1
|
Hieromnimon M, Regan DP, Lokken RP, Schook LB, Gaba RC, Schachtschneider KM. Single and multi-omic characterization of a porcine model of ethanol-induced hepatic fibrosis. Epigenetics 2025; 20:2471127. [PMID: 40040391 PMCID: PMC11901410 DOI: 10.1080/15592294.2025.2471127] [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: 07/18/2024] [Revised: 02/10/2025] [Accepted: 02/18/2025] [Indexed: 03/06/2025] Open
Abstract
Cirrhosis is a form of end-stage liver disease characterized by extensive hepatic fibrosis and loss of liver parenchyma. It is most commonly the result of long-term alcohol abuse in the United States. Large animal models of cirrhosis, as well as of one of its common long-term sequelae, HCC, are needed to study novel and emerging therapeutic interventions. In the present study, liver fibrosis was induced in the Oncopig cancer model, a large animal HCC model, via intrahepatic, intra-arterial ethanol infusion. Liver sections from five fibrosis induced and five age-matched controls were harvested for RNA-seq (mRNA and lncRNA), small RNA-seq (miRNA), and reduced representation bisulfite sequencing (RRBS; DNA methylation). Single- and multi-omic analysis was performed to investigate the transcriptomic and epigenomic mechanisms associated with fibrosis deposition in this model. A total of 3,439 genes, 70 miRNAs, 452 lncRNAs, and 7,715 methylation regions were found to be differentially regulated through individual single-omic analysis. Pathway analysis indicated differentially expressed genes were associated with collagen synthesis and turnover, hepatic metabolic functions such as ethanol and lipid metabolism, and proliferative and anti-proliferative pathways including PI3K and BAX/BCL signaling pathways. Multi-omic latent variable analysis demonstrated significant concordance with the single-omic analysis. lncRNA's associated with UHRF1BP1L and S1PR1 genes were found to reliably discriminate the two arms of the study. These genes were previously implicated in human cancer development and vasculogenesis, respectively. These findings support the validity and translatability of this model as a useful preclinical tool in the study of alcoholic liver disease and its treatment.
Collapse
Affiliation(s)
- Mark Hieromnimon
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA
| | - Daniel P. Regan
- Flint Animal Cancer Center, Colorado State University, Fort Collins, CO, USA
| | - R. Peter Lokken
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Lawrence B. Schook
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Sus Clinicals Inc, Chicago, IL, USA
| | - Ron C. Gaba
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA
| | - Kyle M. Schachtschneider
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Sus Clinicals Inc, Chicago, IL, USA
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
2
|
Zhang X, Shao W, Gao Y, Wang X. Macrophage polarization-mediated PKM2/mTORC1/YME1L signaling pathway activation in fibrosis associated with Cardiorenal syndrome. Cell Signal 2025; 131:111664. [PMID: 39961408 DOI: 10.1016/j.cellsig.2025.111664] [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: 10/23/2024] [Revised: 12/16/2024] [Accepted: 02/14/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND Cardiorenal syndrome (CRS) is a complex condition characterized by the interplay between cardiac and renal dysfunction, often culminating in renal fibrosis. The role of macrophage polarization and its downstream effects in CRS-induced renal fibrosis remains an area of active investigation. METHODS Single-cell RNA sequencing (scRNA-seq) and immune infiltration analyses were employed to identify key immune cells and genes involved in renal fibrosis in CRS. Meta-analysis and pseudo-time analysis were conducted to validate the functional relevance of these genes. Functional studies utilizing CRISPR/Cas9 gene editing and lentiviral vectors assessed macrophage polarization and epithelial-to-mesenchymal transition (EMT). In vivo, a CRS mouse model was established, and fibrosis progression was tracked using histological and imaging methods. RESULTS The PKM2/mTORC1/YME1L signaling axis was identified as a critical pathway driving renal fibrosis, mediated by HIF-1α-induced M1 macrophage polarization. Inhibition of HIF-1α significantly alleviated renal fibrosis by restricting M1 polarization and suppressing the PKM2/mTORC1/YME1L axis. Co-culture models further demonstrated the involvement of EMT and metabolic reprogramming in affected cells. CONCLUSION Targeting the HIF-1α signaling pathway offers a promising therapeutic strategy for renal fibrosis by modulating macrophage polarization and the PKM2/mTORC1/YME1L axis.
Collapse
Affiliation(s)
- Xuefeng Zhang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan 030032, China.
| | - Wen Shao
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Yun Gao
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Xiaojun Wang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan 030032, China
| |
Collapse
|
3
|
Xu F, Qiu J, Liu N, Wei H, Gao Y, Fei Y, Xi J, Yu Z, Fan X, Chen L, Xia Y, Dou X. Therapeutic Potential of Raspberry Extract in High-Fat Diet-Induced Liver Injury via Apoptosis and AMPK/PPARα Pathways. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:9408-9423. [PMID: 40168586 DOI: 10.1021/acs.jafc.4c09593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
This study aimed to explore the efficacy and mechanisms of raspberry (Rubus idaeus L. fruit) aqueous extract (RE) in alleviating high-fat diet (HFD)-induced metabolic-associated fatty liver disease (MAFLD). The MAFLD mouse model was established to examine the effects of RE through liver transcriptome and metabolomics analysis. In this study, RE supplementation significantly alleviated HFD-induced liver injury, hepatosteatosis, inflammation, and insulin resistance. Liver transcriptome analysis demonstrated that RE supplementation favorably regulated signaling pathways involved in fatty acid metabolism and inflammation, including the AMPK signaling pathway, PPAR signaling pathway, apoptosis, etc. Furthermore, the injection of compound C, an antagonist of AMPK, notably reversed the hepatoprotective effects of RE, evidenced by increased lipid profile levels, accelerated fatty acid-related gene disorder, and increased positive tunnel staining area. Furthermore, liver metabolomics analysis demonstrated that RE treatment led to substantial enrichment of the liver tissue metabolite umbelliferone (UMB), which has the potential to ameliorate lipid accumulation and hepatocyte injury through the AMPK signaling pathway. In summary, RE intervention mitigated HFD-induced liver dysfunction in mice, with UMB likely being the primary component responsible for its therapeutic efficacy in the liver. In addition, this study provided new insights, suggesting that RE could be used as a promising therapeutic approach for modulating MAFLD via apoptosis and the AMPK/PPARα signaling pathway.
Collapse
Affiliation(s)
- Fangying Xu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Jiannan Qiu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Nian Liu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Huaxin Wei
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Yanyan Gao
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Yang Fei
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Jiale Xi
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Zhiling Yu
- Consun Chinese Medicines Research Centre for Renal Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 852, China
| | - Xiaohui Fan
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lin Chen
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Yongliang Xia
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Xiaobing Dou
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| |
Collapse
|
4
|
Miao YR, Yang XJ. Hepatocellular carcinoma resistance to tyrosine kinase inhibitors: Current status and perspectives. World J Gastrointest Oncol 2025; 17:101528. [PMID: 40235904 PMCID: PMC11995346 DOI: 10.4251/wjgo.v17.i4.101528] [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: 09/18/2024] [Revised: 12/31/2024] [Accepted: 01/08/2025] [Indexed: 03/25/2025] Open
Abstract
The study conducted by Wang et al, focuses on the role of Rho GTPase activating protein 12 (ARHGAP12), in hepatocellular carcinoma (HCC). This research reveals that ARHGAP12 expression, markedly elevated in malignant cells of HCC, correlates strongly with adverse outcomes for patients. Furthermore, the study illustrates that ARHGAP12 enhances the ability of HCC cells to invade and contributes to their resistance to tyrosine kinase inhibitors (TKIs) through modulation of the focal adhesion pathway. To comprehensively investigate the relationship between ARHGAP12 and TKI resistance, this study integrates single-cell and bulk RNA sequencing methodologies along with data from tumor immune single-cell hub 2, Gene Expression Omnibus, The Cancer Genome Atlas, CellMiner, Genomics of Drug Sensitivity in Cancer 2, as well as immunohistochemical staining and proteomic analyses. Statistical analyses, including the Wilcoxon rank-sum test and receiver operating characteristic curve analysis, were employed to evaluate the correlation between ARHGAP12 expression levels and clinical parameters, as well as drug sensitivity. It is evident that a more profound exploration of the molecular dynamics of HCC, especially those related to resistance against TKIs, is essential.
Collapse
Affiliation(s)
- Yu-Run Miao
- The First Clinical Medical School, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
- Second Ward of General Surgery, Gansu Province People Hospital, Lanzhou 730000, Gansu Province, China
| | - Xiao-Jun Yang
- Second Ward of General Surgery, Gansu Province People Hospital, Lanzhou 730000, Gansu Province, China
| |
Collapse
|
5
|
Wang Y, Wang H, Li Q, Zhang Y, Dai R, Wu J, Zhang Y, Zhang X, Zhao L, Liu J. Identification of Novel Cyclobutane-Based Derivatives as Potent Acetyl-CoA Carboxylase Allosteric Inhibitors for Nonalcoholic Steatohepatitis Drug Discovery. J Med Chem 2025. [PMID: 40227434 DOI: 10.1021/acs.jmedchem.5c00259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Nonalcoholic steatohepatitis (NASH) has become a leading cause of liver fibrosis and hepatocellular carcinoma; however, there are no efficient drugs for NASH therapy. Acetyl-CoA carboxylase (ACC) is a crucial enzyme regulating lipid metabolism that is considered as a potential target for NASH treatment. Allosteric inhibitors target nonfunctional sites, which tend to be highly variable in protein families; thus, allosteric inhibitors are explored as an important source of drug candidates. Herein, several hotspot residues are initially identified by utilizing molecular dynamic simulation, MM-GBSA calculation, and alanine mutation. Then, focusing on the interaction with hotspot residues, several cyclobutane-based ACC allosteric inhibitors are designed, synthesized, and biologically evaluated. Among them, B1 demonstrates potent ACC inhibitory activity in vitro, a higher distribution in liver than in other tissues, and a potent therapeutic effect for NASH in vivo, making it a promising candidate for the treatment of NASH.
Collapse
Affiliation(s)
- Yazhou Wang
- R & D Center, Nanjing Sanhome Pharmaceutical Co. Ltd., Nanjing 210049, China
| | - Hai Wang
- R & D Center, Nanjing Sanhome Pharmaceutical Co. Ltd., Nanjing 210049, China
| | - Qingqing Li
- R & D Center, Nanjing Sanhome Pharmaceutical Co. Ltd., Nanjing 210049, China
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ying Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Rupeng Dai
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jun Wu
- R & D Center, Nanjing Sanhome Pharmaceutical Co. Ltd., Nanjing 210049, China
| | - Yanan Zhang
- R & D Center, Nanjing Sanhome Pharmaceutical Co. Ltd., Nanjing 210049, China
| | - Xiaomeng Zhang
- R & D Center, Nanjing Sanhome Pharmaceutical Co. Ltd., Nanjing 210049, China
| | - Liwen Zhao
- R & D Center, Nanjing Sanhome Pharmaceutical Co. Ltd., Nanjing 210049, China
| | - Jian Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- School of Artificial Intelligence and Information Technology, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| |
Collapse
|
6
|
Zhu JF, Wang YQ, Yang SM, Wang YL, Hu Y, Dai XY. Exploring the mechanism of Bruceine D against cervical cancer by network pharmacology and the effect of Bruceine D on the EGFR pathway. J Pharm Biomed Anal 2025; 262:116887. [PMID: 40239560 DOI: 10.1016/j.jpba.2025.116887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/25/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
Cervical cancer (CC) remains a formidable challenge in oncology due to its high incidence and mortality rates. Despite recent advances in treatment, an immediate necessity exists for innovating advanced pharmacological interventions boasting augmented effectiveness. Bruceine D (BD), a quassinoid derived from the traditional Chinese medicinal plant Brucea javanica, has been demonstrated to possess notable anticancer properties against a range of malignant conditions, including lung, liver, leukemia, and pancreatic cancers. However, its specific effects on CC have not been thoroughly explored. This study sought to decode the effects of BD on CC through a combined method involving molecular docking analysis, network pharmacology, and data mining. From the PharmMapper database, we identified 58 potential targets of BD, and through GeneCards, we pinpointed 14 intersecting targets relevant to CC. A protein-protein interaction (PPI) network highlighted pivotal targets such as ESR1, HSP90AA1, ANXA5, EGFR, CASP7, and CCNA2. GO and KEGG enrichment analyses underscored significant biological processes and pathways, notably the EGFR signaling pathway. Molecular docking analysis revealed a strong binding affinity of BD to EGFR. Cell-based assays demonstrated that BD potently curtailed the viability, colony formation, adhesion, and mobility of Hela and Caski cells, escalating apoptosis in a dose-proportional manner. Supplementary evidence via western blot evaluations underscored BD's capability to obstruct the EGFR signaling pathway. These findings suggest that BD exerts potent anticancer effects against CC through multiple mechanisms, positioning it as a promising therapeutic agent for further investigation and clinical validation.
Collapse
Affiliation(s)
- Ju-Fan Zhu
- Institute of Organoid Technology, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Yuan-Qiu Wang
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Si-Meng Yang
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yu-Li Wang
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yan Hu
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| | - Xin-Yue Dai
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
7
|
Li K, Dai YJ, Zhang H, Zhang Z. YAP1 activates SLC2A1 transcription and augments the malignant behavior of colorectal cancer cells by activating the Wnt/β-catenin signaling pathway. Cell Div 2025; 20:8. [PMID: 40186232 PMCID: PMC11969700 DOI: 10.1186/s13008-025-00148-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 03/18/2025] [Indexed: 04/07/2025] Open
Abstract
OBJECTIVE This paper examined the role of solute carrier family 2 member 1 (SLC2A1) in colorectal cancer (CRC) progression, focusing on its expression levels, functional implications, and regulatory mechanisms involving Yes-associated protein 1 (YAP1) and the Wnt signaling pathway. METHODS GEO datasets (GSE14297, GSE18462, GSE40367) were analyzed to identify genes linked to metastasis in CRC, and TCGA-COAD system was used to analyze the expression pattern and prognostic values of SLC2A1 in CRC. Functional studies were conducted using CRC cell lines (Caco-2 and SW480). Cell viability, migration and invasion, and apoptosis were examined using EdU assays, Transwell assays, and flow cytometry. YAP1's regulatory role on SLC2A1 was investigated using ChIP-qPCR and luciferase reporter assays. The Wnt/β-catenin agonist SKL2001 was used for functional rescue experiments. RESULTS SLC2A1 was upregulated in CRC cells, and its upregulation was associated with tumor metastasis and unfavorable outcomes according to bioinformatics. Knockdown of SLC2A1 resulted in reduced cell viability, decreased migration, and increased apoptosis in Caco-2 and SW480 cells. Additionally, YAP1 was identified as a transcriptional activator of SLC2A1. Knockdown of YAP1 decreased SLC2A1 expression and reduced expression of Wnt target genes, thus suppressing malignant behavior of tumor cells. However, further overexpression of SLC2A1 restored cell viability and migration in YAP1-deficient cells. The YAP1- SLC2A1 axis activated the Wnt/β-catenin by reducing GSK3β activity. CONCLUSION SLC2A1 is critical in CRC progression, with YAP1 serving as a key regulator of its expression and function. The YAP1-SLC2A1-Wnt axis represents a potential therapeutic target for CRC, providing insights into metabolic adaptations that support tumor growth and metastasis.
Collapse
Affiliation(s)
- Kunpeng Li
- Zhongda Hospital of Southeast University, No 87 Dingjiaqiao, Nanjing, 210009, Jiangsu, PR China
| | - Ya-Jie Dai
- Department of General Surgery, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu, PR China
| | - Haifeng Zhang
- Department of General Surgery, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu, PR China
| | - Zhigang Zhang
- Department of General Surgery, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu, PR China.
| |
Collapse
|
8
|
Li Z, Zhang C, Huang G, Zhang Z, Wang Q, Liu X, Qin Y, Zhou H, Hou A, He J, Li L, Hu X, Ding X. Deletion of Tfap2a in hepatocytes and macrophages promotes the progression of hepatocellular carcinoma by regulating SREBP1/FASN/ACC pathway and anti-inflammatory effect of IL10. Cell Death Dis 2025; 16:245. [PMID: 40180937 PMCID: PMC11968862 DOI: 10.1038/s41419-025-07500-8] [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: 11/12/2024] [Revised: 02/13/2025] [Accepted: 03/05/2025] [Indexed: 04/05/2025]
Abstract
The transcription factor AP-2α plays a crucial role in the control of tumor development and progression, and suppresses the proliferation and migration of hepatocellular carcinoma (HCC). However, the detailed function and mechanisms of AP-2α in the pathogenesis of HCC are still elusive. In the current study, we investigated the role of AP-2α regulation in liver injury-mediated HCC development. Downregulation of Tfap2a expression was found in the livers of DEN/CCl4-induced fibrosis and HCC mouse model. Hepatocyte (Alb-Cre), hepatic stellate cell (HSC) (Lrat-Cre) and macrophage (LysM-Cre) specific Tfap2a knockout mice were generated, respectively. Conditional knockout of Tfap2a was able to promote hepatic steatosis in Tfap2aΔHep and Tfap2aΔMΦ mice, but not in Tfap2aΔHSC mice fed with normal chow. Tfap2aΔHep and Tfap2aΔMΦ mice treated with DEN/CCl4 for 6 months increased tumor burden compared to Tfap2a flox controls. Tfap2a-deleted macrophages or hepatocytes could enhance lipid droplet (LD) accumulation in hepatocytes. Mechanistically, AP-2α binds to the promoter regions of SREBP1/ACC/FASN and inhibits hepatic lipid de novo synthesis. Deletion of Tfap2a in macrophages enhances polarization of M1 macrophages with increased iNOS expression but decreased CD206 expression, which resulted in increased pro-inflammatory cytokines and decreased anti-inflammatory factors, especially the hepatoprotective factor IL-10. The m6A modification writer WTAP could reduce the mRNA stability of AP-2α in a reader YTHDC1-dependent manner, whereas knockdown of WTAP or YTHDC1 enhances AP-2α expression and decreases lipid accumulation in HCC cells. Clinically, AP-2α expression negatively correlates with the expression of FASN, WTAP, YTHDC1 and the development of liver disease. Taken together, hepatocyte- or macrophage-specific deletion of Tfap2a promotes hepatic steatosis, fibrosis, and the development of HCC. These results suggest that AP-2α has been identified as a novel therapeutic target in fibrosis and inflammation-related HCC, exerting anti-lipogenesis, anti-inflammatory, and anti-tumor multi-roles.
