1
|
Borah K, Das HS, Budhathoki RK, Aurangzeb K, Mallik S. DOMSCNet: a deep learning model for the classification of stomach cancer using multi-layer omics data. Brief Bioinform 2025; 26:bbaf115. [PMID: 40178281 PMCID: PMC11966610 DOI: 10.1093/bib/bbaf115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/31/2025] [Accepted: 02/20/2025] [Indexed: 04/05/2025] Open
Abstract
The rapid advancement of next-generation sequencing (NGS) technology and the expanding availability of NGS datasets have led to a significant surge in biomedical research. To better understand the molecular processes, underlying cancer and to support its development, diagnosis, prediction, and therapy; NGS data analysis is crucial. However, the NGS multi-layer omics high-dimensional dataset is highly complex. In recent times, some computational methods have been developed for cancer omics data interpretation. However, various existing methods face challenges in accounting for diverse types of cancer omics data and struggle to effectively extract informative features for the integrated identification of core units. To address these challenges, we proposed a hybrid feature selection (HFS) technique to detect optimal features from multi-layer omics datasets. Subsequently, this study proposes a novel hybrid deep recurrent neural network-based model DOMSCNet to classify stomach cancer. The proposed model was made generic for all four multi-layer omics datasets. To observe the robustness of the DOMSCNet model, the proposed model was validated with eight external datasets. Experimental results showed that the SelectKBest-maximum relevancy minimum redundancy-Boruta (SMB), HFS technique outperformed all other HFS techniques. Across four multi-layer omics datasets and validated datasets, the proposed DOMSCNet model outdid existing classifiers along with other proposed classifiers.
Collapse
Affiliation(s)
- Kasmika Borah
- Department of Computer Science and Information Technology, Cotton University, Hem Baruah Rd, Panbazar, Guwahati, Kamrup Metropolitan district, Assam 781001, India
| | - Himanish Shekhar Das
- Department of Computer Science and Information Technology, Cotton University, Hem Baruah Rd, Panbazar, Guwahati, Kamrup Metropolitan district, Assam 781001, India
| | - Ram Kaji Budhathoki
- Department of Electrical and Electronics Engineering, School of Engineering, Kathmandu University, Kavrepalanchok district, Dhulikhel 45200, Nepal
| | - Khursheed Aurangzeb
- Department of Computer Engineering, College of Computer and Information Sciences, King Saud University, P. O. Box 51178, Riyadh district, 11543, Saudi Arabia
| | - Saurav Mallik
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA 02115, United States
- Department of Pharmacology & Toxicology, University of Arizona, 1295 N Martin Ave, Pima district, Tucson, AZ 85721, United States
| |
Collapse
|
2
|
Wang L, He H, Zhai R, Gao R, Su M, Duan R, Tu Z, Huang R. Investigation of the mechanism by which FOXJ2 inhibits proliferation, metastasis and cell cycle progression of ovarian cancer cells through the PI3K/AKT signaling pathway. Eur J Med Res 2025; 30:152. [PMID: 40038842 PMCID: PMC11881463 DOI: 10.1186/s40001-025-02270-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/01/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND As one member of the Forkhead Box transcription factor, Forkhead Box J2 (FOXJ2) is involved in diverse cancers. At present, the specific role and mechanism of FOXJ2 in ovarian cancer (OC) have not been fully addressed, which allows us to fill the blank. MATERIALS AND METHODS Accordingly, the expression of FOXJ2 in OC cells and ovarian epithelial cells was quantified via real-time qPCR. Following the transfection, cell counting kit-8, Transwell, wound healing and flow cytometry assays were performed to measure the proliferation, metastasis, apoptosis and cell cycle of OC cells A2780 and HEY. Further, real-time qPCR and Western blotting were both employed for the quantification assays on the expression levels of FOXJ2 as well as phosphoinositide 3-kinase (PI3K) and protein kinase B (AKT) (in both unphosphorylated and phosphorylated forms). RESULTS Based on the results, FOXJ2 were highly-expressed in OC cells (P < 0.05). Silencing of FOXJ2 resulted in attenuated OC cell proliferation, reduced numbers of migrating and invading OC cells, decreased apoptotic capacity, and cell cycle arrest in G1/S phase (P < 0.05). In addition, the knockdown of FOXJ2 caused the downward trend on the phosphorylation level of both PI3K and AKT in OC cells (P < 0.05). CONCLUSION The silencing of FOXJ2 could repress the growth and metastasis potentials and cause the cell cycle G1/S arrest of OC cells in vitro, which was possibly achieved via targeting the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Liyuan Wang
- Reproductive Center, The First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Han He
- Department of Urology and Reproductive Oncology, The First People's Hospital of Foshan, Foshan, 528000, China
| | - Ruifang Zhai
- Gynecology Department, The First Hospital of Shanxi Medical University, Taiyuan, 03001, China
| | - Ruifan Gao
- Reproductive Center, The First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Min Su
- Reproductive Center, The First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Ruiyun Duan
- Reproductive Center, The First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Zengrong Tu
- Reproductive Center, The First Hospital of Shanxi Medical University, Taiyuan, 030001, China.
