growth suppression. Furthermore, POU3F2 promoted the transcription of METTL16 in HCC cells. CONCLUSIONS Our findings define the crucial role of the POU3F2/METTL16/PFKM axis in HCC pathogenesis, offering the potential opportunity to combat HCC.
Collapse
Affiliation(s)
- Ming Chen
- Department of Gastroenterology and Hepatology, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang City, 421001, Hunan, China
| | - Yuan Yang
- Department of Gastroenterology and Hepatology, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang City, 421001, Hunan, China
| | - Guangsheng Hu
- Department of Gastroenterology and Hepatology, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang City, 421001, Hunan, China
| | - Zhong Peng
- Department of Gastroenterology and Hepatology, Yiyang Central Hospital, Yiyang City, 413099, Hunan, China
| | - Wu Wen
- Department of Hepato-Biliary-Pancreatic Surgery, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang City, 421001, Hunan, China.
| |
Collapse
|
39
|
Xie JW, Wang HL, Lin LQ, Guo YF, Wang M, Zhu XZ, Niu JJ, Lin LR. Telomere-methylation genes: Novel prognostic biomarkers for hepatocellular carcinoma. Clin Res Hepatol Gastroenterol 2025; 49:102516. [PMID: 39675625 DOI: 10.1016/j.clinre.2024.102516] [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: 08/09/2024] [Revised: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND Since telomere length and DNA methylation both correlate with hepatocellular carcinoma (HCC) prognosis, telomere-methylation genes could be novel prognostic markers for HCC. METHOD This study first investigated the interaction between telomere length and DNA methylation in HCC through Mendelian randomization analysis. Then, this study identified telomere-methylation genes in HCC by employing the TCGA-LIHC cohort, and explored the expression patterns of these genes in the tumor microenvironment of HCC and potential underlying mechanisms. Finally, the HCC risk-scoring model and prognostic model based on these genes were established, and the performance of the model was assessed. RESULT The findings revealed a bidirectional relationship between telomere length and DNA methylation in HCC. Fifty telomere-methylation genes were identified, and the prognosis-related telomere-methylation genes were closely associated with Treg and Tprolif cell subsets within the HCC tumor microenvironment. Telomere-methylation genes could potentially impact the prognosis of HCC patients by modulating chromosome stability and regulating the cell cycle. Additionally, the constructed risk scoring model and prognostic prediction model showcased compelling clinical applicability, as evidenced by the receiver operating characteristic curve, the decision curve analysis, and the calibration curves. CONCLUSION This study elucidated the potential of telomere-methylation genes as prognostic biomarkers for HCC and paves the way for novel approaches in prognostication and treatment management for HCC patients.
Collapse
Affiliation(s)
- Jia-Wen Xie
- Center of Clinical Laboratory, Zhongshan Hospital Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China; Institute of Infectious Disease, School of Medicine, Xiamen University, Xiamen, China
| | - Hui-Ling Wang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Ling-Qing Lin
- Center of Clinical Laboratory, Zhongshan Hospital Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China
| | - Yin-Feng Guo
- Center of Clinical Laboratory, Zhongshan Hospital Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China; Institute of Infectious Disease, School of Medicine, Xiamen University, Xiamen, China
| | - Mao Wang
- Center of Clinical Laboratory, Zhongshan Hospital Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China; Institute of Infectious Disease, School of Medicine, Xiamen University, Xiamen, China; Department of Pathology, Chengdu Wenjiang District People's Hospital, Chengdu, China
| | - Xiao-Zhen Zhu
- Center of Clinical Laboratory, Zhongshan Hospital Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China; Institute of Infectious Disease, School of Medicine, Xiamen University, Xiamen, China.
| | - Jian-Jun Niu
- Center of Clinical Laboratory, Zhongshan Hospital Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China; Institute of Infectious Disease, School of Medicine, Xiamen University, Xiamen, China.
| | - Li-Rong Lin
- Center of Clinical Laboratory, Zhongshan Hospital Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China; Institute of Infectious Disease, School of Medicine, Xiamen University, Xiamen, China.
