1
|
Huang Y, Min G, Wang H, Jiang L. A Prognostic Model for Senescence-Related LncRNA in a Novel Colon Adenocarcinoma Based on WGCNA and LASSO Regression. Biomedicines 2025; 13:1088. [PMID: 40426916 PMCID: PMC12109385 DOI: 10.3390/biomedicines13051088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/12/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025] Open
Abstract
Objective: This study aims to develop a prognostic model based on senescence-related long non-coding RNAs (lncRNAs) to predict the prognosis of patients with colon cancer and enhance their survival rates. Method: Differential expression analysis and Pearson correlation were employed to identify senescence-related lncRNAs in colon cancer. A risk prognosis model was constructed using univariate Cox regression analysis and Least Absolute Shrinkage and Selection Operator (LASSO) regression analysis. The reliability of this model was validated through survival analysis, receiver operating characteristic (ROC) curves, bar charts, and calibration curves. Additionally, the relationship between the prognostic model, immune microenvironment, and drug sensitivity was explored. Results: A risk prognosis model comprising eight senescence-related lncRNAs (LINC02257, AL138921.1, ATP2B1-AS1, AC005332.7, AC007728.3, AC018755.4, AL390719.3, and THCAT158) was successfully established, demonstrating strong performance in predicting the overall survival rates of colon cancer patients (AUC = 0.733). A significant correlation was observed between the senescence-related lncRNA prognostic model and the tumor microenvironment, immune cell infiltration, and drug sensitivity (p < 0.05). Conclusions: The senescence-related lncRNA prognostic model developed in this work can accurately forecast the prognosis of colon cancer patients, offering new insights for personalized treatment approaches in colon cancer.
Collapse
Affiliation(s)
- Yichu Huang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, China
| | - Guangtao Min
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China; (G.M.)
| | - Hongpeng Wang
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China; (G.M.)
| | - Lei Jiang
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China; (G.M.)
| |
Collapse
|
2
|
Wang T, Chen Z, Wang W, Wang H, Li S. Single-cell and spatial transcriptomic analysis reveals tumor cell heterogeneity and underlying molecular program in colorectal cancer. Front Immunol 2025; 16:1556386. [PMID: 40145096 PMCID: PMC11936967 DOI: 10.3389/fimmu.2025.1556386] [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: 01/06/2025] [Accepted: 02/24/2025] [Indexed: 03/28/2025] Open
Abstract
Background Colorectal cancer (CRC) is a highly heterogeneous tumor, with significant variation in malignant cells, posing challenges for treatment and prognosis. However, this heterogeneity offers opportunities for personalized therapy. Methods The consensus non-negative matrix factorization algorithm was employed to analyze single-cell transcriptomic data from CRC, which helped identify malignant cell expression programs (MCEPs). Subsequently, a crosstalk network linking MCEPs with immune/stromal cell trajectory development was constructed using Monocle3 and NicheNet. Additionally, bulk RNA-seq data were utilized to systematically explore the relationships between MCEPs, clinical features, and genetic mutations. A prognostic model was then established through Lasso and Cox regression analyses, integrating clinical data into a nomogram for personalized risk prediction. Furthermore, key genes associated with MCEPs and their potential therapeutic targets were identified using protein-protein interaction networks, followed by molecular docking to predict drug-binding affinity. Results We classified CRC malignant cell transcriptional states into eight distinct MCEPs and successfully constructed crosstalk networks between these MCEPs and immune or stromal cells. A prognostic model containing 15 genes was developed, demonstrating an AUC greater than 0.8 for prognostic evaluation over 1 to 10 years when combined with clinical features. A key drug target gene TIMP1 was identified, and several potential targeted drugs were discovered. Conclusion This study demonstrated that characterization of the malignant cell transcriptional programs could effectively reveal the biological features of highly heterogeneous tumors like CRC and exhibit significant potential in tumor prognosis assessment. Our research provides new theoretical and practical directions for CRC prognosis and targeted therapy.
