1
|
Zhou W, Hu Z, Wu J, Liu Q, Jie Z, Sun H, Zhang W. Integrated analysis of single‑cell and bulk RNA sequencing data to construct a risk assessment model based on plasma cell immune‑related genes for predicting patient prognosis and therapeutic response in lung adenocarcinoma. Oncol Lett 2025; 29:271. [PMID: 40235679 PMCID: PMC11998079 DOI: 10.3892/ol.2025.15017] [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: 08/19/2024] [Accepted: 02/28/2025] [Indexed: 04/17/2025] Open
Abstract
Plasma cells serve a crucial role in the human immune system and are important in tumor progression. However, the specific role of plasma cell immune-related genes (PCIGs) in tumor progression remains unclear. Therefore, the present study aimed to establish a risk assessment model for patients with lung adenocarcinoma (LUAD) based on PCIGs. The data used in the present study were obtained from The Cancer Genome Atlas and the Gene Expression Omnibus databases. After identifying nine PCIGs, a risk assessment model was constructed and a nomogram was developed for predicting patient prognosis. To explore the molecular mechanism and clinical significance, gene set enrichment analysis (GSEA), tumor mutational burden (TMB) analysis, tumor microenvironment (TME) analysis and drug sensitivity prediction were performed. Furthermore, the accuracy of the model was validated using reverse transcription-quantitative PCR (RT-qPCR). The present study constructed a risk assessment model consisting of nine PCIGs. Kaplan-Meier survival curves indicated a worse prognosis in the high-risk subgroup (risk score ≥0.982) compared with that in the low-risk subgroup. The nomogram exhibited predictive value for survival prediction (area under the curve=0.727). GSEA enrichment analysis revealed enrichment of the focal adhesion and extracellular matrix-receptor interaction pathways in the high-risk group. Moreover, the high-risk group exhibited a higher TMB, as demonstrated by the TME analysis showing lower ESTIMATE scores. Drug sensitivity prediction facilitated potential drug selection. Subsequently, differential gene expression was validated in multiple LUAD cell lines using RT-qPCR. In conclusion, the risk assessment model based on nine PCIGs may be used to predict the prognosis and drug selection in patients with LUAD.
Collapse
Affiliation(s)
- Weijun Zhou
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhuozheng Hu
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jiajun Wu
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Qinghua Liu
- Department of Thoracic Surgery, Ganzhou People's Hospital, Ganzhou, Jiangxi 341099, P.R. China
| | - Zhangning Jie
- Department of Thoracic Surgery, Ganzhou People's Hospital, Ganzhou, Jiangxi 341099, P.R. China
| | - Hui Sun
- Department of Thoracic Surgery, Ganzhou People's Hospital, Ganzhou, Jiangxi 341099, P.R. China
| | - Wenxiong Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
2
|
Xiao D, Chen T, Yu X, Song Y, Liu Y, Yan W. The MYC/TXNIP axis mediates NCL-Suppressed CD8 +T cell immune response in lung adenocarcinoma. Mol Med 2025; 31:180. [PMID: 40346484 PMCID: PMC12063364 DOI: 10.1186/s10020-025-01224-3] [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: 10/11/2024] [Accepted: 04/22/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Lung adenocarcinoma is a deadly malignancy with immune evasion playing a key role in tumor progression. Glucose metabolism is crucial for T cell function, and the nucleolar protein NCL may influence T cell glucose metabolism. This study aims to investigate NCL's role in T cell glucose metabolism and immune evasion by lung adenocarcinoma cells. METHODS Utilizing single-cell RNA sequencing (scRNA-seq) data from the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA), we analyzed cell clustering, annotation, and prognosis. In vitro experiments involved manipulating NCL expression in CD8+ T cells to study immune function and glucose metabolism. In vivo studies using an orthotopic transplant mouse model monitored NCL's impact on CD8+ T cell glucose metabolism and anti-tumor immune function. RESULTS NCL was associated with T cell dysfunction and glucose metabolism. NCL silencing enhanced CD8+ T cell glucose metabolism, cytotoxicity, and infiltration, while NCL overexpression had the opposite effect. NCL overexpression relieved MYC-mediated transcriptional repression of TXNIP, reducing CD8+ T cell glucose metabolism. In vivo, NCL inhibited CD8+ T cell glucose metabolism through the MYC/TXNIP axis, hindering anti-tumor immune function. CONCLUSIONS NCL overexpression suppresses CD8+ T cell glucose metabolism and anti-tumor immune function, promoting lung adenocarcinoma progression via the MYC/TXNIP axis.
Collapse
Affiliation(s)
- Dan Xiao
- Department of Thoracic Oncology, Jiangxi Cancer Hospital&Institute, Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Key Laboratory of Oncology, No. 519 Beijing East Road, Nanchang, 330029, Jiangxi, China
| | - Tanxiu Chen
- Jiangxi Academy of Clinical Medical Sciences, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Xinlin Yu
- Department of Medical Laboratory, Jiangxi Cancer Hospital&Institute, Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Key Laboratory of Oncology, No. 519 Beijing East Road, 330029, Nanchang, China
| | - Ying Song
- Department of Medical Laboratory, Jiangxi Cancer Hospital&Institute, Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Key Laboratory of Oncology, No. 519 Beijing East Road, 330029, Nanchang, China
| | - Yigang Liu
- Department of Ultrasound Medicine, Jiangxi Cancer Hospital&Institute, Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Key Laboratory of Oncology, No. 519 Beijing East Road, Nanchang, 330029, Jiangxi, China.
| | - Wei Yan
- Department of Thoracic Oncology, Jiangxi Cancer Hospital&Institute, Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Key Laboratory of Oncology, No. 519 Beijing East Road, Nanchang, 330029, Jiangxi, China.
| |
Collapse
|
3
|
Jiang W, Jiang L, Zhao X, Liu Y, Sun H, Zhou X, Liu Y, Huang S. Bioinformatics Analysis Reveals HIST1H2BH as a Novel Diagnostic Biomarker for Atrial Fibrillation-Related Cardiogenic Thromboembolic Stroke. Mol Biotechnol 2025; 67:2111-2126. [PMID: 38825608 DOI: 10.1007/s12033-024-01187-6] [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: 10/27/2023] [Accepted: 04/29/2024] [Indexed: 06/04/2024]
Abstract
Atrial fibrillation (AF) is a significant precursor to cerebral embolism. Our study sought to unearth new diagnostic biomarkers for atrial fibrillation-related cerebral embolism (AF-CE) by meticulously examining multiple GEO datasets and meta-analysis. The gene expression omnibus (GEO) database provided RNA sequencing data associated with AF and stroke. We began by pinpointing genes with varied expressions in AF-CE patient blood samples. A meta-analysis was subsequently undertaken using several RNA sequencing datasets to verify these genes. LASSO regression discerned key genes for AF-CE, with their diagnostic prowess verified through ROC curve examination. Active signaling pathways within stroke patients were discerned via GO and KEGG enrichment, with PPI interactions detailing gene interplay. Differential gene analysis revealed an upregulation of sixteen genes and a downregulation of four in stroke patient blood samples. Eight genes showcased varied expression in the meta-analysis. LASSO regression zeroed in on five of these, culminating in HIST1H2BH's identification as a characteristic gene. HIST1H2BH's prowess in predicting AF-CE was confirmed through ROC. Integrin signaling, platelet activation, ECM interactions, and the PI3K-Akt pathway were found active in stroke victims. HIST1H2BH's interaction with the notably upregulated ITGA2B was spotlighted by PPI. Additionally, HIST1H2BH exhibited links with NK cells and eosinophils. HIST1H2BH emerges as an insightful diagnostic beacon for AF-CE. Its presence, post AF, potentially modulates pathways, accentuating platelet activation and consequent thrombus generation, leading to cerebral embolism.
Collapse
Affiliation(s)
- Wenbing Jiang
- Department of Cardiology, Wenzhou Integrated Traditional Chinese and Western Medicine Hospital, No.75 Jinxiu Road, Lucheng District, Wenzhou, 325000, Zhejiang Province, People's Republic of China.
| | - Lelin Jiang
- Second Clinical College of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Xiaoli Zhao
- Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Yiying Liu
- Postgraduate Training Base Allianceof Wenzhou Medical University (Wenzhou Central Hosptial), Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Huanghui Sun
- The Dingli Clinical College of Wenzhou Medical University, Heart Function Examination Room, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Xinlang Zhou
- Department of Cardiology, Wenzhou Integrated Traditional Chinese and Western Medicine Hospital, No.75 Jinxiu Road, Lucheng District, Wenzhou, 325000, Zhejiang Province, People's Republic of China
| | - Yin Liu
- Department of Cardiology, Wenzhou Integrated Traditional Chinese and Western Medicine Hospital, No.75 Jinxiu Road, Lucheng District, Wenzhou, 325000, Zhejiang Province, People's Republic of China
| | - Shu'se Huang
- Department of Cardiology, Wenzhou Integrated Traditional Chinese and Western Medicine Hospital, No.75 Jinxiu Road, Lucheng District, Wenzhou, 325000, Zhejiang Province, People's Republic of China
| |
Collapse
|
4
|
Hu Y, Huang J, Wang S, Sun X, Wang X, Yu H. Deciphering Autoimmune Diseases: Unveiling the Diagnostic, Therapeutic, and Prognostic Potential of Immune Repertoire Sequencing. Inflammation 2025; 48:676-695. [PMID: 38914737 DOI: 10.1007/s10753-024-02079-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/31/2024] [Accepted: 06/08/2024] [Indexed: 06/26/2024]
Abstract
Autoimmune diseases (AIDs) are immune system disorders where the body exhibits an immune response to its own antigens, causing damage to its own tissues and organs. The pathogenesis of AIDs is incompletely understood. However, recent advances in immune repertoire sequencing (IR-seq) technology have opened-up a new avenue to study the IR. These studies have revealed the prevalence in IR alterations, potentially inducing AIDs by disrupting immune tolerance and thereby contributing to our comprehension of AIDs. IR-seq harbors significant potential for the clinical diagnosis, personalized treatment, and prognosis of AIDs. This article reviews the application and progress of IR-seq in diseases, such as multiple sclerosis, systemic lupus erythematosus, rheumatoid arthritis, and type 1 diabetes, to enhance our understanding of the pathogenesis of AIDs and offer valuable references for the diagnosis and treatment of AIDs.
Collapse
Affiliation(s)
- Yuelin Hu
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China
| | - Jialing Huang
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China
| | - Shuqing Wang
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China
| | - Xin Sun
- School of Basic Medical Sciences, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China
| | - Xin Wang
- School of Basic Medical Sciences, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China
| | - Hongsong Yu
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China.
| |
Collapse
|
5
|
Fan Y, Yang J, Yang X, Xie Y, Li H, Yang S, Sun G, Ge G, Ding X, Lai S, Liao Y, Ji S, Yang R, Zhang X. Unveiling the power of Treg.Sig: a novel machine-learning derived signature for predicting ICI response in melanoma. Front Immunol 2025; 16:1508638. [PMID: 40226609 PMCID: PMC11985843 DOI: 10.3389/fimmu.2025.1508638] [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: 10/09/2024] [Accepted: 03/03/2025] [Indexed: 04/15/2025] Open
Abstract
Background Although immune checkpoint inhibitor (ICI) represents a significant breakthrough in cancer immunotherapy, only a few patients benefit from it. Given the critical role of Treg cells in ICI treatment resistance, we explored a Treg-associated signature in melanoma, which had never been elucidated yet. Methods A new Treg signature, Treg.Sig, was created using a computational framework guided by machine learning, utilizing transcriptome data from both single-cell RNA-sequencing (scRNA-seq) and bulk RNA-sequencing (bulk-seq). Among the 10 Treg.Sig genes, hub gene STAT1's function was further validated in ICI resistance in melanoma mice receiving anti-PD-1 treatment. Results Treg.Sig, based on machine learning, was able to forecast survival outcomes for melanoma across training dataset and external test dataset, and more importantly, showed superior predictive power than 51 previously established signatures. Analysis of the immune profile revealed that groups with high Treg.Sig levels exhibited immune-suppressive conditions, with inverse correlations observed between Treg.Sig and anti-cancer immune responses. Notably, among the 10 Treg.Sig genes, hub gene STAT1 mutation harbored lower response rate in ICIs-treated cohort. Mechanistically, STAT1 impinged on ICI resistances by modulating the phenotypic switch in N2 neutrophil polarization in melanoma mice receiving anti-PD-1 therapy, which affects overall survival. Conclusion The study developed a promising Treg.Sig signature that predicts ICI response of melanomas and could be used for selecting patients for immunotherapy. Meanwhile, our study potentially paves the way for overcoming immune resistance by targeting Treg-associated genes.
Collapse
Affiliation(s)
- Yunlong Fan
- Department of Dermatology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
- Chinese PLA Medical School, Beijing, China
| | - Jiaman Yang
- Zhujiang Hospital, Southern Medical University or The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Xin Yang
- Department of Dermatology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yulin Xie
- Zhujiang Hospital, Southern Medical University or The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Haiyang Li
- Chinese PLA Medical School, Beijing, China
| | - Shuo Yang
- Department of Spine Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | | | - Ge Ge
- Department of Dermatology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiao Ding
- Department of Dermatology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | | | - Yong Liao
- Department of Dermatology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | | | - Rongya Yang
- Department of Dermatology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
- Chinese PLA Medical School, Beijing, China
| | - Xingyue Zhang
- Department of Dermatology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
6
|
Li M, Zhu W, Lu Y, Shao Y, Xu F, Liu L, Zhao Q. Identification and validation of a CD4 + T cell-related prognostic model to predict immune responses in stage III-IV colorectal cancer. BMC Gastroenterol 2025; 25:153. [PMID: 40069612 PMCID: PMC11895157 DOI: 10.1186/s12876-025-03716-2] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 02/19/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND CD4+ T cells play an indispensable role in anti-tumor immunity and shaping tumor development. We sought to explore the characteristics of CD4+ T cell marker genes and construct a CD4+ T cell-related prognostic signature for stage III-IV colorectal cancer (CRC) patients. METHOD We combined scRNA and bulk-RNA sequencing to analyze stage III-IV CRC patients and identified the CD4+ T cell marker genes. Unsupervised cluster analysis was performed to divide patients into two clusters. The LASSO and multivariate Cox regression were performed to establish a prognostic-related signature. RT-qpcr and immunofluorescence staining were performed to examine the expression of ANXA2 in CRC tissue. RESULT We found a higher infiltration abundance of activated memory CD4+ T cells was associated with improved prognosis in stage III-IV CRC patients. Patients were divided into two subgroups with distinct clinical and immunological behaviors based on CD4+ T cell marker genes. And then a prognostic signature consisting of six CD4+ T cell marker genes was established, which stratified patients into high- and low-risk groups. Immune spectrum showed that the low-risk group had higher immune cell infiltration than the high-risk group. Furthermore, the risk score of this signature could predict the susceptibility of stage III-IV CRC patients to immune checkpoint inhibitors and chemotherapy drugs. Finally, we validated that ANXA2 was enriched in Tregs and was associated with infiltration of Tregs in CRC tumor microenvironment. CONCLUSION The CD4+ T cell-related prognostic signature established in the study can predict the prognosis and the response to immunotherapy in stage III-IV CRC patients. Our findings provide new insights for tumor immunotherapy of advanced CRC patients.
Collapse
Affiliation(s)
- Mengting Li
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Weining Zhu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Yuanyuan Lu
- Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
- Department of Gastroenterology, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, China
| | - Yu Shao
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China
| | - Fei Xu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China.
| | - Lan Liu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China.
| | - Qiu Zhao
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, China.
| |
Collapse
|
7
|
Kong Z, Zhou P, Xu J, Zhang Y, Wang Y. RFX2 downregulates RASSF1 expression and YAP phosphorylation through Hippo signaling to promote immune escape in lung adenocarcinoma. Cell Div 2025; 20:7. [PMID: 40069841 PMCID: PMC11895337 DOI: 10.1186/s13008-025-00147-z] [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: 10/16/2024] [Accepted: 02/27/2025] [Indexed: 03/15/2025] Open
Abstract
OBJECTIVE Regulatory Factor X (RFX) transcription factors have been implicated in different cancers. Ras association domain family (RASSF) has been shown clinical significance in lung cancer. This paper was to investigate the interaction of RFX2 and RASSF1 in lung adenocarcinoma (LUAD). METHODS The transcriptome differences of LUAD patients in GSE32863, GSE43458, and GSE21933 datasets were analyzed. A-549 and NCI-H358 cell lines after overexpression of RFX2 were co-cultured with activated CD8+ T cells, and the release of IFN-γ, GZMB, PRF1 by CD8+ T cells, and PD-L1 in the LUAD cells were detected. Cell viability, invasion, and apoptosis were analyzed by CCK-8, Transwell, and TUNEL assays. Dual-luciferase assay and ChIP were conducted to detect the interaction between RFX2 and RASSF1 promoter. An in vivo tumor model was constructed to monitor tumor growth. YAP protein levels and phosphorylation were measured. A-549 and NCI-H358 cells treated with DMSO or PY-60 after RFX2 overexpression were co-cultured with activated CD8+ T cells. RESULTS RFX2 was notably downregulated in LUAD. RFX2 overexpression increased infiltrating CD8+ T cells within transplanted tumors and inhibited immune escape, proliferation, and invasion of LUAD cells. RFX2 was enriched in the RASSF1 promoter, and RFX2 activated RASSF1 transcription by binding to the RASSF1 promoter. RASSF1 knockdown reversed the ability of RFX2 overexpression to inhibit immune escape. RFX2 depletion downregulated RASSF1, which reduced YAP phosphorylation, thus affecting the Hippo pathway to promote the immune escape. CONCLUSION RFX2 Loss in LUAD downregulates RASSF1 expression and YAP phosphorylation, thereby promoting immune escape through Hippo signaling.