| |
Collapse
|
40
|
Miao G, Zhang Z, Wang M, Gu X, Xiang D, Cao H. Berberine in combination with anti-PD-L1 suppresses hepatocellular carcinoma progression and metastasis via Erk signaling pathway. Ann Med Surg (Lond) 2025; 87:103-112. [PMID: 40109642 PMCID: PMC11918555 DOI: 10.1097/ms9.0000000000002746] [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: 07/11/2024] [Accepted: 11/05/2024] [Indexed: 03/22/2025] Open
Abstract
Background Berberine (BBR) is an isoquinoline alkaloid extracted from Huang Lian and other herbal medicines. It has been reported to play a crucial role in multiple metabolic diseases and cancers. Programmed cell death-1 (PD-L1) is known as the immune checkpoint; immunotherapy targeting PD1/PD-L1 axis can effectively block its pro-tumor activity. However, the effect of the combined use of BBR and anti-PD-L1 on hepatocellular carcinoma (HCC) has not been reported. Methods Hep-3B and HCCLM3 cells were chosen as the experimental objects. To determine the potential anti-cancer activity of the combination of BBR and anti-PD-L1, we first treated v cells with BBR. The cell viability of Hep-3B and HCCLM3 with BBR treatment was measured by Cell Count Kit 8 assay. Cytometry by time-of-flight was performed to analyze tumor tissues after treatment with BBR and/or anti-PD-L1. Proliferation-, migration-, and invasion-related markers were measured by western blotting and immunohistochemistry. Results The results showed that BBR significantly inhibited the proliferation of Hep-3B and HCCLM3.The combination treatment of BBR and anti-PD-L1 had a prominent inhibitory effect on HCC tumorigenesis. Cytometry by time-of-flight analysis indicated that BBR affects the immune subsets in the tumors. Besides, BBR and anti-PD-L1 inhibited the migration and invasion of HCC by inactivating the phosphorylation of Erk. Conclusion Our study proposed that the combination treatment of BBR and anti-PD-L1 markedly inhibited the tumorigenesis of HCC by Erk signaling pathway. We hope our research can provide a new strategy for the potential of BBR as a therapeutic agent in the treatment of HCC.
Collapse
Affiliation(s)
- Ganggang Miao
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of General Surgery, The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Danyang, Zhenjiang, China
| | - Zhiyu Zhang
- Department of General Surgery, The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Danyang, Zhenjiang, China
| | - Meiyan Wang
- Suzhou Industrial Park Institute of Services Outsourcing, Suzhou, China
| | - Xingwei Gu
- Department of General Surgery, The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Danyang, Zhenjiang, China
| | - Dongxiao Xiang
- Department of Pediatrics, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hongyong Cao
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
41
|
Du J, Luo H, Ye S, Zhang H, Zheng Z, Liu K. Unraveling IFI44L's biofunction in human disease. Front Oncol 2024; 14:1436576. [PMID: 39737399 PMCID: PMC11682996 DOI: 10.3389/fonc.2024.1436576] [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: 05/22/2024] [Accepted: 11/26/2024] [Indexed: 01/01/2025] Open
Abstract
Interferon-induced protein 44-like (IFI44L) is regarded as an immune-related gene and is a member of interferon-stimulated genes (ISGs). They participate in network transduction, and its own epigenetic modifications, apoptosis, cell-matrix formation, and many other pathways in tumors, autoimmune diseases, and viral infections. The current review provides a comprehensive overview of the onset and biological mechanisms of IFI44L and its potential clinical applications in malignant tumors and non-neoplastic diseases.
