1
|
Wang Z, Liu T, He K, Wang L, Ma X, Yang Z, Zhang Y, Zhao L. Knockdown of HGH1 in breast cancer cell lines can inhibit the viability, invasion and migration of tumor cells. Cell Adh Migr 2025; 19:1-14. [PMID: 39691959 DOI: 10.1080/19336918.2024.2442349] [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: 06/21/2024] [Revised: 10/19/2024] [Accepted: 12/09/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND Research on the function of HGH1 in breast cancer remains lacking. METHODS TCGAand GEO (GSE45827) datasets investigated discrepancies in HGH1 expression in BC. An aggregate of 106 clinical samples were gathered through immunohistochemistry, KM curves were drawn for prognostic analysis, and the function of HGH1 of BC was predicted. Finally, the effects of HGH1 knockdown on MDA-MB-231 and MCF-7 BC cells were verified via CCK8, invasion, wound healing and colony formation assays. RESULTS HGH1 is highly expressed in BC and is linked to unfavorable prognosis. HGH1 overexpression is connected to keratinization and the cell cycle and is closely related to ER and PR expression and tumor stage in BC patients. Knocking down HGH1 in BC cells inhibited the viability, invasion and migration. CONCLUSION Knockdown of HGH1 in breast cancer cell lines can inhibit the viability, invasion and migration of tumor cells.
Collapse
Affiliation(s)
- Zeyu Wang
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Taiyuan Liu
- Department of Breast Surgery, Second Hospital of Jilin University, Changchun, China
| | - Kang He
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Longyun Wang
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Xiaoxuan Ma
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Zhaoyun Yang
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Yingchao Zhang
- Department of Breast Surgery, Second Hospital of Jilin University, Changchun, China
| | - Lijing Zhao
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| |
Collapse
|
2
|
Wang H, Mei Q, Mei P. Comprehensive analysis of the role of Caspases in glioma. Brain Res 2025; 1855:149529. [PMID: 40032044 DOI: 10.1016/j.brainres.2025.149529] [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: 09/15/2024] [Revised: 02/17/2025] [Accepted: 02/21/2025] [Indexed: 03/05/2025]
Abstract
Caspases (CASPs) are attractive targets for cancer therapy. Many prognostic models based on gene signatures include genes from the CASPs family in diffuse glioma. CASP3, CASP4 and CASP6 in glioma have been studied individually. However, specialized comprehensive analysis of the roles of CASPs family in glioma is lacking. Therefore, this study utilized bioinformatics methods to investigate this issue. CASP1-10 expressionlevels were significantly up-regulated in LGG and GBM and glioma, and varied significantly across different clinical subgroups of glioma and LGG and various cell types, and most of CASP1-10 members showed significant differences in recurrence status of LGG. 10 signatures (CASP1-10) were associated with poor overall survival (OS) in glioma and LGG and GBM. However, pan-cancer survival analysis showed that CASP1-10 were associated with the prognosis of LGG, but not GBM. CASP1-10 were related to poor prognosis of glioma and LGG, except for CASP9, which was the opposite of a protective factor. CASP1-10 were independent prognostic factors for OS in glioma and LGG, except for CASP5, and also for recurrence-free survival (RFS) in LGG. Most of CASP1-10 were also independent prognostic factors for disease-specific survival (DSS) and progression-free interval (PFI) and had diagnostic value in glioma and LGG. Genetic alterations of CASP1-10 genes set were associated with poor prognosis in LGG. CASP1-10 were involved in immune infiltration and programmed cell death in glioma and LGG and GBM, and might promote the apoptosis of immune cells. Compared to GBM, CASP1-10 had a more significant impact on the prognosis, cancer-related pathways, and immune infiltration in LGG, indicating that CASP1-10 might play important roles in the recurrence and progression of LGG, and might be promising therapeutic targets for LGG. Therefore, it is speculated that natural caspase inhibitor p35 may be a promising drug for the treatment of glioma, especially for LGG.
Collapse
Affiliation(s)
- Heming Wang
- Hainan Key Laboratory for Sustainable Utilization of Tropical Bioresource, Hainan University, Haikou 570228, China
| | - Qunfang Mei
- Fujian Provincial Key Laboratory of Plant Functional Biology, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Pengying Mei
- Fujian Provincial Key Laboratory of Plant Functional Biology, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| |
Collapse
|
3
|
Lv C, Wu H, Yin D, Fang W, Zhou L. Integrative analysis of ceRNA networks and immune cell infiltration in thyroid cancer for enhanced diagnostic and prognostic insights. Sci Rep 2025; 15:12190. [PMID: 40204789 DOI: 10.1038/s41598-025-96287-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 03/27/2025] [Indexed: 04/11/2025] Open
Abstract
Thyroid cancer, the most common endocrine malignancy, has seen a significant rise in incidence, necessitating improved diagnostic and prognostic methods. Despite advancements in fine-needle aspiration biopsy (FNAB) and molecular mutation detection, these techniques have limitations, particularly in large nodules. This study aims to identify molecular markers and construct a comprehensive ceRNA regulatory network to enhance thyroid cancer diagnosis and prognosis. Using transcriptomic data from TCGA, GTEx, and GEO datasets, we performed differential expression analysis and WGCNA to identify key lncRNAs, miRNAs, and mRNAs involved in thyroid cancer. Gene Ontology and KEGG pathway analyses elucidated the biological functions and pathways of these genes. A ceRNA network was constructed, highlighting the interactions between 32 lncRNAs, 18 miRNAs, and 56 mRNAs. Survival analysis and the Cibersort algorithm further revealed the relationship between ceRNA regulatory networks and immune cell infiltration. A prognostic risk model was developed, incorporating key prognostic genes (PRR15, HCP5, and DUXAP8) and immune cells (resting NK cells, monocytes, M0 macrophages, and activated mast cells). DUXAP8 was positively correlated with activated mast cells and monocytes, while HCP5 was negatively correlated with resting NK cells. This study provides new insights into thyroid cancer pathogenesis, suggesting potential molecular markers for early diagnosis and personalized treatment. Integrating ceRNA regulatory mechanisms with immune cell analysis offers a novel perspective on the tumor microenvironment's role in thyroid cancer progression.
Collapse
Affiliation(s)
- Chang Lv
- College of Life Sciences, North China University of Science and Technology, Tangshan, 063000, Hebei, China
| | - Huazhe Wu
- College of Life Sciences, North China University of Science and Technology, Tangshan, 063000, Hebei, China
| | - Dejun Yin
- College of Life Sciences, North China University of Science and Technology, Tangshan, 063000, Hebei, China
| | - Wei Fang
- College of Life Sciences, North China University of Science and Technology, Tangshan, 063000, Hebei, China.
| | - Liming Zhou
- College of Life Sciences, North China University of Science and Technology, Tangshan, 063000, Hebei, China.
| |
Collapse
|
4
|
Xu Q, Ma L, Streuer A, Altrock E, Schmitt N, Rapp F, Klär A, Nowak V, Obländer J, Weimer N, Palme I, Göl M, Zhu HH, Hofmann WK, Nowak D, Riabov V. Machine learning-based in-silico analysis identifies signatures of lysyl oxidases for prognostic and therapeutic response prediction in cancer. Cell Commun Signal 2025; 23:169. [PMID: 40186284 PMCID: PMC11971788 DOI: 10.1186/s12964-025-02176-1] [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: 09/25/2024] [Accepted: 03/26/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Lysyl oxidases (LOX/LOXL1-4) are crucial for cancer progression, yet their transcriptional regulation, potential therapeutic targeting, prognostic value and involvement in immune regulation remain poorly understood. This study comprehensively evaluates LOX/LOXL expression in cancer and highlights cancer types where targeting these enzymes and developing LOX/LOXL-based prognostic models could have significant clinical relevance. METHODS We assessed the association of LOX/LOXL expression with survival and drug sensitivity via analyzing public datasets (including bulk and single-cell RNA sequencing data of six datasets from Gene Expression Omnibus (GEO), Chinese Glioma Genome Atlas (CGGA) and Cancer Genome Atlas Program (TCGA)). We performed comprehensive machine learning-based bioinformatics analyses, including unsupervised consensus clustering, a total of 10 machine-learning algorithms for prognostic prediction and the Connectivity map tool for drug sensitivity prediction. RESULTS The clinical significance of the LOX/LOXL family was evaluated across 33 cancer types. Overexpression of LOX/LOXL showed a strong correlation with tumor progression and poor survival, particularly in glioma. Therefore, we developed a novel prognostic model for glioma by integrating LOX/LOXL expression and its co-expressed genes. This model was highly predictive for overall survival in glioma patients, indicating significant clinical utility in prognostic assessment. Furthermore, our analysis uncovered a distinct LOXL2-overexpressing malignant cell population in recurrent glioma, characterized by activation of collagen, laminin, and semaphorin-3 pathways, along with enhanced epithelial-mesenchymal transition. Apart from glioma, our data revealed the role of LOXL3 overexpression in macrophages and in predicting the response to immune checkpoint blockade in bladder and renal cancers. Given the pro-tumor role of LOX/LOXL genes in most analyzed cancers, we identified potential therapeutic compounds, such as the VEGFR inhibitor cediranib, to target pan-LOX/LOXL overexpression in cancer. CONCLUSIONS Our study provides novel insights into the potential value of LOX/LOXL in cancer pathogenesis and treatment, and particularly its prognostic significance in glioma.
Collapse
Affiliation(s)
- Qingyu Xu
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany.
- Department of Hematology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| | - Ling Ma
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Alexander Streuer
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Eva Altrock
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Nanni Schmitt
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Felicitas Rapp
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Alessa Klär
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Verena Nowak
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Julia Obländer
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Nadine Weimer
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Iris Palme
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Melda Göl
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Hong-Hu Zhu
- Department of Hematology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Chinese Institutes for Medical Research, Beijing, China
| | - Wolf-Karsten Hofmann
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Daniel Nowak
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| | - Vladimir Riabov
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, 68169, Germany
| |
Collapse
|
5
|
Xiong X, Du Y, Liu P, Li X, Lai X, Miao H, Ning B. Unveiling EIF5A2: A Multifaceted Player in Cellular Regulation, Tumorigenesis and Drug Resistance. Eur J Pharmacol 2025:177596. [PMID: 40194645 DOI: 10.1016/j.ejphar.2025.177596] [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: 02/11/2025] [Revised: 03/28/2025] [Accepted: 04/02/2025] [Indexed: 04/09/2025]
Abstract
The eukaryotic initiation factor 5A2 gene (EIF5A2) is a highly conserved and multifunctional gene that significantly influences various cellular processes, including translation elongation, RNA binding, ribosome binding, protein binding and post-translational modifications. Overexpression of EIF5A2 is frequently observed in multiple cancers, where it functions as an oncoprotein. Additionally, EIF5A2 is implicated in drug resistance through the regulation of various molecular pathways. In the review, we describe the structure and functions of EIF5A2 in normal cells and its role in tumorigenesis. We also elucidate the molecular mechanisms associated with EIF5A2 in the context of tumorigenesis and drug resistance. We propose that the biological roles of EIF5A2 in regulating diverse cellular processes and tumorigenesis are clinically significant and warrant further investigation.
Collapse
Affiliation(s)
- Xifeng Xiong
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou 510220, Guangdong, China; Guangzhou Institute of Burn Clinical Medicine, Guangzhou Red Cross Hospital of Jinan University, Guangzhou 510220, Guangdong, China
| | - Yanli Du
- Guangdong Medical University, Zhanjiang 524023, Guangdong, China; Department of Orthopedic, Guangzhou Red Cross Hospital of Jinan University, Guangzhou 510220, Guangdong, China
| | - Peng Liu
- Departments of Burn and Plastic, Guangzhou Red Cross Hospital of Jinan University, Guangzhou 510220, Guangdong, China
| | - Xinye Li
- Guangdong Medical University, Zhanjiang 524023, Guangdong, China; Department of Orthopedic, Guangzhou Red Cross Hospital of Jinan University, Guangzhou 510220, Guangdong, China
| | - Xudong Lai
- Department of infectious disease, Guangzhou Red Cross Hospital of Jinan University, Guangzhou 510220, Guangdong, China
| | - Haixiong Miao
- Department of Orthopedic, Guangzhou Red Cross Hospital of Jinan University, Guangzhou 510220, Guangdong, China.
| | - Bo Ning
- Department of Neurosurgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou 510220, Guangdong, China.
| |
Collapse
|
6
|
Zhang Y, Jia Z, Cao D, Zhong Y, Wu Y, Fu Y, Cui Y, Yu X, Liu Y, Jiang J. RGS1 can serve as a long-term prognostic marker in gastric cancer by promoting the infiltration and polarization of macrophages. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167711. [PMID: 39933597 DOI: 10.1016/j.bbadis.2025.167711] [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: 10/27/2024] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 02/13/2025]
Abstract
Gastric cancer (GC) remains a prevalent and aggressive malignancy worldwide, characterized by significant morbidity and mortality. The regulator of G-protein signaling 1 (RGS1) plays an oncogenic role in various cancers, including GC, but its clinical relevance and mechanisms remain underexplored. In this pilot study, we investigated RGS1 expression in GC tissues and its potential as a prognostic marker, laying the groundwork for future research. Our analysis of patient data from the TCGA data and our cohort of 375 surgically resected GC patients revealed that RGS1 was upregulated in GC tissues and had prognostic significance (TCGA: adjusted HR:1.49, 95%CI: 1.02-2.18; GC cohort: adjusted HR: 1.38, 95%CI: 1.02-1.85). GO function and KEGG enrichment analyses suggest that RGS1 is involved in macrophage-mediated immune responses in GC. We observed a positive correlation between RGS1 expression and M2 macrophage infiltration. Furthermore, co-occurrence of elevated RGS1 expression and M2 macrophage infiltration predicts a worse prognosis (adjusted HR: 1.73, 95%CI: 1.24-2.42 in our cohort). In vitro, RGS1 upregulation and the presence of M2 macrophages enhanced malignant phenotypes of GC cells. Additionally, we confirmed that RGS1 promoted macrophage recruitment and M2 polarization via upregulation of CCL4 expression in vivo. In conclusion, this study suggests that RGS1 could serve as a promising prognostic marker for GC and a potential target for immunotherapy. However, further investigation with more advanced experimental models is needed to confirm these preliminary findings.
Collapse
Affiliation(s)
- Yuzheng Zhang
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, China; Department of Hospital Infection Management, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zhifang Jia
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, China
| | - Donghui Cao
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, China
| | - Yanping Zhong
- Division of Pathology, First Hospital of Jilin University, Changchun, China
| | - Yanhua Wu
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, China
| | - Yingli Fu
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, China
| | - Yingnan Cui
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Xinyi Yu
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, China
| | - Yu Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, China
| | - Jing Jiang
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
7
|
Yan H, Wu X, Li H, Yu Z, Jin X. Pan-Cancer Analysis Identifies BCLAF1 as a Potential Biomarker for Renal Cell Carcinoma. Biochem Genet 2025; 63:1479-1508. [PMID: 38573525 DOI: 10.1007/s10528-024-10773-x] [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: 07/04/2023] [Accepted: 03/04/2024] [Indexed: 04/05/2024]
Abstract
B-cell lymphoma-2-associated transcription factor 1 (BCLAF1) is a versatile protein involved in the regulation of gene transcription and post-transcriptional processing. Although BCLAF1 exerts a broad tumor suppressor effect or tumor promoter effect in many cancer types, the specific roles concerning its expression levels, and its impact on tumorigenesis in Renal cell carcinoma (RCC) remain unclear. Here, we utilized the Cancer Genome Atlas (TCGA) and Genotype Tissue Expression (GTEx) datasets alongside R software and online tools to unravel the specific roles of BCLAF1 in 33 cancer types, including its expression levels, tumor immune and molecular subtypes, and its correlation with prognosis, diagnosis, DNA methylation, and immune microenvironment. Additionally, we carried out cell biology experiments to independently investigate the expression of BCLAF1 in RCC and its effects on tumor progression. BCLAF1 was differentially expressed in tumor tissues compared to normal tissues across various cancer types and was also differentially expressed in different immune and molecular subtypes. In RCC, patients with high BCLAF1 expression had a better prognosis and BCLAF1 was tightly correlated with the stage, gender, and histological grade of patients. Furthermore, BCLAF1 had higher DNA methylation levels and higher immune infiltration levels in tumor tissues. Additionally, cell functional experiments confirmed the low expression of BCLAF1 in RCC and that BCLAF1 significantly inhibited the proliferation, migration, and invasion, while inducing apoptosis and cell cycle arrest in RCC cells in vitro. Our study under-scored the potential of BCLAF1 as an important actor in tumorigenesis, especially concerning RCC where it may serve as an effective prognostic marker.