Collapse
Affiliation(s)
- Zhiwei Li
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha, 410081, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Chun Zhang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha, 410081, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Guixiang Huang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha, 410081, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Zixin Zhang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha, 410081, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Qinghao Wang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha, 410081, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Xiran Liu
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha, 410081, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Yanling Qin
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha, 410081, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Hao Zhou
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha, 410081, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Anyi Hou
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha, 410081, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
| | - Jun He
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defects Prevention and Control, Changsha Hospital for Maternal & Child Health Care Affiliated to Hunan Normal University, Changsha, 410007, China
| | - Limin Li
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
- College of Engineering and Design, Hunan Normal University, Changsha, 410081, China
| | - Xiang Hu
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha, 410081, China
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China
- Peptide and small molecule drug R&D platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, China
| | - Xiaofeng Ding
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha, 410081, China.
- Institute of Interdisciplinary Studies, Hunan Normal University, Changsha, 410081, China.
- Peptide and small molecule drug R&D platform, Furong Laboratory, Hunan Normal University, Changsha, 410081, China.
| |
Collapse
|
9
|
Peng Z, Xu S, Wang H, Huang Y, Liu S, Jiao Z, Lin M, Zhu P, Chen Y, Shi Y, Wang Y, Li Y, Yuan W, Wu X, Jiang Z, Li F, Fan X. Identification of GDP as a small inhibitory molecule in HepG2 cells by non‑targeted metabolomics analysis. Oncol Lett 2025; 29:178. [PMID: 39990806 PMCID: PMC11843412 DOI: 10.3892/ol.2025.14924] [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: 07/22/2024] [Accepted: 12/12/2024] [Indexed: 02/25/2025] Open
Abstract
Identifying the mechanism by which lipid metabolism regulates cancer may offer a novel approach for therapeutic intervention. It has previously been identified that a lipid metabolism-related factor, namely fatty acid hydroxylase domain containing 2 (FAXDC2), is downregulated in various types of cancer, and inhibits the proliferation and migration of liver cancer cells through a mechanism associated with ERK. The liver is important for lipid metabolism, and FAXDC2 is involved in the synthesis of cholesterol and sphingomyelin. However, the functional mechanism by which FAXDC2 influences liver cancer cells through metabolic processes and ERK signaling remains unclear. Therefore, the present study induced the overexpression of FAXDC2 in HepG2 liver cancer cells and performed a metabolomics analysis. This identified guanosine diphosphate (GDP) as a significantly altered metabolite. Using AlphaFold3, a robust interaction was predicted between FAXDC2 and GDP, which lead to the hypothesis that GDP may mediate the inhibitory effects of FAXDC2 on liver cancer cells by directly modulating the functional properties of the cells, thereby influencing their behavior and progression. Cell Counting Kit-8 assays were used to study the impact of elevated GDP concentrations on HepG2 cell growth. The results revealed a gradual reduction in the viability of HepG2 cells as the GDP concentration increased. In addition, western blotting showed that GDP treatment was accompanied by a significant downregulation of cyclin dependent kinase 4 and cyclin D1 expression levels, and Transwell experiments revealed that GDP treatment significantly decreased the invasion of HepG2 cells. Treatment with GDP also significantly inhibited the expression of ERK. In summary, the present study is the first to indicate that GDP is a metabolic small molecule with inhibitory activity in cancer cells, which has previously been overlooked in tumor metabolic reprogramming. The study findings offer new insights and strategies for the diagnosis and treatment of liver cancer, and potentially other types of cancer.
Collapse
Affiliation(s)
- Zhilin Peng
- The Center for Heart Development, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Siting Xu
- The Center for Heart Development, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Haocheng Wang
- The Center for Heart Development, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Yanli Huang
- The Center for Heart Development, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Siyuan Liu
- The Center for Heart Development, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Zhongbei Jiao
- The Center for Heart Development, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Mei Lin
- The Center for Heart Development, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Ping Zhu
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangdong Provincial People's Hospital Affiliated to Southern Medical University, Guangzhou, Guangdong 510100, P.R. China
| | - Yu Chen
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangdong Provincial People's Hospital Affiliated to Southern Medical University, Guangzhou, Guangdong 510100, P.R. China
| | - Yan Shi
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangdong Provincial People's Hospital Affiliated to Southern Medical University, Guangzhou, Guangdong 510100, P.R. China
| | - Yuequn Wang
- The Center for Heart Development, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Yongqing Li
- The Center for Heart Development, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Wuzhou Yuan
- The Center for Heart Development, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Xiushan Wu
- The Center for Heart Development, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Zhigang Jiang
- The Center for Heart Development, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Fang Li
- The Center for Heart Development, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| | - Xiongwei Fan
- The Center for Heart Development, College of Life Science, Hunan Normal University, Changsha, Hunan 410081, P.R. China
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, P.R. China
| |
Collapse
|
10
|
Yang J, Zhang H, Yang L, Yi S, Zhang T. Investigating the Role of Scd1 in OSAHS-Induced Vascular Changes. J Biochem Mol Toxicol 2025; 39:e70221. [PMID: 40233246 DOI: 10.1002/jbt.70221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 01/16/2025] [Accepted: 03/03/2025] [Indexed: 04/17/2025]
Abstract
This study investigates the role of Stearoyl-CoA Desaturase-1 (Scd1) in vascular remodeling associated with Obstructive Sleep Apnea-Hypopnea Syndrome (OSAHS) using multi-omics analysis. Transcriptomic and metabolomic datasets of OSAHS mouse models were analyzed to identify differentially expressed genes and metabolites, followed by functional enrichment analysis. Key genes were screened using weighted gene correlation network analysis (WGCNA) and machine learning, and a PPI network was constructed. An OSAHS mouse model was developed via intermittent hypoxia exposure. Human aortic smooth muscle cells (HASMCs) were subjected to hypoxia/reoxygenation cycles to simulate OSAHS in vitro. Blood pressure, plasma lipid profiles, histological changes in the thoracic aorta, and Scd1 protein expression were assessed. CCK-8 and Transwell assays evaluated HASMC proliferation and migration. Scd1 was identified as a critical factor in OSAHS-related vascular remodeling, with its expression significantly upregulated in vascular tissues of OSAHS mice. Metabolomic analysis revealed changes in fatty acid metabolism. Scd1 knockdown reduced blood pressure, lipid levels, aortic wall thickness, collagen deposition, elastic fiber accumulation, and mucin deposition in vivo. In vitro, hypoxia/reoxygenation cycles elevated Scd1 expression, while Scd1 knockdown inhibited HASMC proliferation and migration. Multi-omics analyses highlight Scd1 as a key regulator in OSAHS-associated vascular remodeling, driving pathological changes through its upregulation. These findings suggest Scd1 as a potential therapeutic target for managing OSAHS-related vascular pathologies.
Collapse
MESH Headings
- Stearoyl-CoA Desaturase/metabolism
- Stearoyl-CoA Desaturase/genetics
- Animals
- Mice
- Humans
- Vascular Remodeling
- Sleep Apnea, Obstructive/pathology
- Sleep Apnea, Obstructive/metabolism
- Sleep Apnea, Obstructive/genetics
- Sleep Apnea, Obstructive/enzymology
- Male
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Cell Proliferation
- Mice, Inbred C57BL
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
- Disease Models, Animal
- Cell Movement
Collapse
Affiliation(s)
- Jing Yang
- Department of Respiratory and Critical Care Medicine, South China Hospital Affiliated to Shenzhen University, Shenzhen, China
| | - Hui Zhang
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Lulu Yang
- Department of General Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Shen Yi
- Department of General Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ting Zhang
- Department II of Respiratory and Critical Care in Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| |
Collapse
|
11
|
Eslam M, Fan JG, Yu ML, Wong VWS, Cua IH, Liu CJ, Tanwandee T, Gani R, Seto WK, Alam S, Young DY, Hamid S, Zheng MH, Kawaguchi T, Chan WK, Payawal D, Tan SS, Goh GBB, Strasser SI, Viet HD, Kao JH, Kim W, Kim SU, Keating SE, Yilmaz Y, Kamani L, Wang CC, Fouad Y, Abbas Z, Treeprasertsuk S, Thanapirom K, Al Mahtab M, Lkhagvaa U, Baatarkhuu O, Choudhury AK, Stedman CAM, Chowdhury A, Dokmeci AK, Wang FS, Lin HC, Huang JF, Howell J, Jia J, Alboraie M, Roberts SK, Yoneda M, Ghazinian H, Mirijanyan A, Nan Y, Lesmana CRA, Adams LA, Shiha G, Kumar M, Örmeci N, Wei L, Lau G, Omata M, Sarin SK, George J. The Asian Pacific association for the study of the liver clinical practice guidelines for the diagnosis and management of metabolic dysfunction-associated fatty liver disease. Hepatol Int 2025; 19:261-301. [PMID: 40016576 DOI: 10.1007/s12072-024-10774-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 12/28/2024] [Indexed: 03/01/2025]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) affects over one-fourth of the global adult population and is the leading cause of liver disease worldwide. To address this, the Asian Pacific Association for the Study of the Liver (APASL) has created clinical practice guidelines focused on MAFLD. The guidelines cover various aspects of the disease, such as its epidemiology, diagnosis, screening, assessment, and treatment. The guidelines aim to advance clinical practice, knowledge, and research on MAFLD, particularly in special groups. The guidelines are designed to advance clinical practice, to provide evidence-based recommendations to assist healthcare stakeholders in decision-making and to improve patient care and disease awareness. The guidelines take into account the burden of clinical management for the healthcare sector.
Collapse
Affiliation(s)
- Mohammed Eslam
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Westmead, NSW, 2145, Australia.
| | - Jian-Gao Fan
- Center for Fatty Liver, Department of Gastroenterology, Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming-Lung Yu
- Hepatobiliary Division, Department of Internal MedicineCollege of Medicine and Center for Liquid Biopsy and Cohort ResearchFaculty of Internal Medicine and Hepatitis Research Center, School of Medicine, College of MedicineSchool of Medicine and Doctoral Program of Clinical and Experimental Medicine, College of Medicine and Center of Excellence for Metabolic Associated Fatty Liver Disease, Kaohsiung Medical University, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Vincent Wai-Sun Wong
- Medical Data Analytics Centre, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, Chinese University of Hong Kong, Hong Kong, China
| | - Ian Homer Cua
- Institute of Digestive and Liver Diseases, St. Luke's Medical Center, Global City, Philippines
| | - Chun-Jen Liu
- Division of Gastroenterology and Hepatology, Department of Internal MedicineHepatitis Research CenterGraduate Institute of Clinical Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Tawesak Tanwandee
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Rino Gani
- Department of Internal Medicine, Hepatobiliary Division, Dr. Cipto Mangunkusumo National General Hospital, Universitas Indonesia, Pangeran Diponegoro Road No. 71St, Central Jakarta, 10430, Indonesia
| | - Wai-Kay Seto
- Department of Medicine, School of Clinical Medicine, State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Shahinul Alam
- Department of Hepatology, Bangabandhu Sheikh Mujib Medical University, Shahbag, Dhaka, Bangladesh
| | - Dan Yock Young
- Department of Medicine, Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore
| | - Saeed Hamid
- Department of Medicine, Aga Khan University, Karachi, Pakistan
| | - Ming-Hua Zheng
- MAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Diagnosis and Treatment for The Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, China
| | - Takumi Kawaguchi
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Wah-Kheong Chan
- Gastroenterology and Hepatology Unit, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Diana Payawal
- Department of Medicine, Cardinal Santos Medical Center, Mandaluyong, Philippines
| | - Soek-Siam Tan
- Department of Hepatology, Selayang Hospital, Batu Caves, Malaysia
| | - George Boon-Bee Goh
- Department of Gastroenterology and Hepatology, Singapore General Hospital, Singapore, Singapore
- Medicine Academic Clinical Program, Duke-NUS Medical School, Singapore, Singapore
| | - Simone I Strasser
- AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Hang Dao Viet
- Internal Medicine Faculty, Hanoi Medical University, Hanoi, Vietnam
| | - Jia-Horng Kao
- Graduate Institute of Clinical MedicineDepartment of Internal MedicineHepatitis Research CenterDepartment of Medical Research, National Taiwan University College of Medicine, National Taiwan University, National Taiwan University Hospital, 1 Chang-Te Street, 10002, Taipei, Taiwan
| | - Won Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul Metropolitan Government Boramae Medical Center, Seoul, Republic of Korea
| | - Seung Up Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Severance Hospital, 50-1, Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea
| | - Shelley E Keating
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Yusuf Yilmaz
- Department of Gastroenterology, School of Medicine, Recep Tayyip Erdoğan University, Rize, Turkey
| | | | - Chia-Chi Wang
- Buddhist Tzu Chi Medical Foundation and School of Medicine, Taipei Tzu Chi Hospital, Tzu Chi University, Taipei, Taiwan
| | - Yasser Fouad
- Department of Gastroenterology, Hepatology and Endemic Medicine, Faculty of Medicine, Minia University, Cairo, Egypt
| | - Zaigham Abbas
- Department of Hepatogastroenterology, Dr.Ziauddin University Hospital, Clifton, Karachi, Pakistan
| | | | | | - Mamun Al Mahtab
- Department of Hepatology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - Undram Lkhagvaa
- Department of Health Policy, School of Public Health, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Oidov Baatarkhuu
- Department of Infectious Diseases, School of Medicine, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Ashok Kumar Choudhury
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, 110070, India
| | | | - Abhijit Chowdhury
- Department of Hepatology, School of Digestive and Liver Diseases, Institute of Post Graduate Medical Education and Research, Kolkata, India
| | - A Kadir Dokmeci
- Department of Medicine, Ankara University School of Medicine, Ankara, Turkey
| | - Fu-Sheng Wang
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Chinese PLA Medical School, Chinese PLA General Hospital, Beijing, 100039, China
| | - Han-Chieh Lin
- Division of Gastroenterology and Hepatology, Department of Medicine, Institute of Clinical Medicine, School of Medicine, Taipei Veterans General Hospital, National Yang-Ming Chiao Tung University, No. 201, Section 2, Shipai RdNo. 155, Section 2, Linong St, Beitou District, Taipei City, 112, Taiwan
| | - Jee-Fu Huang
- Hepatobiliary Division, Department of Internal MedicineCollege of Medicine and Center for Liquid Biopsy and Cohort ResearchFaculty of Internal Medicine and Hepatitis Research Center, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jess Howell
- Burnet Institute, Melbourne, VIC, 3004, Australia
- Department of Epidemiology and Preventive Medicine, Monash University, Clayton, VIC, 3008, Australia
- Department of Medicine, The University of Melbourne, Parkville, VIC, 3050, Australia
- Department of Gastroenterology, St Vincent's Hospital Melbourne, Melbourne, VIC, 3165, Australia
| | - Jidong Jia
- Liver Research Center, Beijing Key Laboratory of Translational Medicine On Liver Cirrhosis, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Mohamed Alboraie
- Department of Internal Medicine, Al-Azhar University, Cairo, 11884, Egypt
| | - Stuart K Roberts
- Department of Gastroenterology and Hepatology, Central Clinical School, The Alfred, Monash University, Melbourne, Australia
| | - Masato Yoneda
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Hasmik Ghazinian
- Gastroenterology and Hepatology Department, Yerevan Medical Scientific Center, Yerevan, Armenia
| | - Aram Mirijanyan
- Gastroenterology and Hepatology Department, Yerevan Medical Scientific Center, Yerevan, Armenia
| | - Yuemin Nan
- Department of Traditional and Western Medical Hepatology, Third Hospital of Hebei Medical University, Shijiazhuang, China
| | | | - Leon A Adams
- Medical School, Faculty of Medicine and Health Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Gamal Shiha
- Hepatology and Gastroenterology Unit, Internal Medicine Department, Faculty of Medicine, Mansoura University, Egyptian Liver Research Institute and Hospital (ELRIAH), Sherbin, El Mansoura, Egypt
| | - Manoj Kumar
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Necati Örmeci
- Department of Gastroenterohepatology, Istanbul Health and Technology University, Istanbul, Turkey
| | - Lai Wei
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - George Lau
- Humanity and Health Medical Group, Humanity and Health Clinical Trial Center, Hong Kong SAR, China
- The Fifth Medical Center of Chinese, PLA General Hospital, Beijing, 100039, China
| | - Masao Omata
- Department of Gastroenterology, Yamanashi Central Hospital, Yamanashi, Japan
- University of Tokyo, Tokyo, Japan
| | - Shiv K Sarin
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India.