Collapse
|
42
|
Liu G, Long J, Liu C, Chen J. Development and verification of a nomogram for predicting portal vein tumor thrombosis in hepatocellular carcinoma. Am J Transl Res 2024; 16:7511-7520. [PMID: 39822560 PMCID: PMC11733390 DOI: 10.62347/plqf5135] [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: 10/31/2024] [Accepted: 12/07/2024] [Indexed: 01/19/2025]
Abstract
OBJECTIVE To develop a nomogram to predict the risk of portal vein tumor thrombosis (PVTT) in hepatocellular carcinoma (HCC) patients. METHODS Patients diagnosed with HCC at Hunan Provincial People's Hospital between January 2010 and January 2022 were enrolled. Data on demographic characteristics, comorbidities, and laboratory tests were collected. Multivariate logistic regression was used to identify independent risk factors for PVTT, which were then incorporated into a predictive nomogram. The nomogram's discriminative ability was evaluated using the area under the receiver operating characteristic (AUC) curve. Clinical utility was assessed through decision curve analysis (DCA). RESULTS Being male (OR 1.991, 95% CI 1.314-3.017, P = 0.001), Barcelona Clinic Liver Cancer (BCLC) staging (stage C: OR 8.043, 95% CI 4.334-14.926, P<0.001; stage D: OR 7.977, 95% CI 3.532-18.017, P<0.001), tumor size >5 cm (OR 1.792, 95% CI 1.116-2.876, P = 0.016), and D-dimer (OR 1.126, 95% CI 1.083-1.171, P<0.001) were identified as independent risk factors for PVTT. The nomogram formula is: Logit = -2.8961 + 0.6586 (male) + BCLC staging (-0.1922 for B, 1.9251 for C, or 1.7938 for D) + 0.5418 (tumor size >5 cm) + 0.1051 DDi. The nomogram achieved an AUC of 0.798 (95% CI 0.774-0.822) in the training set and 0.822 (95% CI 0.782-0.862) in the validation set. Sensitivities were 86.6% and 90.7%, while specificies were 68.2% and 71.8% in the training and validation sets, respectively, demonstrating strong discrimination and predictive accuracy. DCA indicated a favorable risk threshold probability. CONCLUSION A nomogram incorporating male sex, BCLC staging, tumor size, and D-dimer demonstrated good predictive performance for PVTT. This tool may aid in the early comprehensive assessment of PVTT risk in HCC patients.
Collapse
Affiliation(s)
- Guanghua Liu
- Department of Blood Transfusion, Laboratory of Hematology, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University)Changsha 410002, Hunan, China
| | - Jiangwen Long
- Department of Blood Transfusion, Laboratory of Hematology, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University)Changsha 410002, Hunan, China
| | - Chaoshui Liu
- Hunan Provincial Key Laboratory of The Research and Development of Novel Pharmaceutical Preparations, The “Double-First Class” Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), Changsha Medical UniversityChangsha 410219, Hunan, China
| | - Jie Chen
- Department of Clinical Laboratory, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University)Changsha 410002, Hunan, China
| |
Collapse
|
43
|
Duan J, Jiang R, Shen H, Xu X, Sun D. Analysis of nitrogen metabolism-related gene expression in hepatocellular carcinoma to establish relevant indicators for prediction of prognosis and guidance of immunotherapy. Comput Methods Biomech Biomed Engin 2024:1-17. [PMID: 39673385 DOI: 10.1080/10255842.2024.2438922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 10/14/2024] [Accepted: 12/02/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND The prognosis of cancers is strongly connected with nitrogen metabolism (NM), which plays a critical role in the microenvironment and growth of tumors. It is unsubstantiated, however, how important NM-related genes are for the prognosis of hepatocellular carcinoma (HCC). METHODS Using publicly available data, we examined potential mechanisms of NM-related genes in HCC, created a predictive model, and assessed immune infiltration and medication sensitivity. RESULTS A prognostic model, which included 12 NM genes (COLQ, GNE, ISCU, MSRA, SARS2, SPHK1, CBS, GOT2, CHST1, EXTL2, GCLM, YARS1), was constructed based on regression analysis. The robustness of the model was validated using multiple methods. The high-risk (HR) and low-risk (LR) groups had varying degrees of immune infiltration, according to an immunology-related study. Of these, B cells and Type_II_IFN_Response were greatly infiltrated in the LR group, whereas aCDs, Macrophages, and Treg were heavily infiltrated in the HR group (p < 0.05). Because of higher immunophenoscore, the low-risk group could benefit from immunotherapy more. Drug sensitivity predictions indicated that people with high CBS expression and low GOT2 and ISCU expression may benefit more from treatment with SCH-772984, Pimasertib, Cobimetinib (isomer1), TAK-733, LY-3214996, ARRY-162, Cladribine, Fludarabine, and Hydroxyurea. CONCLUSION This work created a 12-gene signature based on NM, preliminary investigated immune infiltration in two risk categories, and discovered some possible anti-tumor medications. To sum up, our study findings offer fresh perspectives on the roles played by NM-associated genes in HCC development, prognosis, immunological response, and medication screening.