Collapse
Affiliation(s)
- Huan Yan
- Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xiang Wu
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Hong Li
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Zongdong Yu
- Department of Neurosurgery, Shangrao People's Hospital, Shangrao, 334099, China.
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China.
| | - Xiaofeng Jin
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
8
|
Zhang W, Xie Y, Chen F, Xie B, Yin Z. Development and validation of a neutrophil extracellular traps-related gene signature for lower-grade gliomas. Comput Biol Med 2025; 188:109844. [PMID: 39978096 DOI: 10.1016/j.compbiomed.2025.109844] [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: 07/01/2024] [Revised: 02/09/2025] [Accepted: 02/10/2025] [Indexed: 02/22/2025]
Abstract
There is growing evidence linking neutrophil extracellular traps (NETs) to tumor genesis, growth, distant metastasis, and tumor-related thrombosis. However, the roles of NETs-related genes (NETRGs) on LGG prognosis remain unclear. The purpose of this study was to integrate multiple machine learning techniques and experiment validation to develop a reliable NETs-based signature that opens up novel approaches for assessing the prognosis and treatment response of LGG patients. Consensus clustering, k-means clustering and Nonnegative Matrix Factorization was used for the TCGA-LGG dataset and identified two NETs-related subgroups. The prognostic hallmark and nomogram for LGG were developed, which consist of five differentially expressed NETRGs (FPR1, PTAFR, SLC11A1, ICAM1, LTF) based on nine analytic approaches. The ROC curves and calibration curves of our NETRGs signature and nomogram exhibited strong and robust prognosis prediction abilities in both the TCGA-LGG training set and CGGA-325, CGGA-693 validation sets. The prognosis for LGG individuals in the low-risk category was better. The TISCH was used to examine the five NETRGs at the single-cell level. Common immunological checkpoints were expressed at greater levels in high-risk individuals. LGG individuals in the low-risk category posses a higher likelihood of being sensitive to Carmustine and Vincristine, as indicated by the drug sensitivity analysis. The qRT-PCR experiment and immunohistochemistry images confirmed that the expression of FPR1, PTAFR, SLC11A1 and ICAM1 are higher in low-grade oligodendroglioma. The NETRGs signature and nomogram can accurately and conveniently predict the LGG patients' prognosis, which can facilitate individualized treatment and the improvement of prognosis.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning Province, China
| | - Youlong Xie
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing, 400016, China
| | - Fengming Chen
- Hunan Provincial Key Laboratory of the Traditional Chinese Medicine Agricultural Biogenomics, Changsha Medical University, 410129, China
| | - Biao Xie
- Department of Health Statistics, School of Public Health, Chongqing Medical University, Chongqing, 400016, China
| | - Zhihua Yin
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, 110122, Liaoning Province, China.
| |
Collapse
|
9
|
Lin L, Li Z, Chen K, Shao Y, Li X. Uncovering somatic genetic drivers in prostate cancer through comprehensive genome-wide analysis. GeroScience 2025:10.1007/s11357-025-01623-8. [PMID: 40156736 DOI: 10.1007/s11357-025-01623-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 03/16/2025] [Indexed: 04/01/2025] Open
Abstract
Given that hereditary prostate cancer (PCa) accounts for only a small fraction of PCa phenotypes, there is still a substantial journey ahead in exploring the somatic genetic drivers contributing to sporadic PCa. The expression quantitative trait loci (eQTLs) data were sourced from the GTEx dataset for prostate-specific genes, and the summary statistic information was collected for 5854 genes. Genetic associations with PCa were extracted from three well-established consortiums: the UK Biobank (9131 cases and 173,493 controls), the PRACTICAL study (79,148 cases and 61,106 controls), and the FinnGen cohort (13,216 cases and 119,948 controls). To prioritize potential causal targets, additional analysis, including the protein-protein interaction (PPI), The Cancer Genome Atlas (TCGA) dataset, and the single-cell-type expression analysis, was performed. Generally, a total of 150 common significant genes with the same causal association with PCa were identified. Out of the 150 genes examined, 67.33% (101/150) were found to have protein-coding functions, while only 30.67% (46/150) of these genes had prior mentions in the scientific literature. Notably, the analysis of the TCGA dataset showed that only 44.67% (67/150) of the genes produced consistent results with the Mendelian randomization (MR) analysis. Furthermore, the evaluation of single-cell RNA-seq data and colocalization analysis singled out MSMB as a critical gene associated with the occurrence of PCa. We pinpointed a range of prostate-specific genes that display causal associations with the onset of PCa. Among these, the MSMB gene emerged as a pivotal factor linked to PCa, demonstrating robust consistency across all four assessments, including the MR, TCGA dataset, single-cell RNA-seq data, and colocalization analysis. These findings provided fresh perspectives on the pathogenesis of PCa and presented potential targets for drug development.
Collapse
Affiliation(s)
- Lede Lin
- Department of Urology and Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhen Li
- Department of Urology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Kai Chen
- Department of Urology, The First Hospital of Jiaxing, The Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Yanxiang Shao
- Department of Urology and Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Xiang Li
- Department of Urology and Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
10
|
Lv H, Lv M, Guo X, Zhu X, Chao Y, Li D. Lipopolysaccharide-binding protein (LBP): a prognostic biomarker for gastric cancer linked to immune infiltration. BMC Gastroenterol 2025; 25:205. [PMID: 40155834 PMCID: PMC11951659 DOI: 10.1186/s12876-025-03794-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/17/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Gastric cancer, characterized by rising global incidence and mortality, faces significant challenges due to the lack of effective early detection methods, delaying timely interventions and underscoring the need for novel biomarkers. Lipopolysaccharide-binding protein (LBP), implicated in cancers such as lung, colon, and cervical cancer, has emerged as a promising candidate. However, its specific roles and mechanisms in gastric cancer remain unclear, necessitating further investigation. METHODS This study utilized data from The Cancer Genome Atlas (TCGA), the Gene Expression Omnibus (GEO), and the Human Protein Atlas (HPA) to assess LBP mRNA and protein expression levels in gastric cancer patients and explore their associations with clinical outcomes. Analytical techniques included volcano plots, protein-protein interaction networks, Gene Ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, gene set enrichment analysis (GSEA), and immune infiltration assessments. Furthermore, lentiviral vectors containing interference sequences targeting LBP were used to manipulate its expression in AGS and HGC-27 gastric cancer cell lines, enabling the analysis of gene knockdown effects on malignant behaviors. Western blotting (WB) was performed to validate the impact of LBP knockdown on the expression of key signaling pathway proteins. RESULTS Our pan-cancer comparative analysis across 33 cancer types revealed significant upregulation of LBP in gastric cancer, with diagnostic ROC curve analysis yielding an AUC of 0.765. Univariate and multivariate Cox regression analyses revealed that high LBP expression was inversely related to patient survival. Additionally, immune infiltration and functional enrichment analyses revealed the involvement of LBP in pathways crucial to cancer development, such as immune response modulation and lipid metabolism. LBP knockdown in gastric cancer cell lines reduced proliferation, migration, and invasion. WB confirmed decreased expression of P65, P-P65, STAT3, and P-STAT3 upon LBP knockdown. CONCLUSION LBP is intricately linked to gastric cancer pathogenesis; it influences cell proliferation, migration, and invasion, thereby representing a valuable prognostic and diagnostic biomarker. This study not only highlights the potential of LBP as a therapeutic target but also provides the groundwork for future investigations into its mechanistic pathways in gastric cancer. CLINICAL TRIAL NUMBER Not applicable. I would like to clarify that our research does not fall under clinical studies and therefore does not involve ethical concerns related to human or animal subjects. The cells used in this study are established cell lines purchased from a certified biotechnology company. All experimental procedures comply with standard research protocols and guidelines for cell line studies.
Collapse
Affiliation(s)
- Hao Lv
- Department of Gastroenterology Medicine (Endoscopy Center), China-Japan, Union Hospital of Jilin University, Changchun, Jilin, 130033, PR China
- Department of Gastroenterology, University of Health and Rehabilitation Sciences Affiliated, Qingdao Municipal Hospital Jiaozhou Road 1#, Qingdao, 266071, PR China
| | - Mei Lv
- Department of Gastroenterology, University of Health and Rehabilitation Sciences Affiliated, Qingdao Municipal Hospital Jiaozhou Road 1#, Qingdao, 266071, PR China
| | - Xuyang Guo
- Department of Gastroenterology Medicine (Endoscopy Center), China-Japan, Union Hospital of Jilin University, Changchun, Jilin, 130033, PR China
| | - Xiaoman Zhu
- Department of Gastroenterology Medicine (Endoscopy Center), China-Japan, Union Hospital of Jilin University, Changchun, Jilin, 130033, PR China
| | - Yue Chao
- Department of Gastroenterology Medicine (Endoscopy Center), China-Japan, Union Hospital of Jilin University, Changchun, Jilin, 130033, PR China
| | - Dandan Li
- Department of Gastroenterology Medicine (Endoscopy Center), China-Japan, Union Hospital of Jilin University, Changchun, Jilin, 130033, PR China.
| |
Collapse
|
11
|
Zhang H, Li H, Qi Y, He J, Deng L, Chen S, Pan H, Guo H. IL17A/F secreted by ASCT2-overexpression ovarian cancer cells contributes to immune escape through the suppression of natural killer (NK) cells cytotoxicity by the activation of c-JUN/ PTGS2 pathway. Int Immunopharmacol 2025; 150:114226. [PMID: 39954656 DOI: 10.1016/j.intimp.2025.114226] [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: 10/30/2024] [Revised: 01/23/2025] [Accepted: 02/02/2025] [Indexed: 02/17/2025]
Abstract
Ovarian cancer (OC) is a deadly gynecologic cancer associated with metastasis, recurrence, and treatment resistance. The expression of the alanine-serine-cysteine transporter 2 (ASCT2) has been linked to poor prognosis and immune cell infiltration in OC tumors, but the underlying mechanisms are unclear. Lentiviral constructs were used to manipulate ASCT2 expression in OC cells (SKOV-3). The effects of ASCT2 on SKOV-3 behaviors including proliferation, invasion, migration, apoptosis, and cell cycle were assessed using various assays. The correlation between ASCT2 expression and immune infiltration in OC was analyzed using the Cancer Genome Atlas (TCGA) database. Co-culture experiments were conducted to evaluate the impact of ASCT2 overexpression in SKOV-3 on NK cells, followed by transcriptomics and cytokine analysis. ASCT2 expression and cytokine levels were characterized using qPCR and western blotting. ASCT2 overexpression significantly promoted cell proliferation, invasion, migration, and the percentage of G1-phase cells, while inhibiting apoptosis. ASCT2 silencing had the opposite effect. The expression of ASCT2 was negatively associated with NK cells in OC. ASCT2 overexpression in SKOV-3 cells led to excessive IL-17A/F production and inhibited the antitumor activity of NK cells, possibly through activating the IL-17 signaling pathway. The core regulatory genes c-JUN/PTGS2 in this pathway were upregulated, and antitumor cytokines were decreased in co-cultured NK cells, resulting in decreased antitumor activity and immune infiltration within the tumor. Our results suggest that overexpression of ASCT2 may play a predominant role in OC and NK cell immune infiltration within the tumor.
Collapse
Affiliation(s)
- Huixiang Zhang
- Department of Gynaecology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China; Library, Kunming Medical University, No.1168 West Chunrong Road, Chenggong District, Kunming, Yunnan 650500, China
| | - Haohan Li
- Department of Gynaecology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yanyan Qi
- Department of Gynaecology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Junhan He
- Department of Gynaecology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Langping Deng
- Department of Gynaecology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Shipu Chen
- Department of Gynaecology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Hong Pan
- Department of Gynaecology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Huiming Guo
- Department of Gynaecology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.
| |
Collapse
|
12
|
Vasquez YA, Beale HC, Sanders L, Lyle AG, Kephart ET, Learned K, Thompson D, Peralez J, Li A, Huang M, Pyke-Grimm KA, Salama SR, Haussler D, Bjork I, Sheri LS, Vaske OM. Comparative analysis of RNA expression in a single institution cohort of pediatric cancer patients. NPJ Precis Oncol 2025; 9:81. [PMID: 40119139 PMCID: PMC11928651 DOI: 10.1038/s41698-025-00852-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 02/26/2025] [Indexed: 03/24/2025] Open
Abstract
With the low incidence of mutations in pediatric cancers, alternate genomic approaches are needed to identify therapeutic targets. Our study, the Comparative Analysis of RNA Expression to Improve Pediatric and Young Adult Cancer Treatment, was conducted by the UC Santa Cruz Treehouse Childhood Cancer Initiative and Stanford University School of Medicine. RNA sequencing data from 33 children and young adults with a relapsed, refractory or rare cancer underwent CARE analysis to reveal activated cancer driver pathways and nominate treatments. We compare our pipeline to other gene expression outlier detection approaches and discuss challenges for clinical implementation. Of our 33 patients, 31 (94%) had findings of potential clinical significance. Findings were implemented in 5 patients, 3 of which had defined clinical benefit. We demonstrate that comparator cohort composition determines which outliers are detected. This study highlights the clinical utility and challenges of implementing comparative RNA sequencing analysis in the clinic.
Collapse
Affiliation(s)
- Yvonne A Vasquez
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA, USA
- UC Santa Cruz Genomics Institute, Santa Cruz, CA, USA
| | - Holly C Beale
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA, USA
- UC Santa Cruz Genomics Institute, Santa Cruz, CA, USA
| | - Lauren Sanders
- UC Santa Cruz Genomics Institute, Santa Cruz, CA, USA
- Department of Biomolecular Engineering, School of Engineering, University of California, Santa Cruz, CA, USA
- Blue Marble Space Institute of Science, NASA Ames GeneLab, Silicon Valley, CA, USA
| | - A Geoffrey Lyle
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA, USA
- UC Santa Cruz Genomics Institute, Santa Cruz, CA, USA
| | | | | | - Drew Thompson
- Department of Biomolecular Engineering, School of Engineering, University of California, Santa Cruz, CA, USA
| | | | - Amy Li
- Stanford University School of Medicine, Stanford, CA, USA
| | - Min Huang
- Stanford University School of Medicine, Stanford, CA, USA
| | - Kimberly A Pyke-Grimm
- Stanford University School of Medicine, Stanford, CA, USA
- Department of Nursing Research and Evidence-Based Practice at Stanford Medicine Children's Health, Stanford, USA
| | - Sofie R Salama
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA, USA
- UC Santa Cruz Genomics Institute, Santa Cruz, CA, USA
| | - David Haussler
- UC Santa Cruz Genomics Institute, Santa Cruz, CA, USA
- Department of Biomolecular Engineering, School of Engineering, University of California, Santa Cruz, CA, USA
| | - Isabel Bjork
- UC Santa Cruz Genomics Institute, Santa Cruz, CA, USA
- Foundation to Advance Vascular Cures, Redwood City, CA, USA
| | - L Spunt Sheri
- Stanford University School of Medicine, Stanford, CA, USA
| | - Olena M Vaske
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, CA, USA.
