1
|
Zhu XR, Zhu JQ, Gu QH, Liu N, Lu JJ, Li XH, Liu YY, Zheng X, Chen MB, Ji Y. A novel identified epithelial ligand-receptor-associated gene signature highlights POPDC3 as a potential therapy target for non-small cell lung cancer. Cell Death Dis 2025; 16:114. [PMID: 39971925 PMCID: PMC11840029 DOI: 10.1038/s41419-025-07410-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 01/13/2025] [Accepted: 01/30/2025] [Indexed: 02/21/2025]
Abstract
The tumor microenvironment (TME) is pivotal in non-small cell lung cancer (NSCLC) progression, influencing drug resistance and immune cell behavior through complex ligand-receptor (LR) interactions. This study developed an epithelial LR-related prognostic risk score (LRrisk) to identify biomarkers and targets in NSCLC. We identified twenty epithelial LRs with significant prognostic implications and delineated three molecular NSCLC subtypes with distinct outcomes, pathological characteristics, biological pathways, and immune profiles. The LRrisk model was constructed using twelve differentially expressed ligand-receptor interaction-related genes (LRGs), with a focus on POPDC3 (popeye domain-containing protein 3), which was overexpressed in NSCLC cells. Functional assays revealed that POPDC3 knockdown reduced cell proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT), while its overexpression promoted cancerous activities. In vivo, POPDC3 silencing hindered, and its overexpression accelerated the growth of NSCLC xenografts in nude mice. Additionally, high expression levels of POPDC3 in NSCLC tissues were associated with enhanced CD4+ T cell infiltration and increased PD-1 expression within the TME. Moreover, ectopic POPDC3 overexpression in C57BL/6 J mouse Lewis lung carcinoma (LLC) xenografts enhanced CD4+ T cell infiltration and PD-1 expression in the TME. This research establishes a robust epithelial LR-related signature, highlighting POPDC3 as a critical facilitator of NSCLC progression and a potential therapeutic target.
Collapse
Affiliation(s)
- Xiao-Ren Zhu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
- Clinical Research and Lab Center, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
- Medical School of Jiangsu University, Zhenjiang, China
| | - Jia-Qi Zhu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Qian-Hui Gu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Na Liu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
- Medical School of Jiangsu University, Zhenjiang, China
| | - Jing-Jing Lu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
- Medical School of Jiangsu University, Zhenjiang, China
| | - Xiao-Hong Li
- Department of Clinical Laboratory, The First People's Hospital of Taicang, Taicang, China
| | - Yuan-Yuan Liu
- Clinical Research and Lab Center, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
- Medical School of Jiangsu University, Zhenjiang, China
| | - Xian Zheng
- Medical School of Jiangsu University, Zhenjiang, China.
- Department of Pharmacy, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China.
| | - Min-Bin Chen
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China.
- Medical School of Jiangsu University, Zhenjiang, China.
| | - Yong Ji
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China.
| |
Collapse
|
2
|
Wu Z, Chen Y, Jiang D, Pan Y, Tang T, Ma Y, Shapaer T. Mitochondrial-related drug resistance lncRNAs as prognostic biomarkers in laryngeal squamous cell carcinoma. Discov Oncol 2024; 15:785. [PMID: 39692950 DOI: 10.1007/s12672-024-01690-x] [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: 09/14/2024] [Accepted: 12/09/2024] [Indexed: 12/19/2024] Open
Abstract
Laryngeal squamous cell carcinoma (LSCC) is a common malignant tumor of the head and neck that significantly impacts patients' quality of life, with chemotherapy resistance notably affecting prognosis. This study aims to identify prognostic biomarkers to optimize treatment strategies for LSCC. Using data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO), combined with mitochondrial gene database analysis, we identified mitochondrial lncRNAs associated with drug resistance genes. Key long non-coding RNAs (lncRNAs) were selected through univariate Cox regression and Lasso regression, and a multivariate Cox regression model was constructed to predict prognosis. We further analyzed the differences in immune function and biological pathway enrichment between high- and low-risk groups, developed a nomogram, and compared drug sensitivity. Results showed that the prognostic model based on seven mitochondrial lncRNAs could serve as an independent prognostic factor, with Area Under the Curve (AUC) values of 0.746, 0.827, and 0.771 at 1, 3, and 5 years, respectively, outperforming some existing models, demonstrating high predictive performance. Significant differences were observed in immune function and drug sensitivity between the high- and low-risk groups. The risk prediction model incorporating seven drug resistance-related mitochondrial lncRNAs can accurately and independently predict the prognosis of LSCC patients.