Collapse
Affiliation(s)
- Jianwen Duan
- Department of Hepatobiliary Surgery, Quzhou Hospital Affiliated of Wenzhou Medical University (Quzhou People's Hospital), Quzhou, Zhejiang, China
| | - Renya Jiang
- Department of Hepatobiliary Surgery, Quzhou Hospital Affiliated of Wenzhou Medical University (Quzhou People's Hospital), Quzhou, Zhejiang, China
| | - Hongbo Shen
- Department of Hepatobiliary Surgery, Quzhou Hospital Affiliated of Wenzhou Medical University (Quzhou People's Hospital), Quzhou, Zhejiang, China
| | - Xiaofang Xu
- Department of Oncology, The First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Da Sun
- Department of Hepatobiliary Surgery, Quzhou Hospital Affiliated of Wenzhou Medical University (Quzhou People's Hospital), Quzhou, Zhejiang, China
| |
Collapse
|
44
|
Chen H, Lin Y, Chen J, Luo X, Kan Y, He Y, Zhu R, Jin J, Li D, Wang Y, Han Z. Targeting caspase-8: a new strategy for combating hepatocellular carcinoma. Front Immunol 2024; 15:1501659. [PMID: 39726605 PMCID: PMC11669555 DOI: 10.3389/fimmu.2024.1501659] [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: 09/25/2024] [Accepted: 11/29/2024] [Indexed: 12/28/2024] Open
Abstract
Hepatocellular carcinoma (HCC) represents the most prevalent form of primary liver cancer and has a high mortality rate. Caspase-8 plays a pivotal role in an array of cellular signaling pathways and is essential for the governance of programmed cell death mechanisms, inflammatory responses, and the dynamics of the tumor microenvironment. Dysregulation of caspase-8 is intricately linked to the complex biological underpinnings of HCC. In this manuscript, we provide a comprehensive review of the regulatory roles of caspase-8 in apoptosis, necroptosis, pyroptosis, and PANoptosis, as well as its impact on inflammatory reactions and the intricate interplay with critical immune cells within the tumor microenvironment, such as tumor-associated macrophages, T cells, natural killer cells, and dendritic cells. Furthermore, we emphasize how caspase-8 plays pivotal roles in the development, progression, and drug resistance observed in HCC, and explore the potential of targeting caspase-8 as a promising strategy for HCC treatment.
Collapse
Affiliation(s)
- Haoran Chen
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Yumeng Lin
- Health Management Center, Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Jie Chen
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Xuemei Luo
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Yubo Kan
- Sichuan Provincial Woman’s and Children’s Hospital/The Affiliated Women’s and Children’s Hospital of Chengdu Medical College, Chengdu, China
| | - Yuqi He
- Department of Blood Transfusion, Lu’an People’s Hospital, the Affiliated Hospital of Anhui Medical University, Lu’an, China
| | - Renhe Zhu
- Department of Blood Transfusion, Lu’an People’s Hospital, the Affiliated Hospital of Anhui Medical University, Lu’an, China
| | - Jiahui Jin
- Department of gastroenterology, Baoji Central Hospital, Baoji, China
| | - Dongxuan Li
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Yi Wang
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Zhongyu Han
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| |
Collapse
|
45
|
Rashidipour M, Abbaszadeh S, Birjandi M, Pajouhi N, Ahmadi Somaghian S, Goudarzi G, Shahryarhesami S, Moradi Sarabi M, Babaeenezhad E. Antimicrobial activity and cytotoxic and epigenetic effects of tannic acid-loaded chitosan nanoparticles. Sci Rep 2024; 14:30405. [PMID: 39638815 PMCID: PMC11621443 DOI: 10.1038/s41598-024-80771-x] [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: 08/12/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024] Open
Abstract