- UC Santa Cruz Genomics Institute, Santa Cruz, CA, USA.
| |
Collapse
|
13
|
Zhong F, Mao S, Peng S, Li J, Xie Y, Xia Z, Chen C, Sun A, Zhang S, Wang S. Exploration of SUSD3 in pan-cancer: studying its role, predictive analysis, and biological significance in various malignant tumors in humans. Front Immunol 2025; 16:1521965. [PMID: 40191190 PMCID: PMC11968365 DOI: 10.3389/fimmu.2025.1521965] [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: 11/03/2024] [Accepted: 02/17/2025] [Indexed: 04/09/2025] Open
Abstract
Background The SUSD3 protein, marked by the Sushi domain, plays a key role in cancer progression, with its expression linked to tumor advancement and patient prognosis. Altered SUSD3 levels could serve as a predictive biomarker for cancer progression. Recognized as a novel susceptibility marker, SUSD3 presents a promising target for antibody-based therapies, offering a potential approach for the prevention, diagnosis, and treatment of breast cancer. Methods Using the HPA and GeneMANIA platforms, the distribution of SUSD3 protein across tissues was analyzed, while expression levels in tumor and healthy tissues were compared using The Cancer Genome Atlas data. The TISCH and STOmics DB databases facilitated the mapping of SUSD3 expression in different cell types and its spatial relationship with cancer markers. Univariate Cox regression assessed the prognostic significance of SUSD3 expression in various cancers. Genomic alterations of SUSD3 were explored through the cBioPortal database. The potential of SUSD3 as a predictor of immunotherapy response was investigated using TIMER2.0, and GSEA/GSVA identified related biological pathways. Drugs targeting SUSD3 were identified through CellMiner, CTRP, and GDSC databases, complemented by molecular docking studies. In vitro experiments demonstrated that SUSD3 knockdown in breast cancer cell lines significantly reduced proliferation and migration. Results SUSD3 expression variations in pan-cancer cohorts are closely linked to the prognosis of various malignancies. In the tumor microenvironment (TME), SUSD3 is predominantly expressed in monocytes/macrophages and CD4+ T cells. Research indicates a strong correlation between SUSD3 expression and key cancer immunotherapy biomarkers, immune cell infiltration, and immunomodulatory factors. To explore its immune regulatory role, StromalScore, ImmuneScore, ESTIMATE, and Immune Infiltration metrics were employed. Molecular docking studies revealed that selumetinib inhibits tumor cell proliferation. Finally, SUSD3 knockdown reduced cancer cell proliferation and migration. Conclusion These findings provide valuable insights and establish a foundation for further exploration of SUSD3's role in pan-carcinomas. Additionally, they offer novel perspectives and potential targets for the development of innovative therapeutic strategies in cancer treatment.
Collapse
Affiliation(s)
- Fei Zhong
- Department of Laboratory Medicine, The Affiliated Huai’an Hospital of Xuzhou Medical University, The Second People’s Hospital of Huai’an, Huai’an, Jiangsu, China
| | - Shining Mao
- Faculty of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian, Jiangsu, China
| | - Shuangfu Peng
- Department of Laboratory Medicine, The Affiliated Huai’an Hospital of Xuzhou Medical University, The Second People’s Hospital of Huai’an, Huai’an, Jiangsu, China
| | - Jiaqi Li
- Faculty of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian, Jiangsu, China
| | - YanTeng Xie
- Faculty of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian, Jiangsu, China
| | - Ziqian Xia
- Faculty of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian, Jiangsu, China
| | - Chao Chen
- Faculty of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian, Jiangsu, China
| | - Aijun Sun
- Department of Laboratory Medicine, The Affiliated Huai’an Hospital of Xuzhou Medical University, The Second People’s Hospital of Huai’an, Huai’an, Jiangsu, China
| | - Shasha Zhang
- Key Laboratory of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shiyan Wang
- Faculty of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian, Jiangsu, China
| |
Collapse
|
14
|
Chen F, Xiang W, Qiang G. Tanshinone IIA affects the proliferation of A549/Tax by affecting the expression of MMP7 through the PI3K-AKT-mTOR signaling pathway. Discov Oncol 2025; 16:369. [PMID: 40113621 PMCID: PMC11926296 DOI: 10.1007/s12672-025-02152-8] [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: 12/10/2024] [Accepted: 03/14/2025] [Indexed: 03/22/2025] Open
Abstract
OBJECTIVE This study aims to explore whether tanshinone IIA can act on paclitaxel-resistant non-small cell lung cancer A549/Tax and analyze the possible mechanisms involved. METHODS Using the Cell Counting Kit-8 (CCK-8), we preliminarily analyzed whether tanshinone IIA has an inhibitory effect on A549/Tax cells. We utilized public datasets, self-collected transcriptome datasets, and drug target analysis to identify potential targets. We employed real-time fluorescent quantitative polymerase chain reaction (RT-qPCR) to detect the expression of core genes before and after drug treatment to analyze potential target genes and validated them using data from The Cancer Genome Atlas (TCGA). We conducted enrichment analysis on co-expressed genes of the target genes to explore potential mechanisms. Furthermore, we employed molecular docking and western blot to verify the possible mechanisms involved. RESULTS The CCK8 results indicated that tanshinone IIA has a significant inhibitory effect on A549/Tax cells. The qPCR results and the analysis of TCGA data indicated that MMP7 is the target gene. Enrichment results of MMP7 co-expressed genes suggested that the PI3K-AKT signaling pathway might play a key role. Molecular docking results indicated that tanshinone IIA has strong binding activity with PI3K, AKT, mTOR, and MMP7. Western blotting results showed that tanshinone IIA might inhibit MMP7 through the PI3K-AKT-mTOR signaling pathway. CONCLUSIONS Tanshinone IIA may affect the proliferation of A549/Tax by influencing the expression of MMP7 through the PI3K-AKT-mTOR signaling pathway.
Collapse
Affiliation(s)
- Fangjun Chen
- Department of Thoracic Surgery, China Japan Friendship Institute of Clinical Medicine Research, Beijing, China
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Wenqiong Xiang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guangliang Qiang
- Department of Thoracic Surgery, Peking University Third Hospital, No.49 Huayuan North Road, Haidian District, Beijing, 100191, China.
| |
Collapse
|
15
|
Du S, Wang T, Li Z, Li T, Miao Z, Chen Y, Zhu S, Wei W, Deng H. Therapeutic Potential of Qilianxiaopi Formula: Targeting ADAM17-Mediated Chronic Inflammation in Atrophic Gastritis. Pharmaceuticals (Basel) 2025; 18:435. [PMID: 40143211 PMCID: PMC11944831 DOI: 10.3390/ph18030435] [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: 02/13/2025] [Revised: 03/09/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
Background: Gastric cancer (GC) is a leading cause of mortality worldwide, particularly in China. Chronic atrophic gastritis (CAG) and intestinal metaplasia (IM) are recognized as precancerous conditions contributing to GC development. Qilianxiaopi formula (QLXP), a traditional Chinese medicine (TCM), has demonstrated significant therapeutic effects on CAG and IM; however, its underlying mechanisms remain poorly understood. Methods: This study utilized chromatography-mass spectrometry to identify the major compounds in QLXP. Network pharmacology was used to predict the associated targets of these components. Thermal proteome profiling (TPP) pinpointed the potential binding proteins of QLXP, which were validated by bioinformatic analyses. Bio-layer interferometry (BLI) was used to analyze the interactions between QLXP and its key target proteins, thereby determining their binding components. Molecular docking predicted the binding modes between the components and proteins. Results: ADAM17 was identified as a key binding protein for QLXP. Further investigation revealed that QLXP inhibits the enzymatic activity of ADAM17, thereby reducing the secretion of the pro-inflammatory cytokine TNF-α, contributing to the anti-inflammatory properties of QLXP. BLI confirmed direct and reversible binding interactions between QLXP and ADAM17. Narirutin, isolated from the ADAM17 binding fraction, displayed the highest affinity for QLXP. Conclusions: This study highlights ADAM17 as a key molecular target of QLXP and narirutin as its principal binding component. The integrated approach combining chromatography-mass spectrometry, network pharmacology, TPP, BLI, and molecular docking provides a robust framework for elucidating the mechanisms of action of TCM.
Collapse
Affiliation(s)
- Sijing Du
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; (S.D.)
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Tianxiang Wang
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; (S.D.)
| | - Zhiqiang Li
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; (S.D.)
| | - Ting Li
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; (S.D.)
| | - Zelong Miao
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; (S.D.)
| | - Yuling Chen
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; (S.D.)
| | - Songbiao Zhu
- Chinese Institutes for Medical Research (CIMR), Beijing 100069, China
| | - Wei Wei
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Haiteng Deng
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; (S.D.)
| |
Collapse
|
16
|
Liang Z, Zhang T, Huang J, Huang Z, Zhao Z, Cai S, Ma J. A comprehensive prognostic and immunological analysis of hexokinase domain containing protein-1 (HKDC1) in pan-cancer. PeerJ 2025; 13:e19083. [PMID: 40124623 PMCID: PMC11929506 DOI: 10.7717/peerj.19083] [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: 10/24/2024] [Accepted: 02/10/2025] [Indexed: 03/25/2025] Open
Abstract
Background Currently, research on the role of hexokinase domain-containing protein-1 (HKDC1) in neoplasm metabolism remains sparse. This study seeks to conduct a thorough investigation of HKDC1's potential functions across thirty-three different tumor types, utilizing data obtained from The Cancer Genome Atlas (TCGA). Method We conducted a thorough data extraction from the TCGA database, subsequently employing R (version 4.2.2) and its associated software packages for detailed analysis. Our investigation centered on evaluating the differential expression and prognostic significance of HKDC1, while also examining its connections to tumor heterogeneity, mutation profiles, and RNA modifications. Furthermore, we analyzed the relationship between HKDC1 expression and tumor immunity utilizing the TIMER analysis approach. Results A comprehensive analysis of various tumor types has revealed that HKDC1 is significantly upregulated in many malignant tumors. Importantly, patients with elevated HKDC1 levels in their tumor tissues often experience poorer prognoses. The association between HKDC1 expression, immune cell infiltration, and the existence of immune checkpoints suggests a possible connection between the tumor microenvironment and HKDC1, alongside tumor advancement. Gene set enrichment analysis (GSEA) further substantiates the idea that HKDC1 may play a role in several critical pathways and biological processes associated with neoplasm. Additionally, the overexpression of HKDC1 is influenced by promoter methylation and alterations in DNA copy number amplification. Furthermore, in vitro experiments demonstrated that silencing HKDC1 resulted in a marked reduction in the proliferation, migration, and invasion capabilities of neoplasm cells. Conclusion Our initial pan-cancer analysis provided a comprehensive understanding of the oncogenic roles of HKDC1 across diverse cancer types. Moreover, HKDC1 has the potential to serve as a significant prognostic biomarker.
Collapse
Affiliation(s)
- Zhi Liang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Tianhao Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Jiajia Huang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Zhixin Huang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Zeyu Zhao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Shirong Cai
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Jinping Ma
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
17
|
Li Q, Wang J, Liu Q, Gan M, Yan J, Yu X, Shao Y. Downregulated STAT3 and STAT5B are prognostic biomarkers for colorectal cancer and are associated with immune infiltration. Discov Oncol 2025; 16:343. [PMID: 40100436 PMCID: PMC11920514 DOI: 10.1007/s12672-025-02085-2] [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: 09/11/2024] [Accepted: 03/06/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Colorectal cancer has high incidence and mortality rates. The signal transducer and activator of transcription (STAT) family plays vital roles in the tumorigenesis and development of colorectal cancer. The expression, prognostic value, and immune function of the STAT family are becoming much more clearly. METHODS Our study collected data from several public data portals such as TCGA (644 samples) and GTEx database (308 samples) and clinical samples (30 samples, China). Then we systematically assessed the expression level and prognostic value of the STAT family in colorectal cancer samples. Moreover, the immune function and immune infiltration levels of prognosis-related STAT members were explored via single cell RNA-seq and spatial transcriptomics technology data. Several useful portals and tools have been utilized such as CancerSEA and TISIDB in single-cell analysis, CBio Cancer Genomics in multidimensional alterations, MethSurv in DNA methylation, and related R packages. RESULTS Our study found that STAT3 and STAT5B were significantly lower in colorectal cancer via multi-omics (P < 0.001). Higher STAT3 and STAT5B level were correlated with better future outcome. Nomograms were developed to predict the distal survival time (C-index = 0.724). The functions of STAT3 and STAT5B are associated with inflammation, the JAK/STAT pathway and the immune response. The major cell types of colorectal cancer were CD4Tconv, CD8T, CD8Tex, Tprolif, Treg and STAT3 and STAT5B widely expressed in these cells. STAT3 and STAT5B both correlated with CD244 and KDR for immune checkpoints. CONCLUSION STAT3 and STAT5B are downregulated in colorectal cancer and have great potential as prognostic biomarkers and novel immunotherapy targets.
Collapse
Affiliation(s)
- Qier Li
- Department of Gastroenterology, the First Affiliated Hospital of Ningbo University, Ningbo, 315020, Zhejiang, China
| | - Jingzhi Wang
- Yancheng First Hospital, Yancheng, Jiangsu, China
| | - Qingqing Liu
- Department of Gastroenterology, the First Affiliated Hospital of Ningbo University, Ningbo, 315020, Zhejiang, China
| | - Min Gan
- Department of Gastroenterology, the First Affiliated Hospital of Ningbo University, Ningbo, 315020, Zhejiang, China
| | - Jianing Yan
- Department of Gastroenterology, the First Affiliated Hospital of Ningbo University, Ningbo, 315020, Zhejiang, China
| | - Xuan Yu
- Department of Gastroenterology, the First Affiliated Hospital of Ningbo University, Ningbo, 315020, Zhejiang, China.
| | - Yongfu Shao
- Department of Gastroenterology, the First Affiliated Hospital of Ningbo University, Ningbo, 315020, Zhejiang, China.
| |
Collapse
|
18
|
Tian L, Wang Y, Guan J, Zhang L, Fan J. The Prognostic Value and Immunomodulatory Role of Spsb2, a Novel Immune Checkpoint Molecule, in Hepatocellular Carcinoma. Genes (Basel) 2025; 16:346. [PMID: 40149497 PMCID: PMC11941779 DOI: 10.3390/genes16030346] [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: 02/21/2025] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Liver cancer, specifically hepatocellular carcinoma (LIHC), ranks as the second most common cause of cancer-related fatalities globally. Moreover, the occurrence rate of LIHC is steadily increasing. A recently identified gene, SPSB2, has been implicated in cell signaling, impacting the development and progression of non-small cell lung cancer. Nevertheless, studies on the role of SPSB2 in the pathogenesis of LIHC are lacking. METHODS Using the TCGA, GTEx, and GEO databases, we obtained differentially expressed genes that affect the prognosis of patients with LIHC. We utilized the Kruskal-Wallis test, along with univariate and multivariate COX regression analyses, to determine the correlation between SPSB2 and patient clinical indicators. Potential biological functions of SPSB2 in LIHC were explored by enrichment analysis, ssGSEA, and Spearman correlation analysis. Finally, LIHC cell lines Huh7 and SMMC-7721 were used to validate the biological function of SPSB2. RESULTS The results showed LIHC patients with higher SPSB2 expression had a poorer prognosis, and SPSB2 expression was significantly correlated with LIHC patients' Histologic grade, Pathologic T stage, Prothrombin time, Pathologic stage, BMI, weight, adjacent hepatic tissue inflammation, AFP level, and OS event (p < 0.05). SPSB2 shows notable enrichment in pathways linked to tumorigenesis and the immune system. Moreover, its expression is strongly connected to immune cells and immune checkpoints. Knockdown of SPSB2 expression in Huh7 cells and SMMC-7721 cells inhibits SPSB2's biological functions, including proliferation, invasion, metastasis, and other phenotypes. CONCLUSIONS SPSB2 plays a crucial role in the development of LIHC. It is related to the immune response and unfavorable outcomes. SPSB2 may function as a clinical biomarker for prognosis.