Collapse
Affiliation(s)
- Zhimin Wu
- Department of Otorhinolaryngology Head and Neck Surgery, The Maternal and Child Health Care Hospital of Guizhou Medical University, Guiyang, 550000, Guizhou, China
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, 550003, Guizhou, China
| | - Yi Chen
- Department of Breast and Thyroid Surgery, the Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, 830011, Xinjiang Uygur Autonomous Region, China
| | - Dizhi Jiang
- Department of Radiation Oncology, Cheeloo College of Medicine, Qilu Hospital of Shandong University, Shandong University, Jinan, 250012, Shandong, China
| | - Yipeng Pan
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310020, Zhejiang, China
| | - Tuoxian Tang
- Department of Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Yifei Ma
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, 550003, Guizhou, China.
| | - Tiannake Shapaer
- Department of Gastrointestinal Surgery, the Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, 830011, Xinjiang Uygur Autonomous Region, China.
| |
Collapse
|
3
|
Xu K, Li D, Ji K, Zhang Y, Zhang M, Zhou H, Hou X, Jiang J, Zhang Z, Dai H, Sun H. Disulfidptosis-associated LncRNA signature predicts prognosis and immune response in kidney renal clear cell carcinoma. Biol Direct 2024; 19:71. [PMID: 39175011 PMCID: PMC11340127 DOI: 10.1186/s13062-024-00517-7] [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: 04/27/2024] [Accepted: 08/08/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND Kidney renal clear cell carcinoma (KIRC) represents a significant proportion of renal cell carcinomas and is characterized by high aggressiveness and poor prognosis despite advancements in immunotherapy. Disulfidptosis, a novel cell death pathway, has emerged as a critical mechanism in various cellular processes, including cancer. This study leverages machine learning to identify disulfidptosis-related long noncoding RNAs (DRlncRNAs) as potential prognostic biomarkers in KIRC, offering new insights into tumor pathogenesis and treatment avenues. RESULTS Our analysis of data from The Cancer Genome Atlas (TCGA) led to the identification of 431 DRlncRNAs correlated with disulfidptosis-related genes. Five key DRlncRNAs (SPINT1-AS1, AL161782.1, OVCH1-AS1, AC131009.3, and AC108673.3) were used to develop a prognostic model that effectively distinguished between low- and high-risk patients with significant differences in overall survival and progression-free survival. The low-risk group had a favorable prognosis associated with a protective immune microenvironment and a better response to targeted drugs. Conversely, the high-risk group displayed aggressive tumor features and poor immunotherapy outcomes. Validation through qRT‒PCR confirmed the differential expression of these DRlncRNAs in KIRC cells compared to normal kidney cells, underscoring their potential functional significance in tumor biology. CONCLUSIONS This study established a robust link between disulfidptosis-related lncRNAs and patient prognosis in KIRC, underscoring their potential as prognostic biomarkers and therapeutic targets. The differential expression of these lncRNAs in tumor versus normal tissue further highlights their relevance in KIRC pathogenesis. The predictive model not only enhances our understanding of KIRC biology but also provides a novel stratification tool for precision medicine approaches, improving treatment personalization and outcomes in KIRC patients.
Collapse
Affiliation(s)
- Kangjie Xu
- Central Laboratory Department, Binhai County People's Hospital, Clinical Medical College of Yangzhou University, Yancheng, China
| | - Dongling Li
- Nephrology Department, Binhai County People's Hospital, Yancheng, China
| | - Kangkang Ji
- Central Laboratory Department, Binhai County People's Hospital, Clinical Medical College of Yangzhou University, Yancheng, China
| | - Yanhua Zhang
- Obstetrics and Gynecology Department, Binhai County People's Hospital, Yancheng, China
| | - Minglei Zhang
- Oncology Department, Binhai County People's Hospital, Yancheng, China
| | - Hai Zhou
- Science and Education Department, Binhai County People's Hospital, Yancheng, China
| | - Xuefeng Hou
- Central Laboratory Department, Binhai County People's Hospital, Clinical Medical College of Yangzhou University, Yancheng, China
| | - Jian Jiang
- Central Laboratory Department, Binhai County People's Hospital, Clinical Medical College of Yangzhou University, Yancheng, China
| | - Zihang Zhang
- Pathology Department, Binhai County People's Hospital, Yancheng, China
| | - Hua Dai
- Jiangsu Key Laboratory of Experimental & Translational Noncoding RNA Research, Yangzhou University Clinical Medical College, Yangzhou, China
| | - Hang Sun
- Urology Department, Binhai County People's Hospital, Yancheng, China.