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Westmead, NSW, 2145, Australia
| |
Collapse
|
12
|
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
|
13
|
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
|
14
|
Gu XY, Zhou ZJ, Yao H, Yang JL, Gu J, Mu R, Zhao LJ. The role of transketolase in the immunotherapy and prognosis of hepatocellular carcinoma: a multi-omics approach. Front Immunol 2025; 16:1529029. [PMID: 40230848 PMCID: PMC11994433 DOI: 10.3389/fimmu.2025.1529029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 03/12/2025] [Indexed: 04/16/2025] Open
Abstract
Objective To explore the role of transketolase (TKT) in the immunotherapy and prognosis of hepatocellular carcinoma (HCC). Materials and methods TKT expression across various cancers and its associations with tumor immunity and prognosis were analyzed using nomogram models. A multi-omics approach was employed, including bulk RNA-seq analysis, methylation profiling, single-cell analysis, and spatial transcriptomics. Experimental methods included RT-qPCR, siRNA transfection, luciferase reporter assay, and chromatin immunoprecipitation. Results TKT was significantly upregulated in multiple cancers and correlated with immune cell infiltration, particularly in HCC. Elevated TKT expression was associated with poor overall survival (OS) in HCC and was an independent prognostic factor (p < 0.05). Drug sensitivity analysis suggested that higher TKT expression was associated with reduced sensitivity to several chemotherapeutic agents, including sorafenib (p < 0.01). Furthermore, hypermethylation of the TKT promoter and low TKT expression were linked to improved OS in HCC (log-rank test p = 0.005). Single-cell analysis revealed that TKT was predominantly expressed in the monocyte/macrophage cluster associated with HCC, and pseudo-time series analysis highlighted TKT's role in cell differentiation within this cluster. Spatial transcriptomics confirmed the close association between TKT and macrophage distribution in HCC. Moreover, STAT3 was found to directly regulate TKT expression by binding to its promoter region. Conclusion Our findings suggest that TKT may play a role in tumor immunity and prognosis in HCC. Although these results provide insights into the potential involvement of TKT in immune cell infiltration and survival outcomes, further studies are required to fully elucidate its role in immunotherapy.
Collapse
Affiliation(s)
- Xuan-Yu Gu
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zheng-Jun Zhou
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Hua Yao
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jia-Li Yang
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jin Gu
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Rui Mu
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Li-Jin Zhao
- Department of General Surgery, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
15
|
Wang YY, Yang WX, Cai JY, Wang FF, You CG. Comprehensive molecular characteristics of hepatocellular carcinoma based on multi-omics analysis. BMC Cancer 2025; 25:573. [PMID: 40159482 PMCID: PMC11956240 DOI: 10.1186/s12885-025-13952-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 03/17/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND The high heterogeneity of hepatocellular carcinoma (HCC) poses challenges for precision treatment strategies. This study aims to use multi-omics methodologies to better understand its pathogenesis and discover biomarkers. METHODS Quantitative proteomics was used to investigate hepatocellular carcinoma tissues (HCT) and their corresponding adjacent non-tumor tissues (DNT), obtained from six HCC patients. Untargeted metabolomics was applied to analyze the metabolic profiles of HCT and DNT of ten HCC patients. Statistical analyses, such as the Student's t-test, were performed to identify differentially expressed proteins (DEPs) and metabolites (DEMs) between the two groups. The functions and metabolic pathways involving DEPs and DEMs were annotated and enriched using the gene ontology (GO) and kyoto encyclopedia of genes and genomes (KEGG) databases. Bioinformatics methods were then utilized to analyze consistency between proteomics and metabolomics results, leading to identification of potential biomarkers along with key altered pathways associated with HCC. RESULTS This study identified 1556 DEPs between HCT and DNT samples. These DEPs were primarily enriched in crucial biological pathways such as amino acid degradation, fatty acid metabolism, and DNA replication. Subsequently, the analysis of metabolomics identified 500 DEMs that mainly participated in glycerophospholipid metabolism, the phospholipase D signaling pathway, and choline metabolism related to cancer. Integrated analysis of proteomics and metabolomics data unveiled significant dysfunctions in bile secretion, multiple amino acid and fatty acid metabolic pathways among HCC patients. Further investigation revealed that five proteins (PTP4A3, B4GALT5, GAB1, ME2, and PKM) along with seven metabolites (PI(6 keto-PGF1alpha/16:0), 13, 16, 19-docosatrienoic acid, PA(18:2(9Z, 12Z)/20:1(11Z)), Citric Acid, PG(20:3(6, 8, 11)-OH(5)/18:2(9Z, 12Z)), Spermidine, and N2-Acetylornithine) exhibited excellent diagnostic efficiency for HCC and could serve as its potential biomarkers. CONCLUSION Our integrated proteome and metabolome analysis revealed 10 key HCC-related pathways and proposed 12 potential biomarkers, which may enhance our understanding of HCC pathophysiology and be helpful in facilitating early diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Ying-Ying Wang
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, 730030, China
| | - Wan-Xia Yang
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, 730030, China
| | - Jiang-Ying Cai
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, 730030, China
| | - Fang-Fang Wang
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, 730030, China
| | - Chong-Ge You
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, Gansu Province, 730030, China.
| |
Collapse
|
16
|
Zhao QY, Liu WJ, Wang JG, Li H, Lv JL, Wang Y, Wang C. Increasing cisplatin exposure promotes small-cell lung cancer transformation after a shift from glucose metabolism to fatty acid metabolism. J Cancer Res Clin Oncol 2025; 151:126. [PMID: 40155472 PMCID: PMC11953189 DOI: 10.1007/s00432-025-06164-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 03/10/2025] [Indexed: 04/01/2025]
Abstract
OBJECTIVES Lung cancer is a leading cause of global cancer mortality. Clinical observations reveal that histological transformation from non-small cell lung cancer (NSCLC) to small cell lung cancer (SCLC) is accompanied by mutations in TP53 and RB1. By applying gradually increasing cisplatin concentrations to mimic the escalating drug pressure within the tumor microenvironment, this study investigated the link between phenotypic transformation to SCLC in cisplatin-resistant human lung adenocarcinoma cells and alterations in cellular energy production pathways. MATERIALS AND METHODS We established two cisplatin-resistant NSCLC cell lines with varying resistance levels. RNAseq analyses identified TP53 and RB1 gene mutations. Comprehensive functional assays were performed to characterize A549/DDP1 μg/mL and A549/DDP3 μg/mL cells, focusing on proliferation and migratory capabilities. Cellular bioenergetics were assessed through glycolysis and oxidative phosphorylation analyses. Western blotting was employed to examine epithelial-mesenchymal transition (EMT), glucose metabolism, and lipid metabolism markers. Cell cycle distribution was analyzed by flow cytometry. Additionally, a xenograft mouse model was developed for in vivo validation. RESULTS TP53 and RB1 mutations were associated with cisplatin concentration-dependent phenotypic transformation, with A549/DDP cells acquiring a more aggressive SCLC-like phenotype (In the article we call the A549/DDPSCLC cells). Analysis of cell bioenergetics profiling and Western blot analyses revealed enhanced glucose metabolism in A549/DDP1 μg/mL cells, while A549/DDPSCLC cells exhibited predominant lipid metabolism. Compound3K and Etomoxir specifically inhibit the activity of PKM2 and CPT1A, respectively, with Etomoxir demonstrating substantially inhibited A549/DDPSCLC cells growth and more cell cycle arrest in the G0/G1 phase. Combinatorial of Compound3K and Etomoxir effectively induced cell death in A549/DDPSCLC phenotype cells in vitro. Etomoxir alone or combined with Compound3K significantly inhibited tumor growth in vivo, with enhanced efficacy in the combination group. CONCLUSIONS This study provides the first evidence of cisplatin concentration-dependent metabolic reprogramming during NSCLC-to-SCLC transformation. We identified a phenotypic transition from NSCLC to SCLC accompanied by a metabolic shift from glucose to fatty acid metabolism, offering new insights into therapeutic strategies for treatmentresistant lung cancer.
Collapse
MESH Headings
- Cisplatin/pharmacology
- Humans
- Lung Neoplasms/metabolism
- Lung Neoplasms/drug therapy
- Lung Neoplasms/pathology
- Lung Neoplasms/genetics
- Animals
- Small Cell Lung Carcinoma/pathology
- Small Cell Lung Carcinoma/metabolism
- Small Cell Lung Carcinoma/drug therapy
- Small Cell Lung Carcinoma/genetics
- Mice
- Glucose/metabolism
- Fatty Acids/metabolism
- Cell Transformation, Neoplastic/drug effects
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/genetics
- Xenograft Model Antitumor Assays
- Antineoplastic Agents/pharmacology
- Drug Resistance, Neoplasm
- Tumor Suppressor Protein p53/metabolism
- Tumor Suppressor Protein p53/genetics
- Cell Proliferation/drug effects
- Mutation
- Mice, Nude
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/genetics
- Cell Line, Tumor
- Epithelial-Mesenchymal Transition/drug effects
- A549 Cells
Collapse
Affiliation(s)
- Qiu-Yu Zhao
- College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, People's Republic of China
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, People's Republic of China
| | - Wen-Jun Liu
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, People's Republic of China
- Teaching and Experimental Center, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, People's Republic of China
| | - Jian-Guang Wang
- College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, People's Republic of China
| | - He Li
- College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, People's Republic of China
| | - Jia-Lu Lv
- College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, People's Republic of China
| | - Yumeng Wang
- School of Biomedical Engineering, Shanghai Tech University, Shanghai, 201210, China
| | - Chun Wang
- College of Integrated Chinese and Western Medical, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning, People's Republic of China.
| |
Collapse
|
17
|
Xi D, Yang Y, Guo J, Wang M, Yan X, Li C. Single-cell sequencing and spatial transcriptomics reveal the evolution of glucose metabolism in hepatocellular carcinoma and identify G6PD as a potential therapeutic target. Front Oncol 2025; 15:1553722. [PMID: 40201344 PMCID: PMC11975570 DOI: 10.3389/fonc.2025.1553722] [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: 12/31/2024] [Accepted: 03/04/2025] [Indexed: 04/10/2025] Open
Abstract
Background Glucose metabolism reprogramming provides significant insights into the development and progression of malignant tumors. This study aims to explore the temporal-spatial evolution of the glucose metabolism in HCC using single-cell sequencing and spatial transcriptomics (ST), and validates G6PD as a potential therapeutic target for HCC. Methods We collected single-cell sequencing data from 7 HCC and adjacent non-cancerous tissues from the GSE149614 database, and ST data from 4 HCC tissues from the HRA000437 database. Pseudotime analysis was performed on the single-cell data, while ST data was used to analyze spatial metabolic activity. High-throughput sequencing and experiments, including wound healing, CCK-8, and transwell assays, were conducted to validate the role and regulatory mechanisms of G6PD in HCC. Results Our study identified a progressive upregulation of PPP-related genes during tumorigenesis. ST analysis revealed elevated PPP metabolic scores in the central and intermediate tumor regions compared to the peripheral zones. High-throughput sequencing and experimental validation further suggested that G6PD-mediated regulation of HCC cell proliferation, migration, and invasion is likely associated with glutathione metabolism and ROS production. Finally, Cox regression analysis cofirmed G6PD as an independent prognostic factor for overall survival in HCC patients. Conclusion Our study provides novel insights into the changes in glucose metabolism in HCC from both temporal and spatial perspectives. We experimentally demonstrated that G6PD regulates proliferation, migration, and invasion in HCC and propose G6PD as a prognostic marker and therapeutic metabolic target for the HCC.
Collapse
Affiliation(s)
- Deyang Xi
- Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Infectious Diseases, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yinshuang Yang
- Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jiayi Guo
- Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Infectious Diseases, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Mengjiao Wang
- Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Infectious Diseases, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xuebing Yan
- Department of Infectious Diseases, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Chunyang Li
- Department of Infectious Diseases, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
18
|
He S, Lv Y, Qiu J, Cui S, Gao Z, Peng L. Ta 4C 3 MXene Slows Progression of Fatty Liver Disease through Its Anti-Inflammatory and ROS-Scavenging Effects. ACS APPLIED MATERIALS & INTERFACES 2025; 17:17217-17229. [PMID: 40051029 DOI: 10.1021/acsami.4c20945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/21/2025]
Abstract
Treating metabolic dysfunction-associated fatty liver disease (MAFLD) and reducing the occurrence of MAFLD-associated liver cancer remain challenging. Two-dimensional (2D) tantalum carbide (Ta4C3) MXene nanozymes (MXenzymes) exhibit antioxidant and anti-inflammatory activities and have thus attracted considerable attention in the fields of oncology and engineering. However, the potential mechanism of action and bioactive properties of Ta4C3 in MAFLD remain uncertain. In our study, Ta4C3 not only inhibited lipid accumulation and disrupted lipid metabolism in hepatocytes but also reduced cell death caused by fatty acids by decreasing intracellular reactive oxygen species (ROS) levels, which significantly promoted the polarization of M1 macrophages to M2 macrophages by alleviating oxidative stress and further suppressing inflammatory factor expression. In mice fed a methionine-choline-deficient (MCD) diet, Ta4C3 reduced lipid accumulation, the infiltration of inflammatory cells, and liver cell apoptosis by modulating the cellular microenvironment through its anti-inflammatory and antioxidant properties. Therefore, Ta4C3 can be used as a multifunctional bioactive material to alleviate hepatic steatosis and inflammation in individuals with MAFLD/metabolic dysfunction-associated steatohepatitis (MASH) because of its robust antioxidant and anti-inflammatory effects.
Collapse
Affiliation(s)
- Shuying He
- Department of Gastroenterology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Yuerong Lv
- Department of Gastroenterology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Jingnan Qiu
- Department of Gastroenterology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Shudan Cui
- Department of Gastroenterology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Zixian Gao
- Department of Gastroenterology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Liang Peng
- Department of Gastroenterology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510120, China
- Department of Medicine, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| |
Collapse
|
19
|
Liu W, Hu K, Fu Y, Zhou T, Zhong Q, Wang W, Gui Y, Zhang P, Yao D, Yang X, Zhu W, Liu Z, Luo D, Xiao Y. Identification of methionine metabolism related prognostic model and tumor suppressive functions of BHMT in hepatocellular carcinoma. Sci Rep 2025; 15:9250. [PMID: 40102459 PMCID: PMC11920202 DOI: 10.1038/s41598-025-93650-w] [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: 09/13/2024] [Accepted: 03/07/2025] [Indexed: 03/20/2025] Open
Abstract
Given the resistance to conventional treatments and limitations of immune checkpoint blockade therapy in hepatocellular carcinoma (HCC), it is imperative to explore novel prognostic models and biomarkers. The dependence of cancer cell on exogenous methionine, known as Hoffman effect, is a hallmark of HCC, with numerous studies reporting a strong correlation between methionine metabolism and tumor development. Betaine-homocysteine S-methyltransferase (BHMT), a critical component of methionine metabolism pathway, has polymorphisms linking to poor prognosis in multiple cancers. Nevertheless, there is little literature regarding the relationship between methionine metabolism and incidence, mortality of HCC, as well as the function of BHMT in HCC progression. In this study, by analyzing multiple datasets, we constructed a methionine metabolism-related prognostic model and thoroughly investigated the influence of BHMT on the prognosis of HCC. Bioinformatics analysis revealed a marked decrease in BHMT expression in HCC, which was linked to adverse clinical outcomes. CIBERSORT results suggest that BHMT promotes infiltration of M1 macrophages. Our results suggest its potential as an ideal prognostic biomarker for anti PD-L1 immunotherapy. In summary, this study innovatively provides first methionine metabolism-related prognostic model and unveils the tumor suppressive function of BHMT in HCC, providing potential mechanism by which BHMT exert its function.