Collapse
Affiliation(s)
- Zhenzhen Kong
- Department of Laboratory, Wujin Hospital Affiliated With Jiangsu University, No. 2 of Yongning North Road, Changzhou, 213002, Jiangsu, People's Republic of China
- The Wujin Clinical College of Xuzhou Medical University, Xuzhou, 221006, Jiangsu, People's Republic of China
| | - Ping Zhou
- Department of Medical Laboratory, Xuzhou Mining Group General Hospital, Xuzhou, 221011, Jiangsu, People's Republic of China
| | - Jiahao Xu
- Department of Laboratory, Wujin Hospital Affiliated With Jiangsu University, No. 2 of Yongning North Road, Changzhou, 213002, Jiangsu, People's Republic of China
- The Wujin Clinical College of Xuzhou Medical University, Xuzhou, 221006, Jiangsu, People's Republic of China
| | - Ying Zhang
- Department of Laboratory, Wujin Hospital Affiliated With Jiangsu University, No. 2 of Yongning North Road, Changzhou, 213002, Jiangsu, People's Republic of China
- The Wujin Clinical College of Xuzhou Medical University, Xuzhou, 221006, Jiangsu, People's Republic of China
| | - Yong Wang
- Department of Laboratory, Wujin Hospital Affiliated With Jiangsu University, No. 2 of Yongning North Road, Changzhou, 213002, Jiangsu, People's Republic of China.
- The Wujin Clinical College of Xuzhou Medical University, Xuzhou, 221006, Jiangsu, People's Republic of China.
| |
Collapse
|
8
|
Guo X, Deng Y, Jiang W, Li H, Luo Y, Zhang H, Wu H. Single cell transcriptomic analysis reveals tumor immune infiltration by macrophage cells gene signature in lung adenocarcinoma. Discov Oncol 2025; 16:261. [PMID: 40029500 DOI: 10.1007/s12672-025-01834-7] [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: 08/20/2024] [Accepted: 01/20/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Tumor-associated macrophages (TAMs) play pivotal roles in innate immunity and contribute to the advancement of lung cancer. We aimed to identify novel TAM-related biomarkers and significance of macrophage infiltration in lung adenocarcinoma (LUAD) through an integrative analysis of single-cell RNA-sequencing (scRNA-seq) data. To describe the cell atlas and construct a novel prognostic signature in LUAD. METHODS The gene signature linked to TAMs was identified utilizing Scanpy from the scRNA-seq dataset GSE131907. Subsequent analysis involved evaluating the expression levels of these genes, their potential molecular mechanisms, and prognostic significance in LUAD using data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. We also constructed a risk score models through LASSO Cox regression for these genes. The underlying mechanism was further elucidated through the application of GSEA, ESTIMATE, TIDE, and other bioinformatic algorithms. RESULTS Single-cell atlas was described by analyze 29 scRNA-seq samples from 19 LUAD patients. The TAMs-related gene signature (TGS) was identified as an independent prognostic factor by LASSO Cox regression analysis using differential expression genes (DEGs) derived from pro- and anti-inflammatory macrophage cells. Risk score model including nine TAMs-related genes (FOSL1, ZNF697, ADM, UBE2S, TICAM1, S100P, BIRC3, TLE1, and DEFB1) were obtained for prognosis construction. Moreover, the risk model underwent additional validation in four external GEO cohorts: GSE31210, GSE72094, GSE26939, and GSE30219. Interestingly, TGS-high tumors revealed enrichments in TGF-β signaling and hypoxia pathways, which shown low immune infiltration and immunosuppression by ESTIMATE and TIDE algorithm. The TGS-high risk group exhibited lower richness and diversity in the T-cell receptor (TCR) repertoire. CONCLUSION This study introduces a novel TGS score developed through LASSO Cox regression analysis, utilizing DEGs in pro- and anti-inflammatory macrophage cells. High TGS tumors exhibited enrichment in TGF-β signaling and hypoxia pathways, suggesting their potential utility in predicting prognosis and immune responses in patients with LUAD. These results offer promising implications for the development of therapeutic strategies for LUAD.
Collapse
Affiliation(s)
- Xiaotong Guo
- Department of Thoracic Surgery, Cancer Hospital Chinese Academy of Medical Sciences, Shenzhen Center Shenzhen Cancer Hospital, Shenzhen, China
| | - Youjun Deng
- Department of Thoracic Surgery, Cancer Hospital Chinese Academy of Medical Sciences, Shenzhen Center Shenzhen Cancer Hospital, Shenzhen, China
| | - Wenjun Jiang
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital, Chengdu, China
| | - Heng Li
- Department of Thoracic Surgery, Yunnan Hospital of Oncology, Kunming, China
| | - Yisheng Luo
- Department of Thoracic Surgery, Shenzhen Second People's Hospital, Shenzhen, China
| | - Huachuan Zhang
- Department of Thoracic Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China.
| | - Hao Wu
- Department of Thoracic Surgery, Shenzhen Second People's Hospital, Shenzhen, China.
| |
Collapse
|
9
|
Zhu W, Zhang Y, Yang L, Chen L, Chen C, Shi Q, Xu Z. Construction of a lung adenocarcinoma prognostic model based on KEAP1/NRF2/HO‑1 mutation‑mediated upregulated genes and bioinformatic analysis. Oncol Lett 2025; 29:155. [PMID: 39911153 PMCID: PMC11795234 DOI: 10.3892/ol.2025.14902] [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: 08/27/2024] [Accepted: 01/06/2025] [Indexed: 02/07/2025] Open
Abstract
Lung adenocarcinoma (LUAD) is a prevalent malignant tumor of the respiratory tract. The Kelch like ECH associated protein 1 (KEAP1)/nuclear factor erythroid 2-related factor 2 (NRF2)/heme oxygenase 1 (HO-1) axis serves a pivotal role in the occurrence and progression of LUAD. The present study aimed to identify specific genes regulated by mutations of the KEAP1/NRF2/HO-1 axis and to investigate their prognostic potential in LUAD, as well as their association with the tumor microenvironment. Immunohistochemistry was performed to assess the expression levels of KEAP1, NRF2 and HO-1 in LUAD tissues and to evaluate their association with clinical pathology. Sequencing data and clinical information were obtained from The Cancer Genome Atlas (TCGA)-LUAD and Gene Expression Omnibus (GSE68465) databases, whilst mutation information was sourced from the cBio Cancer Genomics Portal website. The R package 'limma' and Venn diagram were utilized to identify upregulated differentially expressed genes. Subsequently, a prognostic model was constructed using univariate Cox regression analysis and 101 machine learning methods. A nomogram of the prognostic model was generated to assess its efficacy in evaluating survival among patients with LUAD. The 'ImmuCellAI' and 'oncoPredict' R packages were used to compare and evaluate differences in immune cell infiltration and immunotherapy between high- and low-risk groups, as well as the sensitivity of LUAD to chemotherapy drugs. Compared with the group with negative expression, the results revealed that the group with positive expression of NRF2 and HO-1 exhibited advanced tumor, lymph node and clinical stages and a worse prognosis. A predictive model incorporating four genes (kynureninase, serpin family B member 5, insulin like 4 and γ-aminobutyric acid type A receptor subunit α3) was constructed based on KEAP1/NRF2/HO-1 mutation-mediated upregulated genes (KNHMUGs). Risk score was an independent prognostic factor for patients with LUAD (hazard ratio, 1.038; 95% confidence interval, 1.034-1.043; P<0.001). A nomogram was developed to predict the prognosis of patients with LUAD, which was validated as a reliable prognostic tool. The low-risk group exhibited higher immune cell infiltration, including CD4+ T, CD8+ T, natural killer (NK) and NKT cells, compared with the high-risk group. In addition, it demonstrated increased expression levels of immune checkpoint inhibitory genes such as cytotoxic T-lymphocyte associated protein 4, T cell immunoreceptor with Ig and ITIM domains, hepatitis A virus cellular receptor 2 and B and T lymphocyte associated protein. Moreover, it displayed enhanced sensitivity to immunotherapy. Drug sensitivity analysis revealed that the high-risk group exhibited increased sensitivity towards vinblastine, docetaxel and cisplatin, whereas the low-risk group showed increased sensitivity to BMS_754807, SB505124_1194 and JQ1_2172. In conclusion, a KNHMUGs-based gene signature was constructed in the present study, which holds promise as a biomarker for evaluating patient prognosis and guiding treatment by effectively assessing immunotherapy response and chemotherapy sensitivity in patients with LUAD.
Collapse
Affiliation(s)
- Wei Zhu
- Department of Pathology, Xishan People's Hospital of Wuxi City, Wuxi, Jiangsu 214105, P.R. China
| | - Ye Zhang
- Department of Pathology, Xishan People's Hospital of Wuxi City, Wuxi, Jiangsu 214105, P.R. China
| | - Lingyun Yang
- Department of Renal and Rheumatology, Affiliated Children's Hospital of Jiangnan University (Wuxi Children's Hospital), Wuxi, Jiangsu 214000, P.R. China
| | - Lu Chen
- Department of Pathology, Xishan People's Hospital of Wuxi City, Wuxi, Jiangsu 214105, P.R. China
| | - Chaobo Chen
- Department of General Surgery, Xishan People's Hospital of Wuxi City, Wuxi, Jiangsu 214105, P.R. China
- Department of Hepatobiliary and Transplantation Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210000, P.R. China
| | - Qifeng Shi
- Department of Pathology, Xishan People's Hospital of Wuxi City, Wuxi, Jiangsu 214105, P.R. China
| | - Zipeng Xu
- Department of General Surgery, Xishan People's Hospital of Wuxi City, Wuxi, Jiangsu 214105, P.R. China
| |
Collapse
|
10
|
Yu P, Xiao L, Hu K, Ling J, Chen Y, Liang R, Liu X, Zhang D, Liu Y, Weng T, Jiang H, Zhang J, Wang W. Comprehensive exploration of programmed cell death landscape in lung adenocarcinoma combining multi-omic analysis and experimental verification. Sci Rep 2025; 15:5364. [PMID: 39948103 PMCID: PMC11825851 DOI: 10.1038/s41598-025-87982-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: 11/02/2024] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
The mortality and therapeutic failure in lung adenocarcinoma (LUAD) are mainly resulted from the wide metastasis and chemotherapy resistance. Up to now, accurate and stable predictive prognostic indicator for revealing the progress and novel therapeutic strategies of LUAD is infrequent, nonetheless. Diversified programmed cell death (PCD) has been widely confirmed that participated in the occurrence and development of various malignant tumors, respectively. In this research, we integrated fourteen types of PCD, bulk multi-omic data from TCGA-LUAD and other cohorts in gene expression omnibus (GEO) and clinical LUAD patients to develop our analysis. Consequently, pivotal fourteen PCD genes, especially CAMP, CDK5R1, CTSW, DAPK2, GAB2, GAPDH, GATA2, HGF, MAPT, NAPSA, NUPR1, PIK3CG, PLA2G3, and SLC7A11, were utilized to establish the prognostic signature, namely cell death index (CDI). The validation in several external cohorts indicated that CDI can be regarded as a potential risk factor of LUAD patients. Combined with other common clinical information, a nomogram with potential predictive ability was constructed. Besides, according to the CDI signature, the tumor microenvironment (TME) and sensitivity to some potential chemotherapeutic drugs were further and deeply explored. Notably, verification and functional experiments further demonstrated the remarkable correlation between CDI and unfold protein response. Given all the above, a novel CDI gene signature was indicated to predict the prognosis and exploit precision therapeutic strategies of LUAD patients.
Collapse
Affiliation(s)
- Peng Yu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Leyang Xiao
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- The Second Clinical Medical College, Nanchang University, Nanchang, China
| | - Kaibo Hu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- The Second Clinical Medical College, Nanchang University, Nanchang, China
| | - Jitao Ling
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yixuan Chen
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ruiqi Liang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xinyu Liu
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, USA
| | - Deju Zhang
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Pok Fu Lam, Hong Kong
| | - Yuzhen Liu
- Department of Thoracic Surgery, Jiangxi Provincial Chest Hospital, Nanchang, China
| | - Tongchun Weng
- Department of Thoracic Surgery, Jiangxi Provincial Chest Hospital, Nanchang, China
| | - Hongfa Jiang
- Department of Thoracic Surgery, Jiangxi Provincial Chest Hospital, Nanchang, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wuming Wang
- Department of Thoracic Surgery, Jiangxi Provincial Chest Hospital, Nanchang, China.
| |
Collapse
|
11
|
Lu X, Du W, Zhou J, Li W, Fu Z, Ye Z, Chen G, Huang X, Guo Y, Liao J. Integrated genomic analysis of the stemness index signature of mRNA expression predicts lung adenocarcinoma prognosis and immune landscape. PeerJ 2025; 13:e18945. [PMID: 39959839 PMCID: PMC11830367 DOI: 10.7717/peerj.18945] [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: 10/29/2024] [Accepted: 01/16/2025] [Indexed: 02/18/2025] Open
Abstract
mRNA expression-based stemness index (mRNAsi) has been used for prognostic assessment in various cancers, but its application in lung adenocarcinoma (LUAD) is limited, which is the focus of this study. Low mRNAsi in LUAD predicted a better prognosis. Eight genes (GNG7, EIF5A, ANLN, FKBP4, GAPDH, GNPNAT1, E2F7, CISH) associated with mRNAsi were screened to establish a risk model. The differentially expressed genes between the high and low risk groups were mainly enriched in the metabolism, cell cycle functions pathway. The low risk score group had higher immune cell scores. Patients with lower TIDE scores in the low risk group had better immunotherapy outcomes. In addition, risk score was effective in assessing drug sensitivity of LUAD. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) data showed that eight genes were differentially expressed in LUAD cell lines, and knockdown of EIF5A reduced the invasion and migration ability of LUAD cells. This study designed a risk model based on the eight mRNAsi-related genes for predicting LUAD prognosis. The model accurately predicted the prognosis and survival of LUAD patients, facilitating the assessment of the sensitivity of patients to immunotherapy and chemotherapy.
Collapse
Affiliation(s)
- Xingzhao Lu
- Thoracic Surgery Department, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
- Department of Medical Oncology, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
| | - Wei Du
- Thoracic Surgery Department, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
| | - Jianping Zhou
- Thoracic Surgery Department, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
| | - Weiyang Li
- Thoracic Surgery Department, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
| | - Zhimin Fu
- Thoracic Surgery Department, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
| | - Zhibin Ye
- Thoracic Surgery Department, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
| | - Guobiao Chen
- Thoracic Surgery Department, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
| | - Xian Huang
- Thoracic Surgery Department, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
| | - Yuliang Guo
- Thoracic Surgery Department, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
| | - Jingsheng Liao
- Department of Medical Oncology, The Tenth Affiliated Hospital of Southern Medical University, Dongguan Institute of Clinical Cancer Research, Affiliated Dongguan People’s Hospital, Southern Medical University, Dongguan, China
| |
Collapse
|
12
|
Liu X, Zhang W, Wang H, Yang W. Identification of CKAP2 as a Potential Target for Prevention of Gastric Cancer Progression: A Multi-Omics Study. Int J Mol Sci 2025; 26:1557. [PMID: 40004022 PMCID: PMC11855583 DOI: 10.3390/ijms26041557] [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: 01/04/2025] [Revised: 02/10/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Gastric cancer (GC) ranks as one of the most prevalent malignant tumors globally. The subtle manifestation of its early-stage symptoms often results in many GC patients being diagnosed at a late or advanced stage, thereby posing significant obstacles to the effectiveness of chemotherapy treatments. Therefore, identifying early biomarkers for GC is crucial. In recent years, an increasing number of studies have highlighted the pivotal role that aging plays in the progression of cancer. Among the various proteins involved, Cytoskeleton-associated protein 2 (CKAP2) emerges as a crucial player in controlling cell proliferation, regulating mitosis and cell division, and exerting a significant influence on the aging process. We employed a bioinformatics approach to assess the causal association between aging-related genes and GC and explore the potential significance of CKAP2 in GC by analyzing data sourced from various repositories, including Genotype Tissue Expression (GTEx), GWAS Catalog, The Database of Cell Senescence Genes (CellAge), The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO), Human Protein Atlas (HPA), and the Comparative Toxicology Genome Database (CTD). Our research summarized the causal relationship between CKAP2 expression and the development risk of GC, differential expression in GC, the relationship with the prognosis of GC, genetic correlation, functional analysis, and immune cell infiltration, and explored the interaction of CKAP2 and chemical substances. The findings revealed that an elevation in CKAP2 expression correlated with a reduced likelihood of developing GC. There was a significant difference in the expression of CKAP2 between GC and normal patients. Specifically, there was higher expression in GC compared to normal patients. In addition, CKAP2 has been proven to have diagnostic value in GC, and elevated levels of CKAP2 expression are indicative of a more favorable prognosis. Immune infiltration analysis revealed the relationship between CKAP2 and tumor immune microenvironment, while the Comparative Toxicology Genome Database (CTD) identified a small molecule compound that may target CKAP2. In summary, through comprehensive multivariate analyses, we identified and validated the potential role that CKAP2 may play in GC. Therefore, CKAP2 shows potential as an indicator for both the diagnosis and prognosis of GC, making it worthy of further clinical investigation.
Collapse
Affiliation(s)
- Xueyi Liu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China; (X.L.); (W.Z.)
- Science Island Branch, Graduate School of University of Science and Technology of China, Hefei 230031, China
| | - Wenyu Zhang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China; (X.L.); (W.Z.)
| | - Hui Wang
- Laboratory of Molecular Genetics of Aging & Tumor, Medicine School, Kunming University of Science and Technology, Kunming 650500, China;
| | - Wulin Yang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China; (X.L.); (W.Z.)
| |
Collapse
|
13
|
Qin X, Xu W, Wu J, Li M. Integration of single-cell and bulk RNA-sequencing data to construct and validate a signature based on NK cell marker genes to predict immunotherapy response and prognosis in colorectal cancer. Discov Oncol 2025; 16:134. [PMID: 39920524 PMCID: PMC11805743 DOI: 10.1007/s12672-025-01842-7] [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: 09/10/2024] [Accepted: 01/20/2025] [Indexed: 02/09/2025] Open
Abstract
We aimed to create a NK cell marker genes-based signature to predict immunotherapy response and prognosis in colorectal cancer. We integrated scRNA-seq data from four Gene Expression Omnibus (GEO) samples and performed Weighted gene correlation network analysis (WGCNA) based on 587 the Cancer Genome Atlas (TCGA) colorectal cancer samples to uncover NK cell-related genes. We identified 1080 NK cell-related core genes and 276 NK cell-related feature genes based on WGCNA and clustering and annotation of scRNA-seq data, respectively. Six key NK cell-related prognostic signature genes were obtained by univariate and LASSO regression analyses, including ADAM8, CTSD, CCL4, IL2RB, TTC38, and PLEK. Two validation cohorts from the GEO dataset, comprising 124 and 201 samples respectively, were used. The signature was significantly associated with overall survival and correlated with immune cell infiltration, immune and stromal scores, and immune checkpoint genes. Furthermore, the signature was associated with the homologous recombination deficiency (HRD) and T-cell receptor (TCR) scores. In conclusion, our study proposes a new prognostic signature based on NK cell marker genes, which may serve as a potential tool to predict overall survival and immunotherapy response for CRC patients.