| | - Rong Huang
- Department of Urology and Reproductive Oncology, The First People's Hospital of Foshan, Foshan, 528000, China.
| |
Collapse
|
3
|
Zhou JW, Zhang YB, Huang ZY, Yuan YP, Jin J. Identification of differentially expressed mRNAs as novel predictive biomarkers for gastric cancer diagnosis and prognosis. World J Gastrointest Oncol 2024; 16:1947-1964. [PMID: 38764850 PMCID: PMC11099425 DOI: 10.4251/wjgo.v16.i5.1947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/04/2024] [Accepted: 03/14/2024] [Indexed: 05/09/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) has a high mortality rate worldwide. Despite significant progress in GC diagnosis and treatment, the prognosis for affected patients still remains unfavorable. AIM To identify important candidate genes related to the development of GC and identify potential pathogenic mechanisms through comprehensive bioinformatics analysis. METHODS The Gene Expression Omnibus database was used to obtain the GSE183136 dataset, which includes a total of 135 GC samples. The limma package in R software was employed to identify differentially expressed genes (DEGs). Thereafter, enrichment analyses of Gene Ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were performed for the gene modules using the clusterProfile package in R software. The protein-protein interaction (PPI) networks of target genes were constructed using STRING and visualized by Cytoscape software. The common hub genes that emerged in the cohort of DEGs that was retrieved from the GEPIA database were then screened using a Venn Diagram. The expression levels of these overlapping genes in stomach adenocarcinoma samples and non-tumor samples and their association with prognosis in GC patients were also obtained from the GEPIA database and Kaplan-Meier curves. Moreover, real-time quantitative polymerase chain reaction (RT-qPCR) and western blotting were performed to determine the mRNA and protein levels of glutamic-pyruvic transaminase (GPT) in GC and normal immortalized cell lines. In addition, cell viability, cell cycle distribution, migration and invasion were evaluated by cell counting kit-8, flow cytometry and transwell assays. Furthermore, we also conducted a retrospective analysis on 70 GC patients diagnosed and surgically treated in Wenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, The Second Affiliated Hospital of Shanghai University between January 2017 to December 2020. The tumor and adjacent normal samples were collected from the patients to determine the potential association between the expression level of GPT and the clinical as well as pathological features of GC patients. RESULTS We selected 19214 genes from the GSE183136 dataset, among which there were 250 downregulated genes and 401 upregulated genes in the tumor samples of stage III-IV in comparison to those in tumor samples of stage I-II with a P-value < 0.05. In addition, GO and KEGG results revealed that the various upregulated DEGs were mainly enriched in plasma membrane and neuroactive ligand-receptor interaction, whereas the downregulated DEGs were primarily enriched in cytosol and pancreatic secretion, vascular smooth muscle contraction and biosynthesis of the different cofactors. Furthermore, PPI networks were constructed based on the various upregulated and downregulated genes, and there were a total 15 upregulated and 10 downregulated hub genes. After a comprehensive analysis, several hub genes, including runt-related transcription factor 2 (RUNX2), salmonella pathogenicity island 1 (SPI1), lysyl oxidase (LOX), fibrillin 1 (FBN1) and GPT, displayed prognostic values. Interestingly, it was observed that GPT was downregulated in GC cells and its upregulation could suppress the malignant phenotypes of GC cells. Furthermore, the expression level of GPT was found to be associated with age, lymph node metastasis, pathological staging and distant metastasis (P < 0.05). CONCLUSION RUNX2, SPI1, LOX, FBN1 and GPT were identified key hub genes in GC by bioinformatics analysis. GPT was significantly associated with the prognosis of GC, and its upregulation can effectively inhibit the proliferative, migrative and invasive capabilities of GC cells.