Collapse
Affiliation(s)
- Wenli Liu
- Department of Pathology, Infectious Diseases Hospital of Nanchang University, Nanchang, 330001, Jiangxi, China
| | - Kaiheng Hu
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Yaqing Fu
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Tianmin Zhou
- Department of Pathology, Infectious Diseases Hospital of Nanchang University, Nanchang, 330001, Jiangxi, China
| | - Qingmei Zhong
- Department of Pathology, Infectious Diseases Hospital of Nanchang University, Nanchang, 330001, Jiangxi, China
| | - Wu Wang
- Department of Pathology, Infectious Diseases Hospital of Nanchang University, Nanchang, 330001, Jiangxi, China
| | - Yang Gui
- Department of Pathology, Infectious Diseases Hospital of Nanchang University, Nanchang, 330001, Jiangxi, China
| | - Ping Zhang
- Department of Pathology, Infectious Diseases Hospital of Nanchang University, Nanchang, 330001, Jiangxi, China
| | - Di Yao
- Department of Pathology, Infectious Diseases Hospital of Nanchang University, Nanchang, 330001, Jiangxi, China
| | - Xiaohong Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Weifeng Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Zhuoqi Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| | - Daya Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| | - Yingqun Xiao
- Department of Pathology, Infectious Diseases Hospital of Nanchang University, Nanchang, 330001, Jiangxi, China.
| |
Collapse
|
20
|
Wang M, Huang X, Zhang D, Liu Y, Liu P. The role of fructose-1,6-bisphosphatase 1 on regulating the cancer progression and drug resistance. Discov Oncol 2025; 16:346. [PMID: 40100307 PMCID: PMC11920503 DOI: 10.1007/s12672-025-02112-2] [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: 12/09/2024] [Accepted: 03/10/2025] [Indexed: 03/20/2025] Open
Abstract
Fructose-1,6-bisphosphatase 1 (FBP1) is the enzyme that limits the process of gluconeogenesis as it facilitates the hydrolysis of fructose-1,6-bisphosphate(F-1,6-BP) to produce fructose-6-phosphate(F6P) and inorganic phosphate. Gluconeogenesis is the production of glucose from small carbohydrate substrates. The gluconeogenic process is typically suppressed in cancer because it inhibits glycolysis. Apart from its involvement in cellular glucose metabolism, FBP1 also plays a role in gene transcription, mRNA translation and stability regulation, and the immune microenvironment of tumors. Because of its multifaceted functions, the mechanisms by which FBP1 is involved in tumor development are complex. Moreover, FBP1 deficiency is associated with radiation and chemotherapy resistance and poor prognosis in cancer patients. Restoration of FBP1 expression in cancer cells is expected to hold promise for cancer therapy. However, up to now few reviews have systematically summarized the important functional mechanisms of FBP1 in tumorigenesis and the small molecule compounds that restore FBP1 expression. Therefore, this article addresses the question "How does FBP1 contribute to cancer progression, and can targeting FBP1 be a potential therapeutic approach?" by summarizing the effects of FBP1 on cancer development and progression as well as its mediated drug resistance and the future clinical applications of potential small molecule modulators targeting FBP1.
Collapse
Affiliation(s)
- Mengmeng Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Xiaoju Huang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Dan Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Yisan Liu
- Department of Urology, People's Hospital of Cili, Cili, 427200, Hunan, China.
| | - Pian Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China.
| |
Collapse
|
21
|
Chen J, Wei C, Huang W, Huang T, Zhou L, Xu Y, Qin Y, Lin Q, Liu F, Pan X, Tang Z, Yang W, Fang M. Clonorchis sinensis-infected hepatocellular carcinoma exhibits distinct tumor microenvironment and molecular features. Front Immunol 2025; 16:1526699. [PMID: 40165955 PMCID: PMC11955701 DOI: 10.3389/fimmu.2025.1526699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/26/2025] [Indexed: 04/02/2025] Open
Abstract
Objectives Clonorchis sinensis (Cs)-infected hepatocellular carcinoma (HCC) patients have a poorer prognosis than non-Cs-infected HCCs. However, the molecular mechanisms of Cs-infected HCC remain unclear. To address this, this study aims to uncover the tumor microenvironment and molecular features that may contribute to these poor outcomes. Methods The research involved bulk RNA sequencing of paired tumor and adjacent tissue samples from 10 Cs + HCC and 10 Cs - HCC patients. Differentially expressed genes were identified, followed by enrichment analyses to reveal functional changes. Survival analysis of the top 10 up- and down-regulated genes in Cs + HCC tumors was performed using TCGA database. Additionally, clinical data from 1,461 HCC patients were retrospectively analyzed to assess the impact of Cs infection on microvascular invasion and metastasis rates. In vitro assays were also conducted using Cs excretory/secretory products (CsESPs) to examine their effect on HCC cells and HUVECs. Results We identified 785 up-regulated and 675 down-regulated genes in Cs + HCC tumors compared to Cs - HCC tumors, enriched in pathways related to extracellular matrix remodeling and immunosuppression. Survival analysis revealed that the top 10 up-regulated genes are associated with HCC poor prognosis. Clinical data from 1,461 HCC patients showed Cs infection increased microvascular invasion and metastasis rates. In vitro, CsESPs products enhanced migration and invasion in HCC cells and promoted tube formation in human umbilical vein endothelial cells. Conclusions This study provides novel insights into the molecular landscape of Cs-infected HCC and underscores the Cs infection's role in enhancing tumor migration, invasion and angiogenesis. The findings contribute to the understanding of parasitic infections in cancer progression and suggest potential prognostic markers for Cs + HCC.
Collapse
Affiliation(s)
- Junxian Chen
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Caibiao Wei
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Wencheng Huang
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Taijun Huang
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Lingling Zhou
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yulong Xu
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yuling Qin
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Qiumei Lin
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Fengfei Liu
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xiaolan Pan
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Zeli Tang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
| | - Weilong Yang
- Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
- Institute of Advanced Biotechnology and School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Min Fang
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
- Engineering Research Center for Tissue and Organ Injury and Repair Medicine, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
22
|
Wang C, Zhou J, Jia P, Yang Y, Song R, Zheng X, Zhang H, Li Y. Joint proteomic and metabolomic analysis reveals renal metabolic remodeling of chronic heart failure mice. J Pharm Biomed Anal 2025; 255:116641. [PMID: 39731929 DOI: 10.1016/j.jpba.2024.116641] [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: 10/08/2024] [Revised: 12/01/2024] [Accepted: 12/15/2024] [Indexed: 12/30/2024]
Abstract
Pharmacologic intervention in chronic heart failure (HF) with renal insufficiency is one of the clinical challenges due to the fact that the mechanisms of cardio-renal interactions in chronic heart failure (CHF) progressing have not been fully revealed. In this paper, C57BL/6 mice were applied thoracic aortic narrowing surgery to establish pressure overload CHF model. Cardiac function, serum markers, renal pathologic changes and kidney metabolism were analyzed at 4th, 8th, 12th, and 16th week after surgery respectively to evaluate the heart-Kidney pathologic overlap. Kidney proteomic analysis was performed at 16th week after operation. As a result, renal hypofiltration and exacerbation of pathological damage was observed accompanying cardiac function deterioration after 12th week. 66 differentially expressed proteins and 13 differential metabolites were found to be involved in the cardio-renal pathological overlap. Joint proteomic and metabolomic analysis revealed that signal pathways like Phosphatidylinositol signaling system, Glucagon signaling pathway, the Glyoxylate and dicarboxylate metabolism; DEPs of Pten, Mtmr4, PLC and CPT1, differential metabolites like aspartic acid and isocitrate deserve further investigation.
Collapse
Affiliation(s)
- Chunliu Wang
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, Shaanxi, China; Key Laboratory of TCM Drug Delivery, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, Shaanxi, China
| | - Jie Zhou
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Pu Jia
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Science, Northwest University, Xi'an, Shaanxi, China
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Science, Northwest University, Xi'an, Shaanxi, China
| | - Ruixue Song
- Xi'an Research Institute of Chinese Lacquer, Xi'an, Shaanxi, China
| | - Xiaohui Zheng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Science, Northwest University, Xi'an, Shaanxi, China
| | - Hong Zhang
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, Shaanxi, China; Key Laboratory of TCM Drug Delivery, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, Shaanxi, China.
| | - Ye Li
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, Shaanxi, China; Key Laboratory of TCM Drug Delivery, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, Shaanxi, China.
| |
Collapse
|
23
|
Zhang B, Zheng J, Zheng S. Cirsiliol suppresses malignant progression of hepatocellular carcinoma via regulation of glutamine metabolism. Am J Transl Res 2025; 17:2145-2153. [PMID: 40226041 PMCID: PMC11982850 DOI: 10.62347/aoty4308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 12/06/2024] [Indexed: 04/15/2025]
Abstract
BACKGROUND To investigate the therapeutic potential of cirsiliol in hepatocellular carcinoma (HCC), focusing on its impact on glutamine metabolism. METHODS HCC cell lines HCCLM3 and Huh7 were treated with cirsiliol, and cell viability and proliferation were assessed using CCK-8 assay. Intracellular concentrations of glutamine, α-ketoglutaric acid (α-KG), and adenosine triphosphate (ATP) were measured to evaluate glutamine metabolism. A xenograft tumor model was employed to examine the in vivo effects of cirsiliol. Additionally, network pharmacological analysis was used to identify potential targets of cirsiliol in HCC. Western blotting was conducted to analyze the modulation of the PI3K/AKT signaling pathway by cirsiliol. RESULTS Cirsiliol significantly inhibited HCC cell growth both in vitro and in vivo while reducing levels of glutamine, α-KG, and ATP, indicating suppression of glutamine metabolism. Activation of the PI3K signaling pathway reversed the inhibitory effects of cirsiliol on HCC cell growth and metabolism. CONCLUSION Cirsiliol suppresses glutamine metabolism and inhibits the growth of HCC cells by modulating the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Bin Zhang
- Hepatopancreatobiliary Surgery Department, The First Affiliated Hospital of Ningbo University No. 59, Liuting Street, Haishu District, Ningbo 315000, Zhejiang, China
| | - Jianbo Zheng
- Hepatopancreatobiliary Surgery Department, The First Affiliated Hospital of Ningbo University No. 59, Liuting Street, Haishu District, Ningbo 315000, Zhejiang, China
| | - Siming Zheng
- Hepatopancreatobiliary Surgery Department, The First Affiliated Hospital of Ningbo University No. 59, Liuting Street, Haishu District, Ningbo 315000, Zhejiang, China
| |
Collapse
|
24
|
Wang LL, Zhang FC, Xu HX, Deng DD, Ren BJ, Tan Q, Liu YX, Zhao WH, Lu JL. Advances in imaging techniques for tumor microenvironment evaluation in hepatocellular carcinoma. World J Gastroenterol 2025; 31:103454. [PMID: 40093677 PMCID: PMC11886532 DOI: 10.3748/wjg.v31.i10.103454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/31/2024] [Accepted: 02/11/2025] [Indexed: 02/26/2025] Open
Abstract
The tumor microenvironment (TME) plays a critical role in the development and treatment of liver cancer, which ranks sixth in incidence and third in mortality worldwide, according to the "Global Cancer Statistics 2022". Hepatocellular carcinoma (HCC), the most common form of liver cancer, is heavily influenced by the TME, which affects tumor growth, invasion, metastasis, and the response to various treatments. Despite advancements in surgery, liver transplantation, targeted therapies, and immunotherapy, the complexity of the TME often limits treatment efficacy, especially in advanced-stage HCC cases. The TME consists of a dynamic interaction between tumor cells, immune cells, fibroblasts, blood vessels, and signaling molecules, all of which contribute to cancer progression and therapy resistance. Assessing the HCC TME is essential for designing effective, personalized treatments and improving patient outcomes. Recent research highlights the value of imaging technologies as non-invasive tools to evaluate the TME, offering new possibilities for more targeted therapies and better prognosis monitoring in HCC patients.
Collapse
Affiliation(s)
- Li-Li Wang
- Department of Radiology, The First Hospital of Lanzhou University, Lanzhou 730030, Gansu Province, China
| | - Fa-Chang Zhang
- First Clinical Medical School of Lanzhou University, Lanzhou University, Lanzhou 730030, Gansu Province, China
| | - Han-Xin Xu
- First Clinical Medical School of Lanzhou University, Lanzhou University, Lanzhou 730030, Gansu Province, China
| | - Dian-Dian Deng
- First Clinical Medical School of Lanzhou University, Lanzhou University, Lanzhou 730030, Gansu Province, China
| | - Bing-Jie Ren
- First Clinical Medical School of Lanzhou University, Lanzhou University, Lanzhou 730030, Gansu Province, China
| | - Qi Tan
- First Clinical Medical School of Lanzhou University, Lanzhou University, Lanzhou 730030, Gansu Province, China
| | - Ya-Xin Liu
- First Clinical Medical School of Lanzhou University, Lanzhou University, Lanzhou 730030, Gansu Province, China
| | - Wen-Hui Zhao
- First Clinical Medical School of Lanzhou University, Lanzhou University, Lanzhou 730030, Gansu Province, China
| | - Jia-Le Lu
- First Clinical Medical School of Lanzhou University, Lanzhou University, Lanzhou 730030, Gansu Province, China
| |
Collapse
|
25
|
Li S, Wang X, Xiao J, Yi J. SLC7A11, a disulfidptosis-related gene, correlates with multi-omics prognostic analysis in hepatocellular carcinoma. Eur J Med Res 2025; 30:161. [PMID: 40069889 PMCID: PMC11900568 DOI: 10.1186/s40001-025-02411-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND This study sought to establish a risk score signature based on disulfidptosis-related genes (DRGs) to predict the prognosis of hepatocellular carcinoma (HCC) patients. METHODS The expression data of DRGs from the Cancer Genome Atlas (TCGA) and the International Cancer Genome Consortium (ICGC) was analyzed to develop and validate a DRG prognostic signature (DRGPS). In vitro, experiments were conducted to explore DRG expressions and roles in HCC tissues and cell lines. HCC tissue microarrays were employed to analyze SLC7A11 expression and its association with clinicopathological characteristics. RESULTS The DRGPS consisted of 5 DRGs (SLC7A11, MATN3, CLEC3B, CCNJL, and PON1). The survival rate of HCC patients in high-risk group was significantly lower than that in low-risk group. The DRGPS was also associated with the modulation of tumor microenvironment (TME), tumor mutation burden (TMB), stemness and chemosensitivity. Furthermore, pan-cancer analysis suggested that the DRGPS risk score was associated with immune infiltration and stemness in multiple cancers. Moreover, our DRGPS had potential for predicting treatment efficacy in HCC patients. Finally, we confirmed that downregulation of SLC7A11, a DRG, inhibited the proliferation and migration of HCC cells, while its high expression correlated with advanced TNM clinical stage and larger tumor size. CONCLUSIONS This study systematically describes a novel DRGPS constructed for predicting HCC prognosis, providing a new approach to risk stratification and treatment options. It also investigates the expression and function of SLC7A11, contributing to further exploration of the molecular mechanism underlying disulfidptosis in HCC, as well as its prognostic and therapeutic implications.
Collapse
Affiliation(s)
- Shizhe Li
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, 410008, Hunan, China
| | - Xiaotong Wang
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, 410008, Hunan, China
| | - Junbo Xiao
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, 410008, Hunan, China.
| | - Jun Yi
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, 410008, Hunan, China.
| |
Collapse
|
26
|
Li S, Lu Z, Jiang W, Xu Y, Chen R, Wang J, Jiao B, Lu X. Chaetocin, a Natural Inhibitor of Transketolase, Suppresses the Non-Oxidative Pentose Phosphate Pathway and Inhibits the Growth of Drug-Resistant Non-Small Cell Lung Cancer. Antioxidants (Basel) 2025; 14:330. [PMID: 40227333 PMCID: PMC11939327 DOI: 10.3390/antiox14030330] [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: 12/26/2024] [Revised: 02/16/2025] [Accepted: 03/09/2025] [Indexed: 04/15/2025] Open
Abstract
Worldwide, lung cancer is the most common cause of cancer-related death, which is made worse by the development of drug resistance during treatment. It is urgent to develop new therapeutic methods and small molecule drugs for tumor resistance. Chaetocin, extracted from Chaetomium minutum, is a natural compound with good antitumor activity. However, there are few studies on its tumor resistance. In this paper, firstly, chaetotocin significantly inhibited the viability and migration of cisplatin-resistant non-small cell lung cancer (NSCLC) cells and inhibited the xenograft growth of nude mice. Chaetocin at 4 mg/kg significantly inhibited A549/DDP xenograft growth with an inhibition rate of 70.43%. Subsequently, the underlying mechanism behind the actions of chaetocin was explored. It was discovered that chaetocin can inhibit transketolase (TKT), thereby inhibiting the growth of NSCLC cells and inducing cell death. Compared with cisplatin-sensitive cells, a lower concentration of chaetocin can inhibit cisplatin-resistance cell viability and migration. Mechanistically, TKT was identified as a potential target for chaetocin. The KD value of the interaction between chaetocin and TKT was 63.2 μM. An amount of 0.2 μM chaetocin may suppress the enzyme activity and expression level of TKT. We found the TKT expression is higher in cisplatin-resistant cells, which further explains why these cells were more vulnerable to chaetocin in terms of cell phenotype. Additionally, the muti-omics analysis and RNA interference suggested that chaetocin can inhibit the PI3K/Akt signaling pathway through TKT. In conclusion, chaetocin could directly bind to TKT, inhibiting its enzyme activity and expression, which interfered with intracellular metabolism and oxidation-reduction balance, and then regulated the PI3K/Akt signaling pathway to inhibit the growth of NSCLC and induce apoptosis.