Collapse
Affiliation(s)
- Xiaoyu Qin
- Department of Gastroenterology, Shanghai Pudong New Area Gongli Hospital, Shanghai, 200135, China
| | - Wenjuan Xu
- Department of General Surgery, Shanghai Punan Hospital, Pudong New District, Shanghai, 200125, China
| | - Jinxiu Wu
- Department of General Surgery, Shanghai Punan Hospital, Pudong New District, Shanghai, 200125, China
| | - Ming Li
- Department of General Surgery, Shanghai Punan Hospital, Pudong New District, Shanghai, 200125, China.
| |
Collapse
|
14
|
Tang Z, Pan Y, Li W, Ma R, Wang J. Unlocking the future: mitochondrial genes and neural networks in predicting ovarian cancer prognosis and immunotherapy response. Transl Cancer Res 2025; 14:512-521. [PMID: 39974375 PMCID: PMC11833377 DOI: 10.21037/tcr-24-1233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 11/13/2024] [Indexed: 02/21/2025]
Abstract
Background Mitochondrial genes are involved in the tumor metabolism of ovarian cancer (OC), affecting immune cell infiltration and treatment response. We aimed to utilize mitochondrial genes to predict OC prognosis and immunotherapy response. Methods The prognosis data, immunotherapy efficacy and next generation sequencing data of OC patients were downloaded from The Cancer Genome Atlas Program (TCGA) and Gene Expression Omnibus (GEO). Mitochondrial genes were sourced from the MitoCarta3.0 database. Seventy percent of the patients were randomly selected as the discovery cohort for model construction, while the remaining 30% constituted the validation cohort for model assessment. Using the expression of mitochondrial genes as the predictor variable and based on the neural network algorithm, the overall survival (OS) time and immunotherapy efficacy (complete or partial response) of the included patients were predicted. Results There were 375 OC patients included to construct the prognostic model, and 26 patients were included to construct the immune efficacy model. The average area under the receiver operating characteristic curve (AUC) of the prognostic model was: 0.7268 [95% confidence interval (CI), 0.7258-0.7278] in the discovery cohort and 0.6475 (95% CI: 0.6466-0.6484) in the validation cohort. The average AUC of the immunotherapy efficacy model was: 0.9444 (95% CI: 0.8333-1.0000) in the discovery cohort and 0.9167 (95% CI: 0.6667-1.0000) in the validation cohort. Conclusions The application of mitochondrial genes and neural networks shows potential in predicting the prognosis and immunotherapy response in OC patients. And this approach could provide valuable insights for personalized treatment strategies.
Collapse
Affiliation(s)
- Zhijian Tang
- Department of Obstetrics and Gynecology, Peking University People’s Hospital, Beijing, China
| | - Yuanming Pan
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Wei Li
- Department of Thoracic Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Ruiqiong Ma
- Department of Obstetrics and Gynecology, Peking University People’s Hospital, Beijing, China
| | - Jianliu Wang
- Department of Obstetrics and Gynecology, Peking University People’s Hospital, Beijing, China
| |
Collapse
|
15
|
Tang ZJ, Pan YM, Li W, Ma RQ, Wang JL. Unlocking the future: Mitochondrial genes and neural networks in predicting ovarian cancer prognosis and immunotherapy response. World J Clin Oncol 2025; 16:94813. [PMID: 39867736 PMCID: PMC11528894 DOI: 10.5306/wjco.v16.i1.94813] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/17/2024] [Accepted: 06/05/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Mitochondrial genes are involved in tumor metabolism in ovarian cancer (OC) and affect immune cell infiltration and treatment responses. AIM To predict prognosis and immunotherapy response in patients diagnosed with OC using mitochondrial genes and neural networks. METHODS Prognosis, immunotherapy efficacy, and next-generation sequencing data of patients with OC were downloaded from The Cancer Genome Atlas and Gene Expression Omnibus. Mitochondrial genes were sourced from the MitoCarta3.0 database. The discovery cohort for model construction was created from 70% of the patients, whereas the remaining 30% constituted the validation cohort. Using the expression of mitochondrial genes as the predictor variable and based on neural network algorithm, the overall survival time and immunotherapy efficacy (complete or partial response) of patients were predicted. RESULTS In total, 375 patients with OC were included to construct the prognostic model, and 26 patients were included to construct the immune efficacy model. The average area under the receiver operating characteristic curve of the prognostic model was 0.7268 [95% confidence interval (CI): 0.7258-0.7278] in the discovery cohort and 0.6475 (95%CI: 0.6466-0.6484) in the validation cohort. The average area under the receiver operating characteristic curve of the immunotherapy efficacy model was 0.9444 (95%CI: 0.8333-1.0000) in the discovery cohort and 0.9167 (95%CI: 0.6667-1.0000) in the validation cohort. CONCLUSION The application of mitochondrial genes and neural networks has the potential to predict prognosis and immunotherapy response in patients with OC, providing valuable insights into personalized treatment strategies.
Collapse
Affiliation(s)
- Zhi-Jian Tang
- Department of Obstetrics and Gynecology, Peking University People’s Hospital, Beijing 100044, China
| | - Yuan-Ming Pan
- Cancer Research Center, Beijing Chest Hospital, Beijing 101149, China
| | - Wei Li
- Cancer Research Center, Beijing Chest Hospital, Beijing 101149, China
- Department of Thoracic Surgery, Sichuan Provincial People's Hospital, Chengdu 610072, Sichuan Province, China
| | - Rui-Qiong Ma
- Department of Obstetrics and Gynecology, Peking University People’s Hospital, Beijing 100044, China
| | - Jian-Liu Wang
- Department of Obstetrics and Gynecology, Peking University People’s Hospital, Beijing 100044, China
| |
Collapse
|
16
|
Li X, Wang K, Liu J, Li Y. A comprehensive analysis to reveal the underlying molecular mechanisms of natural killer cell in thyroid carcinoma based on single-cell RNA sequencing data. Discov Oncol 2025; 16:44. [PMID: 39808350 PMCID: PMC11732816 DOI: 10.1007/s12672-025-01779-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: 08/12/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Thyroid carcinoma (THCA) is the most common cancer of the endocrine system. Natural killer (NK) cell play an important role in tumor immune surveillance. The aim of this study was to explore the possible molecular mechanisms involved in NK cell in THCA to help the management and treatment of the disease. METHODS All data were downloaded from public databases. Candidate hub genes associated with NK cell in THCA were identified by limma, WGCNA and singleR packages. Functional enrichment analysis was performed on the candidate hub genes. Hub genes associated with NK cell were identified by Pearson correlation analysis. The mRNA-miRNA-lncRNA and transcription factors (TF) networks were constructed and the drug was predicted. RESULTS The infiltration level of NK cell in THCA tissues was higher than that in paracancerous tissues. KEGG functional enrichment analysis only obtained two signaling pathways, thyroid hormone synthesis and mineral absorption. CTSC, FN1, SLC34A2 and TMSB4X identified by Pearson correlation analysis were considered as the hub genes. Receiver operating characteristic analysis suggested that hub genes may be potential diagnostic biomarkers. In mRNA-miRNA-lncRNA network, FN1 had the highest correlation with IQCH-AS1, and IQCH-AS1 was also correlated with hsa-miR-543. In addition, FN1 and RUNX1 were also found to have the highest correlation in TF network. Finally, NK cell-related drugs belinostat and vorinostat were identified based on ASGARD. CONCLUSION The identification of important signaling pathways, molecules and drugs provides potential research directions for further research in THCA and contributes to the development of diagnostic and therapeutic approaches for this disease.
Collapse
Affiliation(s)
- Xiaoqiong Li
- The Department of Experimental Medicine, Meishan City People's Hospital, No. 288, South Fourth Section, Dongpo Avenue, Meishan, 620000, Sichuan, China.
| | - Kejiang Wang
- The Department of Experimental Medicine, Meishan City People's Hospital, No. 288, South Fourth Section, Dongpo Avenue, Meishan, 620000, Sichuan, China
| | - Jiaxin Liu
- The Department of Experimental Medicine, Meishan City People's Hospital, No. 288, South Fourth Section, Dongpo Avenue, Meishan, 620000, Sichuan, China
| | - Yan Li
- The Department of Experimental Medicine, Meishan City People's Hospital, No. 288, South Fourth Section, Dongpo Avenue, Meishan, 620000, Sichuan, China
| |
Collapse
|
17
|
Luo J, An J, Jia R, Liu C, Zhang Y. Identification and Verification of Metabolism-related Immunotherapy Features and Prognosis in Lung Adenocarcinoma. Curr Med Chem 2025; 32:1423-1441. [PMID: 38500277 DOI: 10.2174/0109298673293414240314043529] [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: 11/30/2023] [Revised: 02/21/2024] [Accepted: 03/04/2024] [Indexed: 03/20/2024]
Abstract
BACKGROUND Lung cancer is a frequent malignancy with a poor prognosis. Extensive metabolic alterations are involved in carcinogenesis and could, therefore, serve as a reliable prognostic phenotype. AIMS Our study aimed to develop a prognosis signature and explore the relationship between metabolic characteristic-related signature and immune infiltration in lung adenocarcinoma (LUAD). OBJECTIVE TCGA-LUAD and GSE31210 datasets were used as a training set and a validation set, respectively. METHODS A total of 513 LUAD samples collected from The Cancer Genome Atlas database (TCGA-LUAD) were used as a training dataset. Molecular subtypes were classified by consensus clustering, and prognostic genes related to metabolism were analyzed based on Differentially Expressed Genes (DEGs), Protein-Protein Interaction (PPI) network, the univariate/multivariate- and Lasso- Cox regression analysis. RESULTS Two molecular subtypes with significant survival differences were divided by the metabolism gene sets. The DEGs between the two subtypes were identified by integrated analysis and then used to develop an 8-gene signature (TTK, TOP2A, KIF15, DLGAP5, PLK1, PTTG1, ECT2, and ANLN) for predicting LUAD prognosis. Overexpression of the 8 genes was significantly correlated with worse prognostic outcomes. RiskScore was an independent factor that could divide LUAD patients into low- and high-risk groups. Specifically, high-risk patients had poorer prognoses and higher immune escape. The Receiver Operating Characteristic (ROC) curve showed strong performance of the RiskScore model in estimating 1-, 3- and 5-year survival in both training and validation sets. Finally, an optimized nomogram model was developed and contributed the most to the prognostic prediction in LUAD. CONCLUSION The current model could help effectively identify high-risk patients and suggest the most effective drug and treatment candidates for patients with LUAD.
Collapse
Affiliation(s)
- Junfang Luo
- Department of Geriatric Respiratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jinlu An
- Department of Geriatric Respiratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Rongyan Jia
- Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Cong Liu
- Department of Geriatric Respiratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yang Zhang
- Department of Geriatric Respiratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| |
Collapse
|
18
|
Ouyang M, Gui Y, Li N, Zhao L. Prognostic model based on tumor stemness genes for triple-negative breast cancer. Sci Rep 2024; 14:30855. [PMID: 39730613 DOI: 10.1038/s41598-024-81503-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: 08/05/2024] [Accepted: 11/27/2024] [Indexed: 12/29/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive disease with a poor prognosis and lack of effective treatment. In this study, TNBCs were analyzed from the perspective of tumor stemness based on scRNA-seq data. The analysis showed that tumor cells of TNBC were divided into 4 subtypes, with subtype 2 having the highest stemness score. A prognostic model of 7 tumor stemness-related genes (AP2S1, CHML, FABP7, FADS2, PAXX, SDC1 and TOP2A) was developed based on marker genes of this subtype and TCGA data, and the predictive power of this feature was well validated in different clinical subgroups. TNBC patients in the low TS group had a better prognosis. In addition, drug sensitivity analysis showed that patients in the high TS (tumor stemness) score group were more sensitive to PD-L1 inhibitors and the chemotherapeutic agents. In conclusion, our study developed a prognostic model based on TNBC tumor stemness cell marker genes, which has a good ability to predict the prognosis of TNBC patients and the effect of response to drug therapy.
Collapse
Affiliation(s)
- Min Ouyang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
| | - Yajun Gui
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, 410000, Human, China
| | - Namei Li
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, 410000, Human, China
| | - Lin Zhao
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, 410000, Hunan, China.
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, 410000, Human, China.
| |
Collapse
|
19
|
Li L, He S. Programmed cell death pathways in lung adenocarcinoma: illuminating tumor drug resistance and therapeutic opportunities through single-cell analysis. Discov Oncol 2024; 15:828. [PMID: 39714518 DOI: 10.1007/s12672-024-01736-0] [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: 11/25/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024] Open
Abstract
Lung adenocarcinoma (LUAD) is a major contributor to cancer-related deaths, distinguished by its pronounced tumor heterogeneity and persistent challenges in overcoming drug resistance. In this study, we utilized single-cell RNA sequencing (scRNA-seq) to dissect the roles of programmed cell death (PCD) pathways, including apoptosis, necroptosis, pyroptosis, and ferroptosis, in shaping LUAD heterogeneity, immune infiltration, and prognosis. Among these, ferroptosis and pyroptosis were most significantly associated with favorable survival outcomes, highlighting their potential roles in enhancing anti-tumor immunity. Distinct PCD-related LUAD subtypes were identified, characterized by differential pathway activation and immune cell composition. Subtypes enriched with cytotoxic lymphocytes and dendritic cells demonstrated improved survival outcomes and increased potential responsiveness to immunotherapy. Drug sensitivity analysis revealed that these subtypes exhibited heightened sensitivity to targeted therapies and immune checkpoint inhibitors, suggesting opportunities for personalized treatment strategies. Our findings emphasize the interplay between PCD pathways and the tumor microenvironment, providing insights into the mechanisms underlying tumor drug resistance and immune evasion. By linking molecular and immune features to clinical outcomes, this study highlights the potential of targeting PCD pathways to enhance therapeutic efficacy and overcome resistance in LUAD. These results contribute to a growing framework for developing precise and adaptable cancer therapies tailored to specific tumor characteristics.
Collapse
Affiliation(s)
- Long Li
- Department of Critical Care Medicine, The Fifth People's Hospital of Ganzhou City, Ganzhou, 341000, China
- Ganzhou Key Laboratory of Respiratory Diseases, Ganzhou, 341000, China
- Ganzhou Institute for the Prevention and Treatment of Respiratory Diseases, Ganzhou, 341000, China
| | - Shancheng He
- Department of Critical Care Medicine, The Fifth People's Hospital of Ganzhou City, Ganzhou, 341000, China.
- Ganzhou Key Laboratory of Respiratory Diseases, Ganzhou, 341000, China.
- Ganzhou Institute for the Prevention and Treatment of Respiratory Diseases, Ganzhou, 341000, China.
| |
Collapse
|
20
|
Dong L, Li L, Zhu L, Xu F, Zhang R, Li Q, Zhu Y, Zeng Z, Ding K. Multiomics analysis of homologous recombination deficiency across cancer types. BIOMOLECULES & BIOMEDICINE 2024; 25:71-81. [PMID: 39073402 PMCID: PMC11647252 DOI: 10.17305/bb.2024.10448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
There remains ongoing debate regarding the association of homologous recombination deficiency (HRD) with patient survival across various malignancies, highlighting the need for a comprehensive understanding of HRD's role in different cancer types. Based on data from databases, we conducted a multivariable omics analysis on HRD in 33 cancer types, focusing mainly on 23 cancers in which HRD was significantly associated with patient overall survival (OS) rates. This analysis included the mechanisms related to patient prognosis, gene expression, gene mutation, and signaling pathways. In this study, HRD was found to be significantly associated with patient prognosis, but its impact varied among different cancers. HRD was linked to different outcomes for patients with distinct tumor subtypes and was correlated with clinical features such as clinical stage and tumor grade. Driver gene mutations, including TP53, MUC4, KRAS, HRAS, FLG, ANK3, BRCA2, ATRX, FGFR3, NFE2L2, MAP3K1, PIK3CA, CIC, FUBP1, ALB, CTNNB1, and MED12, were associated with HRD across specific cancer types. We also analyzed differentially expressed genes (DEGs) and differentially methylated regions (DMRs) in relation to HRD levels in these cancers. Furthermore, we explored the correlation between HRD and signaling pathways, as well as immune cell infiltration. Overall, our findings contribute to a comprehensive understanding of HRD's multifaceted role in cancer.
