1
|
Tan X, Liang X, Feng Y, Xie M, Zhong K, Luo W, Wang Y, Yin Y, Cai Y. NEK2 promotes the progression of osteosarcoma through the AKT/p-AKT pathway and interacts with FoxM1. Med Oncol 2025; 42:120. [PMID: 40106063 DOI: 10.1007/s12032-025-02657-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: 12/13/2024] [Accepted: 03/03/2025] [Indexed: 03/22/2025]
Abstract
Osteosarcoma is a highly invasive and metastatic primary malignant bone tumor, and resistance to chemotherapy remains a major therapeutic challenge. Our previous studies showed that increased Forkhead box protein M1 (FoxM1) expression promotes osteosarcoma progression. While NIMA-related kinase 2 (NEK2) has emerged as a potential oncogenic factor, its functional role and molecular mechanisms in osteosarcoma remain poorly understood. Pearson's correlation analysis was performed to assess the relationship between FoxM1 and NEK2 expression using the GSE33382 dataset from GEO. Coimmunoprecipitation (Co-IP) was employed to investigate FoxM1-NEK2 interactions. NEK2 expression was modulated in the HOS and U2OS osteosarcoma cell lines through pharmacological inhibition (MBM-55), siRNA-mediated knockdown, and plasmid-mediated overexpression. Cellular proliferation was evaluated via CCK-8 and colony formation assays. Transwell migration/invasion assays and flow cytometry were performed to assess the metastatic potential and apoptosis, respectively. The protein levels of FoxM1, NEK2, and AKT/p-AKT were analyzed by Western blotting. Western blot analyses of FoxM1-overexpressing cell lines and RCM-1-treated cells revealed a positive correlation between NEK2 and FoxM1 levels. Co-IP confirmed their interaction. NEK2 knockdown significantly suppressed proliferation, migration, and invasion; enhanced cisplatin sensitivity (reduced the IC50); and promoted apoptosis. Conversely, NEK2 overexpression exacerbated malignant phenotypes and decreased chemosensitivity. Mechanistically, NEK2 activation was shown to drive osteosarcoma progression via AKT/p-AKT pathway activation. This study revealed that NEK2 promotes osteosarcoma proliferation, invasion, migration, and chemoresistance while inhibiting apoptosis, likely through AKT/p-AKT signaling. These effects may be regulated by FoxM1.
Collapse
Affiliation(s)
- Xin Tan
- Department of Pathology, School of Basic Medical Science, Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Xiaojing Liang
- Department of Pathology, School of Basic Medical Science, Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Yi Feng
- Department of Pathology, School of Basic Medical Science, Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Ming Xie
- Department of Pathology, School of Basic Medical Science, Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Kun Zhong
- Department of Pathology, School of Basic Medical Science, Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Wenwu Luo
- Department of Pathology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Yurao Wang
- The Second School of Clinical Medicine, Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China
| | - Yu Yin
- Department of Pathology, School of Basic Medical Science, Anhui Medical University, Hefei, 230032, People's Republic of China.
- Department of Pathology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China.
| | - Yongping Cai
- Department of Pathology, School of Basic Medical Science, Anhui Medical University, Hefei, 230032, People's Republic of China.
- Department of Pathology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China.