Tannic acid (TA) is a potent antitumor agent, but its low bioavailability and absorption limit its use. In this study, it was loaded into chitosan-based nanoparticles (Chi-NPs) to overcome these limitations and to improve its antimicrobial and anticancer activities. TA-loaded Chi-NPs (Chi-TA-NPs) were synthesized using the ionic gelation method and physicochemically characterized by FE-SEM, FTIR, XRD, PDI, DLS, and zeta potential analysis. Additionally, the antimicrobial activity of Chi-TA-NPs against two G+ bacterial strains, two G- bacterial strains, and a fungal strain (Candida albicans) was investigated using the microbroth dilution method. MTT assay was used to examine the cytotoxic effects of Chi-TA-NPs on HepG2 cells. The expression of DNA methyltransferase 1 (DNMT1), DNMT3A, and DNMT3B was examined in HepG2 cells using RT-qPCR. The amount of 5-methylcytosine in the HepG2 cell-derived genomic DNA was measured using ELISA. FE-SEM micrographs showed the loading of TA into the chitosan-based formulation. The peaks detected in the XRD and FTIR analyses confirmed the formation of the Chi-TA-NPs. The PDI value (0.247 ± 0.03), size (567.0 ± 25.84 nm), and zeta potential (17.0 ± 5.86 mV) confirmed the relative stability of Chi-TA-NPs. A constant release profile in line with the Korsmeyer-Peppas model was detected for Chi-TA-NPs, such that approximately 44% of TA was released after 300 min. In addition, Chi-TA-NPs exhibited effective antimicrobial activity against the studied microbial strains, as manifested by MIC values ranging from 250 to 1000 µg/mL. Chi-TA-NPs induced cytotoxicity in liver tumor cell line, with an IC50 value of 500 µg/mL. Furthermore, Chi-TA-NPs considerably decreased the expression of DNMT1 (2.52-fold; p = 0.01), DNMT3A (2.96-fold; p = 0.004), and DNMT3B (2.94-fold; p < 0.0001). However, 5-methylcytosine levels in HepG2 cells were unaffected by Chi-TA-NPs treatment (p = 0.62). Finally, the antimicrobial, cytotoxic, and epigenetic effects of Chi-TA-NPs were more pronounced than those of free TA and the unloaded Chi-NPs. In conclusion, Chi-TA-NPs exhibit promising potential for reducing microbial growth and promoting cytotoxicity in liver cancer cells.
Collapse
Affiliation(s)
- Marzieh Rashidipour
- Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Saber Abbaszadeh
- Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mehdi Birjandi
- Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Naser Pajouhi
- Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | | | - Gholamreza Goudarzi
- Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Soroosh Shahryarhesami
- Functional Genome Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120, Heidelberg, Germany
| | - Mostafa Moradi Sarabi
- Hepatities Research Center, Department of Biochemistry and Genetics, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran.
| | - Esmaeel Babaeenezhad
- Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran.
| |
Collapse
|
46
|
Han SH, Ko JY, Jung S, Oh S, Kim DY, Kang E, Kim MS, Chun KH, Yoo KH, Park JH. VIM-AS1, which is regulated by CpG methylation, cooperates with IGF2BP1 to inhibit tumor aggressiveness via EPHA3 degradation in hepatocellular carcinoma. Exp Mol Med 2024; 56:2617-2630. [PMID: 39617786 DOI: 10.1038/s12276-024-01352-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 12/28/2024] Open
Abstract
Early tumor recurrence in hepatocellular carcinoma (HCC) remains a challenging area, as the mechanisms involved are not fully understood. While microvascular invasion is linked to early recurrence, established biomarkers for diagnosis and prognostication are lacking. In this study, our objective was to identify DNA methylation sites that can predict the outcomes of liver cancer patients and elucidate the molecular mechanisms driving HCC aggressiveness. Using DNA methylome data from HCC patient samples from the CGRC and TCGA databases, we pinpointed hypermethylated CpG sites in HCC. Our analysis revealed that cg02746869 acts as a crucial regulatory site for VIM-AS1 (vimentin antisense RNA1), a 1.8 kb long noncoding RNA. RNA sequencing of HCC cells with manipulated VIM-AS1 expression revealed EPHA3 as a pathogenic target of VIM-AS1, which performs an oncogenic function in HCC. Hypermethylation-induced suppression of VIM-AS1 significantly impacted HCC cell dynamics, particularly impairing motility and invasiveness. Mechanistically, reduced VIM-AS1 expression stabilized EPHA3 mRNA by enhancing the binding of IGF2BP1 to EPHA3 mRNA, leading to increased expression of EPHA3 mRNA and the promotion of HCC progression. In vivo experiments further confirmed that the VIM-AS1‒EPHA3 axis controlled tumor growth and the tumor microenvironment in HCC. These findings suggest that the downregulation of VIM-AS1 due to hypermethylation at cg02746869 increased EPHA3 mRNA expression via a m6A-dependent mechanism to increase HCC aggressiveness.