Collapse
Affiliation(s)
- Lin Dong
- Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Lin Li
- Department of Pathology, Tongling People’s Hospital, Tongling, Anhui, China
| | - Linyan Zhu
- Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Fei Xu
- Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
- Department of Pathology, Anhui Provincial Children’s Hospital, Hefei, Anhui, China
| | - Rumeng Zhang
- Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Qiushuang Li
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Yong Zhu
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Zhutian Zeng
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Keshuo Ding
- Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
21
|
Xie Y, Chen H, Zhang X, Zhang J, Zhang K, Wang X, Min S, Wang X, Lian C. Integration of the bulk transcriptome and single-cell transcriptome reveals efferocytosis features in lung adenocarcinoma prognosis and immunotherapy by combining deep learning. Cancer Cell Int 2024; 24:388. [PMID: 39580462 PMCID: PMC11585238 DOI: 10.1186/s12935-024-03571-3] [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: 04/17/2024] [Accepted: 11/10/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND Efferocytosis (ER) refers to the process of phagocytic clearance of programmed dead cells, and studies have shown that it is closely related to tumor immune escape. METHODS This study was based on a comprehensive analysis of TCGA, GEO and CTRP databases. ER-related genes were collected from previous literature, univariate Cox regression was performed and consistent clustering was performed to categorize lung adenocarcinoma (LUAD) patients into two subgroups. Lasso regression and multivariate Cox regression analyses were used to construct ER-related prognostic features, and multiple immune infiltration algorithms were used to assess the correlation between the extracellular burial-related risk score (ERGRS) and tumor microenvironment (TME). And the key gene HAVCR1 was identified by deep learning, etc. Finally, pan-cancer analysis of the key genes was performed and in vitro experiments were conducted to verify the promotional effect of HAVCR1 on LUAD progression. RESULTS A total of 33 ER-related genes associated with the prognosis of LUAD were identified, and the prognostic signature of ERGRS was successfully constructed to predict the overall survival (OS) and treatment response of LUAD patients. The high-risk group was highly enriched in some oncogenic pathways, while the low-ERGRS group was highly enriched in some immune-related pathways. In addition, the high ERGRS group had higher TMB, TNB and TIDE scores and lower immune scores. The low-risk group had better immunotherapeutic response and less likelihood of immune escape. Drug sensitivity analysis revealed that BRD-K92856060, monensin and hexaminolevulinate may be potential therapeutic agents for the high-risk group. And ERGRS was validated in several cohorts. In addition, HAVCR1 is one of the key genes, and knockdown of HAVCR1 in vitro significantly reduced the proliferation, migration and invasion ability of lung adenocarcinoma cells. CONCLUSION Our study developed a novel prognostic signature of efferocytosis-related genes. This prognostic signature accurately predicted survival prognosis as well as treatment outcome in LUAD patients and explored the role of HAVCR1 in lung adenocarcinoma progression.
Collapse
Affiliation(s)
- Yiluo Xie
- Anhui Province Key Laboratory of Respiratory Tumor and Infectious Disease, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233030, China
- Department of Clinical Medicine, Bengbu Medical University, Bengbu, 233030, China
| | - Huili Chen
- Research Center of Clinical Laboratory Science, Bengbu Medical University, Bengbu, 233030, China
| | - Xueying Zhang
- Anhui Province Key Laboratory of Respiratory Tumor and Infectious Disease, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233030, China
| | - Jing Zhang
- Department of Genetics, School of Life Sciences, Bengbu Medical University, Bengbu, 233030, China
| | - Kai Zhang
- Department of Clinical Medicine, Bengbu Medical University, Bengbu, 233030, China
| | - Xinyu Wang
- Department of Clinical Medicine, Bengbu Medical University, Bengbu, 233030, China
| | - Shengping Min
- Anhui Province Key Laboratory of Respiratory Tumor and Infectious Disease, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233030, China
| | - Xiaojing Wang
- Anhui Province Key Laboratory of Respiratory Tumor and Infectious Disease, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233030, China.
| | - Chaoqun Lian
- Research Center of Clinical Laboratory Science, Bengbu Medical University, Bengbu, 233030, China.
| |
Collapse
|
22
|
Pan M, Yuan X, Peng J, Wu R, Chen X. Identification and validation of a novel innate lymphoid cell-based signature to predict prognosis and immune response in liver cancer by integrated single-cell RNA analysis and bulk RNA sequencing. Transl Cancer Res 2024; 13:5395-5416. [PMID: 39525025 PMCID: PMC11543044 DOI: 10.21037/tcr-24-725] [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: 04/29/2024] [Accepted: 08/22/2024] [Indexed: 11/16/2024]
Abstract
Background Innate lymphoid cells (ILCs) exert tumor suppressive and tumor promoting effects. However, the prognostic significance of ILC-associated genes remains unclear in hepatocellular carcinoma (HCC). Hence, the aim of this research was to develop an innovative predictive risk classification system using bioinformatics examination. Methods We explored the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) databases to gather data pertaining to HCC and its clinical details. Significantly different ILC-associated genes were investigated by Seurat analysis. The number of signaling interactions of ILCs with other cells was discovered by CellPhoneDB analysis. ClusterProfiler and Metascape were utilized to perform Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses on ILC genes. In order to identify potential ILC predictors, we utilized univariate Cox regression and least absolute shrinkage and selection operator (LASSO) analyses, subsequently validating these predictors in TCGA and GEO groups. The multi-omics ILC signature model's clinical predictive capabilities, along with drug sensitivity and immune factor relations, were assessed using Cell-type Identification by Estimating Relative Subsets of RNA Transcripts (CIBERSORT) and pRRophetic. We investigated the possible molecular pathways in our predictive ILC signature through the utilization of gene set enrichment analysis (GSEA) and gene set variation analysis (GSVA). Five key genes were screened out by constructing a competing endogenous RNA (ceRNA) network using Cytoscape and their values in clinical indexes were demonstrated. Immunohistochemistry (IHC) in HCC cases confirmed the expression of these genes. Results ILC cell subsets were identified, and cell-cell communication analysis revealed that the signaling pathways involving ILC cell subsets were mostly abundant in the HCC microenvironment. Subsequently, 270 marker genes of the ILC clusters were subjected to GO and KEGG enrichment analysis. Furthermore, a total of 58 prognostically relevant genes were screened as features for prognostic prediction models. Next, the models were validated and clinically evaluated (P values of Kaplan-Meier survival curves below 0.05). Five key genes (C2, STK4, CALM1, IL7R, and RORA) were further screened by multi omics analysis of immune cell and factor and drug sensitivity and correlation analysis of tumor regulatory genes in liver cancer. Furthermore, the potential clinical value of the 5 key genes was confirmed in HCC patients. Finally, the IHC results confirmed the expression of C2, STK4, CALM1, IL7R, and RORA in HCC. Our experimental results provided preliminary evidence supporting the oncogenic roles of STK4 and CALM1, as well as the tumor-suppressive roles of C2, RORA, and IL7R in HCC. Conclusions A novel prognostic feature of ILC potentially involved in HCC was discovered. It showed high values in predicting patient overall survival (OS) as well as good differences in immunity and drug sensitivity. Therefore, targeting these ILC signatures may be a potential effective approach in HCC treatment.
Collapse
Affiliation(s)
- Meng Pan
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Xiaolong Yuan
- Department of Pharmacy, The Second Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Junlu Peng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Ruiqi Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Xiaopeng Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| |
Collapse
|
23
|
Hou Q, Li C, Chong Y, Yin H, Guo Y, Yang L, Li T, Yin S. Comprehensive single-cell and bulk transcriptomic analyses to develop an NK cell-derived gene signature for prognostic assessment and precision medicine in breast cancer. Front Immunol 2024; 15:1460607. [PMID: 39507529 PMCID: PMC11537931 DOI: 10.3389/fimmu.2024.1460607] [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: 07/06/2024] [Accepted: 10/07/2024] [Indexed: 11/08/2024] Open
Abstract
Background Natural killer (NK) cells play crucial roles in mediating anti-cancer activity in breast cancer (BRCA). However, the potential of NK cell-related molecules in predicting BRCA outcomes and guiding personalized therapy remains largely unexplored. This study focused on developing a prognostic and therapeutic prediction model for BRCA by incorporating NK cell-related genes. Methods The data analyzed primarily originated from the TCGA and GEO databases. The prognostic role of NK cells was evaluated, and marker genes of NK cells were identified via single-cell analysis. Module genes closely associated with immunotherapy resistance were identified by bulk transcriptome-based weighted correlation network analysis (WGCNA). Following taking intersection and LASSO regression, NK-related genes (NKRGs) relevant to BRCA prognosis were screened, and the NK-related prognostic signature was subsequently constructed. Analyses were further expanded to clinicopathological relevance, GSEA, tumor microenvironment (TME) analysis, immune function, immunotherapy responsiveness, and chemotherapeutics. Key NKRGs were screened by machine learning and validated by spatial transcriptomics (ST) and immunohistochemistry (IHC). Results Tumor-infiltrating NK cells are a favorable prognostic factor in BRCA. By combining scRNA-seq and bulk transcriptomic analyses, we identified 7 NK-related prognostic NKRGs (CCL5, EFHD2, KLRB1, C1S, SOCS3, IRF1, and CCND2) and developed an NK-related risk scoring (NKRS) system. The prognostic reliability of NKRS was verified through survival and clinical relevance analyses across multiple cohorts. NKRS also demonstrated robust predictive power in various aspects, including TME landscape, immune functions, immunotherapy responses, and chemotherapeutic sensitivity. Additionally, KLRB1 and CCND2 emerged as key prognostic NKRGs identified through machine learning and external validation, with their expression correlation with NK cells confirmed in BRCA specimens by ST and IHC. Conclusions We developed a novel NK-related gene signature that has proven valuable for evaluating prognosis and treatment response in BRCA, expecting to advance precision medicine of BRCA.
Collapse
Affiliation(s)
- Qianshan Hou
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Naval Medical University, Shanghai, China
| | - Chunzhen Li
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Naval Medical University, Shanghai, China
| | - Yuhui Chong
- School of Pharmacy, Naval Medical University, Shanghai, China
| | - Haofeng Yin
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Naval Medical University, Shanghai, China
| | - Yuchen Guo
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Naval Medical University, Shanghai, China
| | - Lanjie Yang
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Naval Medical University, Shanghai, China
| | - Tianliang Li
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Naval Medical University, Shanghai, China
| | - Shulei Yin
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Naval Medical University, Shanghai, China
| |
Collapse
|
24
|
Li X, Sun T, Li H, Liu J, Huang N, Liu S. The Novel-B-Cell-Related Gene Signature Predicts the Prognosis and Immune Status of Patients with Esophageal Carcinoma. J Gastrointest Cancer 2024; 55:1313-1323. [PMID: 38963643 PMCID: PMC11347472 DOI: 10.1007/s12029-024-01083-x] [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] [Accepted: 06/15/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND The current understanding of the prognostic significance of B cells and their role in the tumor microenvironment (TME) in esophageal carcinoma (ESCA) is limited. METHODS We conducted a screening for B-cell-related genes through the analysis of single-cell transcriptome data. Subsequently, we developed a B-cell-related gene signature (BRGrisk) using LASSO regression analysis. Patients from The Cancer Genome Atlas cohort were divided into a training cohort and a test cohort. Patients were categorized into high- and low-risk groups based on their median BRGrisk scores. The overall survival was assessed using the Kaplan-Meier method, and a nomogram based on BRGrisk was constructed. Immune infiltration profiles between the risk groups were also compared. RESULTS The BRGrisk prognostic model indicated significantly worse outcomes for patients with high BRGrisk scores (p < 0.001). The BRGrisk-based nomogram exhibited good prognostic performance. Analysis of immune infiltration revealed that patients in the high-BRGrisk group had notably higher levels of immune cell infiltration and were more likely to be in an immunoresponsive state. Enrichment analysis showed a strong correlation between the prognostic gene signature and cancer-related pathways. IC50 results indicated that patients in the low-BRGrisk group were more responsive to common drugs compared to those in the high-BRGrisk group. CONCLUSIONS This study presents a novel BRGrisk that can be used to stratify the prognosis of ESCA patients and may offer guidance for personalized treatment strategies aimed at improving prognosis.
Collapse
Affiliation(s)
- Xinhong Li
- Department of Oncohematology, Norinco General Hospital, Xi'an, Shaanxi, 710061, China
| | - Tongyu Sun
- Hepatobiliary and Vascular Surgery, Norinco General Hospital, Xi'an, Shaanxi, 710061, China
| | - Hongyan Li
- Department of Radiology, Norinco General Hospital, Xi'an, Shaanxi, 710061, China
| | - Juan Liu
- Department of Oncohematology, Norinco General Hospital, Xi'an, Shaanxi, 710061, China
| | - Na Huang
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Surong Liu
- Department of Oncohematology, Norinco General Hospital, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
25
|
Maimaitiyiming A, An H, Xing C, Li X, Li Z, Bai J, Luo C, Zhuo T, Huang X, Maimaiti A, Aikemu A, Wang Y. Machine learning-driven mast cell gene signatures for prognostic and therapeutic prediction in prostate cancer. Heliyon 2024; 10:e35157. [PMID: 39170129 PMCID: PMC11336432 DOI: 10.1016/j.heliyon.2024.e35157] [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: 02/01/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/23/2024] Open
Abstract
Background The role of Mast cells has not been thoroughly explored in the context of prostate cancer's (PCA) unpredictable prognosis and mixed immunotherapy outcomes. Our research aims to employs a comprehensive computational methodology to evaluate Mast cell marker gene signatures (MCMGS) derived from a global cohort of 1091 PCA patients. This approach is designed to identify a robust biomarker to assist in prognosis and predicting responses to immunotherapy. Methods This study initially identified mast cell-associated biomarkers from prostate adenocarcinoma (PRAD) patients across six international cohorts. We employed a variety of machine learning techniques, including Random Forest, Support Vector Machine (SVM), Lasso regression, and the Cox Proportional Hazards Model, to develop an effective MCMGS from candidate genes. Subsequently, an immunological assessment of MCMGS was conducted to provide new insights into the evaluation of immunotherapy responses and prognostic assessments. Additionally, we utilized Gene Set Enrichment Analysis (GSEA) and pathway analysis to explore the biological pathways and mechanisms associated with MCMGS. Results MCMGS incorporated 13 marker genes and was successful in segregating patients into distinct high- and low-risk categories. Prognostic efficacy was confirmed by survival analysis incorporating MCMGS scores, alongside clinical parameters such as age, T stage, and Gleason scores. High MCMGS scores were correlated with upregulated pathways in fatty acid metabolism and β-alanine metabolism, while low scores correlated with DNA repair mechanisms, homologous recombination, and cell cycle progression. Patients classified as low-risk displayed increased sensitivity to drugs, indicating the utility of MCMGS in forecasting responses to immune checkpoint inhibitors. Conclusion The combination of MCMGS with a robust machine learning methodology demonstrates considerable promise in guiding personalized risk stratification and informing therapeutic decisions for patients with PCA.
Collapse
Affiliation(s)
- Abudukeyoumu Maimaitiyiming
- The First Affiliated Hospital, Xinjiang Medical University, Urumqi, China
- Department of Urological, Urology Centre, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Hengqing An
- The First Affiliated Hospital, Xinjiang Medical University, Urumqi, China
- Department of Urological, Urology Centre, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
- Xinjiang Clinical Research Center of Urogenital Diseases, Urumqi, China
| | - Chen Xing
- The First Affiliated Hospital, Xinjiang Medical University, Urumqi, China
- Department of Urological, Urology Centre, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
- Xinjiang Clinical Research Center of Urogenital Diseases, Urumqi, China
| | - Xiaodong Li
- The First Affiliated Hospital, Xinjiang Medical University, Urumqi, China
- Department of Urological, Urology Centre, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
- Xinjiang Clinical Research Center of Urogenital Diseases, Urumqi, China
| | - Zhao Li
- Department of Abdominal Ultrasonography, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Junbo Bai
- Department of Pediatric Urology, Urology Centre, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Cheng Luo
- The First Affiliated Hospital, Xinjiang Medical University, Urumqi, China
- Department of Urological, Urology Centre, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Tao Zhuo
- The First Affiliated Hospital, Xinjiang Medical University, Urumqi, China
- Department of Urological, Urology Centre, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Xin Huang
- The First Affiliated Hospital, Xinjiang Medical University, Urumqi, China
- Department of Urological, Urology Centre, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Aierpati Maimaiti
- The First Affiliated Hospital, Xinjiang Medical University, Urumqi, China
- Department of Neurosurgery, Neurosurgery Centre, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | | | - Yujie Wang
- The First Affiliated Hospital, Xinjiang Medical University, Urumqi, China
- Department of Urological, Urology Centre, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
- Xinjiang Clinical Research Center of Urogenital Diseases, Urumqi, China
| |
Collapse
|
26
|
Zhang Y, Zhang C, He J, Lai G, Li W, Zeng H, Zhong X, Xie B. Comprehensive analysis of single cell and bulk RNA sequencing reveals the heterogeneity of melanoma tumor microenvironment and predicts the response of immunotherapy. Inflamm Res 2024; 73:1393-1409. [PMID: 38896289 DOI: 10.1007/s00011-024-01905-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/07/2024] [Accepted: 06/09/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Tumor microenvironment (TME) heterogeneity is an important factor affecting the treatment response of immune checkpoint inhibitors (ICI). However, the TME heterogeneity of melanoma is still widely characterized. METHODS We downloaded the single-cell sequencing data sets of two melanoma patients from the GEO database, and used the "Scissor" algorithm and the "BayesPrism" algorithm to comprehensively analyze the characteristics of microenvironment cells based on single-cell and bulk RNA-seq data. The prediction model of immunotherapy response was constructed by machine learning and verified in three cohorts of GEO database. RESULTS We identified seven cell types. In the Scissor+ subtype cell population, the top three were T cells, B cells and melanoma cells. In the Scissor- subtype, there are more macrophages. By quantifying the characteristics of TME, significant differences in B cells between responders and non-responders were observed. The higher the proportion of B cells, the better the prognosis. At the same time, macrophages in the non-responsive group increased significantly. Finally, nine gene features for predicting ICI response were constructed, and their predictive performance was superior in three external validation groups. CONCLUSION Our study revealed the heterogeneity of melanoma TME and found a new predictive biomarker, which provided theoretical support and new insights for precise immunotherapy of melanoma patients.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, Chongqing, 400016, China
- Research Center for Medicine and Social Development, Chongqing Medical University, Chongqing, China
| | - Cong Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, Chongqing, 400016, China
- Research Center for Medicine and Social Development, Chongqing Medical University, Chongqing, China
| | - Jing He
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, Chongqing, 400016, China
- Research Center for Medicine and Social Development, Chongqing Medical University, Chongqing, China
| | - Guichuan Lai
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, Chongqing, 400016, China
- Research Center for Medicine and Social Development, Chongqing Medical University, Chongqing, China
| | - Wenlong Li
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, Chongqing, 400016, China
- Research Center for Medicine and Social Development, Chongqing Medical University, Chongqing, China
| | - Haijiao Zeng
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, Chongqing, 400016, China
- Research Center for Medicine and Social Development, Chongqing Medical University, Chongqing, China
| | - Xiaoni Zhong
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, Chongqing, 400016, China.