| |
Collapse
|
4
|
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
|
5
|
Chen L, Lin J, Wen Y, Lan B, Xiong J, Fu Y, Chen Y, Chen CB. A senescence-related lncRNA signature predicts prognosis and reflects immune landscape in HNSCC. Oral Oncol 2024; 149:106659. [PMID: 38134702 DOI: 10.1016/j.oraloncology.2023.106659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 11/15/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023]
Abstract
OBJECTIVE Long noncoding RNAs (lncRNAs) regulate cancer cell senescence in many cancers. However, their specific involvement in head and neck squamous cell carcinoma (HNSCC) remains unclear. We are looking for an ingenious prognostic signature that utilizes senescence-related lncRNAs (SRlncRNAs) to predict prognosis and provide insights into the immune landscape in HNSCC. MATERIALS AND METHODS HNSCC clinical and Cellular senescence genes information were collected from The Cancer Genome Atlas and Human Aging Genomic Resources. Then we performed Cox and Lasso regression to locate SRlncRNAs related to the prognosis of HNSCC and built a predictive signature. Further, prognosis assessment, potential mechanisms, and immune status were assessed by Kaplan-Meier analysis, Gene Set Enrichment Analysis (GSEA), and CIBERSORT, respectively. RESULTS A prognosis prediction model based on sixteen SRlncRNAs was identified and internally validated. Then, patients with high-risk scores suffered an unfavorable overall survival (All p < 0.05). The risk score, age, and stage were independent prognostic parameters (all p < 0.001). Our model has good predictive ability (The AUC (area under the curves) 1-year = 0.707, AUC3-year = 0.748 and AUC5-year = 0.779). Subsequently, GESA revealed SRlncRNAs regulated immune responses. Patients in the high-risk group had higher tumor mutation burden and Tumor Immune Dysfunction and Exclusion but lower levels of 37 immune checkpoint genes, immune scores, and immune cells like CD8 + T cells, follicular helper T cells, and regulatory T cells. CONCLUSIONS A prognostic model based on SRlncRNAs is the potential target for improving immunotherapy outcomes for HNSCC.
Collapse
Affiliation(s)
- Lizhu Chen
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China; Cancer Bio-Immunotherapy Center, Clinical Oncology School of Fujian Medical University & Fujian Cancer Hospital, Fuzhou, Fujian Province, China; Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian Province, China
| | - Jing Lin
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China; Cancer Bio-Immunotherapy Center, Clinical Oncology School of Fujian Medical University & Fujian Cancer Hospital, Fuzhou, Fujian Province, China; Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian Province, China
| | - Yaoming Wen
- Fujian Institute of Microbiology, Fuzhou, Fujian Province, China
| | - Bin Lan
- Cancer Bio-Immunotherapy Center, Clinical Oncology School of Fujian Medical University & Fujian Cancer Hospital, Fuzhou, Fujian Province, China; Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian Province, China
| | - Jiani Xiong
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China; Cancer Bio-Immunotherapy Center, Clinical Oncology School of Fujian Medical University & Fujian Cancer Hospital, Fuzhou, Fujian Province, China; Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian Province, China
| | - Yajuan Fu
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province, China
| | - Yu Chen
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China; Cancer Bio-Immunotherapy Center, Clinical Oncology School of Fujian Medical University & Fujian Cancer Hospital, Fuzhou, Fujian Province, China; Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian Province, China.
| | - Chuan-Ben Chen
- Cancer Bio-Immunotherapy Center, Clinical Oncology School of Fujian Medical University & Fujian Cancer Hospital, Fuzhou, Fujian Province, China; Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian Province, China; Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, China.