Collapse
Affiliation(s)
- Lv Tian
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Yiming Wang
- School of Nursing, Jilin University, Changchun 130021, China
| | - Jiexin Guan
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Lu Zhang
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Jun Fan
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 611731, China
| |
Collapse
|
19
|
Cai F, Mao S, Peng S, Wang Z, Li W, Zhang R, Wang S, Sun A, Zhang S. A comprehensive pan-cancer examination of transcription factor MAFF: Oncogenic potential, prognostic relevance, and immune landscape dynamics. Int Immunopharmacol 2025; 149:114105. [PMID: 39923580 DOI: 10.1016/j.intimp.2025.114105] [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: 11/01/2024] [Revised: 01/05/2025] [Accepted: 01/14/2025] [Indexed: 02/11/2025]
Abstract
AIMS Previous studies indicate that MAF BZIP Transcription Factor F (MAFF) facilitates ectopic metastasis and tumor cell migration. While its role in neoplasm progression is recognized, a thorough pan-cancer analysis of MAFF's impact remains pending. MAIN METHODS MAFF expression across normal and tumor tissues was analyzed using transcriptomic data from Genomic Data Commons (GDC) and UCSC XENA, with protein details from Human Protein Atlas (HPA) and GeneMANIA. Tumor Immune Single-cell Hub (TISCH) and Spatial Transcriptomics Omics DataBase (STOmics DB) identified MAFF expression in the tumor microenvironment (TME). MAFF's prognostic significance and immune-related gene associations were evaluated through univariate Cox regression, TIMER2.0 immune cell infiltration analysis, and Spearman correlation. Critical pathways were identified using Gene Set Enrichment Analysis (GSEA) and Gene Set Variation Analysis (GSVA), while molecular docking explored anticancer agent interactions. KEY FINDINGS MAFF expression varies across cancers, affecting tumor prognosis, notably in monocytes/macrophages and endothelial cells. Copy number variation (CNV) positively correlates with MAFF expression, while methylation shows inverse correlation. MAFF mutations significantly affect LGG patient prognosis and correlate with immune therapy responses. ESTIMATE and immune profiling linked MAFF to immunosuppression pathways. Molecular docking identified MAFF-targeted drugs, with validated effects on breast cancer and endometrial cancer cell survival and migration in vitro. SIGNIFICANCE Multi-omics analysis identified MAFF as a potential prognostic marker correlating with tumor immunity and microenvironment, suggesting its value for personalized cancer immunotherapy, particularly in BRCA and UCEC.
Collapse
Affiliation(s)
- Fengze Cai
- Faculty of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian, Jiangsu, China
| | - Shining Mao
- School of Chemical Engineering, Huaiyin Institute of Technology, Huaian, Jiangsu, China
| | - Shuangfu Peng
- Department of Thyroid and Breast Oncological Surgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, The Second People's Hospital of Huai'an, Huaian, Jiangsu, China
| | - Zirui Wang
- Faculty of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian, Jiangsu, China
| | - Wen Li
- Faculty of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian, Jiangsu, China
| | - Ruixuan Zhang
- Faculty of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian, Jiangsu, China
| | - Shiyan Wang
- Faculty of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian, Jiangsu, China.
| | - Aijun Sun
- Department of Thyroid and Breast Oncological Surgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, The Second People's Hospital of Huai'an, Huaian, Jiangsu, China.
| | - Shasha Zhang
- Key Laboratory of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
20
|
Creux C, Zehraoui F, Radvanyi F, Tahi F. MMnc: multi-modal interpretable representation for non-coding RNA classification and class annotation. Bioinformatics 2025; 41:btaf051. [PMID: 39891346 PMCID: PMC11890286 DOI: 10.1093/bioinformatics/btaf051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/16/2025] [Accepted: 01/29/2025] [Indexed: 02/03/2025] Open
Abstract
MOTIVATION As the biological roles and disease implications of non-coding RNAs continue to emerge, the need to thoroughly characterize previously unexplored non-coding RNAs becomes increasingly urgent. These molecules hold potential as biomarkers and therapeutic targets. However, the vast and complex nature of non-coding RNAs data presents a challenge. We introduce MMnc, an interpretable deep-learning approach designed to classify non-coding RNAs into functional groups. MMnc leverages multiple data sources-such as the sequence, secondary structure, and expression-using attention-based multi-modal data integration. This ensures the learning of meaningful representations while accounting for missing sources in some samples. RESULTS Our findings demonstrate that MMnc achieves high classification accuracy across diverse non-coding RNA classes. The method's modular architecture allows for the consideration of multiple types of modalities, whereas other tools only consider one or two at most. MMnc is resilient to missing data, ensuring that all available information is effectively utilized. Importantly, the generated attention scores offer interpretable insights into the underlying patterns of the different non-coding RNA classes, potentially driving future non-coding RNA research and applications. AVAILABILITY AND IMPLEMENTATION Data and source code can be found at EvryRNA.ibisc.univ-evry.fr/EvryRNA/MMnc.
Collapse
Affiliation(s)
- Constance Creux
- Université Paris-Saclay, Univ Evry, IBISC, Evry-Courcouronnes 91020, France
- Molecular Oncology, PSL Research University, CNRS, UMR 144, Institut Curie, Paris 75248, France
| | - Farida Zehraoui
- Université Paris-Saclay, Univ Evry, IBISC, Evry-Courcouronnes 91020, France
| | - François Radvanyi
- Molecular Oncology, PSL Research University, CNRS, UMR 144, Institut Curie, Paris 75248, France
| | - Fariza Tahi
- Université Paris-Saclay, Univ Evry, IBISC, Evry-Courcouronnes 91020, France
| |
Collapse
|
21
|
Yu M, Liu J, Zhou W, Gu X, Yu S. MRI radiomics based on machine learning in high-grade gliomas as a promising tool for prediction of CD44 expression and overall survival. Sci Rep 2025; 15:7433. [PMID: 40032983 PMCID: PMC11876340 DOI: 10.1038/s41598-025-90128-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 02/11/2025] [Indexed: 03/05/2025] Open
Abstract
We aimed to predict CD44 expression and assess its prognostic significance in patients with high-grade gliomas (HGG) using non-invasive radiomics models based on machine learning. Enhanced magnetic resonance imaging, along with the corresponding gene expression and clinicopathological data, was downloaded from online database. Kaplan-Meier survival curves, univariate and multivariate COX analyses, and time-dependent receiver operating characteristic were used to assess the prognostic value of CD44. Following the screening of radiomic features using repeat least absolute shrinkage and selection operator, two radiomics models were constructed utilizing logistic regression and support vector machine for validation purposes. The results indicated that CD44 protein levels were higher in HGG compared to normal brain tissues, and CD44 expression emerged as an independent biomarker of diminished overall survival (OS) in patients with HGG. Moreover, two predictive models based on seven radiomic features were built to predict CD44 expression levels in HGG, achieving areas under the curves (AUC) of 0.809 and 0.806, respectively. Calibration and decision curve analysis validated the fitness of the models. Notably, patients with high radiomic scores presented worse OS (p < 0.001). In summary, our results indicated that the radiomics models effectively differentiate CD44 expression level and OS in patients with HGG.
Collapse
Affiliation(s)
- Mingjun Yu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
- Department of Medical Affairs, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Jinliang Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Wen Zhou
- Department of Pain Management, Dalian Municipal Central Hospital, Dalian, 116033, People's Republic of China
| | - Xiao Gu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.
| | - Shijia Yu
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.
| |
Collapse
|
22
|
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
|
23
|
Qiu Z, Guo C, Liu X, Gao S, Xiao W, Cheng H, Yin L. Exploring the relationship between MGAT2 and glioblastoma: A Mendelian Randomization and bioinformatics approach. Brain Res 2025; 1850:149449. [PMID: 39788365 DOI: 10.1016/j.brainres.2025.149449] [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: 10/22/2024] [Revised: 12/15/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
BACKGROUND Mannosyl-glycoprotein beta-1,2-N-acetylglucosaminyltransferase 2 (MGAT2) and tumors' relevant research was in full swing recently. Therefore, we employed Mendelian Randomization (MR) alongside bioinformatics to thoroughly investigate the possible relationship between MGAT2 and glioblastoma (GBM). METHODS We utilized the summary statistics of genome-wide association studies (GWAS) for MGAT2 (N = 35,559 from deCODE) and glioblastoma (N = 379,155 from FinnGen). MR was used to assess the causal relationship between MGAT2 and GBM. Bioinformatics was used for a more in-depth exploration of the relationship between MGAT2 and GBM. RESULTS MR analysis demonstrated a causal relationship, showing that elevated levels of MGAT2 are associated with an increased risk of GBM (OR = 2.59, 95 % CI: 1.13-5.91, p = 0.023). Further investigation revealed significant differences in MGAT2 expression across normal tissue, tumor tissue, and gliomas of different types. Additionally, we found that MGAT2 may influence GBM through immune-related pathways, particularly through the role of macrophages. Proteins associated with MGAT2 were also identified in the PPI network. CONCLUSION This study first validated the causal relationship between MGAT2 and glioblastoma, and used bioinformatics to explore the relationship from multiple perspectives. Additionally, we proposed hypotheses for further research to investigate the potential mechanisms underlying this connection.
Collapse
Affiliation(s)
- Zili Qiu
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China; Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou 221002, China
| | - Chengcheng Guo
- Department of Intensive Care Unit, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xuejiao Liu
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China; Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou 221002, China
| | - Shangfeng Gao
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China; Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou 221002, China
| | - Weihan Xiao
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China; Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou 221002, China
| | - Hai Cheng
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China; Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou 221002, China
| | - Luxin Yin
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China; Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou 221002, China.
| |
Collapse
|
24
|
Hu W, Ding X, Wu X, Xi X, Xu J, Dai S, Chen J, Hu S, Zhao Q, Chen F. A Comprehensive Analysis of Epoxide Hydrolase 2 (EPHX2) in Pan-Cancer. Cancer Rep (Hoboken) 2025; 8:e70188. [PMID: 40129060 PMCID: PMC11932960 DOI: 10.1002/cnr2.70188] [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: 11/21/2023] [Revised: 01/28/2025] [Accepted: 03/10/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND AND AIMS Epoxide hydrolase 2 (EPHX2) regulates lipid signaling across various metabolites by encoding soluble epoxide hydrolase. However, its mechanisms and implications in human malignancies remain unknown. This research aimed to detail the prognostic landscape of EPHX2 in pan-cancer and explore its potential relationship with immune infiltration in the tumor microenvironment. METHODS Herein, multiple bioinformatics tools were used to comprehensively evaluate the expression, diagnostic, and prognostic significance of EPHX2 and its roles in the tumor immune microenvironment in human cancers. The underlying EPHX2-associated signaling pathways in cancers were investigated by gene set variation analysis (GSVA). TIDE, GDSC, and CTRP databases were applied to predict the response of EPHX2 to immunotherapy and sensitivity to small molecule drugs. Furthermore, EPHX2 expression was also validated by qPCR experiments in various cancer cell lines. RESULTS Overall results revealed significant down-regulation of EPHX2 mRNA expression in most tumors. Despite its high predictive significance across cancers, EPHX2 played a protective or detrimental effect in distinct types of cancers. EPHX2 proved to be a valuable diagnostic biomarker in a range of tumor types, particularly in kidney renal clear cell carcinoma, cervical squamous cell carcinoma, and endocervical adenocarcinoma. Genetic alterations of EPHX2 in 33 tumors were also investigated. EPHX2 expression was significantly linked to immune cell infiltrations (particularly tumor-associated macrophages), tumor mutation burden, microsatellite instability, immune modulators, and immunotherapeutic biomarkers. Single-cell sequencing and GSVA highlighted the relevance of EPHX2 in regulating various cancer-related biological processes, including cell cycle and apoptosis. In this view, targeting EPHX2-dependent signaling could be a promising therapeutic strategy for tumor immunotherapy. CONCLUSION EPHX2 may serve as a potential molecular biomarker for diagnosis and prognosis in pan-cancer and could become a novel therapeutic target for various cancers.
Collapse
Affiliation(s)
- Weiquan Hu
- Department of Joint SurgeryGanzhou People's HospitalGanzhouJiangxiChina
| | - Xiaoli Ding
- Department of Laboratory MedicineFirst Affiliated Hospital of Gannan Medical UniversityGanzhouJiangxiChina
| | - Xiangsheng Wu
- Department of Laboratory MedicineFirst Affiliated Hospital of Gannan Medical UniversityGanzhouJiangxiChina
| | - Xuxiang Xi
- Department of Laboratory MedicineFirst Affiliated Hospital of Gannan Medical UniversityGanzhouJiangxiChina
| | - Jing Xu
- Department of Orthopaedic SurgerySun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Shengyun Dai
- National Institutes for Food and Drug ControlBeijingChina
| | - Jing Chen
- Department of Laboratory MedicineFirst Affiliated Hospital of Gannan Medical UniversityGanzhouJiangxiChina
| | - Suping Hu
- Department of EmergencyFirst Affiliated Hospital of Gannan Medical UniversityGanzhouJiangxiChina
| | - Qinfei Zhao
- Department of Laboratory MedicineFirst Affiliated Hospital of Gannan Medical UniversityGanzhouJiangxiChina
| | - Fangfang Chen
- The First School of Clinical Medicine, Southern Medical UniversityGuangzhouGuangdongChina
- Jinling Hospital, Affiliated Hospital of Medical School, Nanjing UniversityNanjingJiangsuChina
| |
Collapse
|
25
|
Xu Y, Kang K, Coakley BA, Eisenstein S, Parveen A, Mai S, Wang YS, Zheng J, Boral D, Mai J, Pan W, Zhang L, Aaronson SA, Fang B, Divino C, Zhang B, Song WM, Hung MC, Pan PY, Chen SH. Modulation of tumor inflammatory signaling and drug sensitivity by CMTM4. EMBO J 2025; 44:1866-1883. [PMID: 39948411 PMCID: PMC11914105 DOI: 10.1038/s44318-024-00330-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: 02/06/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 03/19/2025] Open
Abstract
Although inflammation has been widely associated with cancer development, how it affects the outcomes of immunotherapy and chemotherapy remains incompletely understood. Here, we show that CKLF-like MARVEL transmembrane domain-containing member 4 (CMTM4) is highly expressed in multiple human and murine cancer types including Lewis lung carcinoma, triple-negative mammary cancer and melanoma. In lung carcinoma, loss of CMTM4 significantly reduces tumor growth and impairs NF-κB, mTOR, and PI3K/Akt pathway activation. Furthermore, we demonstrate that CMTM4 can regulate epidermal growth factor (EGF) signaling post-translationally by promoting EGFR recycling and preventing its Rab-dependent degradation. Consequently, CMTM4 knockout sensitizes human lung tumor cells to EGFR inhibitors. In addition, CMTM4 knockout tumors stimulated with EGF show a decreased ability to produce inflammatory cytokines including granulocyte colony-stimulating factor (G-CSF), leading to decreased recruitment of polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) and therefore establishing a less suppressive tumor immune environment in both lung and mammary cancers. We also present evidence indicating that CMTM4-targeting siRNA-loaded liposomes reduce lung tumor growth in vivo and prolong animal survival. Knockout of CMTM4 enhances immune checkpoint blockade or chemotherapy to further reduce lung tumor growth. These data suggest that CMTM4 represents a novel target for the inhibition of tumor inflammation, and improvement of the immune response and tumor drug sensitivity.
Collapse
Affiliation(s)
- Yitian Xu
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Neal Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Kyeongah Kang
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Neal Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Brian A Coakley
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Samuel Eisenstein
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Arshiya Parveen
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Neal Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Sunny Mai
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Neal Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Yuan Shuo Wang
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Junjun Zheng
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Neal Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Debasish Boral
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Junhua Mai
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - William Pan
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Licheng Zhang
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Neal Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Stuart A Aaronson
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bingliang Fang
- Department of Thoracic and Cardiovascular Surgery, MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Celia Divino
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Won-Min Song
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
| | - Ping-Ying Pan
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Shu-Hsia Chen
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA.
- Neal Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, 77030, USA.
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Physiology, Biophysics, and Systems Biology, Weill Cornell Medical Science and Graduate School of Medical Sciences, New York, NY, 10065, USA.