Collapse
MESH Headings
- Humans
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Liver Neoplasms/genetics
- DNA Methylation
- Receptor, EphA3/metabolism
- Receptor, EphA3/genetics
- CpG Islands
- Animals
- Gene Expression Regulation, Neoplastic
- RNA-Binding Proteins/metabolism
- RNA-Binding Proteins/genetics
- Mice
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Cell Line, Tumor
- Cell Proliferation
- Cell Movement/genetics
- RNA, Antisense/genetics
- RNA, Antisense/metabolism
Collapse
Affiliation(s)
- Su-Hyang Han
- Laboratory of Biomedical Genomics, Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Je Yeong Ko
- Molecular Medicine Laboratory, Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Sungju Jung
- Laboratory of Biomedical Genomics, Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Sumin Oh
- Laboratory of Biomedical Genomics, Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Do Yeon Kim
- Molecular Medicine Laboratory, Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Eunseo Kang
- Laboratory of Biomedical Genomics, Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Myung Sup Kim
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyung-Hee Chun
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyung Hyun Yoo
- Laboratory of Biomedical Genomics, Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea.
- Research Institute of Women's Health, Sookmyung Women's University, Seoul, 04310, Republic of Korea.
| | - Jong Hoon Park
- Molecular Medicine Laboratory, Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea.
| |
Collapse
|
47
|
Liu Z, Yuan J, Zeng Q, Wu Z, Han J. UBAP2 contributes to radioresistance by enhancing homologous recombination through SLC27A5 ubiquitination in hepatocellular carcinoma. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167481. [PMID: 39186963 DOI: 10.1016/j.bbadis.2024.167481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 08/03/2024] [Accepted: 08/19/2024] [Indexed: 08/28/2024]
Abstract
Radiotherapy stands as an effective method in the clinical treatment of hepatocellular carcinoma (HCC) patients. However, both primary and acquired radioresistance limit its clinical application in HCC. Therefore, investigating the mechanism of radioresistance may provide other options for treating HCC. Based on single-cell RNA sequencing (scRNA-seq) and HCC transcriptome datasets, 227 feature genes with prognostic value were selected to establish the tSNE score. The tSNE score emerged as an independent prognostic factor for HCC and correlated with cell proliferation and radioresistance-related biological functions. UBAP2 was identified as the most relevant gene with the tSNE score, consistently elevated in human HCC samples, and positively associated with patient prognosis. Functionally, UBAP2 knockdown impeded HCC development and reduced radiation resistance in vitro and in vivo. The ectopic expression of SLC27A5 reversed the effects of UBAP2. Mechanically, we uncovered that UBAP2, through the ubiquitin-proteasome system, decreased the homologous recombination-related gene RAD51, not the non-homologous end-joining (NHEJ)-related gene CTIP, by degrading the antioncogene SLC27A5, thereby generating radioresistance in HCC. The findings recapitulated that UBAP2 promoted HCC progression and radioresistance via SLC27A5 stability mediated by the ubiquitin-proteasome pathway. It was also suggested that targeting the UBAP2/SLC27A5 axis could be a valuable radiosensitization strategy in HCC.