- Research Center for Medicine and Social Development, Chongqing Medical University, Chongqing, China.
| | - Biao Xie
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Yixue Road, Chongqing, 400016, China.
- Research Center for Medicine and Social Development, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
27
|
Wang J, Tan Z, Huang Y, Li C, Zhan P, Wang H, Li H. Integrating single-cell RNA-seq to identify fibroblast-based molecular subtypes for predicting prognosis and therapeutic response in bladder cancer. Aging (Albany NY) 2024; 16:11385-11408. [PMID: 39033778 PMCID: PMC11315389 DOI: 10.18632/aging.206021] [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: 01/09/2024] [Accepted: 07/05/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Bladder cancer (BLCA) is a highly aggressive and heterogeneous disease, posing challenges for diagnosis and treatment. Cancer immunotherapy has recently emerged as a promising option for patients with advanced and drug-resistant cancers. Fibroblasts, a significant component of the tumor microenvironment, play a crucial role in tumor progression, but their precise function in BLCA remains uncertain. METHODS Single-cell RNA sequencing (scRNA-seq) data for BLCA were obtained from the Gene Expression Omnibus database. The R package "Seurat" was used for processing scRNA-seq data, with uniform manifold approximation and projection (UMAP) for downscaling and cluster identification. The FindAllMarkers function identified marker genes for each cluster. Differentially expressed genes influencing overall survival (OS) of BLCA patients were identified using the limma package. Differences in clinicopathological characteristics, immune microenvironment, immune checkpoints, and chemotherapeutic drug sensitivity between high- and low-risk groups were investigated. RT-qPCR and immunohistochemistry validated the expression of prognostic genes. RESULTS Fibroblast marker genes identified three molecular subtypes in the testing set. A prognostic signature comprising ten genes stratified BLCA patients into high- and low-score groups. This signature was validated in one internal and two external validation sets. High-score patients exhibited increased immune cell infiltration, elevated chemokine expression, and enhanced immune checkpoint expression but had poorer OS and a reduced response to immunotherapy. Six sensitive anti-tumor drugs were identified for the high-score group. RT-qPCR and immunohistochemistry showed that CERCAM, TM4SF1, FN1, ANXA1, and LOX were highly expressed, while EMP1, HEYL, FBN1, and SLC2A3 were downregulated in BLCA. CONCLUSION A novel fibroblast marker gene-based signature was established, providing robust predictions of survival and immunotherapeutic response in BLCA patients.
Collapse
Affiliation(s)
- Jia Wang
- The Second Clinical Medical College, Kunming Medical University, Kunming, China
- Department of Endocrinology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhiyong Tan
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yinglong Huang
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Charles Li
- Core Facility for Protein Research, Chinese Academy of Sciences, Beijing, China
- Zhongke Jianlan Medical Research Institute, Beijing, China
- Zhejiang Institute of Integrated Traditional and Western Medicine, Hangzhou, China
| | - Peiqin Zhan
- The Second Clinical Medical College, Kunming Medical University, Kunming, China
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Haifeng Wang
- The Second Clinical Medical College, Kunming Medical University, Kunming, China
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Haihao Li
- The Second Clinical Medical College, Kunming Medical University, Kunming, China
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
28
|
Hua T, Liu DX, Zhang XC, Li ST, Wu JL, Zhao Q, Chen SB. Establishment of an ovarian cancer exhausted CD8+T cells-related genes model by integrated analysis of scRNA-seq and bulk RNA-seq. Eur J Med Res 2024; 29:358. [PMID: 38970067 PMCID: PMC11225302 DOI: 10.1186/s40001-024-01948-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 06/24/2024] [Indexed: 07/07/2024] Open
Abstract
Ovarian cancer (OC) was the fifth leading cause of cancer death and the deadliest gynecological cancer in women. This was largely attributed to its late diagnosis, high therapeutic resistance, and a dearth of effective treatments. Clinical and preclinical studies have revealed that tumor-infiltrating CD8+T cells often lost their effector function, the dysfunctional state of CD8+T cells was known as exhaustion. Our objective was to identify genes associated with exhausted CD8+T cells (CD8TEXGs) and their prognostic significance in OC. We downloaded the RNA-seq and clinical data from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. CD8TEXGs were initially identified from single-cell RNA-seq (scRNA-seq) datasets, then univariate Cox regression, the least absolute shrinkage and selection operator (LASSO), and multivariate Cox regression were utilized to calculate risk score and to develop the CD8TEXGs risk signature. Kaplan-Meier analysis, univariate Cox regression, multivariate Cox regression, time-dependent receiver operating characteristics (ROC), nomogram, and calibration were conducted to verify and evaluate the risk signature. Gene set enrichment analyses (GSEA) in the risk groups were used to figure out the closely correlated pathways with the risk group. The role of risk score has been further explored in the homologous recombination repair deficiency (HRD), BRAC1/2 gene mutations and tumor mutation burden (TMB). A risk signature with 4 CD8TEXGs in OC was finally built in the TCGA database and further validated in large GEO cohorts. The signature also demonstrated broad applicability across various types of cancer in the pan-cancer analysis. The high-risk score was significantly associated with a worse prognosis and the risk score was proven to be an independent prognostic biomarker. The 1-, 3-, and 5-years ROC values, nomogram, calibration, and comparison with the previously published models confirmed the excellent prediction power of this model. The low-risk group patients tended to exhibit a higher HRD score, BRCA1/2 gene mutation ratio and TMB. The low-risk group patients were more sensitive to Poly-ADP-ribose polymerase inhibitors (PARPi). Our findings of the prognostic value of CD8TEXGs in prognosis and drug response provided valuable insights into the molecular mechanisms and clinical management of OC.
Collapse
Affiliation(s)
- Tian Hua
- Department of Gynecology, Affiliated Xingtai People Hospital of Hebei Medical University, Xingtai, China
| | - Deng-Xiang Liu
- Department of Oncology, Affiliated Xingtai People Hospital of Hebei Medical University, 16 Hongxing Road, Xingtai, Hebei, 054001, People's Republic of China
| | - Xiao-Chong Zhang
- Department of Oncology, Affiliated Xingtai People Hospital of Hebei Medical University, 16 Hongxing Road, Xingtai, Hebei, 054001, People's Republic of China
| | - Shao-Teng Li
- Department of Oncology, Affiliated Xingtai People Hospital of Hebei Medical University, 16 Hongxing Road, Xingtai, Hebei, 054001, People's Republic of China
| | - Jian-Lei Wu
- Department of Gynecological Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong, 250021, People's Republic of China
| | - Qun Zhao
- The Third Department of Surgery , Hebei Medical University, Fourth Hospital, Road Jiankang No. 12, Hebei, 050001, People's Republic of China.
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, China.
| | - Shu-Bo Chen
- Department of Oncology, Affiliated Xingtai People Hospital of Hebei Medical University, 16 Hongxing Road, Xingtai, Hebei, 054001, People's Republic of China.
| |
Collapse
|
29
|
Huang Y, Zhang Y, Duan X, Hou R, Wang Q, Shi J. Exploring the immune landscape and drug prediction of an M2 tumor-associated macrophage-related gene signature in EGFR-negative lung adenocarcinoma. Thorac Cancer 2024; 15:1626-1637. [PMID: 38886907 PMCID: PMC11260554 DOI: 10.1111/1759-7714.15375] [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: 12/27/2023] [Revised: 05/03/2024] [Accepted: 05/09/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Improving immunotherapy efficacy for EGFR-negative lung adenocarcinoma (LUAD) patients remains a critical challenge, and the therapeutic effect of immunotherapy is largely determined by the tumor microenvironment (TME). Tumor-associated macrophages (TAMs) are the top-ranked immune infiltrating cells in the TME, and M2-TAMs exert potent roles in tumor promotion and chemotherapy resistance. An M2-TAM-based prognostic signature was constructed by integrative analysis of single-cell RNA-seq (scRNA-seq) and bulk RNA-seq data to reveal the immune landscape and select drugs in EGFR-negative LUAD. METHODS M2-TAM-based biomarkers were obtained from the intersection of bulk RNA-seq data and scRNA-seq data. After consensus clustering of EGFR-negative LUAD into different clusters based on M2-TAM-based genes, we compared the prognosis, clinical features, estimate scores, immune infiltration, and checkpoint genes among the clusters. Next, we combined univariate Cox and LASSO regression analyses to establish an M2-TAM-based prognostic signature. RESULTS CCL20, HLA-DMA, HLA-DRB5, KLF4, and TMSB4X were verified as prognostic M2-like TAM-related genes by univariate Cox and LASSO regression analyses. IPS and TMB analyses revealed that the high-risk group responded better to common immunotherapy. CONCLUSION The study shows the potential of the M2-like TAM-related gene signature in EGFR-negative LUAD, explores the immune landscape based on M2-like TAM-related genes, and predict immunotherapy response of patients with EGFR-negative LUAD, providing a new insight for individualized treatment.
Collapse
Affiliation(s)
- Yajie Huang
- Department of Medical OncologyThe Fourth Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Yaozhong Zhang
- Department of Infectious DiseasesThe Fourth Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Xiaoyang Duan
- Department of Medical OncologyThe Fourth Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Ran Hou
- Department of Medical OncologyThe Fourth Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Qi Wang
- Department of EndoscopyThe Fourth Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Jian Shi
- Department of Medical OncologyThe Fourth Hospital of Hebei Medical UniversityShijiazhuangChina
| |
Collapse
|
30
|
Jiao JZ, Zhang Y, Zhang WJ, He MD, Meng M, Liu T, Ma QL, Xu Y, Gao P, Chen CH, Zhang L, Pi HF, Deng P, Wu YZ, Zhou Z, Yu ZP, Deng YC, Lu YH. Radiofrequency radiation reshapes tumor immune microenvironment into antitumor phenotype in pulmonary metastatic melanoma by inducing active transformation of tumor-infiltrating CD8 + T and NK cells. Acta Pharmacol Sin 2024; 45:1492-1505. [PMID: 38538718 PMCID: PMC11192955 DOI: 10.1038/s41401-024-01260-5] [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: 12/25/2023] [Accepted: 03/03/2024] [Indexed: 06/23/2024]
Abstract
Immunosuppression by the tumor microenvironment is a pivotal factor contributing to tumor progression and immunotherapy resistance. Priming the tumor immune microenvironment (TIME) has emerged as a promising strategy for improving the efficacy of cancer immunotherapy. In this study we investigated the effects of noninvasive radiofrequency radiation (RFR) exposure on tumor progression and TIME phenotype, as well as the antitumor potential of PD-1 blockage in a model of pulmonary metastatic melanoma (PMM). Mouse model of PMM was established by tail vein injection of B16F10 cells. From day 3 after injection, the mice were exposed to RFR at an average specific absorption rate of 9.7 W/kg for 1 h per day for 14 days. After RFR exposure, lung tissues were harvested and RNAs were extracted for transcriptome sequencing; PMM-infiltrating immune cells were isolated for single-cell RNA-seq analysis. We showed that RFR exposure significantly impeded PMM progression accompanied by remodeled TIME of PMM via altering the proportion and transcription profile of tumor-infiltrating immune cells. RFR exposure increased the activation and cytotoxicity signatures of tumor-infiltrating CD8+ T cells, particularly in the early activation subset with upregulated genes associated with T cell cytotoxicity. The PD-1 checkpoint pathway was upregulated by RFR exposure in CD8+ T cells. RFR exposure also augmented NK cell subsets with increased cytotoxic characteristics in PMM. RFR exposure enhanced the effector function of tumor-infiltrating CD8+ T cells and NK cells, evidenced by increased expression of cytotoxic molecules. RFR-induced inhibition of PMM growth was mediated by RFR-activated CD8+ T cells and NK cells. We conclude that noninvasive RFR exposure induces antitumor remodeling of the TIME, leading to inhibition of tumor progression, which provides a promising novel strategy for TIME priming and potential combination with cancer immunotherapy.
Collapse
Affiliation(s)
- Jia-Zheng Jiao
- Key Laboratory for Electromagnetic Radiation Medical Protection of Ministry of Education, Army Medical University, Chongqing, 400038, China
- Department of Occupational Health, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Yang Zhang
- Radiation Biology Center, Chongqing University Cancer Hospital, Chongqing, 400030, China
- Radiation Oncology Center, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Wen-Juan Zhang
- Key Laboratory for Electromagnetic Radiation Medical Protection of Ministry of Education, Army Medical University, Chongqing, 400038, China
- Department of Occupational Health, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Min-di He
- Key Laboratory for Electromagnetic Radiation Medical Protection of Ministry of Education, Army Medical University, Chongqing, 400038, China
- Department of Occupational Health, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Meng Meng
- Department of Clinical Hematology, College of Pharmacy and Laboratory Medicine, Army Medical University, Chongqing, 400038, China
| | - Tao Liu
- Department of Clinical Hematology, College of Pharmacy and Laboratory Medicine, Army Medical University, Chongqing, 400038, China
| | - Qin-Long Ma
- Key Laboratory for Electromagnetic Radiation Medical Protection of Ministry of Education, Army Medical University, Chongqing, 400038, China
- Department of Occupational Health, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Ya Xu
- Radiation Biology Center, Chongqing University Cancer Hospital, Chongqing, 400030, China
- Radiation Oncology Center, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Peng Gao
- Key Laboratory for Electromagnetic Radiation Medical Protection of Ministry of Education, Army Medical University, Chongqing, 400038, China
- Department of Occupational Health, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Chun-Hai Chen
- Key Laboratory for Electromagnetic Radiation Medical Protection of Ministry of Education, Army Medical University, Chongqing, 400038, China
- Department of Occupational Health, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Lei Zhang
- Key Laboratory for Electromagnetic Radiation Medical Protection of Ministry of Education, Army Medical University, Chongqing, 400038, China
- Department of Occupational Health, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Hui-Feng Pi
- Key Laboratory for Electromagnetic Radiation Medical Protection of Ministry of Education, Army Medical University, Chongqing, 400038, China
- Department of Occupational Health, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Ping Deng
- Key Laboratory for Electromagnetic Radiation Medical Protection of Ministry of Education, Army Medical University, Chongqing, 400038, China
- Department of Occupational Health, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Yong-Zhong Wu
- Radiation Oncology Center, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Zhou Zhou
- Center for Neurointelligence, School of Medicine, Chongqing University, Chongqing, 400030, China
| | - Zheng-Ping Yu
- Key Laboratory for Electromagnetic Radiation Medical Protection of Ministry of Education, Army Medical University, Chongqing, 400038, China.
- Department of Occupational Health, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China.
| | - You-Cai Deng
- Department of Clinical Hematology, College of Pharmacy and Laboratory Medicine, Army Medical University, Chongqing, 400038, China.
| | - Yong-Hui Lu
- Key Laboratory for Electromagnetic Radiation Medical Protection of Ministry of Education, Army Medical University, Chongqing, 400038, China.
- Department of Occupational Health, College of Preventive Medicine, Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
31
|
Roshan-Zamir M, Khademolhosseini A, Rajalingam K, Ghaderi A, Rajalingam R. The genomic landscape of the immune system in lung cancer: present insights and continuing investigations. Front Genet 2024; 15:1414487. [PMID: 38983267 PMCID: PMC11231382 DOI: 10.3389/fgene.2024.1414487] [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: 04/09/2024] [Accepted: 06/07/2024] [Indexed: 07/11/2024] Open
Abstract
Lung cancer is one of the most prevalent malignancies worldwide, contributing to over a million cancer-related deaths annually. Despite extensive research investigating the genetic factors associated with lung cancer susceptibility and prognosis, few studies have explored genetic predispositions regarding the immune system. This review discusses the most recent genomic findings related to the susceptibility to or protection against lung cancer, patient survival, and therapeutic responses. The results demonstrated the effect of immunogenetic variations in immune system-related genes associated with innate and adaptive immune responses, cytokine, and chemokine secretions, and signaling pathways. These genetic diversities may affect the crosstalk between tumor and immune cells within the tumor microenvironment, influencing cancer progression, invasion, and prognosis. Given the considerable variability in the individual immunegenomics profiles, future studies should prioritize large-scale analyses to identify potential genetic variations associated with lung cancer using highthroughput technologies across different populations. This approach will provide further information for predicting response to targeted therapy and promotes the development of new measures for individualized cancer treatment.