- Graduate and professional school at Texas A&M University, 400 Bizzell St., College Station, TX, 77840, USA.
| |
Collapse
|
26
|
Xu X, Gong C, Wang Y, Yin Z, Wang X, Wu X, Fang Z, Wei S. FOXF1 promotes ovarian cancer metastasis by facilitating HMGA2-mediated USP30-dependent S100A6 deubiquitination. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167633. [PMID: 39694080 DOI: 10.1016/j.bbadis.2024.167633] [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: 02/29/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024]
Abstract
Ovarian cancer is the most common type of gynecological malignant tumor, with the highest mortality rate among female genital malignant tumors. In this study, we initially identified forkhead box F1 (FOXF1) as a potential prognostic biomarker of ovarian cancer through bioinformatics analysis. FOXF1 expression was higher in ovarian cancer tissue samples and served as an unfavorable prognostic factor. In vitro and in vivo experiments demonstrated that FOXF1 enhanced ovarian cancer cell migration and tumor dissemination. Chromatin immunoprecipitation-polymerase chain reaction and luciferase assays revealed that FOXF1 bound directly to the high-mobility group AT-hook 2 (HMGA2) promoter and significantly induced its transcriptional activity. Subsequent co-immunoprecipitation and mass spectrometry analyses demonstrated that HMGA2 stabilized S100 calcium-binding protein A6 (S100A6) protein through recruitment of the deubiquitinase, ubiquitin-specific peptidase 30 (USP30), thereby inhibiting S100A6 degradation. Rescue experiments further illustrated that FOXF1 induced ovarian cancer cell mobility in an HMGA2/S100A6-dependent manner. Additionally, FOXF1, HMGA2, USP30, and S100A6 were clinically relevant in patients with ovarian cancer. This is the first study to reveal the molecular mechanisms underlying FOXF1-mediated ovarian cancer metastasis and demonstrate that FOXF1 represents a potential therapeutic target in patients with metastatic ovarian cancer.
Collapse
Affiliation(s)
- Xi Xu
- Department of Pathology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.
| | - Chaoju Gong
- Central Laboratory, The Municipal Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China.
| | - Yunfeng Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Zhidong Yin
- Department of Pathology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.
| | - Xiaogang Wang
- Department of Pathology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.
| | - Xuebiao Wu
- Center for Molecular Pathology, Department of Pathophysiology, Gannan Medical University, Ganzhou 341000, China.
| | - Zejun Fang
- Central Laboratory, Sanmen People's Hospital, Sanmen 317100, China.
| | - Shumei Wei
- Department of Pathology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.
| |
Collapse
|
27
|
Zhang Y, Cui K, Qiang R, Wang L. FUT10 is related to the poor prognosis and immune infiltration in clear cell renal cell carcinoma. Transl Cancer Res 2025; 14:827-842. [PMID: 40104704 PMCID: PMC11912032 DOI: 10.21037/tcr-24-449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 11/21/2024] [Indexed: 03/20/2025]
Abstract
Background Clear cell renal cell carcinoma (ccRCC), is highly metastatic with unfavorable oncologic outcomes. The metastatic dissemination and underlying mechanisms of ccRCC remain insufficiently understood. The expression of fucosyltransferases (FUTs) has been explored in multiple cancer types, which affect survival of tumor cells and oncology progress. However, the role of fucosyltransferase 10 (FUT10), a member of the FUT family, is still unclear in ccRCC. We aimed to investigate the effects of FUT10 on the prognosis and immune infiltration of ccRCC via The Cancer Genome Atlas (TCGA) database. Methods The relationship between FUT10 expression and clinical-pathologic features was evaluated by Welch's t-test, Wilcoxon signed-rank test, Dunn's test, and logistic regression based on TCGA datasets. The FUT10 expression level was converted into a categorical variable by receiver operating characteristic (ROC) and the area under the curve (AUC). The factors associated with the prognosis were determined by Kaplan-Meier method. The function of FUT10 was identified by functional enrichment analysis, gene set enrichment analysis (GSEA), gene correlation analysis, and immune infiltration analysis. At last, we verified the FUT10 messenger RNA (mRNA) expression in ccRCC and adjacent kidney tissues by quantitative real-time polymerase chain reaction (qRT-PCR). Results Downregulated FUT10 expression in ccRCC was associated with the clinical stage (P<0.001), T stage (P<0.001), M stage (P<0.001), and overall survival (OS) event (P<0.001). The ROC curve suggested that FUT10 had a certain accuracy in the diagnostic ability in ccRCC (AUC =0.787). It was shown that patient survival was prolonged in the FUT10 high-expression group. Meanwhile, multivariate analysis displayed that FUT10 was an independent risk factor for ccRCC patients (P=0.003). Moreover, we uncovered that FUT10 was involved in the phenotype of the immune response, oxidative phosphorylation (OXPHOS), arachidonic acid (AA) metabolism, and primary immunodeficiency (PID) by function enrichment analysis and GSEA. In addition, in the high FUT10 expression group, natural killer (NK) CD56bright cells exhibited lower enrichment scores, and central memory T cells exhibited higher enrichment scores. Especially, ARL8B, a key factor in NK-mediated cytotoxicity, had a certain correlation with FUT10 (r=0.590, P<0.001). Compared to the normal kidney tissues, the FUT10 mRNA expression in the ccRCC was decreased (P=0.004). Conclusions FUT10 might be a promising immune therapy target and prognostic biomarker in ccRCC.
Collapse
Affiliation(s)
- Yuqi Zhang
- Center of Medical Genetics, Northwest Women's and Children's Hospital, Xi'an, China
| | - Ke Cui
- Center of Medical Genetics, Northwest Women's and Children's Hospital, Xi'an, China
| | - Rong Qiang
- Center of Medical Genetics, Northwest Women's and Children's Hospital, Xi'an, China
| | - Lin Wang
- Center of Medical Genetics, Northwest Women's and Children's Hospital, Xi'an, China
| |
Collapse
|
28
|
Zheng M, Huang W, Wang D, Huang L, Ren Y, Gao Q, Huang Y, Lin W, Chen L. Prognostic assessment of cervical cancer based on biomarkers: the interaction of ERRα and immune microenvironment. Virol J 2025; 22:47. [PMID: 39994715 PMCID: PMC11852515 DOI: 10.1186/s12985-025-02664-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Cervical cancer poses a substantial global health challenge. Estrogen-related receptor alpha (ERRα) is a central regulator of cellular energy metabolism associated with poor cancer prognosis. However, the effect of ERRα expression on cervical cancer prognosis and immune infiltration has not been explored. This study aims to clarify the expression pattern and role of ERRα in cervical cancer. METHODS We analyzed ERRα expression and its clinical prognosis in cervical cancer using multiple databases, including The Cancer Genome Atlas (TCGA) and Tumor Immune Estimation Resource (TIMER). The results were further validated through immunohistochemistry (IHC) on 221 cervical cancer tissue samples. Furthermore, Kaplan-Meier and Cox regression analyses were used to assess the clinical significance of ERRα in cervical cancer patients. All calculations were performed using the R package. RESULTS ERRα expression was significantly higher in cervical cancer tissues compared to normal tissues. High ERRα expression was associated with poor overall survival (OS), disease-specific survival (DSS), and progression-free survival (PFS). Multivariate Cox regression analysis confirmed ERRα as an independent prognostic factor. Additionally, ERRα expression correlated with various immune cell types and immune checkpoints, indicating its role in the tumor immune microenvironment. CONCLUSIONS ERRα emerges as a promising prognostic biomarker in cervical cancer, influencing immune cell infiltration and potentially guiding personalized therapeutic approaches. Future investigations are warranted to delineate the mechanistic pathways through which ERRα contributes to cervical cancer progression and to assess its viability as a target for innovative immunotherapy strategies.
Collapse
Affiliation(s)
- Meijin Zheng
- Laboratory of Gynecologic Oncology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Weifeng Huang
- Department of Radiation Oncology, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, 363000, China
| | - Dingjie Wang
- Laboratory of Gynecologic Oncology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital (Fujian Women and Children's Hospital), Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, 350001, Fujian, China
| | - Leyi Huang
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350001, Fujian, China
| | - Yuan Ren
- Laboratory of Gynecologic Oncology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital (Fujian Women and Children's Hospital), Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, 350001, Fujian, China
| | - Qiao Gao
- Laboratory of Gynecologic Oncology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Yuxuan Huang
- Laboratory of Gynecologic Oncology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital (Fujian Women and Children's Hospital), Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, 350001, Fujian, China
| | - Wenyu Lin
- Laboratory of Gynecologic Oncology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China.
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital (Fujian Women and Children's Hospital), Fuzhou, 350001, Fujian, China.
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, 350001, Fujian, China.
| | - Lihua Chen
- Laboratory of Gynecologic Oncology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China.
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital (Fujian Women and Children's Hospital), Fuzhou, 350001, Fujian, China.
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital (Fujian Obstetrics and Gynecology Hospital), Fuzhou, 350001, Fujian, China.
| |
Collapse
|
29
|
Lu J, Chen S, Hu W, Sai K, Li D, Jiang P. m6A regulator-based molecular classification and hub genes associated with immune infiltration characteristics and clinical outcomes in diffuse gliomas. BMC Med Genomics 2025; 18:37. [PMID: 39994800 PMCID: PMC11853526 DOI: 10.1186/s12920-025-02104-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 02/11/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND m6A methylation modification is a new regulatory mechanism involved in tumorigenesis and tumor-immunity interaction. However, its impact on glioma immune microenvironment and clinical outcomes remains unclear. METHODS Comprehensive expression profiles of 18 m6A regulators were used to identify molecular subtypes exhibiting distinct m6A modification patterns in 1673 glioma samples sourced from public datasets. A multi-genes signature was constructed for predicting clinical outcomes and response to immunotherapy in glioma patients. Immunohistochemistry and cellular experiments were performed for validation. RESULTS Two m6A subtypes of gliomas were identified. The m6A-low-risk subtype was characterized by paucity of immune infiltrates; While the m6A-high-risk subtype had higher abundances of multiple immune cells including lymphocyte and macrophage as well as increased expression of PD-L1, corresponding to an immunosuppressive phenotype. The m6A-high-risk subtype had poorer survival than the m6A-low-risk subtype in both the glioblastoma and lower grade gliomas cohorts. Eight m6A-related hub genes of high prognostic significances were identified and selected for developing a scoring signature termed as m6Ascore. Elevated m6Ascore indicated worse survival for glioma patients under standard care, but showed enhanced response to immunotherapy. Moreover, we demonstrated that overexpression of FTO, a m6A demethylase, inhibited the expressions of m6A-related hub genes (PTX3, SPAG4), impaired glioma cell viability and reduced macrophage chemotaxis. CONCLUSION This work develops an immune- and clinical-relevant m6A subtyping and a scoring model, which enhances our understanding of the role of m6A modification in regulating immune infiltration microenvironment in gliomas and helps to identify patients who are more likely to benefit from immunotherapy.
Collapse
Affiliation(s)
- Jie Lu
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, P. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Siyu Chen
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, P. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Wanming Hu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Ke Sai
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, P. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Depei Li
- Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, P. R. China.
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, P. R. China.
| | - Pingping Jiang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, 19 Nongling Road, Guangzhou, 510060, P. R. China.
| |
Collapse
|
30
|
Zhang X, Li N, Chu T, Zhao H, Liu T. Comprehensive pan-cancer analysis of ENOPH1 in human tumors. Discov Oncol 2025; 16:190. [PMID: 39955431 PMCID: PMC11829882 DOI: 10.1007/s12672-025-01965-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 02/10/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND ENOPH1 (Enolase-phosphatase 1), a member of the HAD-like hydrolase superfamily, has been linked to a range of physiological conditions, including neurological disorders. However, its involvement in tumorigenesis remains underexplored. This study is the first to conduct a pan-cancer analysis of ENOPH1, aiming to elucidate its role in multiple cancers through various bioinformatics platforms. METHODS We conducted a thorough analysis using data from UCSC databases. ENOPH1 expression in tumor and normal tissues was evaluated using R language software. Survival analyses, genetic alterations, and RNA modifications were assessed through the GEPIA2 and cBioPortal platforms. The relationships between ENOPH1 and immune infiltration, tumor mutational burden (TMB), microsatellite instability (MSI), and homologous recombination deficiency (HRD) were examined using TIMER2 and R software. ENOPH1-related gene enrichment analysis was performed using the STRING and GEPIA2 databases, followed by Gene Ontology (GO) and KEGG pathway enrichment analyses. RESULTS ENOPH1 expression was significantly upregulated in various cancers, including ACC, BLCA, BRCA, and COAD. High ENOPH1 expression was associated with poor overall survival (OS) in cancers such as KICH, LIHC, BRCA and LUAD. High ENOPH1 expression was associated with poor disease specific survival (DSS) in cancers such as KICH, LIHC, BRCA and MESO. Genetic alterations of ENOPH1, primarily mutations and deep deletions, were identified in UCEC, BLCA, and OV. ENOPH1 showed significant correlations with RNA modifications (m1A, m5C, m6A), immune checkpoints, and immune modulators across multiple cancer types. ENOPH1 was positively correlated with TMB, MSI, and HRD in cancers like BLCA, BRCA, and STAD. Furthermore, enrichment analysis revealed that ENOPH1 interacts with proteins involved in critical pathways such as AMPK, Hippo, and PI3K-AKT, suggesting its role in cancer progression. CONCLUSION This pan-cancer analysis reveals ENOPH1's potential as a prognostic biomarker and its involvement in key signaling pathways across multiple cancers. Our findings provide new insights into the role of ENOPH1 in tumorigenesis and highlight its potential as a therapeutic target in cancer treatment.
Collapse
Affiliation(s)
- Xuezhong Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Department of Laboratory Medicine, Zibo Central Hospital, Zibo, Shandong, China
- Shandong Jincheng Pharmaceutical Group Co., Ltd., Zibo, Shandong, China
| | - Ning Li
- Department of Pulmonary and Critical Care Medicine, Zibo Central Hospital, Zibo, Shandong, China
| | - Tingting Chu
- Department of Laboratory Medicine, Zibo Central Hospital, Zibo, Shandong, China
| | - Haijun Zhao
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Tonggang Liu
- Department of Infectious Diseases, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| |
Collapse
|
31
|
Shadman H, Gomrok S, Litle C, Cheng Q, Jiang Y, Huang X, Ziebarth JD, Wang Y. A machine learning-based investigation of integrin expression patterns in cancer and metastasis. Sci Rep 2025; 15:5270. [PMID: 39939698 PMCID: PMC11821851 DOI: 10.1038/s41598-025-89497-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: 10/09/2024] [Accepted: 02/05/2025] [Indexed: 02/14/2025] Open
Abstract
Integrins, a family of transmembrane receptor proteins, are well known to play important roles in cancer development and metastasis. However, a comprehensive understanding of these roles has not been achieved due to the complex relationships between specific integrins, cancer types, and the stages of cancer progression. Publicly accessible repositories from the Genotype-Tissue Expression (GTEx) and The Cancer Genome Atlas (TCGA) projects provide rich datasets for exploring these relationships using machine learning (ML). In this study, integrin RNA-Seq expression data of ~ 8 healthy tissues in GTEx and corresponding tumors in TCGA were selected. Integrin expression was used to train ML models to distinguish between different healthy tissues, solid tumors, as well as normal and tumor samples from the same tissue type. These ML models can classify samples by tissue origin or disease status with high accuracy, and the integrins essential to these classifiers were identified. In some cases, the expression of only one or two integrins was needed to classify tissue type, tumor type or disease status with accuracy > 0.9. For example, expression of ITGA7 alone can distinguish healthy and cancerous breast tissue. Additionally, integrin co-expression networks in healthy and cancerous breast tissues were compared and were found to change significantly from healthy to cancer, indicating changes in functional involvement of integrins due to cancer. Integrin expression in metastatic tumors were further examined using data from the AURORA project for Metastatic Breast Cancer (MBC), and several integrins such as ITGAD, ITGA4, ITGAL, and ITGA11 were found to have significantly lower expression in metastases than in primary tumors.