Collapse
Affiliation(s)
- Teng Wang
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Zhaoming Chen
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Wang Wang
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing Medical University, Chongqing, China
| | - Heng Wang
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Shenglong Li
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| |
Collapse
|
3
|
Wang M, Li W, Zhou F, Wang Z, Jia X, Han X. A nicotinamide metabolism-related gene signature for predicting immunotherapy response and prognosis in lung adenocarcinoma patients. PeerJ 2025; 13:e18991. [PMID: 40034678 PMCID: PMC11874940 DOI: 10.7717/peerj.18991] [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: 11/07/2024] [Accepted: 01/23/2025] [Indexed: 03/05/2025] Open
Abstract
Background Nicotinamide (NAM) metabolism fulfills crucial functions in tumor progression. The present study aims to establish a NAM metabolism-correlated gene (NMRG) signature to assess the immunotherapy response and prognosis of lung adenocarcinoma (LUAD). Methods The training set and validation set (the GSE31210 dataset) were collected The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO), respectively. Molecular subtypes of LUAD were classified by consensus clustering. Mutation landscape of the top 20 somatic genes was visualized by maftools package. Subsequently, differential expression analysis was conducted using the limma package, and univariate, multivariate and LASSO regression analyses were performed on the screened genes to construct a risk model for LUAD. Next, the MCP-counter, TIMER and ESTIMATE algorithms were utilized to comprehensively assess the immune microenvironmental profile of LUAD patients in different risk groups. The efficacy of immunotherapy and chemotherapy drugs was evaluated by TIDE score and pRRophetic package. A nomogram was created by integrating RiskScore and clinical features. The mRNA expressions of independent prognostic NMRGs and the migration and invasion of LUAD cells were measured by carrying out cellular assays. Results Two subtypes (C1 and C2) of LUAD were classified, with C1 subtype showing a worse prognosis than C2. The top three genes with a high mutation frequency in C1 and C2 subtypes were TTN (45.25%), FLG (25.25%), and ZNF536 (19.8%). Four independent prognostic NMRGs (GJB3, CPA3, DKK1, KRT6A) were screened and used to construct a RiskScore model, which exhibited a strong predictive performance. High-risk group showed low immune cell infiltration, high TIDE score, and worse prognosis, and the patients in this group exhibited a high drug sensitivity to Cisplatin, Erlotinib, Paclitaxel, Saracatini, and CGP_082996. A nomogram was established with an accurate predictive and diagnostic performance. GJB3, DKK1, CPA3, and KRT6A were all high- expressed in LUAD cells, and silencing GJB3 inhibited the migration and invasion of LUAD cells. Conclusion A novel NMRG signature was developed, contributing to the prognostic evaluation and personalized treatment for LUAD patients.
Collapse
Affiliation(s)
- Meng Wang
- Department of Thoracic Surgery, Tianjin Chest Hospital, Tianjin, China
- Chest Hospital, Tianjin University, Tianjin, China
| | - Wei Li
- Department of Thoracic Surgery, Tianjin Chest Hospital, Tianjin, China
- Chest Hospital, Tianjin University, Tianjin, China
| | - Fang Zhou
- Department of Thoracic Surgery, Tianjin Chest Hospital, Tianjin, China
- Chest Hospital, Tianjin University, Tianjin, China
| | - Zheng Wang
- Department of Thoracic Surgery, Tianjin Chest Hospital, Tianjin, China
- Chest Hospital, Tianjin University, Tianjin, China
| | - Xiaoteng Jia
- Clinical School of Thoracic, Tianjin Medical University, Tianjin, China
| | - Xingpeng Han
- Department of Thoracic Surgery, Tianjin Chest Hospital, Tianjin, China
- Chest Hospital, Tianjin University, Tianjin, China
| |
Collapse
|
4
|
Jiang Q, Chen Q, Sun Q, Liu D, Zhu J, Mao W. NADPH oxidase activator 1 (NOXA1) suppresses ferroptosis and radiosensitization in colorectal cancer. Int J Med Sci 2025; 22:1301-1312. [PMID: 40084254 PMCID: PMC11898851 DOI: 10.7150/ijms.107038] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/05/2025] [Indexed: 03/16/2025] Open
Abstract
Radiotherapy is one of the main treatments for colorectal cancer (CRC), but due to the intrinsic resistance of cells or resistance caused by long-term radiotherapy, the effectiveness of this treatment is limited for some CRC patients. Consequently, identifying novel sensitization strategies is essential. This study identifies Noxa1 as a marker linked to radiotherapy resistance in CRC, suggesting its potential as a prognostic biomarker for patients with CRC. The study found that Noxa1 was significantly overexpressed in radiotherapy-resistant colorectal cancer patients, correlating with a poor prognosis. Additionally, we discovered that the high expression of Noxa1 was negatively correlated with ferroptosis and primarily played a role through the glutathione metabolic pathway, as indicated by GSVA analysis. Experimental data indicated that the expression levels of NOXA1, SLC7A11, and GPX4 were significantly elevated in CRC cell lines resistant to radiotherapy. The expression of SLC7A11 and GPX4 decreased after the knockdown of Noxa1, leading to an increase in cellular ROS levels, which induced ferroptosis and sensitized the cells to radiotherapy. Therefore, Noxa1 might influence the radiotherapy sensitivity of CRC via regulating ferroptosis. Targeting Noxa1 could enhance radiotherapy sensitization and improve the prognosis of CRC patients.