| |
Collapse
|
6
|
Wu Z, Wang W, Zhang K, Fan M, Lin R. Epigenetic and Tumor Microenvironment for Prognosis of Patients with Gastric Cancer. Biomolecules 2023; 13:biom13050736. [PMID: 37238607 DOI: 10.3390/biom13050736] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 04/02/2023] [Accepted: 04/12/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND Epigenetics studies heritable or inheritable mechanisms that regulate gene expression rather than altering the DNA sequence. However, no research has investigated the link between TME-related genes (TRGs) and epigenetic-related genes (ERGs) in GC. METHODS A complete review of genomic data was performed to investigate the relationship between the epigenesis tumor microenvironment (TME) and machine learning algorithms in GC. RESULTS Firstly, TME-related differential expression of genes (DEGs) performed non-negative matrix factorization (NMF) clustering analysis and determined two clusters (C1 and C2). Then, Kaplan-Meier curves for overall survival (OS) and progression-free survival (PFS) rates suggested that cluster C1 predicted a poorer prognosis. The Cox-LASSO regression analysis identified eight hub genes (SRMS, MET, OLFML2B, KIF24, CLDN9, RNF43, NETO2, and PRSS21) to build the TRG prognostic model and nine hub genes (TMPO, SLC25A15, SCRG1, ISL1, SOD3, GAD1, LOXL4, AKR1C2, and MAGEA3) to build the ERG prognostic model. Additionally, the signature's area under curve (AUC) values, survival rates, C-index scores, and mean squared error (RMS) curves were evaluated against those of previously published signatures, which revealed that the signature identified in this study performed comparably. Meanwhile, based on the IMvigor210 cohort, a statistically significant difference in OS between immunotherapy and risk scores was observed. It was followed by LASSO regression analysis which identified 17 key DEGs and a support vector machine (SVM) model identified 40 significant DEGs, and based on the Venn diagram, eight co-expression genes (ENPP6, VMP1, LY6E, SHISA6, TMEM158, SYT4, IL11, and KLK8) were discovered. CONCLUSION The study identified some hub genes that could be useful in predicting prognosis and management in GC.
Collapse
Affiliation(s)
- Zenghong Wu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Weijun Wang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Kun Zhang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Mengke Fan
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Rong Lin
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
7
|
Dong R, Chen S, Lu F, Zheng N, Peng G, Li Y, Yang P, Wen H, Qiu Q, Wang Y, Wu H, Liu M. Models for Predicting Response to Immunotherapy and Prognosis in Patients with Gastric Cancer: DNA Damage Response Genes. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4909544. [PMID: 36578802 PMCID: PMC9792237 DOI: 10.1155/2022/4909544] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 10/30/2022] [Accepted: 11/21/2022] [Indexed: 12/23/2022]
Abstract
Objective DNA damage response (DDR) is a complex system that maintains genetic integrity and the stable replication and transmission of genetic material. m6A modifies DDR-related gene expression and affects the balance of DNA damage response in tumor cells. In this study, a risk model based on m6A-modified DDR-related gene was established to evaluate its role in patients with gastric cancer. Methods We downloaded 639 DNA damage response genes from the Gene Set Enrichment Analysis (GSEA) database and constructed risk score models using typed differential genes. We used Kaplan-Meier curves and risk curves to verify the clinical relevance of the model, which was then validated with the univariate and multifactorial Cox analysis, ROC, C-index, and nomogram, and finally this model was used to evaluate the correlation of the risk score model with immune microenvironment, microsatellite instability (MSI), tumor mutational burden (TMB), and immune checkpoints. Results In this study, 337 samples in The Cancer Genome Atlas (TCGA) database were used as training set to construct a DDR-related gene model, and GSE84437 was used as external data set for verification. We found that the prognosis and immunotherapy effect of gastric cancer patients in the low-risk group were significantly better than those in the high-risk group. Conclusion We screened eight DDR-related genes (ZBTB7A, POLQ, CHEK1, NPDC1, RAMP1, AXIN2, SFRP2, and APOD) to establish a risk model, which can predict the prognosis of gastric cancer patients and guide the clinical implementation of immunotherapy.