Collapse
Affiliation(s)
- Mina Roshan-Zamir
- School of Medicine, Shiraz Institute for Cancer Research, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Aida Khademolhosseini
- School of Medicine, Shiraz Institute for Cancer Research, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Kavi Rajalingam
- Cowell College, University of California, Santa Cruz, Santa Cruz, CA, United States
| | - Abbas Ghaderi
- School of Medicine, Shiraz Institute for Cancer Research, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Raja Rajalingam
- Immunogenetics and Transplantation Laboratory, University of California San Francisco, San Francisco, CA, United States
| |
Collapse
|
32
|
Cui H, Li H, Liu J, Zhao P, Liu Y, Zhong R, Li R, Cheng Y. The predictive value of E2F7 in immunotherapy efficacy for lung adenocarcinoma: An observational study. Medicine (Baltimore) 2024; 103:e38574. [PMID: 38905387 PMCID: PMC11191985 DOI: 10.1097/md.0000000000038574] [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: 04/24/2024] [Accepted: 05/23/2024] [Indexed: 06/23/2024] Open
Abstract
Lung adenocarcinoma (LUAD) is the most common pathological type of lung cancer. In recent years, immunotherapy has greatly changed the treatment pattern of advanced LUAD. However, only a small proportion of LUAD patients benefitted from immune checkpoint inhibitor therapy. There is an urgent need to develop a biomarker to predict immune therapy response. E2F7 has been shown to be closely related to immune cell infiltration and immune checkpoint expression in tumors. However, it is unclear whether the E2F7 expression is related to the immunotherapy efficacy in LUAD. Therefore, we conducted this study to investigate the clinical characteristics, function, and immunotherapy responsiveness of E2F7 expression, and to explore the potential of E2F7 as an immunotherapy response biomarker in LUAD. We analyzed the clinical characteristics and biological function of E2F7 expression based on data from the Cancer Genome Atlas and Gene Expression Omnibus database. In addition, we used single-cell sequencing data to analyze the immune regulatory effects of E2F7 in LUAD. Furthermore, we analyzed the immunotherapy response prediction ability of E2F7 expression based on the immunotherapy database. Compared to normal lung tissue, E2F7 was specifically overexpressed in LUAD, and its expression was associated with higher malignancy and poor efficacy. E2F7 high expression was an independent risk factor affecting the prognosis of LUAD. E2F7 was enriched in cell division and cell cycle functions. In addition, the expressions of immune checkpoints were correlated with the E2F7 expression. E2F7 was highly expressed in myeloid cells, and E2F7 highly expressed myeloid cells were associated with immune and inflammatory responses. Moreover, the expression level of E2F7 can effectively distinguish different immune therapy responses in LUAD patients. E2F7 was upregulated in LUAD, and high expression of E2F7 was associated with higher malignancy and poor efficacy. E2F7 high expression was an independent risk factor affecting the prognosis of LUAD. Moreover, E2F7 may exert its immunosuppressive effect by affecting the function of myeloid cells. These results indicated the potential role of E2F7 as a biomarker for predicting LUAD immunotherapy responses.
Collapse
Affiliation(s)
- Heran Cui
- Biobank, Jilin Cancer Hospital, Changchun, China
| | - Hui Li
- Biobank, Jilin Cancer Hospital, Changchun, China
- Translational Oncology Research Lab, Jilin Province and Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun, China
| | - Jingjing Liu
- Biobank, Jilin Cancer Hospital, Changchun, China
- Department of Thoracic Oncology, Jilin Cancer Hospital, Changchun, China
| | - Peiyan Zhao
- Translational Oncology Research Lab, Jilin Province and Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun, China
| | - Yan Liu
- Translational Oncology Research Lab, Jilin Province and Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun, China
| | - Rui Zhong
- Translational Oncology Research Lab, Jilin Province and Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun, China
| | - Rixin Li
- Biobank, Jilin Cancer Hospital, Changchun, China
| | - Ying Cheng
- Translational Oncology Research Lab, Jilin Province and Jilin Provincial Key Laboratory of Molecular Diagnostics for Lung Cancer, Jilin Cancer Hospital, Changchun, China
- Department of Thoracic Oncology, Jilin Cancer Hospital, Changchun, China
| |
Collapse
|
33
|
Huang F, Jin L, Zhang X, Wang M, Zhou C. Integrated pan-cancer analysis reveals the immunological and prognostic potential of RBFOX2 in human tumors. Front Pharmacol 2024; 15:1302134. [PMID: 38881877 PMCID: PMC11176534 DOI: 10.3389/fphar.2024.1302134] [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: 09/26/2023] [Accepted: 05/08/2024] [Indexed: 06/18/2024] Open
Abstract
Background The role of RNA-binding fox one homolog 2 (RBFOX2) in the progression of multiple tumors is increasingly supported by evidence. However, the unclearness pertaining to the expression of RBFOX2, its prognostic potential, and its correlation with the tumor microenvironment (TME) in pan-cancer persists. This study aims to comprehensively investigate the immunological prognostic value of RBFOX2. Methods The Cancer Genome Atlas Gene Expression Omnibus Genotype-Tissue Expression (GTEx), TIMER2.0, Kaplan-Meier (K-M) Plotter, University of Alabama at Birmingham Cancer data analysis Portal (UALCAN), cbioportal, and Gene Expression Profiling Interactive Analysis 2 (GEPIA2) were utilized for a systematic analysis of RBFOX2. This analysis included studying its expression, prognostic value, DNA methylation, enrichment analysis, immune infiltration cells, and immune-related genes. Additionally, qRT-PCR, CCK-8, colony formation, transwell assays, and immunohistochemistry were employed to analyze the expression and biological function of RBFOX2 in liver cancer. Results Variations in RBFOX2 expression have been observed across diverse tumors and have been identified as indicators of unfavorable prognosis. It is closely linked to immune infiltration cells, immune checkpoints, chemokines, and chemokine receptors in the TME. Higher levels of RBFOX2 have been significantly associated with low response and poor prognosis in patients with non-small cell lung cancer (NSCLC) and melanoma who receive immunotherapy. Furthermore, the DNA methylation of RBFOX2 varies across different types of cancer and has shown better prognosis in patients with BLCA, BRCA, CESC, COAD, DLBC, HNSC, LAML, LGG, LUAD, PAAD, SKCM and THYM. Interestingly, RBFOX2 expression was found to be lower in hepatocellular carcinoma (HCC) patients' tumor tissues compared to their paired adjacent tissues. In vitro studies have shown that knockdown of RBFOX2 significantly promotes the growth and metastasis of liver cancer cells. Conclusion This study investigates the correlation between DNA methylation, prognostic value, and immune cell infiltration with the expression of RBFOX2 in pan-cancer and indicates its potential role to inhibit metastasis of liver cancer.
Collapse
Affiliation(s)
- Fengxian Huang
- Department of Radiation Oncology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Long Jin
- Department of Radiation Oncology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Xinyue Zhang
- Department of Radiation Oncology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Min Wang
- Department of Science and Education, Xi'an Children's Hospital Affiliated of Xi'an Jiaotong University, Xi'an, China
| | - Congya Zhou
- Department of Radiation Oncology, Shaanxi Provincial People's Hospital, Xi'an, China
| |
Collapse
|
34
|
Guo Z, Zhang X, Yang D, Hu Z, Wu J, Zhou W, Wu S, Zhang W. Gefitinib metabolism-related lncRNAs for the prediction of prognosis, tumor microenvironment and drug sensitivity in lung adenocarcinoma. Sci Rep 2024; 14:10348. [PMID: 38710798 DOI: 10.1038/s41598-024-61175-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 05/02/2024] [Indexed: 05/08/2024] Open
Abstract
The complete compound of gefitinib is effective in the treatment of lung adenocarcinoma. However, the effect on lung adenocarcinoma (LUAD) during its catabolism has not yet been elucidated. We carried out this study to examine the predictive value of gefitinib metabolism-related long noncoding RNAs (GMLncs) in LUAD patients. To filter GMLncs and create a prognostic model, we employed Pearson correlation, Lasso, univariate Cox, and multivariate Cox analysis. We combined risk scores and clinical features to create nomograms for better application in clinical settings. According to the constructed prognostic model, we performed GO/KEGG and GSEA enrichment analysis, tumor immune microenvironment analysis, immune evasion and immunotherapy analysis, somatic cell mutation analysis, drug sensitivity analysis, IMvigor210 immunotherapy validation, stem cell index analysis and real-time quantitative PCR (RT-qPCR) analysis. We built a predictive model with 9 GMLncs, which showed good predictive performance in validation and training sets. The calibration curve demonstrated excellent agreement between the expected and observed survival rates, for which the predictive performance was better than that of the nomogram without a risk score. The metabolism of gefitinib is related to the cytochrome P450 pathway and lipid metabolism pathway, and may be one of the causes of gefitinib resistance, according to analyses from the Gene Set Enrichment Analysis (GSEA), Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Immunological evasion and immunotherapy analysis revealed that the likelihood of immune evasion increased with risk score. Tumor microenvironment analysis found most immune cells at higher concentrations in the low-risk group. Drug sensitivity analysis found 23 sensitive drugs. Twenty-one of these drugs exhibited heightened sensitivity in the high-risk group. RT-qPCR analysis validated the characteristics of 9 GMlncs. The predictive model and nomogram that we constructed have good application value in evaluating the prognosis of patients and guiding clinical treatment.
Collapse
Affiliation(s)
- Zishun Guo
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College , Nanchang University, 1 Minde Road, Nanchang, 330006, China
| | - Xin Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College , Nanchang University, 1 Minde Road, Nanchang, 330006, China
| | - Dingtao Yang
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College , Nanchang University, 1 Minde Road, Nanchang, 330006, China
| | - Zhuozheng Hu
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College , Nanchang University, 1 Minde Road, Nanchang, 330006, China
| | - Jiajun Wu
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College , Nanchang University, 1 Minde Road, Nanchang, 330006, China
| | - Weijun Zhou
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College , Nanchang University, 1 Minde Road, Nanchang, 330006, China
| | - Shuoming Wu
- Department of Thoracic Surgery, The First People's Hospital of Lianyungang, No. 6, Zhenhua East Road, Lianyungang, 222000, China.
| | - Wenxiong Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College , Nanchang University, 1 Minde Road, Nanchang, 330006, China.
| |
Collapse
|
35
|
Zhong Y, Cao H, Li W, Deng J, Li D, Deng J. An analysis of the prognostic role of reactive oxygen species-associated genes in breast cancer. ENVIRONMENTAL TOXICOLOGY 2024; 39:3055-3148. [PMID: 38319140 DOI: 10.1002/tox.24128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 12/11/2023] [Accepted: 12/25/2023] [Indexed: 02/07/2024]
Abstract
BACKGROUND This study aimed to type breast cancer in relation to reactive oxygen species (ROS), clinical indicators, single nucleotide variant (SNV) mutations, functional differences, immune infiltration, and predictive responses to immunotherapy or chemotherapy, and constructing a prognostic model. METHODS We used uniCox analysis, ConsensusClusterPlus, and the proportion of ambiguous clustering (PAC) to analyze The Cancer Genome Atlas (TCGA) data to determine optimal groupings and obtain differentially expressed ROS-related genes. Clinical indicators were then combined with the classification results and the Chi-square test was used to assess differences. We further examined SNV mutations, and functional differences using gene set enrichment analysis (GSEA) analysis, the Kyoto Encyclopedia of Genes and Genomes (KEGG) database, immune cell infiltration, and response to immunotherapy and chemotherapy. A prognostic model for breast cancer was constructed using these differentially expressed genes, immunotherapy or chemotherapy responses, and survival curves. RT-qPCR was used to detect the differences in the expression of LCE3D, CA1, PIRT and SMR3A in breast cancer cell lines and normal breast epithelial cell line. RESULTS We identified two distinct tumor types with significant differences in ROS-related gene expression, clinical indicators, SNV mutations, functional pathways, and immune infiltration. The response to specific chemotherapy drugs and immunotherapy treatments also documented significant differences. The prognostic model constructed with 16 genes linked to survival could efficiently divide patients into high- and low-risk groups. The high-risk group showed a poorer prognosis, higher tumor purity, distinct immune microenvironment, and lower immunotherapy response. RT-qPCR results showed that LCE3D, CA1, PIRT and SMR3A are highly expressed in breast cancer. CONCLUSION Our methodical examination presented an enhanced insight into the molecular and immunological heterogeneity of breast cancer. It can contribute to the understanding of prognosis and offer valuable insights for personalized treatment strategies. Further, the prognostic model can potentially serve as a powerful tool for risk stratification and therapeutic decision-making in clinical settings.
Collapse
Affiliation(s)
- Yangyan Zhong
- The Second Affiliated Hospital, Department of Breast and Thyroid Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Clinical Research Center for Breast and Thyroid Disease Prevention and Control in Hunan Province, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Hong Cao
- The Second Affiliated Hospital, Department of Breast and Thyroid Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Clinical Research Center for Breast and Thyroid Disease Prevention and Control in Hunan Province, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Wei Li
- The Second Affiliated Hospital, Department of Breast and Thyroid Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Clinical Research Center for Breast and Thyroid Disease Prevention and Control in Hunan Province, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jian Deng
- The Second Affiliated Hospital, Department of Breast and Thyroid Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Clinical Research Center for Breast and Thyroid Disease Prevention and Control in Hunan Province, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Dan Li
- The Second Affiliated Hospital, Department of Breast and Thyroid Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Clinical Research Center for Breast and Thyroid Disease Prevention and Control in Hunan Province, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Junjie Deng
- The Second Affiliated Hospital, Department of Breast and Thyroid Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Clinical Research Center for Breast and Thyroid Disease Prevention and Control in Hunan Province, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
36
|
Lai C, Wu Z, Li Z, Huang X, Hu Z, Yu H, Yuan Z, Shi J, Hu J, Mulati Y, Liu C, Xu K. Single-cell analysis extracted CAFs-related genes to established online app to predict clinical outcome and radiotherapy prognosis of prostate cancer. Clin Transl Oncol 2024; 26:1240-1255. [PMID: 38070051 DOI: 10.1007/s12094-023-03348-6] [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: 09/14/2023] [Accepted: 11/03/2023] [Indexed: 04/20/2024]
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) play a significant role in regulating the clinical outcome and radiotherapy prognosis of prostate cancer (PCa). The aim of this study is to identify CAFs-related genes (CAFsRGs) using single-cell analysis and evaluate their potential for predicting the prognosis and radiotherapy prognosis in PCa. METHODS We acquire transcriptome and single-cell RNA sequencing (scRNA-seq) results of PCa and normal adjacent tissues from The GEO and TCGA databases. The "MCPcounter" and "EPIC" R packages were used to assess the infiltration level of CAFs and examine their correlation with PCa prognosis. ScRNA-seq and differential gene expression analyses were used to extract CAFsRGs. We also applied COX and LASSO analysis to further construct a risk score (CAFsRS) to assess biochemical recurrence-free survival (BRFS) and radiotherapy prognosis of PCa. The predictive efficacy of CAFsRS was evaluated by ROC curves and subgroup analysis. Finally, we integrated the CAFsRS gene signature with relevant clinical features to develop a nomogram, enhancing the predictive accuracy. RESULTS The abundance of CAFs is associated with a poor prognosis of PCa patients. ScRNA-seq and differential gene expression analysis revealed 323 CAFsRGs. After COX and LASSO analysis, we obtained seven CAFsRGs with prognostic significance (PTGS2, FKBP10, ENG, CDH11, COL5A1, COL5A2, and SRD5A2). Additionally, we established a risk score model based on the training set (n = 257). The ROC curve was used to confirm the performance of CAFsRS (The AUC values for 1, 3 and 5-year survival were determined to be 0.732, 0.773, and 0.775, respectively.). The testing set (n = 129), GSE70770 set (n = 199) and GSE116918 set (n = 248) revealed that the model exhibited exceptional predictive performance. This was also confirmed by clinical subgroup analysis. The violin plot demonstrated a statistically significant disparity in the CAFs infiltrations between the high-risk and low-risk groups of CAFsRS. Further analysis confirmed that both CAFsRS and T stage were independent prognostic factors for PCa. The nomogram was then established and its excellent predictive performance was demonstrated through calibration and ROC curves. Finally, we developed an online prognostic prediction app ( https://sysu-symh-cafsnomogram.streamlit.app/ ) to facilitate the practical application of the nomogram. CONCLUSIONS The prognostic prediction risk score model we constructed could accurately predict BRFS and radiotherapy prognosis PCa, which can provide new ideas for clinicians to develop personalized PCa treatment and follow-up programs.
Collapse
Affiliation(s)
- Cong Lai
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China
| | - Zhikai Wu
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China
| | - Zhuohang Li
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, 510000, Guangdong, China
| | - Xin Huang
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China
| | - Zhensheng Hu
- Department of Medical Informatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China
| | - Hao Yu
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, 510000, Guangdong, China
| | - Zhihan Yuan
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China
| | - Juanyi Shi
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, 510000, Guangdong, China
| | - Jintao Hu
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China
| | - Yelisudan Mulati
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China
| | - Cheng Liu
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China.
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, 510000, Guangdong, China.
| | - Kewei Xu
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510000, Guangdong, China.
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, 510000, Guangdong, China.
- Sun Yat-Sen College of Medical Science, Sun Yat-Sen University, Shenzhen, 518000, Guangdong, China.
| |
Collapse
|
37
|
Zhang J, Li Z, Chen Z, Shi W, Xu Y, Huang Z, Lin Z, Dou R, Lin S, Jiang X, Li M, Jiang S. Comprehensive analysis of macrophage-related genes in prostate cancer by integrated analysis of single-cell and bulk RNA sequencing. Aging (Albany NY) 2024; 16:6809-6838. [PMID: 38663915 PMCID: PMC11087116 DOI: 10.18632/aging.205727] [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: 10/30/2023] [Accepted: 01/30/2024] [Indexed: 05/08/2024]
Abstract
Macrophages, as essential components of the tumor immune microenvironment (TIME), could promote growth and invasion in many cancers. However, the role of macrophages in tumor microenvironment (TME) and immunotherapy in PCa is largely unexplored at present. Here, we investigated the roles of macrophage-related genes in molecular stratification, prognosis, TME, and immunotherapeutic response in PCa. Public databases provided single-cell RNA sequencing (scRNA-seq) and bulk RNAseq data. Using the Seurat R package, scRNA-seq data was processed and macrophage clusters were identified automatically and manually. Using the CellChat R package, intercellular communication analysis revealed that tumor-associated macrophages (TAMs) interact with other cells in the PCa TME primarily through MIF - (CD74+CXCR4) and MIF - (CD74+CD44) ligand-receptor pairs. We constructed coexpression networks of macrophages using the WGCNA to identify macrophage-related genes. Using the R package ConsensusClusterPlus, unsupervised hierarchical clustering analysis identified two distinct macrophage-associated subtypes, which have significantly different pathway activation status, TIME, and immunotherapeutic efficacy. Next, an 8-gene macrophage-related risk signature (MRS) was established through the LASSO Cox regression analysis with 10-fold cross-validation, and the performance of the MRS was validated in eight external PCa cohorts. The high-risk group had more active immune-related functions, more infiltrating immune cells, higher HLA and immune checkpoint gene expression, higher immune scores, and lower TIDE scores. Finally, the NCF4 gene has been identified as the hub gene in MRS using the "mgeneSim" function.