Collapse
Affiliation(s)
- Qingyu Jiang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou 310000, Zhejiang, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310000, China
| | - Qianping Chen
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou 310000, Zhejiang, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310000, China
| | - Quanquan Sun
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou 310000, Zhejiang, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310000, China
| | - Dong Liu
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou 310000, Zhejiang, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310000, China
| | - Ji Zhu
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou 310000, Zhejiang, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310000, China
| | - Wei Mao
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou 310000, Zhejiang, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310000, China
| |
Collapse
|
5
|
Napoli M, Bauer J, Bonod C, Vadon-Le Goff S, Moali C. PCPE-2 (procollagen C-proteinase enhancer-2): The non-identical twin of PCPE-1. Matrix Biol 2024; 134:59-78. [PMID: 39251075 DOI: 10.1016/j.matbio.2024.09.001] [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: 07/29/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/11/2024]
Abstract
PCPE-2 was discovered at the beginning of this century, and was soon identified as a close homolog of PCPE-1 (procollagen C-proteinase enhancer 1). After the demonstration that it could also stimulate the proteolytic maturation of fibrillar procollagens by BMP-1/tolloid-like proteinases (BTPs), PCPE-2 did not attract much attention as it was thought to fulfill the same functions as PCPE-1 which was already well-described. However, the tissue distribution of PCPE-2 shows both common points and significant differences with PCPE-1, suggesting that their activities are not fully overlapping. Also, the recently established connections between PCPE-2 (gene name PCOLCE2) and several important diseases such as atherosclerosis, inflammatory diseases and cancer have highlighted the need for a thorough reappraisal of the in vivo roles of this regulatory protein. In this context, the recent finding that, while retaining the ability to bind fibrillar procollagens and to activate their C-terminal maturation, PCPE-2 can also bind BTPs and inhibit their activity has substantially extended its potential functions. In this review, we describe the current knowledge about PCPE-2 with a focus on collagen fibrillogenesis, lipid metabolism and inflammation, and discuss how we could further advance our understanding of PCPE-2-dependent biological processes.
Collapse
Affiliation(s)
- Manon Napoli
- Universite Claude Bernard Lyon 1, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367 Lyon, France
| | - Julien Bauer
- Universite Claude Bernard Lyon 1, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367 Lyon, France
| | - Christelle Bonod
- Universite Claude Bernard Lyon 1, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367 Lyon, France
| | - Sandrine Vadon-Le Goff
- Universite Claude Bernard Lyon 1, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367 Lyon, France
| | - Catherine Moali
- Universite Claude Bernard Lyon 1, CNRS UMR 5305, Tissue Biology and Therapeutic Engineering Laboratory (LBTI), 69367 Lyon, France.