Collapse
Affiliation(s)
- Rui Dong
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Shuran Chen
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Fei Lu
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Ni Zheng
- School of Life Science, Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, China
| | - Guisen Peng
- School of Life Science, Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, China
| | - Yan Li
- Department of Gynecologic Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Pan Yang
- School of Life Science, Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, China
| | - Hexin Wen
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Quanwei Qiu
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Yitong Wang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Huazhang Wu
- School of Life Science, Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, China
| | - Mulin Liu
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| |
Collapse
|
8
|
Liu A, Li Y, Shen L, Li N, Zhao Y, Shen L, Li Z. Molecular subtypes based on cuproptosis regulators and immune infiltration in kidney renal clear cell carcinoma. Front Genet 2022; 13:983445. [PMID: 36338990 PMCID: PMC9635053 DOI: 10.3389/fgene.2022.983445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/13/2022] [Indexed: 11/13/2022] Open
Abstract
Copper toxicity involves the destruction of mitochondrial metabolic enzymes, triggering an unusual mechanism of cell death called cuproptosis, which proposes a novel approach using copper toxicity to treat cancer. However, the biological function of cuproptosis has not been fully elucidated in kidney renal clear cell carcinoma (KIRC). Using the expression profile of 13 cuproptosis regulators, we first identified two molecular subtypes related to cuproptosis defined as “hot tumor” and “cold tumor”, having different levels of biological function, clinical prognosis, and immune cell infiltration. We obtained three gene clusters using the differentially expressed genes between the two cuproptosis-related subtypes, which were associated with different molecular activities and clinical characteristics. Next, we developed and validated a cuproptosis prognostic model that included two genes (FDX1 and DBT). The calculated risk score could divide patients into high- and low-risk groups. The high-risk group had a poorer prognosis, lower level of immune infiltration, higher frequency of gene alterations, and greater levels of FDX1 methylation and limited DBT methylation. The risk score was also an independent predictive factor for overall survival in KIRC. The established nomogram calculating the risk score achieved a high predictive ability for the prognosis of individual patients (area under the curve: 0.860). We then identified small molecular inhibitors as potential treatments and analyzed the sensitivity to chemotherapy of the signature genes. Tumor immune dysfunction and exclusion (TIDE) showed that the high-risk group had a higher level of TIDE, exclusion and dysfunction that was lower than the low-risk group, while the microsatellite instability of the high-risk group was significantly lower. The results of two independent immunotherapy datasets indicated that cuproptosis regulators could influence the response and efficacy of immunotherapy in KIRC. Our study provides new insights for individualized and comprehensive therapy of KIRC.
Collapse
Affiliation(s)
- Aibin Liu
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Yanyan Li
- Department of Nursing, Xiangya Hospital, Central South University, Changsha, China
| | - Lin Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Na Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Yajie Zhao
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Liangfang Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhanzhan Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Zhanzhan Li,
| |
Collapse
|
9
|
Wen Z, Yang C, Zou D, Liu J, Wang S, Liu X, Zhang Y, Zhang Y. Pan-cancer analysis of PSAP identifies its expression and clinical relevance in gastric cancer. Pathol Res Pract 2022; 238:154027. [PMID: 36084426 DOI: 10.1016/j.prp.2022.154027] [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: 11/29/2021] [Revised: 07/09/2022] [Accepted: 07/12/2022] [Indexed: 11/23/2022]
Abstract
Prosaposin (PSAP) plays a critical role in sphingolipid and cancer metabolism. Reports have shown that PSAP was involved in proliferation, tumorigenesis, and metastasis. However, the expression pattern of PSAP and its prognostic roles in gastric cancer remain elusive. PSAP expression pattern and its prognostic roles in gastric cancer (GC) were explored using data from the TCGA and Kaplan-Meier Plotter. Immunohistochemical staining of GC tissues was performed to validate the prognostic role of PSAP. TISIDB was used to analyze its correlation with immunomodulators. PSAP-associated genes, PDCD1, TGFB1, and CSF1R were used to build a risk model to evaluate immunotherapy outcomes of patients with stomach adenocarcinoma (STAD). Results showed that PSAP was highly expressed in GC. High PSAP expression in GC patients also significantly indicated a poor prognosis. The results of immunohistochemical staining showed that PSAP was an independent prognostic factor in GC patients. Based on three PSAP-associated genes, a risk model that could predict the prognosis and immunotherapy outcome of STAD was bulit. PSAP was an independent prognostic factor in GC. Our results have identified three prognosis-related genes which were useful to evaluate immunotherapy outcomes of STAD patients.
Collapse
Affiliation(s)
- Zhenpeng Wen
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, PR China.
| | - Chunjiao Yang
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, PR China.
| | - Dan Zou
- Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Insititute, Shenyang, Liaoning Province 110042, PR China.
| | - Jiaqing Liu
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, PR China.
| | - Song Wang
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, PR China.
| | - Xuqin Liu
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, PR China.
| | - Yi Zhang
- Department of Gynecology, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, PR China.
| | - Ye Zhang
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, Shenyang, Liaoning Province 110001, PR China.