Collapse
Affiliation(s)
- Zijian Liu
- Laboratory of Liquid Biopsy and Single Cell Research, Department of Radiation Oncology and Department of Head and Neck Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Jingsheng Yuan
- Liver Transplant Center, Transplant Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Laboratory of Liver Transplantation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Qiwen Zeng
- Liver Transplant Center, Transplant Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Laboratory of Liver Transplantation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhenru Wu
- Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital of Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiaqi Han
- Laboratory of Liquid Biopsy and Single Cell Research, Department of Radiation Oncology and Department of Head and Neck Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
48
|
Long Y, Wang W, Liu S, Wang X, Tao Y. The survival prediction analysis and preliminary study of the biological function of YEATS2 in hepatocellular carcinoma. Cell Oncol (Dordr) 2024; 47:2297-2316. [PMID: 39718737 DOI: 10.1007/s13402-024-01019-4] [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] [Accepted: 11/24/2024] [Indexed: 12/25/2024] Open
Abstract
PURPOSE Our study aims to develop and validate a novel molecular marker for the prognosis and diagnosis of hepatocellular carcinoma (HCC) MATERIALS & METHODS: We retrospectively analyzed mRNA expression profile and clinicopathological data of HCC patients fetched from The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO) and The International Cancer Genome Consortium (ICGC) datasets. Univariate Cox regression analysis was performed to collect differentially expressed mRNA (DEmRNAs) from HCC and non-tumor tissues, and YEATS2, a prognostic marker, was identified by further analysis. ROC curve, survival analysis and multivariate Cox regression analysis as well as nomograms were used to evaluate the prognosis of this gene. Finally, the biological function of this gene was preliminarily discussed by using single gene Gene Set Enrichment Analysis (GSEA), and the YEATS2 overexpression and knockdown hepatoma cell line was used to verify the results in vitro and in vivo. RESULTS Based on the clinical information of HCC in TCGA, GEO and ICGC databases, the gene YEATS2 with significant differences from HCC was identified. There was a statistical difference in the survival prognosis between the two databases and the ROC curve showed that the survival of HCC in both TCGA, GSE14520 and ICGC groups had a satisfactory predictive effect. Univariate and multivariate Cox regression analysis showed that YEATS2 was an independent prognostic factor for HCC, and Nomograms, which combined this prognostic feature with significant clinical features, provided an important reference for the clinical prognostic diagnosis of HCC. Next, we constructed overexpression and knockdown YEATS2 cell line in Hep3B and LM3 cells, and further proved that overexpression YEATS2 promote the proliferation and migration of HCC cells by CCK8, colony formation experiment, and transwell assays, and knockdown YEATS2 inhibited the proliferation and migration of HCC cells by CCK8, colony formation experiment, and transwell assays. Finally, the biological function of YEATS2 was preliminarily explored through GSEA analysis of a single gene, and it was found that it was significantly correlated with cell cycle and DNA repair, which provided us with ideas for further analysis. Furthermore, the knockdown of YEATS2 promoted radiation-induced DNA damage, enhanced radiosensitivity, and ultimately inhibited the proliferation of hepatocellular carcinoma cells in vitro and in vivo. CONCLUSIONS Our study identified a promising prognostic marker for hepatocellular carcinoma that is useful for clinical decision-making and individualized treatment.
Collapse
Affiliation(s)
- Yao Long
- Cancer Research Institute; School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Wei Wang
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, China
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Shouping Liu
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xiang Wang
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| | - Yongguang Tao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China.
- Cancer Research Institute; School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China.
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| |
Collapse
|
49
|
Chai B, Zhang A, Liu Y, Zhang X, Kong P, Zhang Z, Guo Y. KLF7 Promotes Hepatocellular Carcinoma Progression Through Regulating SLC1A5-Mediated Tryptophan Metabolism. J Cell Mol Med 2024; 28:e70245. [PMID: 39648156 PMCID: PMC11625504 DOI: 10.1111/jcmm.70245] [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: 05/09/2024] [Revised: 10/24/2024] [Accepted: 11/15/2024] [Indexed: 12/10/2024] Open
Abstract
Krüppel-like factor 7 is a transcriptional activator and acts as an oncogene in human cancers, including hepatocellular carcinoma (HCC). Tryptophan metabolism is important for HCC cell proliferation, metastasis, and invasion. It is unclear whether KLF7 could regulate Trp metabolism in HCC. In this study, we found that Trp metabolism was suppressed in HCC cells with KLF7 knockdown. The mRNA and protein levels of SLC1A5, SLC7A5, and TPH1, as well as the content of Trp and serotonin, were reduced after KLF7 knockdown, and were potentiated following KLF7 overexpression. Increasing the content of serotonin could restore the malignancy of tumour cells in vitro and tumour growth in vivo. Conversely, decreasing the content of serotonin suppressed HCC cell proliferation. The binding activity of KLF7 was on the promoter of SLC1A5, and KLF7 positively regulated the expression of SLC1A5. KLF7 contributed to the proliferation and migration of HCC cells by up-regulation of SLC1A5. Collectively, KLF7 promotes the progression of HCC through regulating Trp metabolism. The newly identified axis of KLF7/ SLC1A5 in HCC could represent a potential target for HCC therapy.