| |
Collapse
|
6
|
Thomas MJ, Xu H, Wang A, Beg MA, Sorci-Thomas MG. PCPE2: Expression of multifunctional extracellular glycoprotein associated with diverse cellular functions. J Lipid Res 2024; 65:100664. [PMID: 39374805 PMCID: PMC11567036 DOI: 10.1016/j.jlr.2024.100664] [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: 03/31/2024] [Revised: 09/21/2024] [Accepted: 10/01/2024] [Indexed: 10/09/2024] Open
Abstract
Procollagen C-endopeptidase enhancer 2, known as PCPE2 or PCOC2 (gene name, PCOLCE2) is a glycoprotein that resides in the extracellular matrix, and is similar in domain organization to PCPE1/PCPE, PCOC1 (PCOLCE1/PCOLCE). Due to the many similarities between the two related proteins, PCPE2 has been assumed to have biological functions similar to PCPE. PCPE is a well-established enhancer of procollagen processing activating the enzyme, BMP-1. However, reports show that PCPE2 has a strikingly different tissue expression profile compared to PCPE. With that in mind and given the paucity of published studies on PCPE2, this review examines the current literature citing PCPE2 and its association with specific cell types and signaling pathways. Additionally, this review will present a brief history of PCPE2's discovery, highlighting structural and functional similarities and differences compared to PCPE. Considering the widespread use of RNA sequencing techniques to examine associations between cell-specific gene expression and disease states, we will show that PCPE2 is repeatedly found as a differentially regulated gene (DEG) significantly associated with a number of cellular processes, well beyond the scope of procollagen fibril processing.
Collapse
Affiliation(s)
- Michael J Thomas
- Division of Endocrinology and Molecular Medicine, Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA; Cardiovascular Research Center, Division of Endocrinology and Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Hao Xu
- Division of Endocrinology and Molecular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Angela Wang
- Division of Endocrinology and Molecular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Mirza Ahmar Beg
- Division of Endocrinology and Molecular Medicine, Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA; Cardiovascular Research Center, Division of Endocrinology and Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI, USA; Division of Endocrinology and Molecular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Mary G Sorci-Thomas
- Division of Endocrinology and Molecular Medicine, Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA; Cardiovascular Research Center, Division of Endocrinology and Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI, USA; Division of Endocrinology and Molecular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
7
|
Peng Y, Ouyang C, Wu Y, Ma R, Li H, Li Y, Jing J, Sun L. A novel PCDscore based on programmed cell death-related genes can effectively predict prognosis and therapy responses of colon adenocarcinoma. Comput Biol Med 2024; 170:107933. [PMID: 38217978 DOI: 10.1016/j.compbiomed.2024.107933] [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: 07/27/2023] [Revised: 12/06/2023] [Accepted: 01/01/2024] [Indexed: 01/15/2024]
Abstract
Emerging evidence suggests a correlation between oncogenesis and programmed cell death (PCD). However, comprehensive studies that incorporate all identified PCD-related genes to guide colon adenocarcinoma (COAD) prognosis and precision treatment strategies are lacking. In this study, a series of bioinformatics analyses were comprehensively conducted using data from the TCGA-COAD, GSE17538, and GSE39582 cohorts. A total of 21 PCD-associated prognostic genes were identified through univariate Cox analysis. LASSO and multivariate Cox methods were employed to establish a prognostic gene signature (ALOX12, HSPA1A, IL13, MID2, RFFL, and SLC39A8) and the corresponding scoring system, termed PCDscore, which exhibited robust predictive ability. The ssGSEA and ESTIMATE algorithms were utilized to evaluate the tumor microenvironment of COAD. The high PCDscore group demonstrated a poorer prognosis, characterized by lower CD4+ T cell infiltration and a higher stromal score. In contrast, the low PCDscore group exhibited sensitivity to common chemotherapy drugs such as Cisplatin and 5-Fluorouracil. Single-cell sequencing analysis further revealed that the high-PCDscore group displayed a lower proportion of CD4+ T cells. Colorectal cancer samples from the years 2013-2017 were employed to validate the PCDscore, while those from 2018 to 2019 served as a temporal external validation set for the PCDscore. In vitro experimental results indicated that the overexpression of SLC39A8 inhibited the proliferation and invasion of colorectal cancer cells. The study developed a novel PCDscore system based on the analysis of genes related to all identified PCD types, providing valuable insights into clinical prognosis and drug sensitivity for patients with COAD.