Collapse
Affiliation(s)
- Hossain Shadman
- Department of Chemistry, The University of Memphis, Memphis, TN, 38152, USA
| | - Saghar Gomrok
- Department of Chemistry, The University of Memphis, Memphis, TN, 38152, USA
| | - Christopher Litle
- Department of Chemistry, The University of Memphis, Memphis, TN, 38152, USA
| | - Qianyi Cheng
- Department of Chemistry, The University of Memphis, Memphis, TN, 38152, USA
| | - Yu Jiang
- School of Public Health, The University of Memphis, Memphis, TN, 38152, USA
| | - Xiaohua Huang
- Department of Chemistry, The University of Memphis, Memphis, TN, 38152, USA
| | - Jesse D Ziebarth
- Department of Chemistry, The University of Memphis, Memphis, TN, 38152, USA
| | - Yongmei Wang
- Department of Chemistry, The University of Memphis, Memphis, TN, 38152, USA.
| |
Collapse
|
32
|
Zhang X, Dong M, Zheng G, Sun M, Zhang C, Zhou Z, Tang S. MCM4 Promotes the Progression of Malignant Melanoma by Activating the PI3K/AKT Pathway. ENVIRONMENTAL TOXICOLOGY 2025; 40:306-317. [PMID: 39501995 DOI: 10.1002/tox.24433] [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: 07/21/2024] [Revised: 08/26/2024] [Accepted: 10/17/2024] [Indexed: 01/14/2025]
Abstract
This study aims to elucidate the role of minichromosome maintenance protein 4 (MCM4) in malignant melanoma (MM) and explore the underlying mechanism. Initially, data from The Cancer Genome Atlas (TCGA) database and the Molecular Signature Database (MSigDB) were used to investigate the biological impact of MCM4 on MM. Further, a prognostic model using Cox regression analysis was developed to predict the overall survival (OS) rate in the MM patients. The effects of MCM4 on the proliferation, migration, and invasion abilities of MM (B16F0 and A375) cells were demonstrated using the CCK-8, colony formation, EDU, wound scratch, and Transwell assays. In subcutaneous tumor models in C57BL/6 mice in vivo, the expression levels of MCM4 in MM cells and tumors were detected using Western blot and immunofluorescence approaches. The bioinformatics analysis indicated that MCM4 was expressed higher in MM tissues than in the normal tissues (p < 0.05). The established OS prediction model could significantly contribute to devising follow-up strategies and treating MM patients. MCM4 knockdown resulted in reduced proliferation, migration, and invasion abilities of MM cells, which were reversed by MCM4 overexpression (p < 0.05). Moreover, MCM4 could activate the phosphatidylinositol 3'-kinase (PI3K)/AKT pathway in MM cells. The PI3K inhibitor (LY294002) could reverse the effects of MCM4 on MM cells. MCM4 could substantially prompt the tumor growth of MM in mice through the PI3K/AKT pathway in vivo. In summary, MCM4 prompted the development and metastasis of MM by activating the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Xuewei Zhang
- Department of Plastic Surgery and Burn Center, The Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Mingming Dong
- Department of Orthopedic, The Affiliated Cancer Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Guoxing Zheng
- Department of Orthopedic, The Affiliated Cancer Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Meng Sun
- Department of Plastic Surgery and Burn Center, The Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Chuzhao Zhang
- Department of Plastic Surgery and Burn Center, The Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Zibin Zhou
- Department of Orthopedic, The Affiliated Cancer Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Shijie Tang
- Department of Plastic Surgery and Burn Center, The Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
33
|
Pei S, Jiang Z, Cheng H. Brain gliomas new transcriptomic discoveries from differentially expressed genes to therapeutic targets. Sci Rep 2025; 15:2553. [PMID: 39833228 PMCID: PMC11746978 DOI: 10.1038/s41598-025-86316-0] [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: 09/03/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
Gliomas are a prevalent form of primary malignant brain tumor, yet the intricate molecular mechanisms underlying its pathogenesis remain unclear. This study aimed to identify new genetic targets linked to glioma by analyzing microarray datasets to uncover genetic factors involved in its onset and progression. We obtained two independent glioma datasets from the Gene Expression Omnibus database, processed and normalized them using R software, and evaluated the relationship between differentially expressed genes and glioma by differential expression, expression quantitative trait loci, and Mendelian randomization (MR) analyses. Gene set enrichment analysis and immunocytometric analysis further explored the biological functions and pathways of identified genes, which were validated using The Cancer Genome Atlas and Genotype-Tissue Expression datasets. We identified eight co-expressed genes-C1QB, GPX3, LRRC8B, TRIOBP, SNAPC5, SPI1, TSPYL5, and FBXL16-that are crucial in various biological processes. CIBERSORT analysis revealed significant immune cell-type distributions within gliomas, underscoring the significance of immune cell infiltration. Validation in additional datasets confirmed the MR analysis results and upstream regulatory factors were identified using NetworkAnalyst. Our findings offer fresh perspectives on the molecular underpinnings of glioma and highlight potential targets for therapeutic interventions.
Collapse
Affiliation(s)
- Shiwen Pei
- The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Department of Neurosurgery, The Third People's Hospital of Bengbu, Bengbu, 233000, China
| | - Zhiquan Jiang
- Department of Neurosurgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, China.
| | - Hongwei Cheng
- The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
| |
Collapse
|
34
|
Sehgal P, Naqvi AS, Higgins M, Liu J, Harvey K, Jarroux J, Kim T, Mankaliye B, Mishra P, Watterson G, Fine J, Davis J, Hayer KE, Castro A, Mogbo A, Drummer C, Martinez D, Koptyra MP, Ang Z, Wang K, Farrel A, Quesnel-Vallieres M, Barash Y, Spangler JB, Rokita JL, Resnick AC, Tilgner HU, DeRaedt T, Powell DJ, Thomas-Tikhonenko A. Neuronal cell adhesion molecule (NRCAM) variant defined by microexon skipping is an essential, antigenically distinct, and targetable proteoform in high-grade glioma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.09.631916. [PMID: 39868324 PMCID: PMC11761023 DOI: 10.1101/2025.01.09.631916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
To overcome the paucity of known tumor-specific surface antigens in pediatric high-grade glioma (pHGG), we contrasted splicing patterns in pHGGs and normal brain samples. Among alternative splicing events affecting extracellular protein domains, the most pervasive alteration was the skipping of ≤30 nucleotide-long microexons. Several of these skipped microexons mapped to L1-IgCAM family members, such as NRCAM . Bulk and single-nuclei short- and long-read RNA-seq revealed uniform skipping of NRCAM microexons 5 and 19 in virtually every pHGG sample. Importantly, the Δex5Δex19 (but not the full-length) NRCAM proteoform was essential for pHGG cell migration and invasion in vitro and tumor growth in vivo. We developed a monoclonal antibody selective for Δex5Δex19 NRCAM and demonstrated that "painting" of pHGG cells with this antibody enables killing by T cells armed with an FcRI-based universal immune receptor. Thus, pHGG-specific NRCAM and possibly other L1-IgCAM proteoforms are promising and highly selective targets for adoptive immunotherapies. Statement of significance Existing targets for chimeric antigen receptors (CAR)-armed T cells are often shared by CNS tumors and normal tissues, creating the potential for on-target/off-tumor toxicities. Here we demonstrate that in CNS tumors of glial origin, cell adhesion molecules have alternatively spliced proteoforms, which could be targeted by highly selective therapeutic antibodies.
Collapse
|
35
|
Chen G, Shi X, Jiao R, Qian J, Du X, Liu J, Zeng X. Expression and prognostic value of ferritinophagy-related NCOA4 gene in low-grade glioma: integration of bioinformatics and experimental validation. BMC Neurol 2025; 25:26. [PMID: 39825225 PMCID: PMC11742756 DOI: 10.1186/s12883-025-04036-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 01/14/2025] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND Low-grade glioma (LGG) is a primary brain tumor with relatively low malignancy. NCOA4 is a key regulator of ferritinophagy-related processes and is involved in the occurrence and development of many cancers. However, the role of NCOA4 in LGG remains poorly understood. METHODS This study comprehensively analyzed several mainstream bioinformatics databases to explore the expression, diagnostic efficacy, clinical pathological features, immune infiltration, prognostic value, and biological functions of NCOA4 in LGG. Immunohistochemistry experiments were conducted using LGG tissue samples collected from our hospital to validate the bioinformatics analysis results. RESULTS NCOA4 expression was significantly elevated in LGG (p < 0.05), with an Area Under the Receiver Operating Characteristic Curve (AUC) of 0.973, suggesting it as a potential diagnostic marker. High NCOA4 expression was associated with younger age (21-40 years), lower malignancy (oligodendroglioma), and better prognosis (IDHmut-non-codel and IDHmut-codel subtypes) (all p < 0.05) in LGG. Kaplan-Meier survival curves from three databases showed that high NCOA4-expressing LGG patients had better prognosis (all p < 0.05). NCOA4 correlated weakly with B cells, CD8 + T cells, macrophages, and dendritic cells infiltration (all with correlation coefficients r < 0.3, and p < 0.05) in LGG. Multivariate Cox regression identified NCOA4, age, CD8 T cells, and macrophages as LGG independent prognostic factors (all p < 0.05). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses indicated that NCOA4's primary function in LGG is related to autophagy processes (all p < 0.05). CONCLUSION Our findings suggest that NCOA4 could be a potential prognostic marker and therapeutic target in LGG.
Collapse
Affiliation(s)
- Guangtang Chen
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Xueping Shi
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Rukai Jiao
- Department of Neurosurgery, The Affiliated Jinyang Hospital of Guizhou Medical University, Guiyang, 550081, Guizhou, China
| | - Jiacai Qian
- Department of Neurosurgery, The Affiliated Jinyang Hospital of Guizhou Medical University, Guiyang, 550081, Guizhou, China
| | - Xiaolin Du
- Department of Neurosurgery, The Affiliated Jinyang Hospital of Guizhou Medical University, Guiyang, 550081, Guizhou, China
| | - Jian Liu
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China.
- School of Clinical Medicine, Guizhou Medical University, Guiyang, 550004, Guizhou, China.
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, 550499, Guizhou, China.
| | - Xi Zeng
- Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China.
| |
Collapse
|
36
|
Xing W, Zhou Y, Long Q, Yi N, Wang G, Shi R, Huang J, Yin X, Zhu T, Cao S. Multiomic analysis of lactylation and mitochondria-related genes in hepatocellular carcinoma identified MRPL3 as a new prognostic biomarker. Front Oncol 2025; 14:1511958. [PMID: 39868366 PMCID: PMC11757296 DOI: 10.3389/fonc.2024.1511958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 12/19/2024] [Indexed: 01/28/2025] Open
Abstract
Background Recent research has highlighted lactate's crucial role in epigenetic regulation, particularly by influencing histone modifications that drive the initiation and progression of hepatocellular carcinoma (HCC). While mitochondria are known to regulate tumor behavior, the interaction between lactate metabolism and mitochondrial function in cancer tissues remains underexplored. Understanding this relationship may provide deeper insights into tumor metabolic reprogramming and reveal novel therapeutic targets for HCC and other malignancies. Methods We conducted a comprehensive screening of lactylation- and mitochondria-associated genes (LMRGs) in HCC patients, followed by clustering based on these genes. Prognostic outcomes and pathway enrichment were analyzed across the identified clusters. Additionally, we developed a prognostic model based on LMRGs, examining its implications for survival, immune response, and drug sensitivity. In vitro experiments were performed to validate the expression patterns and functional role of MRPL3 in HCC. Results We developed a prognostic model, named the LMRG model, incorporating three key genes: ACACA, MRPL3, and MRPS23. This model revealed significant differences in survival outcomes, immune responses, and drug sensitivity between patients with high and low LMRG scores. MRPL3 was found to be overexpressed in HCC, playing a critical role in tumor growth and metastasis. These results were further validated through in vitro experiments, confirming MRPL3's role in HCC cell proliferation and invasion. Conclusion We created a predictive model, LMRG, and identified MRPL3 as a key biomarker. Our findings suggest that MRPL3 has significant potential as a reliable predictive biomarker for clinical applications in HCC diagnosis and treatment.
Collapse
Affiliation(s)
- Wenya Xing
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Yuanzi Zhou
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Qiuzi Long
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Nan Yi
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Gaoyuan Wang
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Rongwei Shi
- Department of General Internal Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Jinlong Huang
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Xindong Yin
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Taiyang Zhu
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Shibing Cao
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| |
Collapse
|
37
|
Zhang L, Li YW, Xie T, Sun K, Huang X, Xiong W, Liu RJ. Potential role of P4HB in the tumor microenvironment and its clinical prognostic value: a comprehensive pan-cancer analysis and experimental validation with a focus on KIRC. Cancer Cell Int 2025; 25:1. [PMID: 39754183 PMCID: PMC11697512 DOI: 10.1186/s12935-024-03575-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 11/13/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND Tumor microenvironment (TME) plays a crucial role in tumor growth and metastasis. Exploring biomarkers that are significantly associated with TME can help guide individualized treatment of patients. METHODS We analyzed the expression and survival of P4HB in pan-cancer through the TCGA database, and verified the protein level of P4HB by the HPA database. In addition, we used the Metascape database to construct protein-protein interaction networks and the single-cell Sequencing database for functional analysis. An immune cell infiltration analysis was performed to explore the potential role of P4HB in TME. We further analyze the relationship between P4HB and immune checkpoint molecules to explore the role of P4HB in immune checkpoint blockade therapy. Finally, the oncogenic role of P4HB in RCC cells was validated using colony formation and wound healing assays. RESULTS RNA and protein levels of P4HB were extensively up-regulated in pan-cancer. However, high P4HB expression was associated with poor survival in KIRC. The clinical relevance analyses of P4HB suggested that high P4HB expression was associated with advanced clinical TNM stage. Moreover, multivariate cox regression analysis indicated that P4HB (HR = 1.372, 95% CI 1.047-1.681, P = 0.019) was an independent risk factor for OS in KIRC. Functional analysis revealed that P4HB is involved in hypoxia, TME and immune system processes. Our study also found that high P4HB expression was significantly correlated with elevated infiltration levels in CD8 + T cells and M2 macrophages. The results of colony formation and wound healing assays showed that knockdown of P4HB inhibited the RCC growth and migration. CONCLUSIONS P4HB is a specific biomarker for KIRC prognosis and is significantly associated with clinical characteristics. In addition, P4HB may play an influential role in TME and is a biomarker for ICB therapy.
Collapse
Affiliation(s)
- Linxue Zhang
- School of Materials and Energy, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Yu-Wei Li
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, China
| | - Tianyi Xie
- Department of Neuroscience, Kenneth P. Dietrich School of Arts & Science, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ke Sun
- School of Materials and Energy, University of Electronic Science and Technology of China, Chengdu, 610054, China.
| | - Xiang Huang
- Department of Urology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Wei Xiong
- Department of Urology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Rui-Ji Liu
- Department of Urology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
38
|
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
|
39
|
Wang XJ, Huo YX, Yang PJ, Gao J, Hu WD. Significance of Ribonucleoside-diphosphate Reductase Subunit M2 in Lung Adenocarcinoma. Curr Gene Ther 2025; 25:136-156. [PMID: 38920074 DOI: 10.2174/0115665232286359240611051307] [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: 10/09/2023] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 06/27/2024]
Abstract
INTRODUCTION The Ribonucleoside-diphosphate Reductase subunit M2 (RRM2) is known to be overexpressed in various cancers, though its specific functional implications remain unclear. This aims to elucidate the role of RRM2 in the progression of Lung Adenocarcinoma (LUAD) by exploring its involvement and potential impact. METHODS RRM2 data were sourced from multiple databases to assess its diagnostic and prognostic significance in LUAD. We evaluated the association between RRM2 expression and immune cell infiltration, analyzed its function, and explored the effects of modulating RRM2 expression on LUAD cell characteristics through laboratory experiments. RESULTS RRM2 was significantly upregulated in LUAD tissues and cells compared to normal counterparts (p < 0.05), with rare genetic alterations noted (approximately 2%). This overexpression clearly distinguished LUAD from normal tissue (area under the curve (AUC): 0.963, 95% confidence intervals (CI): 0.946-0.981). Elevated RRM2 expression was significantly associated with adverse clinicopathological characteristics and poor prognosis in LUAD patients. Furthermore, a positive association was observed between RRM2 expression and immune cell infiltration. Pathway analysis revealed a critical connection between RRM2 and the cell cycle signaling pathway within LUAD. Targeting RRM2 inhibition effectively suppressed LUAD cell proliferation, migration, and invasion while promoting apoptosis. This intervention also modified the expression of several crucial proteins, including the downregulation of CDC25A, CDC25C, RAD1, Bcl-2, and PPM1D and the upregulation of TP53 and Bax (p < 0.05). CONCLUSION Our findings highlight the potential utility of RRM2 expression as a biomarker for diagnosing and predicting prognosis in LUAD, shedding new light on the role of RRM2 in this malignancy.