Collapse
Affiliation(s)
- Song Li
- Department of Biochemistry and Molecular Biology, Naval Medical University, Shanghai 200433, China; (S.L.); (W.J.); (Y.X.); (R.C.); (J.W.)
| | - Zhanying Lu
- Experimental Training Center of Basic Medical Science, Naval Medical University, Shanghai 200433, China;
| | - Wenli Jiang
- Department of Biochemistry and Molecular Biology, Naval Medical University, Shanghai 200433, China; (S.L.); (W.J.); (Y.X.); (R.C.); (J.W.)
| | - Yao Xu
- Department of Biochemistry and Molecular Biology, Naval Medical University, Shanghai 200433, China; (S.L.); (W.J.); (Y.X.); (R.C.); (J.W.)
| | - Ran Chen
- Department of Biochemistry and Molecular Biology, Naval Medical University, Shanghai 200433, China; (S.L.); (W.J.); (Y.X.); (R.C.); (J.W.)
| | - Jie Wang
- Department of Biochemistry and Molecular Biology, Naval Medical University, Shanghai 200433, China; (S.L.); (W.J.); (Y.X.); (R.C.); (J.W.)
| | - Binghua Jiao
- Department of Biochemistry and Molecular Biology, Naval Medical University, Shanghai 200433, China; (S.L.); (W.J.); (Y.X.); (R.C.); (J.W.)
| | - Xiaoling Lu
- Department of Biochemistry and Molecular Biology, Naval Medical University, Shanghai 200433, China; (S.L.); (W.J.); (Y.X.); (R.C.); (J.W.)
| |
Collapse
|
27
|
Xie X, Liu W, Yuan Z, Chen H, Mao W. Bridging epigenomics and tumor immunometabolism: molecular mechanisms and therapeutic implications. Mol Cancer 2025; 24:71. [PMID: 40057791 PMCID: PMC11889836 DOI: 10.1186/s12943-025-02269-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 02/11/2025] [Indexed: 04/02/2025] Open
Abstract
Epigenomic modifications-such as DNA methylation, histone acetylation, and histone methylation-and their implications in tumorigenesis, progression, and treatment have emerged as a pivotal field in cancer research. Tumors undergo metabolic reprogramming to sustain proliferation and metastasis in nutrient-deficient conditions, while suppressing anti-tumor immunity in the tumor microenvironment (TME). Concurrently, immune cells within the immunosuppressive TME undergo metabolic adaptations, leading to alterations in their immune function. The complicated interplay between metabolites and epigenomic modulation has spotlighted the significance of epigenomic regulation in tumor immunometabolism. In this review, characteristics of the epigenomic modification associated with tumors are systematically summarized alongside with their regulatory roles in tumor metabolic reprogramming and immunometabolism. Classical and emerging approaches are delineated to broaden the boundaries of research on the crosstalk research on the crosstalk between tumor immunometabolism and epigenomics. Furthermore, we discuss potential therapeutic strategies that target tumor immunometabolism to modulate epigenomic modifications, highlighting the burgeoning synergy between metabolic therapies and immunotherapy as a promising avenue for cancer treatment.
Collapse
Affiliation(s)
- Xiaowen Xie
- Department of Thoracic Surgery, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, 214023, China
| | - Weici Liu
- Department of Thoracic Surgery, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, 214023, China
- Center of Clinical Research, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, 214023, China
| | - Zhiyuan Yuan
- Institute of Science and Technology for Brain-Inspired Intelligence; MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence; MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200433, China.
| | - Hanqing Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Capital Medical University, Beijing, 100069, China.
| | - Wenjun Mao
- Department of Thoracic Surgery, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, 214023, China.
| |
Collapse
|
28
|
Liu P, Huang F, Lin P, Liu J, Zhou P, Wang J, Sun H, Xing F, Ma H. Histidine metabolism drives liver cancer progression via immune microenvironment modulation through metabolic reprogramming. J Transl Med 2025; 23:262. [PMID: 40038727 DOI: 10.1186/s12967-025-06267-y] [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: 12/15/2024] [Accepted: 02/14/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Histidine metabolism is crucial in role in tumor biology, contributing to tumor progression, immune regulation, and metabolic reprogramming. In hepatocellular carcinoma (HCC), dysregulated histidine metabolism may promote tumor growth and immune evasion, although the specific mechanisms remain poorly understood. METHODS Using single-cell RNA sequencing, the expression patterns of histidine metabolism-related genes were evaluated across different cell types in HCC samples. In vivo and in vitro experiments were conducted to validate how histidine treatment affects macrophage and T-cell function. Furthermore, the TCGA database was utilized to construct a prognostic model to identify the key gene BUD23 and to examine its correlation with metabolism and immune infiltration. RESULTS The proportion of parenchymal cells exhibiting high histidine metabolism was significantly increased, accompanied by a general reduction in immune and stromal cell infiltration. Notably, macrophages and T cells demonstrated impaired antitumor functions. In the high histidine metabolism group, multiple critical cell communication pathways (e.g., MIF, CLEC, MHC II) were downregulated, macrophages shifted toward immunosuppressive subpopulations, T cells exhibited an exhaustion phenotype, and CD8 + T-cell activation was diminished. Further in vivo and in vitro co-culture experiments confirmed that elevated histidine concentrations promoted M2 polarization in macrophages and weakened T-cell cytotoxicity, accelerating tumor proliferation. According to TCGA analyses, BUD23 was upregulated in the high histidine metabolism group and significantly negatively correlated with patient survival and immune cell infiltration. Silencing BUD23 boosted immune cell activation and cytotoxic effects, effectively reversing the immunosuppressive microenvironment. A multivariable Cox regression-based prognostic model indicated unfavorable outcomes in patients with high histidine metabolism. CONCLUSION Histidine metabolism drives tumor cell metabolic reprogramming and reshapes the tumor immune microenvironment through intercellular communication, thereby promoting tumor progression. BUD23 shows promise as a biomarker for prognosis and immune response prediction in liver cancer. This study provides new therapeutic targets and theoretical support for liver cancer treatment by targeting histidine metabolism.
Collapse
Affiliation(s)
- Pengcheng Liu
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University School of Medicine, Guangzhou, 510006, China
| | - Fuxin Huang
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Peixu Lin
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University School of Medicine, Guangzhou, 510006, China
| | - Jiayao Liu
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Pincheng Zhou
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Jie Wang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510006, China
| | - Huanhuan Sun
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University School of Medicine, Guangzhou, 510006, China.
| | - Fan Xing
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University School of Medicine, Guangzhou, 510006, China.
| | - Haiqing Ma
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University School of Medicine, Guangzhou, 510006, China.
- Department of Oncology, Heyuan Hospital of Guangdong Provincial People's Hospital, Heyuan People's Hospital, Heyuan, 517000, China.
- School of Medicine, South China University of Technology, Guangzhou, 510006, China.
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510006, China.
| |
Collapse
|
29
|
Karim M, Prey J, Willer F, Leiner H, Yasser M, Dombrowski F, Ribback S. Hepatic Deletion of Carbohydrate Response Element Binding Protein Impairs Hepatocarcinogenesis in a High-Fat Diet-Induced Mouse Model. Int J Mol Sci 2025; 26:2246. [PMID: 40076869 PMCID: PMC11900174 DOI: 10.3390/ijms26052246] [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: 01/20/2025] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 03/14/2025] Open
Abstract
The transcription factor carbohydrate response element binding protein (ChREBP) has emerged as a crucial regulator of hepatic glucose and lipid metabolism. The increased ChREBP activity involves the pro-oncogenic PI3K/AKT/mTOR signaling pathway that induces aberrant lipogenesis, thereby promoting hepatocellular carcinomas (HCC). However, the molecular pathogenesis of ChREBP-related hepatocarcinogenesis remains unexplored in the high-fat diet (HFD)-induced mouse model. Male C57BL/6J (WT) and liver-specific (L)-ChREBP-KO mice were maintained on either a HFD or a control diet for 12, 24, and 48 weeks, starting at the age of 4 weeks. At the end of the feeding period, mice were perfused, and liver tissues were formalin-fixed, paraffin-embedded, sectioned, and stained for histological and immunohistochemical analysis. Biochemical and gene expression analysis were conducted using serum and frozen liver tissue. Mice fed with HFD showed a significant increase (p < 0.05) in body weight from 8 weeks onwards compared to the control. WT and L-ChREBP-KO mice also demonstrated a significant increase (p < 0.05) in liver-to-body weight ratio in the 48-week HFD group. HFD mice exhibited a gradual rise in hepatic lipid accumulation over time, with 24-week mice showing a 20-30% increase in fat content, which further advanced to 80-100% fat accumulation at 48 weeks. Both dietary source and the increased expression of lipogenic pathways at transcriptional and protein levels induced steatosis and steatohepatitis in the HFD group. Moreover, WT mice on a HFD exhibited markedly higher inflammation compared to the L-ChREBP-KO mice. The enhanced lipogenesis, glycolysis, persistent inflammation, and activation of the AKT/mTOR pathway collectively resulted in significant metabolic disturbances, thereby promoting HCC development and progression in WT mice. In contrast, hepatic loss of ChREBP resulted in reduced hepatocyte proliferation in the HFD group, which significantly contributed to the impaired hepatocarcinogenesis and a reduced HCC occurrence in the L-ChREBP-KO mice. Our present study implicates that prolonged HFD feeding contributes to NAFLD/NASH, which in turn progresses to HCC development in WT mice. Collectively, hepatic ChREBP deletion ameliorates hepatic inflammation and metabolic alterations, thereby impairing NASH-driven hepatocarcinogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Silvia Ribback
- Institute of Pathology, University Medicine Greifswald, Friedrich-Loeffler-Str. 23e, 17475 Greifswald, Germany; (M.K.); (J.P.); (F.W.); (M.Y.); (F.D.)
| |
Collapse
|
30
|
Zuo H, Liu X, Wang Y, Ding H, Wan W, Zheng S, Hou S, Hu K. SREBF1 facilitates pathological retinal neovascularization by reprogramming the fatty acid metabolism of endothelial cells. Exp Eye Res 2025; 252:110239. [PMID: 39800283 DOI: 10.1016/j.exer.2025.110239] [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: 10/07/2024] [Revised: 12/03/2024] [Accepted: 01/09/2025] [Indexed: 01/31/2025]
Abstract
Retinopathy of prematurity (ROP) is a proliferative retinal vascular disorder that critically affects the visual development of premature infants, potentially leading to irreversible vision loss or even blindness. Despite its significance, the underlying mechanisms of this disease remain insufficiently understood. In this study, we utilized the oxygen-induced retinopathy (OIR) mouse model and conducted endothelial functional assays to explore the role of Sterol Regulatory Element-Binding Protein 1 (SREBF1) in ROP pathogenesis. SREBF1 expression levels, along with its downstream targets, were investigated through Western blotting, RT-qPCR, and immunofluorescence staining techniques. Furthermore, Co-Immunoprecipitation (Co-IP) was employed to examine the molecular mechanisms involved. Our results demonstrated a significant increase in SREBF1 expression in both the OIR mouse model and hypoxic primary human retinal microvascular endothelial cells (HRMECs). Interventions conducted both in vivo and in vitro showed notable efficacy in reducing pathological neovascularization. Importantly, we discovered that SREBF1 plays a key role in modulating lipid metabolism in HRMECs by regulating the expression of ACC1 and FASN, leading to cellular reprogramming. This reprogramming influences HRMEC proliferation, migration, and tube formation through the HIF-1α/TGF-β signaling pathway, ultimately contributing to pathological retinal neovascularization. These findings provide new insights into the role of SREBF1 in angiogenesis within the context of ROP, offering potential therapeutic targets for the management and treatment of this disease.
Collapse
Affiliation(s)
- Hangjia Zuo
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Prevention and Treatment on Major Blinding Diseases, Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, PR China; Chongqing Medical University, Chongqing, PR China
| | - Xianyang Liu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Prevention and Treatment on Major Blinding Diseases, Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, PR China; Chongqing Medical University, Chongqing, PR China; Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China
| | - Yakun Wang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Prevention and Treatment on Major Blinding Diseases, Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, PR China; Chongqing Medical University, Chongqing, PR China
| | - Huannan Ding
- Chongqing Medical University, Chongqing, PR China
| | - Wenjuan Wan
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Prevention and Treatment on Major Blinding Diseases, Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, PR China; Chongqing Medical University, Chongqing, PR China
| | - Shijie Zheng
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Prevention and Treatment on Major Blinding Diseases, Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, PR China; Chongqing Medical University, Chongqing, PR China
| | - Shengping Hou
- Chongqing Medical University, Chongqing, PR China; Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, China.
| | - Ke Hu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Prevention and Treatment on Major Blinding Diseases, Chongqing Eye Institute, Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, PR China; Chongqing Medical University, Chongqing, PR China.
| |
Collapse
|
31
|
Hu GS, Zheng ZZ, He YH, Wang DC, Nie RC, Liu W. Integrated Analysis of Proteome and Transcriptome Profiling Reveals Pan-Cancer-Associated Pathways and Molecular Biomarkers. Mol Cell Proteomics 2025; 24:100919. [PMID: 39884577 PMCID: PMC11907456 DOI: 10.1016/j.mcpro.2025.100919] [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: 06/12/2024] [Revised: 01/02/2025] [Accepted: 01/24/2025] [Indexed: 02/01/2025] Open
Abstract
Understanding dysregulated genes and pathways in cancer is critical for precision oncology. Integrating mass spectrometry-based proteomic data with transcriptomic data presents unique opportunities for systematic analyses of dysregulated genes and pathways in pan-cancer. Here, we compiled a comprehensive set of datasets, encompassing proteomic data from 2404 samples and transcriptomic data from 7752 samples across 13 cancer types. Comparisons between normal or adjacent normal tissues and tumor tissues identified several dysregulated pathways including mRNA splicing, interferon pathway, fatty acid metabolism, and complement coagulation cascade in pan-cancer. Additionally, pan-cancer upregulated and downregulated genes (PCUGs and PCDGs) were also identified. Notably, RRM2 and ADH1B, two genes which belong to PCUGs and PCDGs, respectively, were identified as robust pan-cancer diagnostic biomarkers. TNM stage-based comparisons revealed dysregulated genes and biological pathways involved in cancer progression, among which the dysregulation of complement coagulation cascade and epithelial-mesenchymal transition are frequent in multiple types of cancers. A group of pan-cancer continuously upregulated and downregulated proteins in different tumor stages (PCCUPs and PCCDPs) were identified. We further constructed prognostic risk stratification models for corresponding cancer types based on dysregulated genes, which effectively predict the prognosis for patients with these cancers. Drug prediction based on PCUGs and PCDGs as well as PCCUPs and PCCDPs revealed that small molecule inhibitors targeting CDK, HDAC, MEK, JAK, PI3K, and others might be effective treatments for pan-cancer, thereby supporting drug repurposing. We also developed web tools for cancer diagnosis, pathologic stage assessment, and risk evaluation. Overall, this study highlights the power of combining proteomic and transcriptomic data to identify valuable diagnostic and prognostic markers as well as drug targets and treatments for cancer.
Collapse
Affiliation(s)
- Guo-Sheng Hu
- Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou, China; State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Zao-Zao Zheng
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yao-Hui He
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Du-Chuang Wang
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Rui-Chao Nie
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, Fujian, China
| | - Wen Liu
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
32
|
Luo J, Cai Y, Jia C, Zhang X, Huang Q, Wei J, Chen Q, Chen T. BefA protein alleviates progression of non-alcoholic fatty liver disease by modulating the AMPK signaling pathway through the gut-liver axis. Int J Biol Macromol 2025; 294:139446. [PMID: 39756723 DOI: 10.1016/j.ijbiomac.2024.139446] [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: 08/30/2024] [Revised: 12/29/2024] [Accepted: 12/31/2024] [Indexed: 01/07/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the leading cause of chronic liver diseases worldwide, necessitating urgent novel oral treatments. In this study, β-cell expansion factor A (BefA) was evaluated in a murine NAFLD model induced by high-fat diet (HFD). Our results revealed that BefA significantly reduced body weight (36.58 ± 1.55 g vs. 42.30 ± 1.96 g, p < 0.01), fat mass-to-body weight ratio (0.023 ± 0.019 vs. 0.300 ± 0.019, p < 0.05), liver weight (1.90 ± 0.07 g vs. 2.31 ± 0.21 g, p < 0.05), and liver function parameters (ALT, AST, ALP levels reduced, p < 0.05). Notably, BefA reversed the pathological features of NAFLD, decreasing hepatic steatosis score from 3.67 ± 0.47 to 1.67 ± 0.47 (p < 0.01). Mechanistically, BefA activated the AMPK signaling pathway, resulting in the suppression of lipogenic gene transcription (ACC, FASN, SREBP-1c) and the enhancement of fatty acid oxidation (CPT-1, PPAR-α). However, AMPK inhibitor and broad-spectrum antibiotics significantly attenuated the benefits observed with BefA treatment, increasing body weight, fat-to-body weight ratio, and liver weight (p < 0.05). Similar detrimental effects were also observed in liver function indices and histopathological characteristics. These findings underscore the pivotal role of both gut microbiota modulation and AMPK signaling in BefA's therapeutic efficacy, making it a promising multitargeted approach for NAFLD treatment.