Collapse
Affiliation(s)
- Jili Zhang
- Department of Urology, The First Navy Hospital of Southern Theater Command, Zhanjiang, Guangdong, China
| | - Zhihao Li
- Center of Reproductive Medicine, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhenlin Chen
- Department of Urology, Fujian Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Wenzhen Shi
- Department of Urology, Fujian Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Yue Xu
- Department of Urology, Fujian Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhangcheng Huang
- Department of Urology, Fujian Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Zequn Lin
- Department of Urology, Fujian Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Ruiling Dou
- Department of Urology, Fujian Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Shaoshan Lin
- Department of Urology, Fujian Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Xin Jiang
- Department of Urology, The First Navy Hospital of Southern Theater Command, Zhanjiang, Guangdong, China
| | - Mengqiang Li
- Department of Urology, Fujian Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Shaoqin Jiang
- Department of Urology, Fujian Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
38
|
Sun JR, Kong CF, Ye YX, Wang Q, Qu XK, Jia LQ, Wu S. Integrated analysis of single-cell and bulk RNA-sequencing reveals a novel signature based on NK cell marker genes to predict prognosis and immunotherapy response in gastric cancer. Sci Rep 2024; 14:7648. [PMID: 38561388 PMCID: PMC10985121 DOI: 10.1038/s41598-024-57714-7] [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: 12/25/2023] [Accepted: 03/21/2024] [Indexed: 04/04/2024] Open
Abstract
Natural killer (NK) cells play essential roles in the tumor development, diagnosis, and prognosis of tumors. In this study, we aimed to establish a reliable signature based on marker genes in NK cells, thus providing a new perspective for assessing immunotherapy and the prognosis of patients with gastric cancer (GC). We analyzed a total of 1560 samples retrieved from the public database. We performed a comprehensive analysis of single-cell RNA-sequencing (scRNA-seq) data of gastric cancer and identified 377 marker genes for NK cells. By performing Cox regression analysis, we established a 12-gene NK cell-associated signature (NKCAS) for the Cancer Genome Atlas (TCGA) cohort, that assigned GC patients into a low-risk group (LRG) or a high-risk group (HRG). In the TCGA cohort, the areas under curve (AUC) value were 0.73, 0.81, and 0.80 at 1, 3, and 5 years. External validation of the predictive ability for the signature was then validated in the Gene Expression Omnibus (GEO) cohorts (GSE84437). The expression levels of signature genes were measured and validated in GC cell lines by real-time PCR. Moreover, NKCAS was identified as an independent prognostic factor by multivariate analysis. We combined this with a variety of clinicopathological characteristics (age, M stage, and tumor grade) to construct a nomogram to predict the survival outcomes of patients. Moreover, the LRG showed higher immune cell infiltration, especially CD8+ T cells and NK cells. The risk score was negatively associated with inflammatory activities. Importantly, analysis of the independent immunotherapy cohort showed that the LRG had a better prognosis and immunotherapy response when compared with the HRG. The identification of NK cell marker genes in this study suggests potential therapeutic targets. Additionally, the developed predictive signatures and nomograms may aid in the clinical management of GC.
Collapse
Affiliation(s)
- Jian-Rong Sun
- School of Clinical Medicine, Beijing University of Chinese Medicine, No. 11, North 3rd East Road, Beijing, 100029, Chaoyang, People's Republic of China
| | - Chen-Fan Kong
- Department of Urology, The affiliated Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, No. 16, Liantangxiantong Road, Shenzhen, 518009, Luohu, People's Republic of China
| | - Yi-Xiang Ye
- School of Clinical Medicine, Beijing University of Chinese Medicine, No. 11, North 3rd East Road, Beijing, 100029, Chaoyang, People's Republic of China
| | - Qin Wang
- School of Clinical Medicine, Beijing University of Chinese Medicine, No. 11, North 3rd East Road, Beijing, 100029, Chaoyang, People's Republic of China
| | - Xiang-Ke Qu
- School of Clinical Medicine, Beijing University of Chinese Medicine, No. 11, North 3rd East Road, Beijing, 100029, Chaoyang, People's Republic of China
| | - Li-Qun Jia
- School of Clinical Medicine, Beijing University of Chinese Medicine, No. 11, North 3rd East Road, Beijing, 100029, Chaoyang, People's Republic of China.
| | - Song Wu
- Department of Urology, The affiliated Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, No. 16, Liantangxiantong Road, Shenzhen, 518009, Luohu, People's Republic of China.
- Department of Urology, South China Hospital, Health Science Center, Shenzhen University, Shenzhen, 518116, People's Republic of China.
| |
Collapse
|
39
|
Wang H, An N, Pei A, Sun Y, Li S, Chen S, Zhang N. Exploration of signature based on T cell-related genes in stomach adenocarcinoma by analysis of single cell sequencing data. Aging (Albany NY) 2024; 16:6035-6053. [PMID: 38536020 PMCID: PMC11042963 DOI: 10.18632/aging.205687] [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: 10/10/2023] [Accepted: 12/29/2023] [Indexed: 04/23/2024]
Abstract
BACKGROUND Gastric cancer (GC) is a leading reason for the death of cancer around the world. The immune microenvironment counts a great deal in immunotherapy of advanced tumors, in which T cells exert an indispensable function. METHODS Single-cell RNA sequencing data were utilized to characterize the expression profile of T cells, followed by T cell-related genes (TCRGs) to construct signature and measure differences in survival time, enrichment pathways, somatic mutation status, immune status, and immunotherapy between groups. RESULTS The complex tumor microenvironment was analyzed by scRNA-seq data of GC patients. We screened for these T cell signature expression genes and the TCRGs-based signature was successfully constructed and relied on the riskscore grouping. In gene set enrichment analysis, it was shown that pro-tumor and suppressive immune pathways were more abundant in the higher risk group. We also found different infiltration of immune cells in two groups, and that the higher risk samples had a poorer response to immunotherapy. CONCLUSION Our study established a prognostic model, in which different groups had different prognosis, immune status, and enriched features. These results have provided additional insights into prognostic evaluation and the development of highly potent immunotherapies in GC.
Collapse
Affiliation(s)
- Huimei Wang
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, China
| | - Nan An
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Aiyue Pei
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, China
| | - Yongxiao Sun
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, China
| | - Shuo Li
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, China
| | - Si Chen
- Department of Colorectal and Anal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Nan Zhang
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
40
|
Lian C, Li F, Xie Y, Zhang L, Chen H, Wang Z, Pan X, Wang X, Zhang J. Identification of T-cell exhaustion-related genes and prediction of their immunotherapeutic role in lung adenocarcinoma. J Cancer 2024; 15:2160-2178. [PMID: 38495503 PMCID: PMC10937285 DOI: 10.7150/jca.92839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 02/06/2024] [Indexed: 03/19/2024] Open
Abstract
Background: Lung adenocarcinoma ranks as the second most widespread form of cancer globally, accompanied by a significant mortality rate. Several studies have shown that T cell exhaustion is associated with immunotherapy of tumours. Consequently, it is essential to comprehend the possible impact of T cell exhaustion on the tumor microenvironment. The purpose of this research was to create a TEX-based model that would use single-cell RNA-seq (scRNA-seq) and bulk-RNA sequencing to explore new possibilities for assessing the prognosis and immunotherapeutic response of LUAD patients. Methods: RNA-seq data from LUAD patients was downloaded from the Cancer Genome Atlas (TCGA) database and the National Center for Biotechnology Information (GEO). 10X scRNA sequencing data, as reported by Bischoff P et al., was utilized for down-sampling clustering and subgroup identification using TSNE. TEX-associated genes were identified through gene set variance analysis (GSVA) and weighted gene correlation network analysis (WGCNA). We utilized LASSO-Cox analysis to establish predicted TEX features. External validation was conducted in GSE31210 and GSE30219 cohorts. Immunotherapeutic response was assessed in IMvigor210, GSE78220, GSE35640 and GSE100797 cohorts. Furthermore, we investigated differences in mutational profiles and immune microenvironment between various risk groups. We then screened TEXRS key regulatory genes using ROC diagnostic curves and KM survival curves. Finally, we verified the differential expression of key regulatory genes through RT-qPCR. Results: Nine TEX genes were identified as highly predictive of LUAD prognosis and strongly correlated with disease outcome. Univariate and multivariate analysis revealed that patients in the low-risk group had significantly better overall survival rates compared with those in the high-risk group, highlighting the model's ability to independently predict LUAD prognosis. Our analysis revealed significant variation in the biological function, mutational landscape, and immune cell infiltration within the tumor microenvironment of both high-risk and low-risk groups. Additionally, immunotherapy was found to have a significant impact on both groups, indicating strong predictive efficacy of the model. Conclusions: The TEX model showed good predictive performance and provided a new perspective for evaluating the efficacy of preimmunization, which provides a new strategy for the future treatment of lung adenocarcinoma.
Collapse
Affiliation(s)
- Chaoqun Lian
- Research Center of Clinical Laboratory Science, Bengbu Medical University, Bengbu 233030, China
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu 233030, China
| | - Feifan Li
- Department of Tumor Radiotherapy, The First Affiliated Hospital of Bengbu Medical University, Bengbu 233030, China
| | - Yiluo Xie
- Department of Clinical Medicine, Bengbu Medical University, Bengbu 233030, China
| | - Linxiang Zhang
- Research Center of Clinical Laboratory Science, Bengbu Medical University, Bengbu 233030, China
| | - Huili Chen
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu 233030, China
| | - Ziqiang Wang
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu 233030, China
| | - Xinyu Pan
- Department of Medical Imaging, Bengbu Medical University, Bengbu 233030, China
| | - Xiaojing Wang
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu 233030, China
| | - Jing Zhang
- Department of Genetics, School of Life Sciences, Bengbu Medical University, Bengbu 233030, China
| |
Collapse
|
41
|
Shi S, Chu Y, Liu H, Yu L, Sun D, Yang J, Tian G, Ji L, Zhang C, Lu X. Predictable regulation of survival by intratumoral microbe-immune crosstalk in patients with lung adenocarcinoma. MICROBIAL CELL (GRAZ, AUSTRIA) 2024; 11:29-40. [PMID: 38375207 PMCID: PMC10876218 DOI: 10.15698/mic2024.02.813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 01/09/2024] [Accepted: 01/16/2024] [Indexed: 02/21/2024]
Abstract
Intratumoral microbiota can regulate the tumor immune microenvironment (TIME) and mediate tumor prognosis by promoting inflammatory response or inhibiting anti-tumor effects. Recent studies have elucidated the potential role of local tumor microbiota in the development and progression of lung adenocarcinoma (LUAD). However, whether intratumoral microbes are involved in the TIME that mediates the prognosis of LUAD remains unknown. Here, we obtained the matched tumor microbiome and host transcriptome and survival data of 478 patients with LUAD in The Cancer Genome Atlas (TCGA). Machine learning models based on immune cell marker genes can predict 1- to 5-year survival with relative accuracy. Patients were stratified into high- and low-survival-risk groups based on immune cell marker genes, with significant differences in intratumoral microbial communities. Specifically, patients in the high-risk group had significantly higher alpha diversity (p < 0.05) and were characterized by an enrichment of lung cancer-related genera such as Streptococcus. However, network analysis highlighted a more active pattern of dominant bacteria and immune cell crosstalk in TIME in the low-risk group compared to the high-risk group. Our study demonstrated that intratumoral microbiota-immune crosstalk was strongly associated with prognosis in LUAD patients, which would provide new targets for the development of precise therapeutic strategies.
Collapse
Affiliation(s)
- Shuo Shi
- The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Yuwen Chu
- Geneis Beijing Co., Ltd., Beijing 100102, China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao 266000, Shandong, China
| | - Haiyan Liu
- College of Information Engineering, Changsha Medical University, Changsha 410219, Hunan, China
- Academician Workstation, Changsha Medical University, Changsha 410219, Hunan, China
| | - Lan Yu
- Clinical Medical Research Center, Inner Mongolian People's Hospital, No. 20, Zhaowuda Road, Hohhot, Inner Mongolia, China
- Inner Mongolia Key Laboratory of Gene Regulation of The Metabolic Disease, Inner Mongolian People's Hospital, No. 20, Zhaowuda Road, Hohhot, Inner Mongolia, China
- Inner Mongolia Academy of Medical Sciences, Inner Mongolian People's Hospital, No. 20, Zhaowuda Road, Hohhot, Inner Mongolia, China
| | - Dejun Sun
- Inner Mongolia Academy of Medical Sciences, Inner Mongolian People's Hospital, No. 20, Zhaowuda Road, Hohhot, Inner Mongolia, China
- Pulmonary and Critical Care Medicine, Inner Mongolian People's Hospital, No. 20, Zhaowuda Road, Saihan District, Hohhot, Inner Mongolia, China
| | - Jialiang Yang
- Geneis Beijing Co., Ltd., Beijing 100102, China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao 266000, Shandong, China
- Academician Workstation, Changsha Medical University, Changsha 410219, Hunan, China
| | - Geng Tian
- Geneis Beijing Co., Ltd., Beijing 100102, China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao 266000, Shandong, China
| | - Lei Ji
- Geneis Beijing Co., Ltd., Beijing 100102, China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao 266000, Shandong, China
| | - Cong Zhang
- Hospital of Chengdu University of Traditional Chinese Medicine/No. 39, 12th Bridge Road, Jinniu District, Chengdu City, Sichuan Province, 610072, China
| | - Xinxin Lu
- Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research
| |
Collapse
|
42
|
Lin MX, Zang D, Liu CG, Han X, Chen J. Immune checkpoint inhibitor-related pneumonitis: research advances in prediction and management. Front Immunol 2024; 15:1266850. [PMID: 38426102 PMCID: PMC10902117 DOI: 10.3389/fimmu.2024.1266850] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 02/02/2024] [Indexed: 03/02/2024] Open
Abstract
The advent of immune-checkpoint inhibitors (ICIs) has revolutionized the treatment of malignant solid tumors in the last decade, producing lasting benefits in a subset of patients. However, unattended excessive immune responses may lead to immune-related adverse events (irAEs). IrAEs can manifest in different organs within the body, with pulmonary toxicity commonly referred to as immune checkpoint inhibitor-related pneumonitis (CIP). The CIP incidence remains high and is anticipated to rise further as the therapeutic indications for ICIs expand to encompass a wider range of malignancies. The diagnosis and treatment of CIP is difficult due to the large individual differences in its pathogenesis and severity, and severe CIP often leads to a poor prognosis for patients. This review summarizes the current state of clinical research on the incidence, risk factors, predictive biomarkers, diagnosis, and treatment for CIP, and we address future directions for the prevention and accurate prediction of CIP.
Collapse
Affiliation(s)
| | | | | | | | - Jun Chen
- Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
43
|
Xu H, Hu Y, Peng X, Chen E. Prediction of prognostic and immune therapy response in lung adenocarcinoma based on MHC-I-related genes. Immunopharmacol Immunotoxicol 2024; 46:93-106. [PMID: 37728543 DOI: 10.1080/08923973.2023.2261146] [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: 05/23/2023] [Accepted: 09/14/2023] [Indexed: 09/21/2023]
Abstract
OBJECTIVES The study investigated the prognostic and immune predictive potential of major histocompatibility complex class I (MHC-I) in lung adenocarcinoma (LUAD). MATERIALS AND METHODS With The Cancer Genome Atlas (TCGA)-LUAD and Gene Expression Omnibus datasets (GSE26939, GSE72094) as the training and validation sets, respectively, we used Cox regression analysis to construct a prognostic model, and verified independence of riskscore. The predictive capacity of the model was assessed in both sets using the receiver operating characteristic curve and Kaplan-Meier survival curves. Immune analysis was performed by using ssGSEA. Additionally, immune checkpoint blockade therapy was assessed by using immunophenoscore, Tumor Immune Dysfunction and Exclusion score. Based on the cMAP database, effective small molecule compounds were predicted. RESULTS A prognostic model was established based on 8 MHC-I-related genes, and the predictive capacity of the model was accurate. Immune analysis results revealed that patients classified as high-risk had lower levels of immune cell infiltration and impaired immune function. The low-risk group possessed a better response to immune checkpoint blockade therapy. Theobromine and pravastatin were identified as having great potential in improving the prognosis of LUAD. CONCLUSION Overall, the study revealed MHC-I-related molecular prognostic biomarkers as robust indicators for LUAD prognosis and immune therapy response.
Collapse
Affiliation(s)
| | | | - Xiuming Peng
- Department of Pulmonary and Critical Care Medicine, Regional Medical Center for National Institute of Respiratory Disease, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Enguo Chen
- Department of Pulmonary and Critical Care Medicine, Regional Medical Center for National Institute of Respiratory Disease, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
44
|
Chen H, Xu N, Xu J, Zhang C, Li X, Xu H, Zhu W, Li J, Liang D, Zhou W. A risk signature based on endoplasmic reticulum stress-associated genes predicts prognosis and immunity in pancreatic cancer. Front Mol Biosci 2023; 10:1298077. [PMID: 38106991 PMCID: PMC10721979 DOI: 10.3389/fmolb.2023.1298077] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 11/15/2023] [Indexed: 12/19/2023] Open
Abstract
Introduction: The involvement of endoplasmic reticulum (ER) stress in cancer biology is increasingly recognized, yet its role in pancreatic cancer (PC) remains unclear. This study aims to elucidate the impact of ER stress on prognosis and biological characteristics in PC patients. Methods: A bioinformatic analysis was conducted using RNA-seq data and clinicopathological information from PC patients in the TCGA and ICGC databases. The ER stress-associated gene sets were extracted from MSigDB. ER stress-associated genes closely linked with overall survival (OS) of PC patients were identified via log-rank test and univariate Cox analysis, and further narrowed by LASSO method. A risk signature associated with ER stress was formulated using multivariate Cox regression and assessed through Kaplan-Meier curves, receiver operating characteristic (ROC) analyses, and Harrell's concordance index. External validation was performed with the ICGC cohort. The single-sample gene-set enrichment analysis (ssGSEA) algorithm appraised the immune cell infiltration landscape. Results: Worse OS in PC patients with high-risk signature score was observed. Multivariate analysis underscored our ER stress-associated signature as a valuable and independent predictor of prognosis. Importantly, these results based on TCGA were further validated in ICGC dataset. In addition, our risk signature was closely associated with homeostasis, protein secretion, and immune regulation in PC patients. In particular, PC microenvironment in the high-risk cluster exhibited a more immunosuppressive status. At last, we established a nomogram model by incorporating the risk signature and clinicopathological parameters, which behaves better in predicting prognosis of PC patients. Discussion: This comprehensive molecular analysis presents a new predictive model for the prognosis of PC patients, highlighting ER stress as a potential therapeutic target. Besides, the findings indicate that ER stress can have effect modulating PC immune responses.