Collapse
Affiliation(s)
- Xiao-Jun Wang
- Department of Respiratory Medicine, Gansu Province People Hospital, Lanzhou, Gansu, PR China
| | - Yun-Xia Huo
- Department of Neurological Surgery, The Second People Hospital of Lanzhou City, Lanzhou, Gansu, PR China
| | - Peng-Jun Yang
- Department of Internal Medicine, The Xigu Hospital of Lanzhou City, Lanzhou, Gansu, PR China
| | - Jing Gao
- Department of Respiratory Medicine, Gansu Province People Hospital, Lanzhou, Gansu, PR China
- Department of Medicine, Respiratory Medicine Unit , Karolinska Institute, Stockholm, Sweden
- Department of Pulmonary Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Wei-Dong Hu
- Department of Respiratory Medicine, Gansu Province People Hospital, Lanzhou, Gansu, PR China
| |
Collapse
|
40
|
Luo J, Zhang Q, Wang S, Zheng L, Liu J, Zhang Y, Wang Y, Wang R, Xiao Z, Li Z. Comprehensive Pan-cancer Analysis of CMPK2 as Biomarker and Prognostic Indicator for Immunotherapy. Curr Cancer Drug Targets 2025; 25:209-229. [PMID: 38486392 DOI: 10.2174/0115680096281451240306062101] [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: 10/12/2023] [Revised: 01/30/2024] [Accepted: 02/14/2024] [Indexed: 02/26/2025]
Abstract
BACKGROUND UMP-CMP kinase 2 (CMPK2) is involved in mitochondrial DNA synthesis, which can be oxidized and released into the cytoplasm in innate immunity. It initiates the assembly of NLRP3 inflammasomes and mediates various pathological processes such as human immunodeficiency virus infection and systemic lupus erythematosus. However, the role of CMPK2 in tumor progression and tumor immunity remains unclear. METHODS We identified CMPK2 expression patterns in the Genotype Tissue-Expression (GTEx), The Cancer Genome Atlas (TCGA), and the Cancer Cell Line Encyclopedia (CCLE) databases. Validation was performed using immunohistochemical staining data from the Human Protein Atlas (HPA) database and qPCR experiments. Receiver operating characteristic curve analysis and Kaplan-Meier survival analysis were conducted to assess the clinical relevance of CMPK2 expression. The Estimation of Stromal and Immune Cells in Malignant Tumor Tissues Using Expression Data (ESTIMATE) algorithm and the Tumor IMmune Estimation Resource (TIMER) database were used to evaluate the correlation between CMPK2 and immune infiltration in tumors. The Tumor Immune Syngeneic Mouse (TISMO) database and other public datasets were utilized to assess the impact of CMPK2 on immune therapy response. MEXPRESS and MethSurv databases were employed to investigate the effects of methylation on CMPK2 expression. RESULTS CMPK2 expression was elevated in 23 cancers and decreased in two cancers. Furthermore, CMPK2 expression had a high diagnostic value for 16 cancers. Elevated CMPK2 expression was associated with lower overall survival (OS), disease-specific survival (DSS), and progression- free interval (PFI) in four cancers. Immune microenvironment-related analysis revealed strong associations between CMPK2 expression and immune cell infiltration, as well as immune checkpoint expression across various tumors. Notably, in four mouse immunotherapy cohorts, CMPK2 expression in treated mouse tumors was higher post-treatment. In five clinical immunotherapy cohorts, patients with high CMPK2 expression show better responses to immunotherapy. Moreover, the methylation level of CMPK2 gene was closely correlated to its expression and tumor prognosis. Among these cancers, the clinical and immunological indications of skin cutaneous melanoma (SKCM) are particularly closely related to CMPK2 expression. CONCLUSION Our analysis preliminarily describes the complex function of CMPK2 in cancer progression and immune microenvironment, highlighting its potential as a diagnostic and therapeutic target for immunotherapy.
Collapse
Affiliation(s)
- Jingyuan Luo
- NHC Key Laboratory of Carcinogenesis, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Clinical Medicine, Xiangya School of Medicine of Central South University, Changsha, China
| | - Qianyue Zhang
- Department of Clinical Medicine, Xiangya School of Medicine of Central South University, Changsha, China
| | - Shutong Wang
- NHC Key Laboratory of Carcinogenesis, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Clinical Medicine, Xiangya School of Medicine of Central South University, Changsha, China
| | - Luojie Zheng
- NHC Key Laboratory of Carcinogenesis, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jie Liu
- NHC Key Laboratory of Carcinogenesis, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Clinical Medicine, Xiangya School of Medicine of Central South University, Changsha, China
| | - Yuchen Zhang
- NHC Key Laboratory of Carcinogenesis, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Clinical Medicine, Xiangya School of Medicine of Central South University, Changsha, China
| | - Yingchen Wang
- NHC Key Laboratory of Carcinogenesis, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Clinical Medicine, Xiangya School of Medicine of Central South University, Changsha, China
| | - Ranran Wang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhigang Xiao
- Department of General Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Zheng Li
- NHC Key Laboratory of Carcinogenesis, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| |
Collapse
|
41
|
Bai W, Zhao X, Ning Q. Development and validation of a radiomic prediction model for TACC3 expression and prognosis in non-small cell lung cancer using contrast-enhanced CT imaging. Transl Oncol 2025; 51:102211. [PMID: 39603208 PMCID: PMC11635781 DOI: 10.1016/j.tranon.2024.102211] [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: 08/07/2024] [Revised: 10/10/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUNDS Non-small cell lung cancer (NSCLC) prognosis remains poor despite treatment advances, and classical prognostic indicators often fall short in precision medicine. Transforming acidic coiled-coil protein-3 (TACC3) has been identified as a critical factor in tumor progression and immune infiltration across cancers, including NSCLC. Predicting TACC3 expression through radiomic features may provide valuable insights into tumor biology and aid clinical decision-making. However, its predictive value in NSCLC remains unexplored. Therefore, we aimed to construct and validate a radiomic model to predict TACC3 levels and prognosis in patients with NSCLC. MATERIALS AND METHODS Genomic data and contrast-enhanced computed tomography (CT) images were sourced from The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO) database, and The Cancer Imaging Archive (TCIA). A total of 320 cases of lung adenocarcinoma from TCGA and 122 cases of NSCLC from GEO were used for prognostic analysis. Sixty-three cases from TCIA and GEO were included for radiomics feature extraction and model development. The radiomics model was constructed using logistic regression (LR) and support vector machine (SVM) algorithms. We predicted TACC3 expression and evaluated its correlation with NSCLC prognosis using contrast-enhanced CT-based radiomics. RESULTS TACC3 expression significantly influenced NSCLC prognosis. High TACC3 levels were associated with reduced overall survival, potentially mediated by immune microenvironment and tumor progression regulation. LR and SVM algorithms achieved AUC of 0.719 and 0.724, respectively, which remained at 0.701 and 0.717 after five-fold cross-validation. CONCLUSION Contrast-enhanced CT-based radiomics can non-invasively predict TACC3 expression and provide valuable prognostic information, contributing to personalized treatment strategies.
Collapse
Affiliation(s)
- Weichao Bai
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710061, China
| | - Xinhan Zhao
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710061, China
| | - Qian Ning
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710061, China.
| |
Collapse
|
42
|
Cheng L, Wei L, Chen Q, Li P, Zhang D. DDR1 in pancreatic adenocarcinoma: unraveling the mechanisms of immune exclusion. Scand J Gastroenterol 2025; 60:81-90. [PMID: 39713856 DOI: 10.1080/00365521.2024.2443505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/07/2024] [Accepted: 12/12/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND Pancreatic adenocarcinoma (PAAD) is a deadly cancer marked by extensive collagen deposition and limited response to immunotherapy. Discoidin domain receptor1 (DDR1), part of the transmembrane receptor tyrosine kinase family, is linked to inflammation regulation and immune cell infiltration. However, its role in controlling cytokines and chemokines in the microenvironment of PAAD is still unclear. METHODS Initially, RNA sequencing data from TCGA were utilized to investigate the expression of DDR1. Subsequently, the relationship between DDR1 and immune infiltration was examined through Gene Ontology (GO) and Gene Set Enrichment Analysis (GSEA) analysis, in conjunction with ssGSEA Immunoanalysis. Lastly, the effect of DDR1 on the malignant characteristics of PAAD cells was examined through in vitro experimentation, employing various techniques such as the CCK8 assay, colony formation assay and reverse transcription-quantitative polymerase chain reaction (RT-qPCR). RESULTS The research revealed a notable increase in the expression level of DDR1 in PAAD. Subsequent analysis indicated a correlation between the differential expression of DDR1 with chemokines and immune infiltration. Additionally, cellular experiments demonstrated that the downregulation of DDR1 led to enhanced expression of chemokines CCL4, CCL5 and CXCL10. CONCLUSION DDR1 is linked to tumor immune infiltration, and the knockout of DDR1 results in the upregulation of chemokines CCL4, CCL5 and CXCL10 in Pan02 PAAD cells.
Collapse
Affiliation(s)
- Long Cheng
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, China
| | - Lina Wei
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, China
| | - Qian Chen
- The Second Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Peirong Li
- Department of Emergency, Lanzhou University Second Hospital, Lanzhou, China
| | - Dekui Zhang
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
43
|
Huang T, Wei L, Zhou H, Liu J. Macrophage Infiltration and ITGB2 Expression in ESCC: A Novel Correlation. Cancer Med 2025; 14:e70604. [PMID: 39825491 PMCID: PMC11742006 DOI: 10.1002/cam4.70604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/22/2024] [Accepted: 01/03/2025] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is one of the most prevalent and lethal malignancies worldwide. Despite progress in immunotherapy for cancer treatment, its application and efficacy in ESCC remain limited. Therefore, there is an ongoing need to explore potential molecules and therapeutic strategies related to tumor immunity in ESCC. METHODS In this study, we integrated high-throughput sequencing data, gene chip data, single-cell sequencing data, and various bioinformatics analysis methods along with experimental approaches to identify key genes involved in immune infiltration in ESCC and investigate their relationship with immune cell development, as well as the potential of these key genes in immunotherapy. RESULTS We discovered and validated a positive correlation between macrophage infiltration and ITGB2 expression in ESCC. ITGB2 is overexpressed in ESCC and has potential as a prognostic biomarker for the disease. We present for the first time the finding that the expression of ITGB2 in infiltrating macrophages increases as these macrophages polarize toward a tumor-promoting phenotype in ESCC. Moreover, during the progression of ESCC, ITGB2 expression in infiltrating macrophages is upregulated. The higher the expression of ITGB2, the more feasible it is to target macrophages. Additionally, we found that evaluating immune therapy responses in ESCC patients through ITGB2 expression is a viable approach. Furthermore, we identified three miRNAs associated with abnormal ITGB2 expression, providing insights into the upstream molecular interactions of ITGB2. CONCLUSIONS Macrophage infiltration in ESCC is closely associated with ITGB2, which holds significant potential for immunotherapy applications in ESCC. Based on our findings and prior studies, we propose a novel hypothesis: inducing M1 macrophages in vitro, knocking out ITGB2, and then reinfusing these ITGB2-knockout M1 macrophages into ESCC patients may represent a promising new immunotherapy strategy, providing a new avenue for ESCC immunotherapy.
Collapse
Affiliation(s)
- Tao Huang
- Department of Cardiothoracic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningPeople's Republic of China
| | - Longqian Wei
- Department of Cardiothoracic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningPeople's Republic of China
| | - Huafu Zhou
- Department of Cardiothoracic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningPeople's Republic of China
| | - Jun Liu
- Department of Cardiothoracic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningPeople's Republic of China
| |
Collapse
|
44
|
Liu Y, Wang H, Zhao S, Wang Z, Yang L, Zhang J, Hou Q, Xiao Z, Wang P, Liu Y. Prognostic value and clinical significance of IL-33 expression in patients with uterine corpus endometrial carcinoma. Cytokine 2025; 185:156828. [PMID: 39657332 DOI: 10.1016/j.cyto.2024.156828] [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: 09/19/2024] [Revised: 11/15/2024] [Accepted: 12/03/2024] [Indexed: 12/12/2024]
Abstract
Uterine corpus endometrial carcinoma (UCEC) is one of the most common malignant tumours of the female genital tract. In the occurrence, progression and prognosis of UCEC, chronic inflammation plays an important role, making it pivotal to identify inflammatory response-related endometrial diseases. The cytokine interleukin-33 (IL-33) plays significant roles in immune responses, and has been associated with inappropriate allergic reactions, autoimmune diseases, and cancer pathology. In the past decade, studies have begun to uncover the pivotal roles of IL-33 in shaping tumour microenvironment (TME), where it may promote or inhibit tumorigenesis and development depending on the specific tumour types. However, the association between IL-33 expression and UCEC remains unclear. Here we investigated the expression profiles of IL-33 in pan-cancer based on TCGA database. Then, differential gene expression analysis and correlation analysis of IL-33 was investigated in UCEC. In addition, functional enrichment analysis and Kaplan-Meier survival analysis were performed to predict the potential function of IL-33 and its role in the prognosis of UCEC patients. Also, a nomogram model was constructed to predict the prognosis of UCEC. The expression of the inflammatory factor NF-κB p65 and the IL-33, along with its receptor ST2, was analyzed in UCEC tumour tissues and normal tissues of clinical specimens through immunohistochemical staining. Meanwhile, we used toluidine blue staining and methanol Congo red staining to observe the infiltration of mast cells and eosinophils in the endometrial tissue. The results of Kaplan-Meier plotter data indicated that patients with lower IL-33 expression had poorer progression-free interval than those with higher expression. Based on the results of multifactor Cox regression, a nomogram was generated to predict UCEC occurrence risk and prognosis. Clinical specimen characteristics also confirmed a negative correlation between IL-33 expression and UCEC staging and grading. This comprehensive analysis of IL-33, based on bioinformatics and immunohistochemistry, revealed that IL-33 has the function of inhibiting UCEC occurrence and progression and can be served as a beneficial prognostic marker in the clinic.
Collapse
Affiliation(s)
- Yuqi Liu
- School of Basic Medical College, Beihua University, Jilin 132013, China
| | - Han Wang
- School of Basic Medical College, Beihua University, Jilin 132013, China
| | - Shihan Zhao
- School of Basic Medical College, Beihua University, Jilin 132013, China
| | - Zhenjiang Wang
- School of Basic Medical College, Beihua University, Jilin 132013, China
| | - Lijuan Yang
- School of Basic Medical College, Beihua University, Jilin 132013, China
| | - Jihong Zhang
- The Pathology Department of Affiliated Hospital, Beihua University, Jilin 132013, China
| | - Qinlong Hou
- School of Basic Medical College, Beihua University, Jilin 132013, China
| | - ZiShen Xiao
- School of Basic Medical College, Beihua University, Jilin 132013, China
| | - Pengmin Wang
- Département des sciences animales, Faculté des sciences de l'agriculture et de l'alimentation, Université Laval, Québec City, Québec, Canada.
| | - Yanbo Liu
- School of Basic Medical College, Beihua University, Jilin 132013, China.
| |
Collapse
|
45
|
Peng P, Zheng J, He K, Wang K, Wang L, Zheng X, Wu H, Yang Z, Zhang S, Zhao L. CENPE is a diagnostic and prognostic biomarker for cervical cancer. Heliyon 2024; 10:e40860. [PMID: 39759304 PMCID: PMC11698922 DOI: 10.1016/j.heliyon.2024.e40860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 11/26/2024] [Accepted: 11/30/2024] [Indexed: 01/07/2025] Open
Abstract
Cervical squamous cell carcinoma (CESC) is a common cancer in women. Despite advancements in early diagnosis through high-risk human papillomavirus (HPV) screening, challenges remain in predicting and treating the disease. Hence, the identification of novel biomarkers for prognosis and therapeutic targets is crucial. CENPE, a microtubule-end directed motor protein that accumulates during the G2 phase, is recognized for its involvement in promoting cancer growth and progression. However, its specific role in CESC remains unclear. This research investigated the expression of CENPE in CESC utilizing data from The Cancer Genome Atlas (TCGA), which was further validated through gene expression profiles, the Human Protein Atlas (HPA), and clinical data. The study utilized Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene Set Enrichment Analysis (GSEA), and immune infiltration analysis to elucidate the role of CENPE in CESC. Additionally, Protein-protein interaction (PPI) networks and competing endogenous RNA (CeRNA) networks involving CENPE and its differentially expressed genes were established. Furthermore, Kaplan-Meier survival analysis was conducted to evaluate the impact of CENPE on patient prognosis. Our study revealed an upregulation of CENPE expression in cervical cancer tissues, which promotes the progression of CESC through IL-6-mediated PI3K-Akt and MAPK signaling pathways. The significant associations with ACNG3, LY6H, and SLC6A7 suggest that CENPE may play a role in tumor growth and metastasis, potentially involving the nervous system. Moreover, the correlations with ARIH1, KDM1A, KDM5B, and NSD3 indicate that CENPE could be a promising target for drug development. Our analysis of the ROC curve demonstrated a high diagnostic accuracy of CENPE in CESC (AUC: 0.997, CI: 0.990-1.000). Subgroup analysis highlighted substantial effects in patients under 50 years old, those with a height under 160 cm, individuals in peri- and post-menopausal stages, and patients in clinical stages 1 and 4. Additionally, COX regression analysis indicated that older age, lower BMI, and higher CENPE expression are associated with decreased 1-year, 3-year, and 5-year survival rates. In conclusion, CENPE emerges as a crucial factor in the initiation and advancement of cervical cancer, showing potential as a novel target for therapeutic interventions.