Collapse
Affiliation(s)
- Yangjie Peng
- Key Laboratory of Gastrointestinal Cancer Etiology and Prevention, Shenyang 110001, Liaoning, China; Department of Anorectal Surgery, The First Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Cheng Ouyang
- Key Laboratory of Gastrointestinal Cancer Etiology and Prevention, Shenyang 110001, Liaoning, China; Tumor Etiology and Screening Department of Cancer Institute, and Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, The First Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Yijun Wu
- Key Laboratory of Gastrointestinal Cancer Etiology and Prevention, Shenyang 110001, Liaoning, China; Tumor Etiology and Screening Department of Cancer Institute, and Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, The First Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Rui Ma
- Key Laboratory of Gastrointestinal Cancer Etiology and Prevention, Shenyang 110001, Liaoning, China; Tumor Etiology and Screening Department of Cancer Institute, and Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, The First Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Hao Li
- Department of Clinical Laboratory, The First Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Yanke Li
- Key Laboratory of Gastrointestinal Cancer Etiology and Prevention, Shenyang 110001, Liaoning, China; Department of Anorectal Surgery, The First Hospital of China Medical University, Shenyang 110001, Liaoning, China.
| | - Jingjing Jing
- Key Laboratory of Gastrointestinal Cancer Etiology and Prevention, Shenyang 110001, Liaoning, China; Tumor Etiology and Screening Department of Cancer Institute, and Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, The First Hospital of China Medical University, Shenyang 110001, Liaoning, China.
| | - Liping Sun
- Key Laboratory of Gastrointestinal Cancer Etiology and Prevention, Shenyang 110001, Liaoning, China; Tumor Etiology and Screening Department of Cancer Institute, and Key Laboratory of Cancer Etiology and Prevention in Liaoning Education Department, The First Hospital of China Medical University, Shenyang 110001, Liaoning, China.
| |
Collapse
|
8
|
Chai R, Zhao Y, Su Z, Liang W. Integrative analysis reveals a four-gene signature for predicting survival and immunotherapy response in colon cancer patients using bulk and single-cell RNA-seq data. Front Oncol 2023; 13:1277084. [PMID: 38023180 PMCID: PMC10644708 DOI: 10.3389/fonc.2023.1277084] [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: 08/14/2023] [Accepted: 09/25/2023] [Indexed: 12/01/2023] Open
Abstract
Background Colon cancer (CC) ranks as one of the leading causes of cancer-related mortality globally. Single-cell transcriptome sequencing (scRNA-seq) offers precise gene expression data for distinct cell types. This study aimed to utilize scRNA-seq and bulk transcriptome sequencing (bulk RNA-seq) data from CC samples to develop a novel prognostic model. Methods scRNA-seq data was downloaded from the GSE161277 database. R packages including "Seurat", "Harmony", and "singleR" were employed to categorize eight major cell types within normal and tumor tissues. By comparing tumor and normal samples, differentially expressed genes (DEGs) across these major cell types were identified. Gene Ontology (GO) enrichment analyses of DEGs for each cell type were conducted using "Metascape". DEGs-based signature construction involved Cox regression and least absolute shrinkage operator (LASSO) analyses, performed on The Cancer Genome Atlas (TCGA) training cohort. Validation occurred in the GSE39582 and GSE33382 datasets. The expression pattern of prognostic genes was verified using spatial transcriptome sequencing (ST-seq) data. Ultimately, an established prognostic nomogram based on the gene signature and age was established and calibrated. Sensitivity to chemotherapeutic drugs was predicted with the "oncoPredict" R package. Results Using scRNA-Seq data, we examined 33,213 cells, categorizing them into eight cell types within normal and tumor samples. GO enrichment analysis revealed various cancer-related pathways across DEGs in these cell types. Among the 55 DEGs identified via univariate Cox regression, four independent prognostic genes emerged: PTPN6, CXCL13, SPINK4, and NPDC1. Expression validation through ST-seq confirmed PTPN6 and CXCL13 predominance in immune cells, while SPINK4 and NPDC1 were relatively epithelial cell-specific. Creating a four-gene prognostic signature, Kaplan-Meier survival analyses emphasized higher risk scores correlating with unfavorable prognoses, confirmed across training and validation cohorts. The risk score emerged as an independent prognostic factor, supported by a reliable nomogram. Intriguingly, drug sensitivity analysis unveiled contrasting anti-cancer drug responses in the two risk groups, suggesting significant clinical implications. Conclusion We developed a novel prognostic four-gene risk model, and these genes may act as potential therapeutic targets for CC.
Collapse
Affiliation(s)
- Ruoyang Chai
- Department of General Practice, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yajie Zhao
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhengjia Su
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wei Liang
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|