Collapse
Affiliation(s)
- Jie Luo
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; Jiangxi Provincial Key Laboratory of Disease Prevention and Public Health, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Yujie Cai
- Jiangxi Province Key Laboratory of Bioengineering Drugs, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Chunjian Jia
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Xinfeng Zhang
- Jiangxi Province Key Laboratory of Bioengineering Drugs, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Qifa Huang
- Department of Obstetrics & Gynecology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jing Wei
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Qi Chen
- Department of Obstetrics & Gynecology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| | - Tingtao Chen
- Jiangxi Province Key Laboratory of Bioengineering Drugs, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China.
| |
Collapse
|
33
|
Zhao Z, Li P, Liu Z, Cui Y, Yao Z, Chen W, Wang M, Yu C, Xia S, Sun Y, Zhang N, Shen W. Exploring the molecular mechanisms by which secretory phospholipase a2 regulates lymphatic endothelial cell dysfunction by activating macrophages. Int J Biol Macromol 2025; 294:139038. [PMID: 39708872 DOI: 10.1016/j.ijbiomac.2024.139038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
This study offers new insights into the dual role of secretory phospholipase A2 (sPLA2) in lymphedema, highlighting its impact on lymphatic endothelial cell (LEC) functionality. Through transcriptomic analyses and co-culture experiments, we observed that sPLA2 has both protective and detrimental effects on human LECs (HLECs), mediated by macrophage activation. Our findings reveal that while low levels of sPLA2 promote LEC health, excessive sPLA2 leads to dysfunction, emphasizing the significance of the sPLA2/PLA2R axis and arachidonic acid metabolism (AA) in lymphedema pathology. The study suggests targeting sPLA2 and its downstream pathways as a novel therapeutic strategy for lymphedema, aiming to mitigate its progression by safeguarding HLEC integrity. This research underscores the importance of balanced sPLA2 activity in maintaining lymphatic vessel health and presents a new avenue for lymphedema management and treatment.
Collapse
Affiliation(s)
- Zimin Zhao
- Department of Lymphatic Surgery, Capital Medical University Affiliated Beijing Shijitan Hospital; Clinical Center for Lymphatic Disorders, CMU, China
| | - Peilin Li
- Department of Lymphatic Surgery, Capital Medical University Affiliated Beijing Shijitan Hospital; Clinical Center for Lymphatic Disorders, CMU, China
| | - Zhong Liu
- Department of Lymphatic Surgery, Capital Medical University Affiliated Beijing Shijitan Hospital; Clinical Center for Lymphatic Disorders, CMU, China
| | - Yonghao Cui
- Department of Lymphatic Surgery, Capital Medical University Affiliated Beijing Shijitan Hospital; Clinical Center for Lymphatic Disorders, CMU, China
| | - Zixuan Yao
- Department of Lymphatic Surgery, Capital Medical University Affiliated Beijing Shijitan Hospital; Clinical Center for Lymphatic Disorders, CMU, China
| | - Weijian Chen
- Center for Obesity and Metabolic Disease, Capital Medical University Affiliated Beijing Shijitan Hospital, Beijing, China
| | - Mengqin Wang
- Center for Obesity and Metabolic Disease, Capital Medical University Affiliated Beijing Shijitan Hospital, Beijing, China
| | - Chengyuan Yu
- Peking University Ninth Clinical School of Medicine, China
| | - Song Xia
- Department of Lymphatic Surgery, Capital Medical University Affiliated Beijing Shijitan Hospital; Clinical Center for Lymphatic Disorders, CMU, China
| | - Yuguang Sun
- Department of Lymphatic Surgery, Capital Medical University Affiliated Beijing Shijitan Hospital; Clinical Center for Lymphatic Disorders, CMU, China
| | - Nengwei Zhang
- Center for Obesity and Metabolic Disease, Capital Medical University Affiliated Beijing Shijitan Hospital, Beijing, China.
| | - Wenbin Shen
- Department of Lymphatic Surgery, Capital Medical University Affiliated Beijing Shijitan Hospital; Clinical Center for Lymphatic Disorders, CMU, China.
| |
Collapse
|
34
|
Ye Z, Sun Y, Yang S, Li L, Li B, Xia Y, Yuan T, Yu W, Chen L, Zhou X, Cheng F. Lgals3 Promotes Calcium Oxalate Crystal Formation and Kidney Injury Through Histone Lactylation-Mediated FGFR4 Activation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413937. [PMID: 39903812 PMCID: PMC11947994 DOI: 10.1002/advs.202413937] [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: 10/29/2024] [Revised: 01/19/2025] [Indexed: 02/06/2025]
Abstract
The incidence of kidney stones is increasing worldwide. However, the underlying mechanism of the process of kidney stone formation and the kidney damage caused are not well understood. Here, it is observed that Lgals3, a β-galactoside-binding protein, is significantly increased in tissues with calcium oxalate (CaOx) stones, and in both in vivo and in vitro models. Lgals3 expression is positively correlated with the deposition of CaOx crystals. Knockout of Lgals3 markedly reduces the deposition of CaOx crystal and renal fibrosis in vivo. Furthermore, Lgals3 deficiency decrease the glycolytic rate and lactate production during the process of CaOx deposition and inhibited histone lactylation of H3K18la. Mechanistic studies shows that Lgals3 directly interacted with the key glycolysis protein pyruvate kinase M2 (PKM2) and promoted its expression by modulating E3 ligase Trim21, preventing the ubiquitination of PKM2. Furthermore, H3K18 lactylation promoted CaOx crystal deposition and kidney injury in vivo and in vitro. Lgals3 deficiency inhibites the transcription, activation, and expression of FGFR4 through inhibition of H3K18la. These findings suggest that Lgals3 may play a key role in CaOx stone formation and kidney injury by interacting with PKM2 and promoting both H3K18la-mediated gene transcription and activation.
Collapse
Affiliation(s)
- Zehua Ye
- Department of UrologyRenmin hospital of Wuhan universityWuhan430060China
| | - Yushi Sun
- Department of UrologyRenmin hospital of Wuhan universityWuhan430060China
| | - Songyuan Yang
- Department of UrologyRenmin hospital of Wuhan universityWuhan430060China
| | - Lei Li
- Department of UrologyRenmin hospital of Wuhan universityWuhan430060China
| | - Bojun Li
- Department of UrologyRenmin hospital of Wuhan universityWuhan430060China
| | - Yuqi Xia
- Department of UrologyRenmin hospital of Wuhan universityWuhan430060China
| | - Tianhui Yuan
- Department of UrologyRenmin hospital of Wuhan universityWuhan430060China
| | - Weimin Yu
- Department of UrologyRenmin hospital of Wuhan universityWuhan430060China
| | - Lijia Chen
- Department of UrologyRenmin hospital of Wuhan universityWuhan430060China
| | - Xiangjun Zhou
- Department of UrologyRenmin hospital of Wuhan universityWuhan430060China
| | - Fan Cheng
- Department of UrologyRenmin hospital of Wuhan universityWuhan430060China
| |
Collapse
|
35
|
Wang L, Duan W, Ruan C, Liu J, Miyagishi M, Kasim V, Wu S. YY2-CYP51A1 signaling suppresses hepatocellular carcinoma progression by restraining de novo cholesterol biosynthesis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167658. [PMID: 39761760 DOI: 10.1016/j.bbadis.2025.167658] [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: 08/22/2024] [Revised: 12/11/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025]
Abstract
Lipid accumulation is a frequently observed characteristic of cancer. Lipid accumulation is closely related to tumor progression, metastasis, and drug resistance; however, the mechanism underlying lipid metabolic reprogramming in tumor cells is not fully understood. Yin yang 2 (YY2) is a C2H2‑zinc finger transcription factor that exerts tumor-suppressive effects. However, its involvement in tumor cell lipid metabolic reprogramming remains unclear. In the present study, we identified YY2 as a novel regulator of cholesterol metabolism. We showed that YY2 suppressed cholesterol accumulation in hepatocellular carcinoma (HCC) cells by downregulating the transcriptional activity of cytochrome P450 family 51 subfamily A member 1 (CYP51A1), a key enzyme in de novo cholesterol biosynthesis. Subsequently, through in vitro and in vivo experiments, we demonstrated that this downregulation is crucial for the YY2 tumor suppressive effect. Together, our findings unraveled a previously unprecedented regulation of HCC cells cholesterol metabolism, and eventually, their tumorigenic potential, through YY2 negative regulation on CYP51A1 expression. This study revealed a novel regulatory mechanism of lipid metabolic reprogramming in tumor cells and provided insights into the molecular mechanism underlying the YY2 the suppressive effect. Furthermore, our findings suggest a potential antitumor therapeutic strategy targeting cholesterol metabolic reprogramming using YY2.
Collapse
Affiliation(s)
- Lingxian Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China; The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Wei Duan
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China; The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Cao Ruan
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China; The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Jingyi Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China; The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Makoto Miyagishi
- Life Science Innovation, School of Integrative and Global Majors, University of Tsukuba, Tsukuba, Ibaraki 305-0006, Japan
| | - Vivi Kasim
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China; The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China; Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing University, Chongqing 400030, China.
| | - Shourong Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China; The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China; Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing University, Chongqing 400030, China.
| |
Collapse
|
36
|
Wu B, Liu Y, Li H, Zhu L, Zeng L, Zhang Z, Peng W. Liver as a new target organ in Alzheimer's disease: insight from cholesterol metabolism and its role in amyloid-beta clearance. Neural Regen Res 2025; 20:695-714. [PMID: 38886936 PMCID: PMC11433892 DOI: 10.4103/1673-5374.391305] [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: 05/31/2023] [Revised: 09/14/2023] [Accepted: 11/07/2023] [Indexed: 06/20/2024] Open
Abstract
Alzheimer's disease, the primary cause of dementia, is characterized by neuropathologies, such as amyloid plaques, synaptic and neuronal degeneration, and neurofibrillary tangles. Although amyloid plaques are the primary characteristic of Alzheimer's disease in the central nervous system and peripheral organs, targeting amyloid-beta clearance in the central nervous system has shown limited clinical efficacy in Alzheimer's disease treatment. Metabolic abnormalities are commonly observed in patients with Alzheimer's disease. The liver is the primary peripheral organ involved in amyloid-beta metabolism, playing a crucial role in the pathophysiology of Alzheimer's disease. Notably, impaired cholesterol metabolism in the liver may exacerbate the development of Alzheimer's disease. In this review, we explore the underlying causes of Alzheimer's disease and elucidate the role of the liver in amyloid-beta clearance and cholesterol metabolism. Furthermore, we propose that restoring normal cholesterol metabolism in the liver could represent a promising therapeutic strategy for addressing Alzheimer's disease.
Collapse
Affiliation(s)
- Beibei Wu
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yuqing Liu
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Hongli Li
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Lemei Zhu
- Academician Workstation, Changsha Medical University, Changsha, Hunan Province, China
| | - Lingfeng Zeng
- Academician Workstation, Changsha Medical University, Changsha, Hunan Province, China
| | - Zhen Zhang
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Yangsheng College of Traditional Chinese Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China
- Qinhuangdao Shanhaiguan Pharmaceutical Co., Ltd, Qinhuangdao, Hebei Province, China
| | - Weijun Peng
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Mental Disorder, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
37
|
Qu H, Wang C, Sun S, Zhang W, Liu C, Du X, Shu Y, Wang X, Pan Q, Luo F, Wu H, Zhang X, Liu M. Bioinformatics Identification of Lactate-Associated Genes in Hepatocellular Carcinoma: G6PD's Role in Immune Modulation. Cancer Med 2025; 14:e70801. [PMID: 40116585 PMCID: PMC11927016 DOI: 10.1002/cam4.70801] [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: 12/26/2024] [Revised: 03/08/2025] [Accepted: 03/11/2025] [Indexed: 03/23/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a major global health issue, with poor prognosis often associated with dysregulated metabolic pathways, especially lactate metabolism. This study explored the prognostic significance of lactate-associated genes in HCC and their potential as therapeutic targets. METHODS We analyzed RNA-seq and clinical data from 374 patients with HCC from The Cancer Genome Atlas (TCGA) database. Using Cox regression, LASSO analysis, and Kaplan-Meier survival curves, we identified key lactate-associated genes associated with patient outcomes. Functional validations, including Western blot, flow cytometry, and molecular docking studies, were performed to confirm the biological impact of these genes. RESULTS G6PD, IK, and CALML5 were identified as significant prognostic markers for HCC. A prognostic model was developed that effectively stratified patients into risk groups, which correlated with survival. G6PD's role in immune modulation and its potential as a drug target were validated through biochemical assays and computational analyses. Functional assays in HepG2 cells confirmed that alterations in G6PD expression affect T cell activity, with knockdown enhancing IFN-γ production and overexpression inhibiting it, demonstrating G6PD's role in immune evasion. CONCLUSIONS This study establishes lactate metabolism genes, particularly G6PD, as key prognostic markers in HCC. The validation of G6PD's immunomodulatory effects further supports its potential as a therapeutic target for strategies aimed at enhancing immune surveillance and treatment outcomes in HCC.
Collapse
Affiliation(s)
- Hao‐ran Qu
- State Key Laboratory of Virology, Hubei Province Key Laboratory of Allergy and Immunology, Department of Immunology, Taikang Medical School (School of Basic Medical Sciences)Wuhan UniversityWuhanChina
- Frontier Science Center for Immunology and Metabolism, Department of Allergy Zhongnan HospitalWuhan University School of MedicineWuhanChina
| | - Chao‐qun Wang
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanChina
- Central LaboratoryRenmin Hospital of Wuhan UniversityWuhanChina
| | - Su‐juan Sun
- State Key Laboratory of Virology, Hubei Province Key Laboratory of Allergy and Immunology, Department of Immunology, Taikang Medical School (School of Basic Medical Sciences)Wuhan UniversityWuhanChina
- Frontier Science Center for Immunology and Metabolism, Department of Allergy Zhongnan HospitalWuhan University School of MedicineWuhanChina
| | - Wen‐wen Zhang
- State Key Laboratory of Virology, Hubei Province Key Laboratory of Allergy and Immunology, Department of Immunology, Taikang Medical School (School of Basic Medical Sciences)Wuhan UniversityWuhanChina
- Frontier Science Center for Immunology and Metabolism, Department of Allergy Zhongnan HospitalWuhan University School of MedicineWuhanChina
| | - Cheng‐hao Liu
- State Key Laboratory of Virology, Hubei Province Key Laboratory of Allergy and Immunology, Department of Immunology, Taikang Medical School (School of Basic Medical Sciences)Wuhan UniversityWuhanChina
| | - Xuan‐shuang Du
- State Key Laboratory of Virology, Hubei Province Key Laboratory of Allergy and Immunology, Department of Immunology, Taikang Medical School (School of Basic Medical Sciences)Wuhan UniversityWuhanChina
- Frontier Science Center for Immunology and Metabolism, Department of Allergy Zhongnan HospitalWuhan University School of MedicineWuhanChina
| | - Yao‐yi‐ao Shu
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanChina
- Central LaboratoryRenmin Hospital of Wuhan UniversityWuhanChina
| | - Xi‐cheng Wang
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanChina
- Central LaboratoryRenmin Hospital of Wuhan UniversityWuhanChina
| | - Qin Pan
- State Key Laboratory of Virology, Hubei Province Key Laboratory of Allergy and Immunology, Department of Immunology, Taikang Medical School (School of Basic Medical Sciences)Wuhan UniversityWuhanChina
- Frontier Science Center for Immunology and Metabolism, Department of Allergy Zhongnan HospitalWuhan University School of MedicineWuhanChina
| | - Feng‐ling Luo
- State Key Laboratory of Virology, Hubei Province Key Laboratory of Allergy and Immunology, Department of Immunology, Taikang Medical School (School of Basic Medical Sciences)Wuhan UniversityWuhanChina
- Frontier Science Center for Immunology and Metabolism, Department of Allergy Zhongnan HospitalWuhan University School of MedicineWuhanChina
| | - Hong‐yan Wu
- Hubei Key Laboratory of Tumor Microenvironment and ImmunotherapyChina Three Gorges UniversityYichangChina
- School of Basic MedicineChina Three Gorges UniversityYichangChina
| | - Xiao‐lian Zhang
- State Key Laboratory of Virology, Hubei Province Key Laboratory of Allergy and Immunology, Department of Immunology, Taikang Medical School (School of Basic Medical Sciences)Wuhan UniversityWuhanChina
- Frontier Science Center for Immunology and Metabolism, Department of Allergy Zhongnan HospitalWuhan University School of MedicineWuhanChina
| | - Min Liu
- State Key Laboratory of Virology, Hubei Province Key Laboratory of Allergy and Immunology, Department of Immunology, Taikang Medical School (School of Basic Medical Sciences)Wuhan UniversityWuhanChina
- Frontier Science Center for Immunology and Metabolism, Department of Allergy Zhongnan HospitalWuhan University School of MedicineWuhanChina
| |
Collapse
|
38
|
Yin Q, Yao Y, Ni J, Zhang Y, Wu J, Zeng H, Wu W, Zhuo W, Ying J, Li J. DLAT activates EMT to promote HCC metastasis by regulating GLUT1-mediated aerobic glycolysis. Mol Med 2025; 31:71. [PMID: 39979835 PMCID: PMC11844032 DOI: 10.1186/s10020-025-01125-5] [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: 10/12/2024] [Accepted: 02/11/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Metabolic reprogramming is a hallmark of hepatocellular carcinoma (HCC) progression, driving aberrant cellular processes in response to pathological stimuli. While dihydrolipoyl transacetylase (DLAT) has been implicated in the development of various cancers, its specific role and underlying mechanisms in HCC remain unclear. This study aimed to investigate the expression, function, and mechanistic impact of DLAT in HCC. METHODS A comprehensive analysis was conducted using RNA sequencing data, tissue microarrays, in vitro and in vivo functional assays, and mechanistic studies to evaluate DLAT expression, its functional role in tumor progression, and associated molecular pathways in HCC. RESULTS Our study revealed a significant upregulation of DLAT expression in HCC, which was linked to a poor prognosis. Furthermore, we discovered that DLAT facilitated tumor metastasis by driving metabolic reprogramming in HCC cells. Mechanistically, DLAT was found to enhance glucose transporter 1 (GLUT1) expression via H3K18 acetylation, thereby promoting aerobic glycolysis and epithelial-to-mesenchymal transition (EMT), which subsequently augmented metastasis of HCC both in vitro and in vivo. Finally, we confirmed a positive correlation between DLAT and GLUT1 expression in HCC tissues. CONCLUSIONS These findings establish DLAT as a key regulator in HCC progression and suggest its potential as a promising predictive biomarker and therapeutic target for improving HCC diagnosis and treatment.