Collapse
Affiliation(s)
- Peiqiang Peng
- Department of Medical Rehabilitation, School of Nursing, Jilin University, 965 Xinjiang Street, Chaoyang District, Changchun, 130021, China
| | - Jingying Zheng
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, No.4026, Yatai Street, Changchun City, 130000, Jilin Province, China
| | - Kang He
- Department of Medical Rehabilitation, School of Nursing, Jilin University, 965 Xinjiang Street, Chaoyang District, Changchun, 130021, China
| | - Kai Wang
- Department of Medical Rehabilitation, School of Nursing, Jilin University, 965 Xinjiang Street, Chaoyang District, Changchun, 130021, China
| | - Longyun Wang
- Department of Medical Rehabilitation, School of Nursing, Jilin University, 965 Xinjiang Street, Chaoyang District, Changchun, 130021, China
| | - Xufei Zheng
- Department of Medical Rehabilitation, School of Nursing, Jilin University, 965 Xinjiang Street, Chaoyang District, Changchun, 130021, China
| | - Hao Wu
- Department of Medical Rehabilitation, School of Nursing, Jilin University, 965 Xinjiang Street, Chaoyang District, Changchun, 130021, China
| | - Zhenning Yang
- Department of Medical Rehabilitation, School of Nursing, Jilin University, 965 Xinjiang Street, Chaoyang District, Changchun, 130021, China
| | - Shuang Zhang
- Department of Medical Rehabilitation, School of Nursing, Jilin University, 965 Xinjiang Street, Chaoyang District, Changchun, 130021, China
| | - Lijing Zhao
- Department of Medical Rehabilitation, School of Nursing, Jilin University, 965 Xinjiang Street, Chaoyang District, Changchun, 130021, China
| |
Collapse
|
46
|
Xu X, Jané P, Taelman V, Jané E, Dumont RA, Garama Y, Kim F, Del Val Gómez M, Gariani K, Walter MA. The Theranostic Genome. Nat Commun 2024; 15:10904. [PMID: 39738156 DOI: 10.1038/s41467-024-55291-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 12/05/2024] [Indexed: 01/01/2025] Open
Abstract
Theranostic drugs represent an emerging path to deliver on the promise of precision medicine. However, bottlenecks remain in characterizing theranostic targets, identifying theranostic lead compounds, and tailoring theranostic drugs. To overcome these bottlenecks, we present the Theranostic Genome, the part of the human genome whose expression can be utilized to combine therapeutic and diagnostic applications. Using a deep learning-based hybrid human-AI pipeline that cross-references PubMed, the Gene Expression Omnibus, DisGeNET, The Cancer Genome Atlas and the NIH Molecular Imaging and Contrast Agent Database, we bridge individual genes in human cancers with respective theranostic compounds. Cross-referencing the Theranostic Genome with RNAseq data from over 17'000 human tissues identifies theranostic targets and lead compounds for various human cancers, and allows tailoring targeted theranostics to relevant cancer subpopulations. We expect the Theranostic Genome to facilitate the development of new targeted theranostics to better diagnose, understand, treat, and monitor a variety of human cancers.
Collapse
Affiliation(s)
- Xiaoying Xu
- University of Lucerne, Lucerne, LU, Switzerland
| | - Pablo Jané
- University of Geneva, Geneva, GE, Switzerland
- Nuclear Medicine and Molecular Imaging Division, Geneva University Hospitals, Geneva, GE, Switzerland
| | | | - Eduardo Jané
- Departamento de Matemática Aplicada a la Ingeniería Aeroespacial - ETSIAE, Universidad Politécnica de Madrid, 28040, Madrid, Spain
| | | | | | | | - María Del Val Gómez
- Servicio de Medicina Nuclear, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Karim Gariani
- Division of Endocrinology, Diabetes, Nutrition and Patient Therapeutic Education, Geneva University Hospitals, Geneva, GE, Switzerland
| | - Martin A Walter
- University of Lucerne, Lucerne, LU, Switzerland.
- St. Anna Hospital, University of Lucerne, Lucerne, LU, Switzerland.
| |
Collapse
|
47
|
Agrawal P, Olgun G, Singh A, Gopalan V, Hannenhalli S. Characterizing the pan-cancer role of exosomal miRNAs in metastasis across cancers. Comput Struct Biotechnol J 2024; 27:252-264. [PMID: 39866667 PMCID: PMC11763893 DOI: 10.1016/j.csbj.2024.12.025] [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: 11/13/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/28/2025] Open
Abstract
Exosomal microRNAs (exomiRs) play a critical role in intercellular communication, especially in cancer, where they regulate key cellular processes like proliferation, angiogenesis, and metastasis, highlighting their significance as potential diagnostic and therapeutic targets. Here, we aimed to characterize the role of exomiRs, derived from seven cancer types (four cell lines and three tumors), in influencing the pre-metastatic niche (PMN). In each cancer type we extracted high confidence exomiRs (LogFC >= 2 in exosomes relative to control), their experimentally validated targets, and the enriched pathways among those targets. We then selected the top100 high-confidence targets based on their frequency of appearance in the enriched pathways. We observed significantly higher GC content in exomiRs relative to genomic background. Gene Ontology analysis revealed both general cancer processes, such as wound healing and epithelial cell proliferation, as well as cancer-specific processes, such as "angiogenesis" in the kidney and "ossification" in the lung. ExomiR targets were enriched for cancer-specific tumor suppressor genes and downregulated in PMN formed in lungs compared to normal. Motif analysis showed high inter-cancer similarity among motifs enriched in exomiRs. Our analysis recapitulated exomiRs associated with M2 macrophage differentiation and chemoresistance, such as miR-21 and miR-222-3p, regulating signaling pathways like PTEN/PI3/Akt, NF-kB, etc. Additionally, Cox regression analysis in TCGA indicated that exomiR targets are significantly associated with better overall survival of patients. Lastly, support vector machine model using exomiR targets gene expression classified responders and non-responders to therapy with an AUROC ranging from 0.72 to 0.96, higher than previously reported gene signatures.
Collapse
Affiliation(s)
- Piyush Agrawal
- Cancer Data Science Laboratory, National Cancer Institute, Bethesda, MD, USA
| | - Gulden Olgun
- Department of Computer Engineering, Hacettepe University, Ankara 06800, Turkey
| | - Arashdeep Singh
- Cancer Data Science Laboratory, National Cancer Institute, Bethesda, MD, USA
| | - Vishaka Gopalan
- Cancer Data Science Laboratory, National Cancer Institute, Bethesda, MD, USA
| | - Sridhar Hannenhalli
- Cancer Data Science Laboratory, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
48
|
Zhai Z, Cui Z, Zhang Y, Song P, Wu J, Tan Z, Lin S, Ma X, Guan F, Kang H. Integrated pan-cancer analysis and experimental verification of the roles of meiotic nuclear divisions 1 in breast cancer. Biochem Biophys Res Commun 2024; 739:150600. [PMID: 39191147 DOI: 10.1016/j.bbrc.2024.150600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/08/2024] [Accepted: 08/22/2024] [Indexed: 08/29/2024]
Abstract
INTRODUCTION The aberrant up-regulation of meiotic nuclear division 1 (MND1) in somatic cells is considered as one of the driving factors of oncogenesis, whereas its expression and role in breast invasive cancer (BRCA) remain unclear. Hence, this study embarked on a comprehensive evaluation of MND1 across various cancers and identified its roles in BRCA. METHODS Based on publicly available databases, including but not limited to UCSC Xena, TCGA, GTEx, GEO, STRING, GeneMANIA, and CancerSEA, we evaluated the expression patterns, genomic features, and biological functions of MND1 from a pan-cancer viewpoint and delved into the implications of MND1 in the prognosis and treatment of BRCA. Further molecular biology experiments were undertaken to identify the role of MND1 in proliferation, migration, and apoptosis in BRCA cells. RESULTS Elevated levels of MND1 were notably observed in a wide array of tumor types, especially in BRCA, COAD, HNSC, LIHC, LUAD, LUSC, STAD, and UCEC. Elevated MND1 expression was markedly associated with shortened OS in several tumors, including BRCA (HR = 1.52 [95%CI, 1.10-2.09], P = 0.011). The up-regulation of MND1 in BRCA was validated in external cohorts and clinical samples. Survival analyses demonstrated that elevated MND1 expression was associated with decreased survival for patients with BRCA. Co-expressed genes of MND1 were identified, and subsequent pathway analyses based on significantly associated genes indicated that MND1 plays key roles in DNA replication, cell cycle regulation, and DNA damage repair. The observed abnormal elevation and activation of MND1 led to increased proliferation and migration, along with decreased apoptosis in BRCA cells. CONCLUSIONS MND1 emerges as a promising biomarker for diagnostic and therapeutic targeting in various cancers, including BRCA. The abnormal up-regulation and activation of MND1 are linked to carcinogenesis and poor prognosis among BRCA patients, which may be attributed to its involvement in HR-dependent ALT, warranting further scrutiny.
Collapse
Affiliation(s)
- Zhen Zhai
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West Fifth Road, Xi'an, China; Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West Fifth Road, Xi'an, China
| | - Zhiwei Cui
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, Yanta West Road, Xi' an, China
| | - Yu Zhang
- Department of Infectious Diseases, Honghui-hospital, Xi'an Jiaotong University, Shanghua Road, Xi'an, China
| | - Ping Song
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, No. 157, West Fifth Road, Xi'an, China
| | - Jinpeng Wu
- College of Life Sciences, Northwest University, No. 229, Taibai North Road, Xi'an, China
| | - Zengqi Tan
- Institute of Hematology, Provincial Key Laboratory of Biotechnology, School of Medicine, Northwest University, No. 229, Taibai North Road, Xi'an, China
| | - Shuai Lin
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West Fifth Road, Xi'an, China; Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West Fifth Road, Xi'an, China
| | - Xiaobin Ma
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West Fifth Road, Xi'an, China; Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West Fifth Road, Xi'an, China
| | - Feng Guan
- College of Life Sciences, Northwest University, No. 229, Taibai North Road, Xi'an, China.
| | - Huafeng Kang
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West Fifth Road, Xi'an, China; Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West Fifth Road, Xi'an, China.
| |
Collapse
|
49
|
Bontonou M, Haget A, Boulougouri M, Audit B, Borgnat P, Arbona JM. A comparative analysis of gene expression profiling by statistical and machine learning approaches. BIOINFORMATICS ADVANCES 2024; 5:vbae199. [PMID: 39897946 PMCID: PMC11783302 DOI: 10.1093/bioadv/vbae199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 10/27/2024] [Accepted: 12/16/2024] [Indexed: 02/04/2025]
Abstract
Motivation Many machine learning (ML) models developed to classify phenotype from gene expression data provide interpretations for their decisions, with the aim of understanding biological processes. For many models, including neural networks, interpretations are lists of genes ranked by their importance for the predictions, with top-ranked genes likely linked to the phenotype. In this article, we discuss the limitations of such approaches using integrated gradient, an explainability method developed for neural networks, as an example. Results Experiments are performed on RNA sequencing data from public cancer databases. A collection of ML models, including multilayer perceptrons and graph neural networks, are trained to classify samples by cancer type. Gene rankings from integrated gradients are compared to genes highlighted by statistical feature selection methods such as DESeq2 and other learning methods measuring global feature contribution. Experiments show that a small set of top-ranked genes is sufficient to achieve good classification. However, similar performance is possible with lower-ranked genes, although larger sets are required. Moreover, significant differences in top-ranked genes, especially between statistical and learning methods, prevent a comprehensive biological understanding. In conclusion, while these methods identify pathology-specific biomarkers, the completeness of gene sets selected by explainability techniques for understanding biological processes remains uncertain. Availability and implementation Python code and datasets are available at https://github.com/mbonto/XAI_in_genomics.
Collapse
Affiliation(s)
- Myriam Bontonou
- CNRS, ENS de Lyon, Inserm, LBMC, UMR5239, U1293, F-69342 Lyon Cedex 07, France
- CNRS, ENS de Lyon, LPENSL, UMR5672, F-69342 Lyon Cedex 07, France
| | - Anaïs Haget
- LTS2 Laboratory, EPFL, 1015 Lausanne, Switzerland
| | | | - Benjamin Audit
- CNRS, ENS de Lyon, LPENSL, UMR5672, F-69342 Lyon Cedex 07, France
| | - Pierre Borgnat
- CNRS, ENS de Lyon, LPENSL, UMR5672, F-69342 Lyon Cedex 07, France
| | - Jean-Michel Arbona
- CNRS, ENS de Lyon, Inserm, LBMC, UMR5239, U1293, F-69342 Lyon Cedex 07, France
| |
Collapse
|
50
|
Ni X, Pan F, Lang YK, Zhang W. Prognostic significance of NUAK1 and its association with immune infiltration in stomach adenocarcinoma. Discov Oncol 2024; 15:800. [PMID: 39692916 DOI: 10.1007/s12672-024-01688-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 12/09/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND Stomach adenocarcinoma (STAD) represents a significant global health burden, accounting for a considerable proportion of cancer-related mortalities, and NUAK1, a protein kinase, plays a crucial role in cellular metabolism, cell cycle regulation, migration, and tumor progression. However, its relationship with prognosis and immune infiltration in STAD has not been thoroughly investigated. METHODS RNA sequencing data from the Cancer Genome Atlas (TCGA) and Genotypic Tissue Expression Project (GTEx) databases were employed to assess disparities in NUAK1 expression between STAD tumour and normal tissues. Additionally, we investigated the correlation between NUAK1 expression and patient prognosis, in addition to the level of immune cell infiltration. The potential functions were elucidated through an examination of the Gene Ontology (GO) Encyclopedia, the Kyoto Encyclopedia of Genes and Genomes (KEGG), and an enrichment analysis (GSEA). The GeneMANIA was used to validate the functions of nuak1-related genes. RESULTS Our analysis demonstrated that NUAK1 expression in tumour tissues exhibited a notable disparity from that observed in normal tissues, with elevated levels detected in STAD tissues. We used the GeneMANIA database to identify functionally similar genes with significantly higher expression for some genes in the unpaired group samples. An elevated NUAK1 expression level was found to correlate with a poorer overall survival (OS), disease-specific survival (DSS), and progression-free intervals (PFI). Additionally, immune infiltration analysis indicated a significant positive correlation between NUAK1 expression and various tumor-infiltrating immune cells, while a negative correlation was observed with T helper cell 17(Th17) cells. Furthermore, enrichment analysis was conducted to identify relevant biological features and pathways. CONCLUSION The expression levels of NUAK1 are significantly increased in STAD, and this heightened expression correlates with diminished OS, DSS, and PFI among affected patients. These observations indicate that NUAK1 has the potential to function as a prognostic biomarker for STAD and may represent a viable therapeutic target for intervention in its management.
Collapse
Affiliation(s)
- Xin Ni
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, No. 438 Jiefang Road, Zhenjiang, Jiangsu, China
| | - Fan Pan
- Department of Articular Surgery, Affiliated Hospital of Jiangsu University, No. 438 Jiefang Road, Zhenjiang, Jiangsu, China
| | | | - Wei Zhang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, No. 438 Jiefang Road, Zhenjiang, Jiangsu, China.
| |
Collapse
|