| |
Collapse
|
2
|
Liu X, Zhang X, Zeng T, Chen Y, Ye L, Wang S, Li Y. FOSL1 drives the malignant progression of pancreatic cancer cells by regulating cell stemness, metastasis and multidrug efflux system. J Transl Med 2025; 23:268. [PMID: 40038751 DOI: 10.1186/s12967-025-06304-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 02/23/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Targeted therapy is an effective strategy for the treatment of advanced and metastatic pancreatic cancer, one of the leading causes for cancer-related death worldwide. To address the limitations of existing targeted drugs, there is an urgently need to find novel targets and therapeutic strategies. Transcription factor FOS like 1 (FOSL1) is a potential therapeutic target for challenging pancreatic cancer, which contributes to the malignant progression and poor gnosis of pancreatic cancer. High mobility group A1 (HMGA1) is a nonhistone chromatin structural protein that contributes to malignant progression and poor prognosis of cancer. METHODS Human FOSL1 complete RNA, shRNA against FOSL1 and shRNA against HMGA1 lentiviral recombination vectors were used to overexpress FOSL1 and knock down FOSL1 and HMGA1. RNA sequencing, Q-PCR and Western blots were used to investigate the mechanism of FOSL1 in regulating the proliferation of pancreatic cancer cells. The relationship between FOSL1 and HMGA1 were analyzed by co-immunoprecipitation Mass spectrometry, Q-PCR of chromatin immunoprecipitation and Western blots. The regulation of FOSL1 and HMGA1 in the invasion and migration, stemness, and multidrug efflux system were determined by transwell assay, sphere formation assay, immunofluorescence, Q-PCR and Western blots. RESULTS We found that FOSL1 promoted the proliferation and progression of pancreatic cancer by trigging stemness, invasion and metastasis, and drug resistance. HMGA1 was a key downstream target regulated by FOSL1 at the transcriptional level and directly interacted with FOSL1. Knockdown of HMGA1 inhibited the proliferation of pancreatic cancer cells by regulating the expression of genes related to stemness, epithelial-mesenchymal transition and multidrug efflux system. Targeted inhibition of FOSL1 and HMGA1 expression significantly inhibited the proliferation of pancreatic cancer cells. CONCLUSION FOSL1 promote the malignant progression of pancreatic cancer by promoting HMGA1 expression. Targeting FOSL1 and HMGA1 in monotherapy or combination therapy is a promising strategy for the treatment of advanced and metastasis pancreatic cancer.
Collapse
Affiliation(s)
- Xiaolong Liu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, PR China
| | - Xueyan Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, PR China
| | - Tingyu Zeng
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, PR China
| | - Yali Chen
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, PR China
| | - Liu Ye
- Medical College of Guizhou University, Guiyang, 550025, Guizhou Province, China
| | - Shuping Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, PR China.
| | - Yulan Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, PR China.
| |
Collapse
|
3
|
Yang TL, Tsai CH, Su YW, Chang YC, Lee F, Huang TY, Li FY, Yang PS. Combining KPNA2 and FOXM1 Expression as Prognostic Markers and Therapeutic Targets in Hormone Receptor-Positive, HER2-Negative Breast Cancer. Cancers (Basel) 2025; 17:671. [PMID: 40002266 PMCID: PMC11853725 DOI: 10.3390/cancers17040671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Breast cancer remains the leading malignancy affecting women worldwide, with significant mortality rates. This study aimed to evaluate the prognostic significance of FOXM1 expression specifically in hormone receptor-positive, HER2-negative (HR+HER2-) breast cancer patients with high KPNA2 expression, and to identify potential FOXM1-targeted therapeutic strategies for this patient subgroup. Methods: We analyzed RNA sequencing and microarray data from three independent cohorts: Mackay Memorial Hospital patient samples, The Cancer Genome Atlas, and Gene Expression Omnibus databases. The expression levels of KPNA2, FOXM1, CCNB1, and CCNB2 were evaluated, with particular emphasis on stratifying patients based on KPNA2 expression levels. Their associations with clinical outcomes were assessed using Gene Set Enrichment Analysis and survival analyses. Results: While KPNA2 expression showed strong positive correlations with FOXM1, CCNB1, and CCNB2 across all datasets, our analysis revealed a distinct prognostic pattern in HR+HER2- breast cancer patients with high KPNA2 expressions. In this specific subgroup, low FOXM1 expression emerged as a favorable prognostic indicator, despite the generally poor prognosis associated with high KPNA2 levels. Gene Set Enrichment Analysis demonstrated significant enrichment of the G2/M checkpoint pathway in high KPNA2-expressing patients, suggesting potential therapeutic vulnerability to FOXM1 inhibition in this subgroup. Conclusions: This study establishes FOXM1 expression as a critical prognostic marker, specifically in KPNA2-high HR+HER2- breast cancer patients, where low FOXM1 levels correlate with improved survival outcomes. These findings suggest that FOXM1 inhibition could be particularly effective in patients with high KPNA2 expression, offering a novel therapeutic strategy for this specific molecular subtype. Several FOXM1 inhibitors, including thiostrepton and FDI-6, warrant investigation as potential targeted treatments for KPNA2-high HR+HER2- breast cancer patients.