Collapse
Affiliation(s)
- Qian Yin
- Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, 310022, Zhejiang, China
- Department of Hepato-Pancreato-Biliary & Gastric Medical Oncology, Hangzhou Institute of Medicine (HIM), Zhejiang Cancer Hospital, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, P. R. China
| | - Yinye Yao
- Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, 310022, Zhejiang, China
- Department of Hepato-Pancreato-Biliary & Gastric Medical Oncology, Hangzhou Institute of Medicine (HIM), Zhejiang Cancer Hospital, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, P. R. China
| | - Jiaojiao Ni
- Department of Hepato-Pancreato-Biliary & Gastric Medical Oncology, Hangzhou Institute of Medicine (HIM), Zhejiang Cancer Hospital, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, P. R. China
| | - Yiwen Zhang
- Zhejiang Provincial Clinical Research Center for Malignant Tumor, Hangzhou, 310014, Zhejiang, P. R. China
| | - Jia Wu
- Hepatobiliary and Pancreatic Surgery Department, Hangzhou Institute of Medicine (HIM), Zhejiang Cancer Hospital, Chinese Academy of Sciences, Hangzhou, Zhejiang, P. R. China
| | - Hui Zeng
- Department of Interventional Radiology, Hangzhou Institute of Medicine (HIM), Zhejiang Cancer Hospital, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, P. R. China
| | - Wei Wu
- Department of Pathology, Hangzhou Institute of Medicine (HIM), Zhejiang Cancer Hospital, Chinese Academy of Sciences, Hangzhou, Zhejiang, P. R. China
| | - Wei Zhuo
- Department of Cell Biology, Department of Colorectal Surgery and Oncology, Center for Medical Research and Innovation in Digestive System Tumors, The Second Affiliated Hospital, Cancer Center, Ministry of Education, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Jieer Ying
- Postgraduate training base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, 310022, Zhejiang, China.
- Department of Hepato-Pancreato-Biliary & Gastric Medical Oncology, Hangzhou Institute of Medicine (HIM), Zhejiang Cancer Hospital, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, P. R. China.
| | - Jingjing Li
- Department of Hepato-Pancreato-Biliary & Gastric Medical Oncology, Hangzhou Institute of Medicine (HIM), Zhejiang Cancer Hospital, Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, P. R. China.
| |
Collapse
|
39
|
Yang H, Li J, Niu Y, Zhou T, Zhang P, Liu Y, Li Y. Interactions between the metabolic reprogramming of liver cancer and tumor microenvironment. Front Immunol 2025; 16:1494788. [PMID: 40028341 PMCID: PMC11868052 DOI: 10.3389/fimmu.2025.1494788] [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: 09/11/2024] [Accepted: 01/29/2025] [Indexed: 03/05/2025] Open
Abstract
Metabolic reprogramming is one of the major biological features of malignant tumors, playing a crucial role in the initiation and progression of cancer. The tumor microenvironment consists of various non-cancer cells, such as hepatic stellate cells, cancer-associated fibroblasts (CAFs), immune cells, as well as extracellular matrix and soluble substances. In liver cancer, metabolic reprogramming not only affects its own growth and survival but also interacts with other non-cancer cells by influencing the expression and release of metabolites and cytokines (such as lactate, PGE2, arginine). This interaction leads to acidification of the microenvironment and restricts the uptake of nutrients by other non-cancer cells, resulting in metabolic competition and symbiosis. At the same time, metabolic reprogramming in neighboring cells during proliferation and differentiation processes also impacts tumor immunity. This article provides a comprehensive overview of the metabolic crosstalk between liver cancer cells and their tumor microenvironment, deepening our understanding of relevant findings and pathways. This contributes to further understanding the regulation of cancer development and immune evasion mechanisms while providing assistance in advancing personalized therapies targeting metabolic pathways for anti-cancer treatment.
Collapse
Affiliation(s)
- Haoqiang Yang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Jinghui Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yiting Niu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Tao Zhou
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Pengyu Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yang Liu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yanjun Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, TongjiShanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
40
|
Chen X, Zhang F, Lu C, Wu R, Yang B, Liao T, Du B, Wu F, Ding J, Fang S, Zhao Z, Chen M, Shu G, Chen W, Ji J. Lactate-Fueled Theranostic Nanoplatforms for Enhanced MRI-Guided Ferroptosis Synergistic with Immunotherapy of Hepatocellular Carcinoma. ACS APPLIED MATERIALS & INTERFACES 2025; 17:9155-9172. [PMID: 39901437 DOI: 10.1021/acsami.4c21890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2025]
Abstract
Treatment for hepatocellular carcinoma (HCC) may be improved with ferroptosis, a regulated form of cell death. However, the sensitivity of HCC to ferroptosis was strongly limited by lactic acid. In this study, a platelet membrane (PM)-engineered nanoparticle loaded with erastin, superparamagnetic iron oxide nanoparticles (SPIO) and lactate oxidase (LOX) (termed PM@ESL NPs) was designed for magnetic resonance imaging (MRI)-guided enhanced ferroptosis-immunotherapy of HCC. It was found that PM@ESL NPs could actively accumulate into the tumor due to the tumor-homing ability of PM. Subsequently, PM@ESL NPs could effectively enhance the sensitivity of HCC to ferroptosis by removing the lactic acid in the tumor. The removal of lactic acid also produces hydrogen peroxide (H2O2), which therefore converted into the cytotoxic hydroxyl radicals by the reaction of H2O2 with Fe2+/Fe3+ released from SPIO. Due to the combined ferroptosis and chemodynamic therapy (CDT), PM@ESL NPS showed a strong ability to induce immunogenic cell death (ICD), which could effectively suppress the growth and metastasis of HCC when combined with αPD-L1 immunotherapy. Furthermore, the incorporation of SPIO endows PM@ESL NPs with an outstanding MRI-T2 monitoring capability for HCC treatment. In conclusion, this study introduces a pioneering MRI-guided approach that enhances ferroptosis in tumors and synergistically improves immunotherapy.
Collapse
Affiliation(s)
| | - Feng Zhang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Chenying Lu
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Ronghua Wu
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Clinical College of The Affiliated Central Hospital, School of Medcine, Lishui University, Lishui 323000, China
| | - Baozhu Yang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Tingting Liao
- College of pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 310015, China
| | - Baojie Du
- Shanxi Bethune Hospital, Third Hospital of Shanxi Medical University, Shanxi Academy of Medical Sciences, Taiyuan 030032, China
| | - Fazong Wu
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Jiayi Ding
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Shiji Fang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Zhongwei Zhao
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Clinical College of The Affiliated Central Hospital, School of Medcine, Lishui University, Lishui 323000, China
| | - Minjiang Chen
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Gaofeng Shu
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Weiqian Chen
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Jiansong Ji
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- Department of radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
- College of pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 310015, China
- Clinical College of The Affiliated Central Hospital, School of Medcine, Lishui University, Lishui 323000, China
| |
Collapse
|
41
|
Feng X, Hu H, Zhong F, Hou Y, Li X, Qin Q, Yang Y, Luo X. Lactiplantibacillus plantarum TCCC11824 exerts hypolipidemic and anti-obesity effects through regulation of NF-κB-HMGCR pathway and gut microbiota in mice and clinical patients. Nutrition 2025; 130:112598. [PMID: 39612553 DOI: 10.1016/j.nut.2024.112598] [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: 04/22/2024] [Revised: 09/22/2024] [Accepted: 09/27/2024] [Indexed: 12/01/2024]
Abstract
A strong association exists between the high-fat diet (HFD) and the incidence of obesity, hyperlipidemia and cardiovascular disease, affecting an increasing number of individuals. More and more research has shown that probiotics and gut microbiota play important roles in dietary absorption, metabolism, and general health of the host. This aim of this study was to evaluate the therapeutic effects and the underlying mechanisms of Lactiplantibacillus plantarum TCCC11824 (CGMCC 8198) on hyperlipidemia and obesity in mice and humans. First, there was a dose-dependent improvement in HFD-induced hyperlipidemia and obesity in mice that had been treated with L. plantarum TCCC11824 for 5 wk, thus restoring the balance of the gut microbiota. Furthermore, it showed that cell lysate of L. plantarum TCCC11824 could directly exhibit protective effects on the hepatocyte steatosis induced by oleic acid, and regulate the expression of HMGCR by inhibiting the NF-κB pathway. Importantly, L.plantarum TCCC11824 ameliorated the expression of indicators of hyperlipidemia and inhibited the synthesis of SCFAs (short-chain fatty acids), as shown by blood and fecal tests in hyperlipidemic patients. In summary, L. plantarum TCCC11824 exerts anti-hyperlipidemic and anti-obesity effects through the regulation of HMGCR via NF-κB and modulating gut microbiota, indicating its potential as a dietary supplement for the treatment of hyperlipidemia and obesity.
Collapse
Affiliation(s)
- Xiaomin Feng
- Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education & Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Haijie Hu
- Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education & Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Feiliang Zhong
- Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education & Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Ying Hou
- Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education & Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China; Harbin Meihua Biotechnolgy Co., Ltd. Haerbin, Heilongjiang, Province, China
| | - Xiujuan Li
- Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education & Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China; Harbin Meihua Biotechnolgy Co., Ltd. Haerbin, Heilongjiang, Province, China
| | - Qi Qin
- Harbin Meihua Biotechnolgy Co., Ltd. Haerbin, Heilongjiang, Province, China
| | - Yang Yang
- Department of Endocrinology, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan Hubei, Province, China
| | - Xuegang Luo
- Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education & Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China.
| |
Collapse
|
42
|
He K, Tao F, Lu Y, Fang M, Huang H, Zhou Y. The Role of HK2 in Tumorigenesis and Development: Potential for Targeted Therapy with Natural Products. Int J Med Sci 2025; 22:790-805. [PMID: 39991762 PMCID: PMC11843137 DOI: 10.7150/ijms.105553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/11/2025] [Indexed: 02/25/2025] Open
Abstract
Hexokinase 2 (HK2) is widely distributed in various tissues, particularly showing significantly elevated expression levels in tumor tissues. As the initial rate-limiting enzyme in the glycolysis process, HK2 is believed to directly participate in the metabolic reprogramming of tumor cells. This phenomenon, known as the "Warburg effect," provides the energy and substances necessary for the rapid proliferation, growth, and division of tumor cells. Furthermore, by enhancing glycolysis, HK2 exerts its influence on various metabolic pathways in tumor cells, such as pentose phosphate metabolism, glutamine metabolism, serine metabolism, and glycine metabolism, thereby playing a role in the occurrence and development of cancer. Therefore, HK2 represents a promising target for cancer therapy. Simultaneously, natural products with effects on inhibiting the expression or activity of HK2, have already been discovered to exhibit significant anticancer potential. Flavonoids, pentacyclic triterpenoids, phenolic compounds, and lignans constitute the majority of these natural products, directly inhibiting HK2 or indirectly downregulating it through protein kinase B (AKT), hypoxia-inducible factor 1 alpha (HIF-1α), and c-Myc signaling pathways. However, several challenges remain, such as further screening for natural products that directly target and inhibit HK2, optimizing the selection of natural product inhibitors for HK2, and elucidating the molecular mechanisms by which natural products indirectly inhibit HK2. In conclusion, the potential of targeting HK2 for cancer therapy is promising, and with these challenges addressed, natural products inhibiting HK2 will play an even greater role in the fight against cancer.
Collapse
Affiliation(s)
- Keren He
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Fangfang Tao
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yangyuxiao Lu
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Mengqi Fang
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Hong Huang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuan Zhou
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Key Laboratory of Blood-Stasis-Toxin Syndrome, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
43
|
Huang Z, Chen J, Liu S, Xiang X, Long Y, Tan P, Fu W. MAP17 is a Novel NASH Progression Biomarker Associated with Macrophage Infiltration, Immunotherapy Response, and Oxidative Stress. J Inflamm Res 2025; 18:601-619. [PMID: 39839187 PMCID: PMC11747966 DOI: 10.2147/jir.s497737] [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: 09/24/2024] [Accepted: 01/08/2025] [Indexed: 01/23/2025] Open
Abstract
Background Nonalcoholic steatohepatitis (NASH) has recently garnered increased attention due to immune infiltration. However, the role of membrane-associated protein 17 (MAP17) in NASH remains unclear, which prompted this study to explore its relationship with immune infiltration and its regulatory mechanisms. Methods We employed weighted correlation network analysis (WGCNA) to construct a gene co-expression network aimed at identifying key genes associated with NASH progression. Our further analyses included differential expression evaluation, protein-protein interaction (PPI) network analysis, and Venn diagram analysis to discover novel targets. The CIBERSORT algorithm assessed the correlation between MAP17 and immune cell infiltration within the tumor microenvironment (TME), while the TIDE algorithm predicted responses to immunotherapy. Additionally, we conducted gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and gene set enrichment analysis (GSEA) to elucidate the mechanisms by which MAP17 operates. The expression of MAP17 was validated using liver tissues obtained from NASH patients and mice with diet-induced NASH or CCl4-induced liver fibrosis. Results Our findings identified MAP17 as a novel target in the progression of NASH. Correlation analyses demonstrated a positive association between MAP17 and M1 macrophage infiltration, as well as a negative association with M2 infiltration. TIDE results positioned MAP17 as a potential biomarker for predicting responses to immune checkpoint blockade. Mechanistic studies revealed that MAP17 induced oxidative stress, which subsequently activated the p53, PI3K-AKT, and Wnt signaling pathways. Validation analyses confirmed that MAP17 levels significantly increased in liver tissues of mice with diet-induced NASH or CCl4-induced liver fibrosis, as well as in NASH patients. Conclusion MAP17 is a novel biomarker linked to macrophage infiltration and immunotherapy responses in NASH patients. The oxidative stress induced by MAP17 activates the p53, PI3K-AKT, and Wnt pathways, all of which contribute to the progression of NASH.