Collapse
Affiliation(s)
- Jian-Wei Zhou
- Department of Gastroenterology, Wenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, The Second Affiliated Hospital of Shanghai University, Wenzhou 325000, Zhejiang Province, China
| | - Yi-Bing Zhang
- Department of Gastroenterology, Wenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, The Second Affiliated Hospital of Shanghai University, Wenzhou 325000, Zhejiang Province, China
| | - Zhi-Yang Huang
- Department of Gastroenterology, Wenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, The Second Affiliated Hospital of Shanghai University, Wenzhou 325000, Zhejiang Province, China
| | - Yu-Ping Yuan
- Department of Gastroenterology, Wenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, The Second Affiliated Hospital of Shanghai University, Wenzhou 325000, Zhejiang Province, China
| | - Jie Jin
- Department of Gastroenterology, Wenzhou Central Hospital, Dingli Clinical College of Wenzhou Medical University, The Second Affiliated Hospital of Shanghai University, Wenzhou 325000, Zhejiang Province, China
| |
Collapse
|
4
|
Chen X, Wang L, Yang M, Zhao W, Tu J, Liu B, Yuan X. RUNX transcription factors: biological functions and implications in cancer. Clin Exp Med 2024; 24:50. [PMID: 38430423 PMCID: PMC10908630 DOI: 10.1007/s10238-023-01281-0] [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: 09/30/2023] [Accepted: 11/10/2023] [Indexed: 03/03/2024]
Abstract
Runt-related transcription factors (RUNX) are a family of transcription factors that are essential for normal and malignant hematopoietic processes. Their most widely recognized role in malignancy is to promote the occurrence and development of acute myeloid leukemia. However, it is worth noting that during the last decade, studies of RUNX proteins in solid tumors have made considerable progress, suggesting that these proteins are directly involved in different stages of tumor development, including tumor initiation, progression, and invasion. RUNX proteins also play a role in tumor angiogenesis, the maintenance of tumor cell stemness, and resistance to antitumor drugs. These findings have led to the consideration of RUNX as a tumor biomarker. All RUNX proteins are involved in the occurrence and development of solid tumors, but the role of each RUNX protein in different tumors and the major signaling pathways involved are complicated by tumor heterogeneity and the interacting tumor microenvironment. Understanding how the dysregulation of RUNX in tumors affects normal biological processes is important to elucidate the molecular mechanisms by which RUNX affects malignant tumors.
Collapse
Affiliation(s)
- Xinyi Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jie Fang Road 1095, Wuhan, Hubei Province, China
| | - Lu Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jie Fang Road 1095, Wuhan, Hubei Province, China
| | - Mu Yang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jie Fang Road 1095, Wuhan, Hubei Province, China
| | - Weiheng Zhao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jie Fang Road 1095, Wuhan, Hubei Province, China
| | - Jingyao Tu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jie Fang Road 1095, Wuhan, Hubei Province, China.
| | - Bo Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jie Fang Road 1095, Wuhan, Hubei Province, China.
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jie Fang Road 1095, Wuhan, Hubei Province, China.
| |
Collapse
|
5
|
Toner J, Gordon JAR, Greenyer H, Kaufman P, Stein JL, Stein GS, Lian JB. RUNX2 as a Prognostic Factor in Human Cancers. Crit Rev Eukaryot Gene Expr 2024; 34:51-66. [PMID: 39072409 DOI: 10.1615/critreveukaryotgeneexpr.2024054162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
The RUNX2 transcription factor was discovered as an essential transcriptional regulator for commitment to osteoblast lineage cells and bone formation. Expression of RUNX2 in other tissues, such as breast, prostate, and lung, has been linked to oncogenesis, cancer progression, and metastasis. In this study, we sought to determine the extent of RUNX2 involvement in other tumors using a pan-cancer analysis strategy. We correlated RUNX2 expression and clinical-pathological parameters in human cancers by interrogating publicly available multiparameter clinical data. Our analysis demonstrated that altered RUNX2 expression or function is associated with several cancer types from different tissues. We identified three tumor types associated with increased RUNX2 expression and four other tumor types associated with decreased RUNX2 expression. Our pan-cancer analysis for RUNX2 revealed numerous other discoveries for RUNX2 regulation of different cancers identified in each of the pan-cancer databases. Both up and down regulation of RUNX2 was observed during progression of specific types of cancers in promoting the distinct types of cancers.
Collapse
Affiliation(s)
- J Toner
- Department of Biochemistry, University of Vermont, Larner College of Medicine, Burlington, VT, 05405, USA
| | - Johnathan A R Gordon
- Department of Biochemistry, University of Vermont, Burlington, Vermont, USA; University of Vermont Cancer Center, Burlington, Vermont, USA
| | - H Greenyer
- Department of Biochemistry, University of Vermont, Larner College of Medicine, Burlington, VT, 05405, USA
| | - Peter Kaufman
- Hematology/Oncology Division, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Janet L Stein
- Department of Biochemistry, University of Vermont Larner College of Medicine, Burlington, VT 05405; University of Vermont Cancer Center, University of Vermont Larner College of Medicine, Burlington, VT 05405
| | - Gary S Stein
- Department of Biochemistry, University of Vermont Larner College of Medicine, Burlington, VT 05405; University of Vermont Cancer Center, University of Vermont Larner College of Medicine, Burlington, VT 05405
| | - Jane B Lian
- Department of Biochemistry, University of Vermont Larner College of Medicine, Burlington, VT 05405; University of Vermont Cancer Center, University of Vermont Larner College of Medicine, Burlington, VT 05405
| |
Collapse
|