Collapse
Affiliation(s)
- Tsen-Long Yang
- Department of General Surgery, Shin Kong Wu Ho-Su Memorial Hospital, Taipei 111045, Taiwan
| | - Chung-Hsin Tsai
- Department of General Surgery, MacKay Memorial Hospital, Taipei 104217, Taiwan
| | - Ying-Wen Su
- Department of Medical Oncology, MacKay Memorial Hospital, Taipei 104217, Taiwan
- Department of Medicine, Mackay Medical College, Taipei 252005, Taiwan
| | - Yuan-Ching Chang
- Department of General Surgery, MacKay Memorial Hospital, Taipei 104217, Taiwan
| | - Fang Lee
- Department of General Surgery, MacKay Memorial Hospital, Taipei 104217, Taiwan
| | - To-Yu Huang
- Department of Medical Research, MacKay Memorial Hospital, Taipei 251404, Taiwan
| | - Fang-Yi Li
- Department of Medical Research, MacKay Memorial Hospital, Taipei 251404, Taiwan
| | - Po-Sheng Yang
- Department of General Surgery, MacKay Memorial Hospital, Taipei 104217, Taiwan
- Department of Medicine, Mackay Medical College, Taipei 252005, Taiwan
| |
Collapse
|
4
|
Tang Z, Qian Y, Wang N, Chen Y, Huang H, Zhang J, Luo H, Lu Z, Li Z, He Z, Tang F. HPV-Associated Gene Signatures in Bladder Cancer: A Comprehensive Prognostic Model and its Implications in Immunotherapy. Int J Med Sci 2025; 22:140-157. [PMID: 39744172 PMCID: PMC11659835 DOI: 10.7150/ijms.98334] [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: 05/11/2024] [Accepted: 11/01/2024] [Indexed: 01/18/2025] Open
Abstract
Background: Evidence increasingly indicates that HPV infection plays a pivotal role in the initiation and progression of bladder cancer (BC). Yet, determining the predictive value of HPV-associated genes in BC remains challenging. Methods: We identified differentially expressed HPV-associated genes of BC patients from the TCGA and GEO databases. We screened prognostic genes using COX and LASSO regression, subsequently establishing a risk prediction model. The model's precision and clinical relevance were gauged using Kaplan-Meier survival analyses and ROC curves. Functional enrichment, immune cell infiltration, and drug sensitivity analyses were performed across both high-risk and low-risk sets. PCR assays were utilized to measure the expression levels of genes. Results: We identified 13 HPV-associated genes for our risk model. Among these, FLRT2, HOXC5, LDLR, SCD, GRM7, DSC1, EMP1, and HMGA1 were identified as risk contributors, while LPA, SERPINA6, ZNF124, ETV7, and SCO2 were deemed protective. Cox regression analysis verified that our model provides an independent prediction of overall survival (OS) in bladder cancer (BC) patients. Gene Ontology (GO) analysis revealed predominant gene enrichment in wound healing, extracellular matrix composition, and collagen-rich extracellular matrices. KEGG pathway analysis highlighted primary enrichment areas, including focal adhesion, the PI3K-Akt signalling pathway, and ECM-receptor interaction. Risk scores were correlated with tumor microenvironment (TME) scores, immune cell infiltration, and sensitivities to both chemotherapy and immunotherapy. Conclusion: We have formulated a risk-assessment model pinpointing 13 central HPV-associated genes in BC. These genes present potential as prognostic indicators and therapeutic targets, emphasizing the intertwined relationship between HPV-induced BC progression and the immune landscape.