Collapse
Affiliation(s)
- Bao Chai
- Department of Gastroenterology, Shanxi Bethune Hospital, Shanxi Academy of Medical sciences, TongilShanxi HospitalThird Hospital of Shanxi Medical UniversityTaiyuanShanxiChina
| | - Anhong Zhang
- Department of SurgeryThe First Affiliated Hospital of Shanxi Medical UniversityTaiyuanShanxiChina
| | - Yang Liu
- Shanxi Medical UniversityTaiyuanShanxiChina
| | - Xi Zhang
- Shanxi Medical UniversityTaiyuanShanxiChina
| | - Pengzhou Kong
- Translational Medicine Research Center, Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Department of PathologyShanxi Medical UniversityTaiyuanShanxiChina
| | - Zhuowei Zhang
- College of Medical ImagingShanxi Medical UniversityTaiyuanShanxiChina
| | - Yarong Guo
- Department of Digestive System Oncology, Shanxi Bethune Hospital, Shanxi Academy of Medical sciences, TongilShanxi HospitalThird Hospital of Shanxi Medical UniversityTaiyuanShanxiChina
- Department of OncologyThe First Affiliated Hospital of Shanxi Medical UniversityTaiyuanShanxiChina
| |
Collapse
|
50
|
Kooragayala K, Wang M, Spitz FJ, Gandhi TV, Dibato J, Hong YK. Unmasking Disparities in Gallbladder Cancer Outcomes in the Disaggregated Asian American Population. Ann Surg Oncol 2024; 31:8699-8711. [PMID: 39259371 PMCID: PMC11549147 DOI: 10.1245/s10434-024-16168-x] [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: 02/08/2024] [Accepted: 08/23/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Gallbladder cancer (GBC) is associated with a high mortality rate. Asian American (AsA) are among the fastest-growing populations in the United States, yet little is known about the disparity of GBC within this cohort. This study identified trends in treatment and outcomes for GBC in a disaggregated fashion, specifically for this population. METHODS A retrospective analysis of the National Cancer Database (NCDB) between 2010 and 2019 examining all patients treated for gallbladder cancer was performed. Basic demographic factors were identified for patients of Caucasian, African American, and disaggregated Asian subpopulations. Survival curves were used to identify differences in median overall survival, and a multivariate analysis was performed to determine which factors impact overall survival. RESULTS A total of 1317 (5%) patients were of AsA origin. Median survival for the overall AsA population is 15.1 months compared with Caucasian (11.5 months) and African Americans (11.4 months) (p < 0.0001). Within the AsA groups, the Korean subpopulation had the lowest survival at 12.6 months, whereas Filipinos had the longest survival at 19.1 months (p < 0.0001). Patients of Filipino descent had the highest rate of surgical resection but lower chemotherapy utilization. Conversely, Korean patients had the highest utilization of multimodality therapy. Multivariate analysis demonstrated that belonging to Chinese, Filipino, or Indian ethnicity was associated with decreased risk of mortality. CONCLUSIONS There are disparate differences in survival for patients with GBC between AsA groups. Socioeconomic, genetic, and epigenetic factors may influence these differences. Further research is needed to delineate the causes of this disparity.
Collapse
Affiliation(s)
| | - Michael Wang
- Department of Surgery, Cooper University Hospital, Camden, NJ, USA
| | - Francis J Spitz
- Department of Surgery, Cooper University Hospital, Camden, NJ, USA
| | | | - John Dibato
- Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Young Ki Hong
- Department of Surgery, Cooper University Hospital, Camden, NJ, USA.
- Cooper Medical School of Rowan University, Camden, NJ, USA.
| |
Collapse
|