Collapse
Affiliation(s)
- Zhiwei Huang
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Jiatong Chen
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Shenglu Liu
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Xin Xiang
- Department of General Surgery, The First People’s Hospital of Neijiang, Neijiang, 641000, People’s Republic of China
| | - Yang Long
- Department of Endocrinology and Metabolism, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Peng Tan
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, Academician (Expert) Workstation of Sichuan Province, Department of General Surgery (Hepatopancreatobiliary surgery), The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Wenguang Fu
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, Academician (Expert) Workstation of Sichuan Province, Department of General Surgery (Hepatopancreatobiliary surgery), The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, People’s Republic of China
| |
Collapse
|
44
|
Li B, Zeng T, Chen C, Wu Y, Huang S, Deng J, Pang J, Cai X, Lin Y, Sun Y, Chong Y, Li X, Gong J, Tang G. Unraveling the potential mechanism and prognostic value of pentose phosphate pathway in hepatocellular carcinoma: a comprehensive analysis integrating bulk transcriptomics and single-cell sequencing data. Funct Integr Genomics 2025; 25:11. [PMID: 39798003 DOI: 10.1007/s10142-024-01521-w] [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: 08/16/2024] [Revised: 11/25/2024] [Accepted: 12/28/2024] [Indexed: 01/13/2025]
Abstract
Hepatocellular carcinoma (HCC) remains a malignant and life-threatening tumor with an extremely poor prognosis, posing a significant global health challenge. Despite the continuous emergence of novel therapeutic agents, patients exhibit substantial heterogeneity in their responses to anti-tumor drugs and overall prognosis. The pentose phosphate pathway (PPP) is highly activated in various tumor cells and plays a pivotal role in tumor metabolic reprogramming. This study aimed to construct a model based on PPP-related Genes for risk assessment and prognosis prediction in HCC patients. We integrated RNA-seq and microarray data from TCGA, GEO, and ICGC databases, along with single-cell RNA sequencing (scRNA-seq) data obtained from HCC patients via GEO. Based on the "Seurat" R package, we identified distinct gene clusters related to the PPP within the scRNA-seq data. Using a penalized Cox regression model with least absolute shrinkage and selection operator (LASSO) penalties, we constructed a risk prognosis model. The validity of our risk prognosis model was further confirmed in external cohorts. Additionally, we developed a nomogram capable of accurately predicting overall survival in HCC patients. Furthermore, we explored the predictive potential of our risk model within the immune microenvironment and assessed its relevance to biological function, particularly in the context of immunotherapy. Subsequently, we performed in vitro functional validation of the key genes (ATAD2 and SPP1) in our model. A ten-gene signature associated with the PPP was formulated to enhance the prediction of HCC prognosis and anti-tumor treatment response. Following this, the ROC curve, nomogram, and calibration curve outcomes corroborated the model's robust clinical predictive capability. Functional enrichment analysis unveiled the engagement of the immune system and notable variances in the immune infiltration landscape across the high and low-risk groups. Additionally, tumor mutation frequencies were observed to be elevated in the high-risk group. Based on our analyses, the IC50 values of most identified anticancer agents demonstrated a correlation with the RiskScore. Additionally, the high-risk and low-risk groups exhibited differential sensitivity to various drugs. Cytological experiments revealed that silencing ATAD2 or SPP1 suppresses malignant phenotypes, including viability and migration, in liver cancer cells. In this study, a novel gene signature related to the PPP was developed, demonstrating favorable predictive performance. This signature holds significant guiding value for assessing the prognosis of HCC patients and directing individualized treatment strategies.
Collapse
Affiliation(s)
- Bin Li
- Department of Infectious Diseases, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Tao Zeng
- Department of Infectious Diseases, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Cui Chen
- Department of Thyroid and Breast Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Yuankai Wu
- Department of Infectious Diseases, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Shuying Huang
- Department of Infectious Diseases, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Jing Deng
- Department of Infectious Diseases, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Jiahui Pang
- Department of Infectious Diseases, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Xiang Cai
- Department of Infectious Diseases, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Yuxi Lin
- Department of Infectious Diseases, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Yina Sun
- Department of Laboratory Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Yutian Chong
- Department of Infectious Diseases, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Xinhua Li
- Department of Infectious Diseases, Key Laboratory of Liver Disease of Guangdong Province, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China.
| | - Jiao Gong
- Department of Laboratory Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China.
| | - Guofang Tang
- Institute of Infectious Diseases, Guangdong Province, Guangzhou Eighth People's Hospital, Guangzhou Medical University, 8 Huaying Road, Baiyun District, Guangzhou, 510440, China.
| |
Collapse
|
45
|
Liang J, Liu H, Lv G, Chen X, Yang Z, Hu K, Sun H. Exploring the molecular mechanisms of tirzepatide in alleviating metabolic dysfunction-associated fatty liver in mice through integration of metabolomics, lipidomics, and proteomics. Lipids Health Dis 2025; 24:8. [PMID: 39794823 PMCID: PMC11720920 DOI: 10.1186/s12944-024-02416-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/19/2024] [Indexed: 01/13/2025] Open
Abstract
Clinical studies have suggested that tirzepatide may also possess hepatoprotective effects; however, the molecular mechanisms underlying this association remain unclear. In our study, we performed biochemical analyses of serum and histopathological examinations of liver tissue in mice. To preliminarily explore the molecular mechanisms of tirzepatide on metabolic dysfunction-associated fatty liver disease (MAFLD), liquid chromatography-mass spectrometry (LC-MS) was employed for comprehensive metabolomic, lipidomic, and proteomic analyses in MAFLD mice fed a high-fat diet (HFD). The results demonstrated that tirzepatide significantly reduced serum levels of alanine transaminase (ALT) and aspartate transaminase (AST), as well as hepatic triglycerides (TG) and total cholesterol (TC), indicating its efficacy in treating MAFLD. Further findings revealed that tirzepatide reduced fatty acid uptake by downregulating Cd36 and Fabp2/4, as well as enhance the mitochondrial-lysosomal function by upregulating Lamp1/2. In addition, tirzepatide promoted cholesterol efflux and reduced cholesterol reabsorption by upregulating the expression of Hnf4a, Abcg5, and Abcg8. These results suggest that tirzepatide exerts its therapeutic effects on MAFLD by reducing fatty acid uptake, promoting cholesterol excretion, and enhancing mitochondrial-lysosomal function, providing a theoretical basis for a comprehensive understanding of tirzepatide.
Collapse
Affiliation(s)
- Jinliang Liang
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Huanyi Liu
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Guo Lv
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Xiaotong Chen
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Zhaoshou Yang
- The First Affiliated Hospital, The First School of Clinical Medicine of Guangdong Pharmaceutical University, Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Kunhua Hu
- Guangdong Provincial Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China.
| | - Hongyan Sun
- The State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
46
|
He Q, Lin J, Mo C, Li G, Lu J, Sun Q, Cao L, Gan H, Sun Q, Yao J, Lian S, Wang W. Endothelin receptor antagonists (ERAs) can potentially be used as therapeutic drugs to reduce hypertension caused by small molecule tyrosine kinase inhibitors (TKIs). Front Pharmacol 2025; 15:1463520. [PMID: 39850566 PMCID: PMC11754196 DOI: 10.3389/fphar.2024.1463520] [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: 07/12/2024] [Accepted: 12/23/2024] [Indexed: 01/25/2025] Open
Abstract
The emergence of targeted anti-tumor drugs has significantly prolonged the lifespan and improved the prognosis of cancer patients. Among these drugs, vascular endothelial growth factor (VEGF) inhibitors, particularly novel small molecule tyrosine kinase inhibitors (TKIs), are extensively employed as VEGF inhibitors; however, they are also associated with a higher incidence of complications, with hypertension being the most prevalent cardiovascular toxic side effect. Currently, it is widely accepted that TKIs-induced hypertension involves multiple mechanisms including dysregulation of the endothelin (ET) axis, reduced bioavailability of nitric oxide (NO), imbalance in NO-ROS equilibrium system, vascular rarefaction, and activation of epithelial sodium calcium channels; nevertheless, excessive activation of ET system appears to be predominantly responsible for this condition. Moreover, studies have demonstrated that ET plays a pivotal role in driving TKIs-induced hypertension. Therefore, this review aims to explore the significance of ET in the pathogenesis of hypertension induced by targeted anti-tumor drugs and investigate the potential therapeutic value of endothelin antagonists in managing hypertension caused by targeted anti-tumor drugs.
Collapse
Affiliation(s)
- Qingjian He
- Department of Breast and Thyroid Surgery, First Affiliated Hospital of Huzhou University, Huzhou, China
| | - Junling Lin
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, Huzhou, China
| | - Chanjuan Mo
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, Huzhou, China
| | - Guodong Li
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, Huzhou, China
| | - Jianzhong Lu
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, Huzhou, China
| | - Qiyin Sun
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, Huzhou, China
| | - Lijun Cao
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, Huzhou, China
| | - Haojian Gan
- School of Medicine, Huzhou University, Huzhou, China
| | - Quan Sun
- School of Medicine, Huzhou University, Huzhou, China
| | - Jiafang Yao
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, Huzhou, China
| | - Shengyi Lian
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, Huzhou, China
| | - WenJuan Wang
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, Huzhou, China
| |
Collapse
|
47
|
Shi H, Yu J, Li L, Ji M, Li R, Peng T, Cheng Y, Wang T, Yang L, Wu Z, Zhang G, Wang F, Lu X. Design, Synthesis, and Antitumor Activity Evaluation of 2-Phenylthiazole-5-Carboxylic Acid Derivatives Targeting Transactivation Response RNA-Binding Protein 2. J Med Chem 2025; 68:421-447. [PMID: 39722648 DOI: 10.1021/acs.jmedchem.4c02041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
Transactivation response (TAR) RNA-binding protein 2 (TRBP) plays a critical role in microRNA (miRNA) biosynthesis, with aberrant expression linked to various cancers. Previously, we identified CIB-3b, a phenyloxazole derivative that disrupts the TRBP-Dicer interaction in hepatocellular carcinoma (HCC). In this study, we optimized this scaffold and substituent, leading to the discovery of CIB-L43, a 2-phenylthiazole-5-carboxylic acid derivative with nanomolar inhibitory activity (EC50 = 0.66 nM). CIB-L43 demonstrated superior TRBP binding affinity (KD = 4.78 nM) and enhanced disruption of TRBP-Dicer interactions (IC50 = 2.34 μM). Mechanistically, CIB-L43 suppressed oncogenic miR-21 biosynthesis, increasing PTEN and Smad7 expression and inhibiting AKT and TGF-β signaling, thereby reducing HCC cell proliferation and migration. In vivo, CIB-L43 exhibited favorable pharmacokinetics, including 53.9% oral bioavailability, and comparable antitumor efficacy to first-line anticancer drug, sorafenib, with lower toxicity. CIB-L43 emerges as a promising HCC treatment candidate with potent TRBP inhibition and favorable drug-like properties.
Collapse
Affiliation(s)
- Hailong Shi
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jialing Yu
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lingyu Li
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Minghui Ji
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Runze Li
- Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Ting Peng
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu 610052, China
| | - Yao Cheng
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tao Wang
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Linhan Yang
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhao Wu
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guolin Zhang
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| | - Fei Wang
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| | - Xiaoxia Lu
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| |
Collapse
|
48
|
Tang Z, Wei C, Deng X, Lin Q, Hu Q, Li S, Wang J, Wu Y, Liu D, Fang M, Zhan T. Serum proteomic and metabolomic profiling of hepatocellular carcinoma patients co-infected with Clonorchis sinensis. Front Immunol 2025; 15:1489077. [PMID: 39840062 PMCID: PMC11746118 DOI: 10.3389/fimmu.2024.1489077] [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: 08/31/2024] [Accepted: 12/10/2024] [Indexed: 01/23/2025] Open
Abstract
Background Clonorchis sinensis (C. sinensis) infection is a significant risk factor for hepatocellular carcinoma (HCC), yet its underlying mechanisms remain poorly understood. This study aimed to investigate the impact of C. sinensis infection on the serum proteomic and metabolomic profiling of HCC patients, focusing on the potential mechanisms. Method A retrospective clinical analysis was conducted on 1121 HCC patients, comparing those with and without C. sinensis infection. The influence of C. sinensis on serum proteome and metabolome in HCC was further assessed. Result C. sinensis infection correlated with a younger age at cancer onset, male predominance, advanced cancer stage, liver cirrhosis, and microvascular invasion in HCC patients. It also associated with shorter overall survival (OS) and recurrence-free survival (RFS). The levels of blood lipids (e.g., APO-A, HDL-C, and TG) were significantly altered after C. sinensis infection. Proteomic and metabolomic analyses revealed metabolic reprogramming caused by C. sinensis, with excessive depletion of argininosuccinate synthase (ASS) and D-glucose as potential factors in C. sinensis-associated HCC malignancy. Key molecules ILF2, CNN2, OLFM4, NOTCH3, and LysoPA were implicated in HCC progression. Furthermore, C. sinensis triggered inflammation, insulin resistance, and pro-tumor immune escape, and exacerbated the complication of degenerative diseases. Conclusion This study not only provides compelling evidence for elucidating the mechanisms underlying C. sinensis-mediated HCC development but also identifies potential therapeutic targets for HCC patients co-infected with C. sinensis.
Collapse
Affiliation(s)
- Zeli Tang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, China
- Key Laboratory of Basic Research on Regional Diseases, Education Department of Guangxi Zhuang Autonomous Region, Guangxi Medical University, Nanning, China
| | - Caibiao Wei
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xueling Deng
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
| | - Qiumei Lin
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Qiping Hu
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, China
- Key Laboratory of Basic Research on Regional Diseases, Education Department of Guangxi Zhuang Autonomous Region, Guangxi Medical University, Nanning, China
| | - Shitao Li
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
| | - Jilong Wang
- Department of Parasitology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
| | - Yuhong Wu
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
| | - Dengyu Liu
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, China
- Key Laboratory of Basic Research on Regional Diseases, Education Department of Guangxi Zhuang Autonomous Region, Guangxi Medical University, Nanning, China
- Department of Parasitology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
| | - Min Fang
- Department of Clinical Laboratory, Guangxi Medical University Cancer Hospital, Nanning, China
- Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Tingzheng Zhan
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning, China
- Key Laboratory of Basic Research on Regional Diseases, Education Department of Guangxi Zhuang Autonomous Region, Guangxi Medical University, Nanning, China
- Department of Parasitology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
| |
Collapse
|
49
|
Wang L, Ma S, Su H, Nie D, Wang L. The molecular mechanism of gemcitabine in inhibiting the HIF-1α/VEGFB/FGF2/FGFR1 signaling pathway for ovarian cancer treatment. Discov Oncol 2025; 16:3. [PMID: 39752011 PMCID: PMC11699178 DOI: 10.1007/s12672-024-01723-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025] Open
Abstract
Ovarian cancer is a common malignant tumor in women, exhibiting a certain sensitivity to chemotherapy drugs like gemcitabine (GEM). This study, through the analysis of ovarian cancer single-cell RNA sequencing (scRNA-seq) data and transcriptome data post-GEM treatment, identifies the pivotal role of hypoxia-inducible factor 1 alpha (HIF-1α) in regulating the treatment process. The results reveal that HIF-1α modulates the expression of VEGF-B, thereby inhibiting the fibroblast growth factor 2 (FGF2)/FGFR1 signaling pathway and impacting tumor formation. In vitro experiments validate the mechanistic role of HIF-1α in GEM treatment, demonstrating that overexpression of HIF-1α reverses the drug's effects on ovarian cancer cells while silencing fibroblast growth factor receptor 1 (FGFR1) can restore treatment efficacy. These findings provide essential molecular targets and a theoretical foundation for the development of novel treatment strategies for ovarian cancer in the future.
Collapse
Affiliation(s)
- Liangliang Wang
- Department of Oncology and Gynecology, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzihu District, Bengbu, Anhui, China
| | - Shanshan Ma
- Department of Oncology and Gynecology, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzihu District, Bengbu, Anhui, China
| | - Huiwen Su
- Department of Oncology and Gynecology, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzihu District, Bengbu, Anhui, China
| | - Dandan Nie
- Department of Oncology and Gynecology, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzihu District, Bengbu, Anhui, China
| | - Lihua Wang
- Department of Oncology and Gynecology, The First Affiliated Hospital of Bengbu Medical University, No. 287, Changhuai Road, Longzihu District, Bengbu, Anhui, China.
| |
Collapse
|
50
|
Yan M, Cui Y, Xiang Q. Metabolism of hepatic stellate cells in chronic liver diseases: emerging molecular and therapeutic interventions. Theranostics 2025; 15:1715-1740. [PMID: 39897543 PMCID: PMC11780521 DOI: 10.7150/thno.106597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 12/10/2024] [Indexed: 02/04/2025] Open
Abstract
Chronic liver diseases, primarily metabolic dysfunction-associated steatotic liver disease (MASLD), metabolic and metabolic dysfunction-associated alcoholic liver disease (MetALD), and viral hepatitis, can lead to liver fibrosis, cirrhosis, and cancer. Hepatic stellate cell (HSC) activation plays a central role in the development of myofibroblasts and fibrogenesis in chronic liver diseases. However, HSC activation is influenced by the complex microenvironments within the liver, which are largely shaped by the interactions between HSCs and various other cell types. Changes in HSC phenotypes and metabolic mechanisms involve glucose, lipid, and cholesterol metabolism, oxidative stress, activation of the unfolded protein response (UPR), autophagy, ferroptosis, senescence, and nuclear receptors. Clinical interventions targeting these pathways have shown promising results in addressing liver inflammation and fibrosis, as well as in modulating glucose and lipid metabolism and metabolic stress responses. Therefore, a comprehensive understanding of HSC phenotypes and metabolic mechanisms presents opportunities for novel therapeutic approaches aimed at halting or even reversing chronic liver diseases.
Collapse
Affiliation(s)
- Mengyao Yan
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, China
| | - Yimin Cui
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, China
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| | - Qian Xiang
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, China
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| |
Collapse
|