Collapse
Affiliation(s)
- Haofei Chen
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Ning Xu
- The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jia Xu
- Wuhan Blood Center, Wuhan, China
| | - Cheng Zhang
- The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xin Li
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Hao Xu
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Weixiong Zhu
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Jinze Li
- Department of Gastrointestinal Surgery, The Third People’s Hospital of Hubei Province, Wuhan, China
| | - Daoming Liang
- The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Wence Zhou
- The Second Clinical Medical School, Lanzhou University, Lanzhou, China
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
45
|
Chen Q, Zhao H, Hu J. A robust six-gene prognostic signature based on two prognostic subtypes constructed by chromatin regulators is correlated with immunological features and therapeutic response in lung adenocarcinoma. Aging (Albany NY) 2023; 15:12330-12368. [PMID: 37938151 PMCID: PMC10683604 DOI: 10.18632/aging.205183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 10/02/2023] [Indexed: 11/09/2023]
Abstract
Accumulating evidence has demonstrated that chromatin regulators (CRs) regulate immune cell infiltration and are correlated with prognoses of patients in some cancers. However, the immunological and prognostic roles of CRs in lung adenocarcinoma (LUAD) are still unclear. Here, we systematically revealed the correlations of CRs with immunological features and the survival in LUAD patients based on a cohort of gene expression datasets from the public TCGA and GEO databases and real RNA-seq data by an integrative analysis using a comprehensive bioinformatics method. Totals of 160 differentially expressed CRs (DECRs) were identified between LUAD and normal lung tissues, and two molecular prognostic subtypes (MPSs) were constructed and evaluated based on 27 prognostic DECRs using five independent datasets (p =0.016, <0.0001, =0.008, =0.00038 and =0.00055, respectively). Six differentially expressed genes (DEGs) (CENPK, ANGPTL4, CCL20, CPS1, GJB3, TPSB2) between two MPSs had the most important prognostic feature and a six-gene prognostic model was established. LUAD patients in the low-risk subgroup showed a higher overall survival (OS) rate than those in the high-risk subgroup in nine independent datasets (p <0.0001, =0.021, =0.016, =0.0099, <0.0001, =0.0045, <0.0001, =0.0038 and =0.00013, respectively). Six-gene prognostic signature had the highest concordance index of 0.673 compared with 19 reported prognostic signatures. The risk score was significantly correlated with immunological features and activities of oncogenic signaling pathways. LUAD patients in the low-risk subgroup benefited more from immunotherapy and were less sensitive to conventional chemotherapy agents. This study provides novel insights into the prognostic and immunological roles of CRs in LUAD.
Collapse
Affiliation(s)
- Qiang Chen
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Hongbo Zhao
- Department of Laboratory Animal Science, Kunming Medical University, Kunming, China
| | - Jing Hu
- Department of Medical Oncology, First People’s Hospital of Yunnan Province, Kunming, China
- Department of Medical Oncology, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
46
|
Zhang L, Guan M, Zhang X, Yu F, Lai F. Machine-learning and combined analysis of single-cell and bulk-RNA sequencing identified a DC gene signature to predict prognosis and immunotherapy response for patients with lung adenocarcinoma. J Cancer Res Clin Oncol 2023; 149:13553-13574. [PMID: 37507593 PMCID: PMC10590321 DOI: 10.1007/s00432-023-05151-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 07/09/2023] [Indexed: 07/30/2023]
Abstract
BACKGROUND Innate immune effectors, dendritic cells (DCs), influence cancer prognosis and immunotherapy significantly. As such, dendritic cells are important in killing tumors and influencing tumor microenvironment, whereas their roles in lung adenocarcinoma (LUAD) are largely unknown. METHODS In this study, 1658 LUAD patients from different cohorts were included. In addition, 724 cancer patients who received immunotherapy were also included. To identify DC marker genes in LUAD, we used single-cell RNAsequencing data for analysis and determined 83 genes as DC marker genes. Following that, integrative machine learning procedure was developed to construct a signature for DC marker genes. RESULTS Using TCGA bulk-RNA sequencing data as the training set, we developed a signature consisting of seven genes and classified patients by their risk status. Another six independent cohorts demonstrated the signature' s prognostic power, and multivariate analysis demonstrated it was an independent prognostic factor. LUAD patients in the high-risk group displayed more advanced features, discriminatory immune-cell infiltrations and immunosuppressive states. Cell-cell communication analysis indicates that tumor cells with lower risk scores communicate more actively with the tumor microenvironment. Eight independent immunotherapy cohorts revealed that patients with low-risk had better immunotherapy responses. Drug sensitivity analysis indicated that targeted therapy agents exhibited greater sensitivity to low-risk patients, while chemotherapy agents displayed greater sensitivity to high-risk patients. In vitro experiments confirmed that CTSH is a novel protective factor for LUAD. CONCLUSIONS An unique signature based on DC marker genes that is highly predictive of LUAD patients' prognosis and response to immunotherapy. CTSH is a new biomarker for LUAD.
Collapse
Affiliation(s)
- Liangyu Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Maohao Guan
- Department of Thoracic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Xun Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Fengqiang Yu
- Department of Thoracic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
| | - Fancai Lai
- Department of Thoracic Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Department of Thoracic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
| |
Collapse
|
47
|
Huang L, Zhong L, Cheng R, Chang L, Qin M, Liang H, Liao Z. Ferroptosis and WDFY4 as novel targets for immunotherapy of lung adenocarcinoma. Aging (Albany NY) 2023; 15:9676-9694. [PMID: 37728413 PMCID: PMC10564425 DOI: 10.18632/aging.205042] [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: 02/22/2023] [Accepted: 08/21/2023] [Indexed: 09/21/2023]
Abstract
BACKGROUND Lung cancer exhibits the world's highest mortality rate among malignant cancers worldwide, thereby presenting a significant global challenge in terms of reducing patient mortality. In the field of oncology, targeted immunotherapy has emerged as a novel therapeutic approach for lung cancer. This study aims to explore potential targets for immunotherapy in lung adenocarcinoma (LUAD) through the analysis of Ferroptosis Index (FPI) and Single Cell RNA-Sequencing (scRNA-seq) data. The findings of this research can potentially offer valuable insights for improving LUAD immunotherapy strategies and informing clinical decision-making. METHODS Firstly, the relationship between survival and ferroptosis in LUAD patients was analyzed by FPI. Subsequently, the association between ferroptosis and infiltration and regulation of immune cells was explored by immune infiltration analysis and correlation statistics. Lastly, the relationship between major infiltrating immune cell populations and related pathways and prognosis of LUAD patients was analyzed by GSEA and GSVA. To screen out core genes regulating infiltration of immune cell populations, scRNA-seq data of cancer and para-cancerous tissues of LUAD patients were downloaded, followed by cell clustering analysis, cell identification of core subpopulations, pseudotime analysis, single-cell GSVA and pathway enrichment analysis, and identification and functional analysis of core regulatory genes. Moreover, the expression levels of core functional genes in LUAD tissue microarray were detected by immunohistochemistry, and its relationship with the prognosis of LUAD patients was verified. Finally, we used lentivirus with WDFY4 to transfect LUAD A549 cells. CCK-8, flow cytometry apoptosis detection, Scratch wound healing assay, Transwell migration assay, Xenograft nude mice model, immunohistochemical analysis and other experimental methods were used to explore the biological effects of WDFY4 on LUAD in vitro and in vivo. RESULTS Survival analysis of FPI values in LUAD patients revealed a positive correlation between smaller FPI values and longer overall survival. Immuno-infiltration analysis and its correlation with FPI values revealed that B cells were most strongly associated with ferroptosis. Ferroptosis of cancer cells could promote infiltration and activation of B cell populations, and LUAD patients with more infiltration of B cell populations had longer long-term survival. scRNA-seq data analysis indicated that the B cell population is one of the major cell populations infiltrated by immune cells in LUAD. During the later phases of B cell differentiation in LUAD, there was a decrease in the expression levels of ACAP1, LINC00926, TLR10, MS4A1, WDFY4, and TRIM22 genes, whereas the expression levels of TMEM59, TP53INP1, and METTL7A genes were elevated. The protein-protein interaction (PPI) network analysis indicated that WDFY4 plays a crucial role in regulating B cell differentiation in LUAD. Immunohistochemical analysis of LUAD tissue microarray revealed a significant downregulation of WDFY4 expression, which was closely related to the occurrence sites of LUAD. Moreover, LUAD patients with a low WDFY4 expression exhibited a poorer prognosis. Additionally, experimental findings demonstrated that the overexpression of WDFY4 could inhibit the proliferation and metastasis of A549 cells while promoting apoptosis. It was also confirmed that WDFY4 could inhibit cancer growth in vivo. CONCLUSIONS The results indicate that promoting infiltration and activation of B cell populations could improve the long-term survival of LUAD patients, thereby offering a potential novel immunotherapeutic approach for LUAD. Besides, the promotion of cancer cell ferroptosis and upregulation of WDFY4 expression have been shown to induce the infiltration and activation of B cell populations. Furthermore, the overexpression of WDFY4 can significantly inhibit the growth of lung adenocarcinoma in vitro and in vivo, highlighting its potential as a target for immunotherapy in LUAD.
Collapse
Affiliation(s)
- Ling Huang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, China
- Hainan Center for Drug and Medical Device Evaluation and Service, Hainan Medical Products Administration, Haikou, China
- School of Hainan Provincial Drug Safety Evaluation Research Center, Hainan Medical University, Haikou, China
| | - Lifan Zhong
- School of Hainan Provincial Drug Safety Evaluation Research Center, Hainan Medical University, Haikou, China
| | - Ruxin Cheng
- Emergency and Trauma College, Hainan Medical University, Haikou, Hainan, China
| | - Limei Chang
- Hainan Center for Drug and Medical Device Evaluation and Service, Hainan Medical Products Administration, Haikou, China
| | - Mingyan Qin
- Hainan Center for Drug and Medical Device Evaluation and Service, Hainan Medical Products Administration, Haikou, China
| | - Huaping Liang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Wound Infection and Drug, Daping Hospital, Army Medical University, Chongqing, China
| | - Zhongkai Liao
- Department of Thoracic Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
48
|
Shi X, Dong A, Yang Y, Zheng G, Wang N, Yang C, Wang Y, Lu J, Jia X. Integrated analysis of single-cell and bulk RNA-sequencing identifies a signature based on T-cell marker genes to predict prognosis and immunotherapy response in bladder cancer. J Cancer Res Clin Oncol 2023; 149:9733-9746. [PMID: 37244876 DOI: 10.1007/s00432-023-04881-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 05/19/2023] [Indexed: 05/29/2023]
Abstract
BACKGROUND T cells have been proven to play important roles in anti-tumor and tumor microenvironment shaping, while these roles have not been explained in bladder cancer (BLCA). METHODS Single-cell RNA-sequencing (scRNA-seq) data were downloaded from the gene expression omnibus (GEO) database to screen T-cell marker genes. Bulk RNA-sequencing data and clinical information from BLCA patients were downloaded from the cancer genome atlas (TCGA) database to develop a prognosis signature. We analyzed the association of different risk groups with survival analysis, gene set enrichment analysis (GSEA), tumor mutational burden (TMB), and immunotherapy response. RESULTS Based on 192 T-cell marker genes identified by scRNA-seq analysis, we constructed a prognostic signature containing 7 genes in the training cohort, which was further validated in the testing cohort and GEO cohort. The areas under the receiver operating characteristic curve at 1-, 3-, and 5 years were 0.734, 0.742 and 0.726 in the training cohort, 0.697, 0.671 and 0.670 in the testing cohort, 0.702, 0.665 and 0.629 in the GEO cohort, respectively. In addition, we constructed a nomogram based on clinical factors and the risk score of the signature. The low-risk group exhibited higher immune-related pathways, immune cell infiltration and TMB levels. Importantly, immunophenotype score and immunotherapy cohort (IMvigor210) analyses showed that the low-risk group had better immunotherapy response and prognosis. CONCLUSIONS Our study reveals a novel prognostic signature based on T-cell marker genes, which provides a new target and theoretical support for BLCA patients.
Collapse
Affiliation(s)
- Xuezhong Shi
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Ani Dong
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Yongli Yang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Guowei Zheng
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Nana Wang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Chaojun Yang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Yuping Wang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Jie Lu
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Xiaocan Jia
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China.
| |
Collapse
|
49
|
Feng Q, Huang Z, Song L, Wang L, Lu H, Wu L. Combining bulk and single-cell RNA-sequencing data to develop an NK cell-related prognostic signature for hepatocellular carcinoma based on an integrated machine learning framework. Eur J Med Res 2023; 28:306. [PMID: 37649103 PMCID: PMC10466881 DOI: 10.1186/s40001-023-01300-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 08/18/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND The application of molecular targeting therapy and immunotherapy has notably prolonged the survival of patients with hepatocellular carcinoma (HCC). However, multidrug resistance and high molecular heterogeneity of HCC still prevent the further improvement of clinical benefits. Dysfunction of tumor-infiltrating natural killer (NK) cells was strongly related to HCC progression and survival benefits of HCC patients. Hence, an NK cell-related prognostic signature was built up to predict HCC patients' prognosis and immunotherapeutic response. METHODS NK cell markers were selected from scRNA-Seq data obtained from GSE162616 data set. A consensus machine learning framework including a total of 77 algorithms was developed to establish the gene signature in TCGA-LIHC data set, GSE14520 data set, GSE76427 data set and ICGC-LIRI-JP data set. Moreover, the predictive efficacy on ICI response was externally validated by GSE91061 data set and PRJEB23709 data set. RESULTS With the highest C-index among 77 algorithms, a 11-gene signature was established by the combination of LASSO and CoxBoost algorithm, which classified patients into high- and low-risk group. The prognostic signature displayed a good predictive performance for overall survival rate, moderate to high predictive accuracy and was an independent risk factor for HCC patients' prognosis in TCGA, GEO and ICGC cohorts. Compared with high-risk group, low-risk patients showed higher IPS-PD1 blocker, IPS-CTLA4 blocker, common immune checkpoints expression but lower TIDE score, which indicated low-risk patients might be prone to benefiting from ICI treatment. Moreover, a real-world cohort, PRJEB23709, also revealed better immunotherapeutic response in low-risk group. CONCLUSIONS Overall, the present study developed a gene signature based on NK cell-related genes, which offered a novel platform for prognosis and immunotherapeutic response evaluation of HCC patients.
Collapse
Affiliation(s)
- Qian Feng
- Department of Emergency, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| | - Zhihao Huang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, 1st min de Road, Nanchang, 330000, China
| | - Lei Song
- Department of General Practice, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| | - Le Wang
- Department of Blood Transfusion, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| | - Hongcheng Lu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, 1st min de Road, Nanchang, 330000, China.
| | - Linquan Wu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, 1st min de Road, Nanchang, 330000, China.
| |
Collapse
|
50
|
Yu Q, Shi X, Wang H, Zhang S, Hu S, Cai T. A Novel Prognostic Signature of comprising Nine NK Cell signatures Based on Both Bulk RNA Sequencing and Single-Cell RNA Sequencing for Hepatocellular Carcinoma. J Cancer 2023; 14:2209-2223. [PMID: 37576389 PMCID: PMC10414035 DOI: 10.7150/jca.85873] [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: 05/05/2023] [Accepted: 07/09/2023] [Indexed: 08/15/2023] Open
Abstract
Background: Hepatocellular carcinoma (HCC) has limited prognostic prediction due to its heterogeneity. Understanding the role of natural killer (NK) cells in HCC is vital for prognosis and immunotherapy guidance. We aimed to identify NK cell marker genes through scRNA-seq and develop a prognostic signature for HCC. Methods: We analyzed scRNA-seq data (GSE149614) from 10 patients and bulk RNA-seq data from 786 patients with clinicopathological information. NK cell marker genes were identified using clustering and marker finding functions. A predictive risk signature was constructed using LASSO-COX algorithm. Functional annotations and immune cell infiltration analysis were performed, and the nomogram's performance was evaluated. Results: We identified 79 NK cell marker genes associated with NK cell-mediated cytotoxicity, apoptosis, and immune response. The multigene signature significantly correlated with overall survival (OS) in TCGA-LIHC cohort and was validated in other cohorts. Low-risk patients exhibited higher immune cell infiltration, including CD8+ T cells. The risk signature was an independent prognostic factor for OS (HR > 1, p < 0.001). The nomogram combining the risk signature and clinical predictors demonstrated robust prognostic ability. Conclusion: We developed a nine-gene signature prognostic model based on NK cell marker genes to accurately assess the prognostic risk of HCC. This model can be a valuable tool for personalized evaluation post-surgery. Our study underscores the potential of NK cells in HCC prognosis and highlights the importance of scRNA-seq analysis in identifying prognostic markers.
Collapse
Affiliation(s)
- Qi Yu
- Department of Experimental Medical Science, Ningbo No.2 Hospital, Ningbo 315010, China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo 315032, China
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo 315010, China
| | - Xuefeng Shi
- Department of Pulmonary and Critical Care Medicine, Qinghai provincial people's hospital, Xining 81000, China
| | - Hongjian Wang
- College of Agricultural, Consumer and Environmental Sciences, University of Illinois at Urbana-Champaign, Champaign 61820, USA
| | - Shun Zhang
- Department of Experimental Medical Science, Ningbo No.2 Hospital, Ningbo 315010, China
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo 315010, China
| | - Songnian Hu
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Ting Cai
- Department of Experimental Medical Science, Ningbo No.2 Hospital, Ningbo 315010, China
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo 315032, China
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo 315010, China
| |
Collapse
|