Collapse
Affiliation(s)
- Zhicheng Tang
- Department of Urology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518033, China
| | - Yuxin Qian
- Department of Urology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518033, China
| | - Ni Wang
- School of Medicine, Sun Yat-sen University, Shenzhen 518107, China
| | - Yinqiu Chen
- School of Medicine, Sun Yat-sen University, Shenzhen 518107, China
| | - Haojun Huang
- School of Medicine, Sun Yat-sen University, Shenzhen 518107, China
| | - Jiahao Zhang
- Department of Urology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518033, China
| | - Hongcheng Luo
- Department of Urology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518033, China
| | - Zechao Lu
- Department of Urology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518033, China
| | - Zhibiao Li
- Department of Urology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518033, China
| | - Zhaohui He
- Department of Urology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518033, China
| | - Fucai Tang
- Department of Urology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518033, China
| |
Collapse
|
5
|
Jeon Y, Choi KC, Park YN, Kim YJ. Identification of Molecular Subtypes and Prognostic Traits Based on Chromosomal Instability Phenotype-Related Genes in Lung Adenocarcinoma. Cancers (Basel) 2024; 16:3818. [PMID: 39594773 PMCID: PMC11592823 DOI: 10.3390/cancers16223818] [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: 10/07/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Lung adenocarcinoma (LUAD) exhibits significant molecular heterogeneity; however, previous studies have not fully explored its classification into distinct molecular subtypes. Here, we identified LUAD-significant chromosomal instability (CIN) phenotype genes (n = 24) using a TCGA-LUAD cohort (n = 592) and evaluated their ability to predict pathologic grade. Unsupervised clustering and principal component analysis revealed that LUAD patients could be classified into CIN phenotype-related subtypes (GroupLow, GroupModerate, and GroupHigh), each exhibiting distinct transcriptomic patterns. Notably, the GroupHigh showed significantly poor overall survival [OS; hazard ratio (HR) = 1.43, p-value < 10-3] and disease-free survival (DFS; HR = 1.27, p-value < 10-3). Univariate and multivariate analysis confirmed that its expression status was an independent prognostic predictor (p-value < 10-3, HR = 2.18, 95% C.I = 1.26-3.76) of the clinical outcomes, outperforming pathologic grade (p-value < 10-3, HR = 1.2, 95% C.I = 1.08-1.33). Moreover, analysis of surfactant metabolism-related genes revealed higher expression in the GroupLow, which was associated with a favorable prognosis. By integrating multiple independent cohorts (n = 779), we validated these findings and confirmed that CIN phenotype gene status serves as a critical prognostic marker in LUAD. Furthermore, genomic profiling showed that the GroupHigh exhibited frequent mutations in key genes such as KEAP1, LYST, SETD2, and TP53, with oncogenes in this group preferentially showing copy number gains. Our study highlights the significance of CIN phenotype gene status as a predictor of LUAD prognosis and its association with transcriptomic and genomic alterations, paving the way for further clinical validation and potential therapeutic interventions.
Collapse
Affiliation(s)
- Youngsic Jeon
- Institute of Natural Products, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea;
| | - Kyung-Chul Choi
- Department of Biochemistry and Molecular Biology, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea;
| | - Young Nyun Park
- Department of Pathology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
| | - Young-Joo Kim
- Institute of Natural Products, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea;
| |
Collapse
|
6
|
Fu D, Zhang B, Fan W, Zeng F, Feng J, Wang X. Fatty acid metabolism prognostic signature predicts tumor immune microenvironment and immunotherapy, and identifies tumorigenic role of MOGAT2 in lung adenocarcinoma. Front Immunol 2024; 15:1456719. [PMID: 39478862 PMCID: PMC11521851 DOI: 10.3389/fimmu.2024.1456719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 10/01/2024] [Indexed: 11/02/2024] Open
Abstract
Background Aberrant fatty acid metabolism (FAM) plays a critical role in the tumorigenesis of human malignancies. However, studies on its impact in lung adenocarcinoma (LUAD) are limited. Methods We developed a prognostic signature comprising 10 FAM-related genes (GPR115, SOAT2, CDH17, MOGAT2, COL11A1, TCN1, LGR5, SLC34A2, RHOV, and DKK1) using data from LUAD patients in The Cancer Genome Atlas (TCGA). This signature was validated using six independent LUAD datasets from the Gene Expression Omnibus (GEO). Patients were classified into high- and low-risk groups, and overall survival (OS) was compared by Kaplan-Meier analysis. The signature's independence as a prognostic indicator was assessed after adjusting for clinicopathological features. Receiver operating characteristic (ROC) analysis validated the signature. Tumor immune microenvironment (TIME) was analyzed using ESTIMATE and multiple deconvolution algorithms. Functional assays, including CCK8, cell cycle, apoptosis, transwell, and wound healing assays, were performed on MOGAT2-silenced H1299 cells using CRISPR/Cas9 technology. Results Low-risk group patients exhibited decreased OS. The signature was an independent prognostic indicator and demonstrated strong risk-stratification utility for disease relapse/progression. ROC analysis confirmed the signature's validity across validation sets. TIME analysis revealed higher infiltration of CD8+ T cells, natural killers, and B cells, and lower tumor purity, stemness index, and tumor mutation burden (TMB) in low-risk patients. These patients also showed elevated T cell receptor richness and diversity, along with reduced immune cell senescence. High-risk patients exhibited enrichment in pathways related to resistance to immune checkpoint blockades, such as DNA repair, hypoxia, epithelial-mesenchymal transition, and the G2M checkpoint. LUAD patients receiving anti-PD-1 treatment had lower risk scores among responders compared to non-responders. MOGAT2 was expressed at higher levels in low-risk LUAD patients. Functional assays revealed that MOGAT2 knockdown in H1299 cells promoted proliferation and migration, induced G2 cell cycle arrest, and decreased apoptosis. Conclusions This FAM-related gene signature provides a valuable tool for prognostic stratification and monitoring of TIME and immunotherapy responses in LUAD. MOGAT2 is identified as a potential anti-tumor regulator, offering new insights into its role in LUAD pathogenesis.
Collapse
Affiliation(s)
- Denggang Fu
- College of Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Biyu Zhang
- Key Laboratory of Green Chemical Engineering Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, China
| | - Wenyan Fan
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, Jiangxi, China
| | - Fanfan Zeng
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, Jiangxi, China
| | - Jueping Feng
- Department of Oncology, Wuhan Fourth Hospital, Wuhan, Hubei, China
| | - Xin Wang
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, Jiangxi, China
| |
Collapse
|
7
|
Wu HY, Luo LF, Wei F, Jiang HM. Comprehensive clinicopathological significance and putative transcriptional mechanisms of Forkhead box M1 factor in hepatocellular carcinoma. World J Surg Oncol 2023; 21:366. [PMID: 38001498 PMCID: PMC10675979 DOI: 10.1186/s12957-023-03250-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND The Forkhead box M1 factor (FOXM1) is a crucial activator for cancer cell proliferation. While FOXM1 has been shown to promote hepatocellular carcinoma (HCC) progression, its transcriptional mechanisms remain incompletely understood. METHODS We performed an in-house tissue microarray on 313 HCC and 37 non-HCC tissue samples, followed by immunohistochemical staining. Gene chips and high throughput sequencing data were used to assess FOXM1 expression and prognosis. To identify candidate targets of FOXM1, we comprehensively reanalyzed 41 chromatin immunoprecipitation followed by sequencing (ChIP-seq) data sets. We predicted FOXM1 transcriptional targets in HCC by intersecting candidate FOXM1 targets with HCC overexpressed genes and FOXM1 correlation genes. Enrichment analysis was employed to address the potential mechanisms of FOXM1 underlying HCC. Finally, single-cell RNA sequencing analysis was performed to confirm the transcriptional activity of FOXM1 on its predicted targets. RESULTS This study, based on 4235 HCC tissue samples and 3461 non-HCC tissue samples, confirmed the upregulation of FOXM1 in HCC at mRNA and protein levels (standardized mean difference = 1.70 [1.42, 1.98]), making it the largest multi-centered study to do so. Among HCC patients, FOXM1 was increased in Asian and advanced subgroups, and high expression of FOXM1 had a strong ability to differentiate HCC tissue from non-HCC tissue (area under the curve = 0.94, sensitivity = 88.72%, specificity = 87.24%). FOXM1 was also shown to be an independent exposure risk factor for HCC, with a pooled hazard ratio of 2.00 [1.77, 2.26]. The predicted transcriptional targets of FOXM1 in HCC were predominantly enriched in nuclear division, chromosomal region, and catalytic activity acting on DNA. A gene cluster encoding nine transcriptional factors was predicted to be positively regulated by FOXM1, promoting the cell cycle signaling pathway in HCC. Finally, the transcriptional activity of FOXM1 and its targets was supported by single-cell analysis of HCC cells. CONCLUSIONS This study not only confirmed the upregulation of FOXM1 in HCC but also identified it as an independent risk factor. Moreover, our findings enriched our understanding of the complex transcriptional mechanisms underlying HCC pathogenesis, with FOXM1 potentially promoting HCC progression by activating other transcription factors within the cell cycle pathway.
Collapse
Affiliation(s)
- Hua-Yu Wu
- Department of Medical Experimental Center, The First People's Hospital of Nanning, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Li-Feng Luo
- Department of Pathology, The First People's Hospital of Nanning, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Fang Wei
- Department of Pathology, The First People's Hospital of Nanning, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Hong-Mian Jiang
- Department of Pathology, The First People's Hospital of Nanning, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China.
| |
Collapse
|