| |
Collapse
|
10
|
Yang X, Wang X, Sun X, Xiao M, Fan L, Su Y, Xue L, Luo S, Hou S, Wang H. Construction of five cuproptosis-related lncRNA signature for predicting prognosis and immune activity in skin cutaneous melanoma. Front Genet 2022; 13:972899. [PMID: 36160015 PMCID: PMC9490379 DOI: 10.3389/fgene.2022.972899] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
Cuproptosis is a newly discovered new mechanism of programmed cell death, and its unique pathway to regulate cell death is thought to have a unique role in understanding cancer progression and guiding cancer therapy. However, this regulation has not been studied in SKCM at present. In this study, data on Skin Cutaneous Melanoma (SKCM) patients were downloaded from the TCGA database. We screened the genes related to cuproptosis from the published papers and confirmed the lncRNAs related to them. We applied Univariate/multivariate and LASSO Cox regression algorithms, and finally identified 5 cuproptosis-related lncRNAs for constructing prognosis prediction models (VIM-AS1, AC012443.2, MALINC1, AL354696.2, HSD11B1-AS1). The reliability and validity test of the model indicated that the model could well distinguish the prognosis and survival of SKCM patients. Next, immune microenvironment, immunotherapy analysis, and functional enrichment analysis were also performed. In conclusion, this study is the first analysis based on cuproptosis-related lncRNAs in SKCM and aims to open up new directions for SKCM therapy.
Collapse
Affiliation(s)
- Xiaojing Yang
- Department of Dermatovenereology, Tianjin Medical University General Hospital, Tianjin, China
- *Correspondence: Xiaojing Yang, ; Huiping Wang,
| | - Xing Wang
- Department of Dermatovenereology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xinti Sun
- Department of Thoracic Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Meng Xiao
- Department of Dermatovenereology, Tianjin Medical University General Hospital, Tianjin, China
| | - Liyun Fan
- Department of Dermatovenereology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yunwei Su
- Department of Dermatovenereology, Tianjin Medical University General Hospital, Tianjin, China
| | - Lu Xue
- Department of Dermatovenereology, Tianjin Medical University General Hospital, Tianjin, China
| | - Suju Luo
- Department of Dermatovenereology, Tianjin Medical University General Hospital, Tianjin, China
| | - Shuping Hou
- Department of Dermatovenereology, Tianjin Medical University General Hospital, Tianjin, China
| | - Huiping Wang
- Department of Dermatovenereology, Tianjin Medical University General Hospital, Tianjin, China
- *Correspondence: Xiaojing Yang, ; Huiping Wang,
| |
Collapse
|
11
|
Geng S, Fu Y, Fu S, Wu K. A tumor microenvironment-related risk model for predicting the prognosis and tumor immunity of breast cancer patients. Front Immunol 2022; 13:927565. [PMID: 36059555 PMCID: PMC9433750 DOI: 10.3389/fimmu.2022.927565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
Background This study aimed to construct a tumor microenvironment (TME)-related risk model to predict the overall survival (OS) of patients with breast cancer. Methods Gene expression data from The Cancer Genome Atlas was used as the training set. Differentially expressed gene analysis, prognosis analysis, weighted gene co-expression network analysis, Least Absolute Shrinkage and Selection Operator regression analysis, and Wald stepwise Cox regression were performed to screen for the TME-related risk model. Three Gene Expression Omnibus databases were used to validate the predictive efficiency of the prognostic model. The TME-risk-related biological function was investigated using the gene set enrichment analysis (GSEA) method. Tumor immune and mutation signatures were analyzed between low- and high-TME-risk groups. The patients’ response to chemotherapy and immunotherapy were evaluated by the tumor immune dysfunction and exclusion (TIDE) score and immunophenscore (IPS). Results Five TME-related genes were screened for constructing a prognostic signature. Higher TME risk scores were significantly associated with worse clinical outcomes in the training set and the validation set. Correlation and stratification analyses also confirmed the predictive efficiency of the TME risk model in different subtypes and stages of breast cancer. Furthermore, immune checkpoint expression and immune cell infiltration were found to be upregulated in the low-TME-risk group. Biological processes related to immune response functions were proved to be enriched in the low-TME-risk group through GSEA analysis. Tumor mutation analysis and TIDE and IPS analyses showed that the high-TME-risk group had more tumor mutation burden and responded better to immunotherapy. Conclusion The novel and robust TME-related risk model had a strong implication for breast cancer patients in OS, immune response, and therapeutic efficiency.
Collapse
|
12
|
Zeng Y, Zhang X, Li F, Wang Y, Wei M. AFF3 is a novel prognostic biomarker and a potential target for immunotherapy in gastric cancer. J Clin Lab Anal 2022; 36:e24437. [PMID: 35478418 PMCID: PMC9169183 DOI: 10.1002/jcla.24437] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/25/2022] [Accepted: 04/04/2022] [Indexed: 12/26/2022] Open
Abstract
Background Gastric cancer (GC) is one of the most common cancers worldwide with a poor prognosis. The tumor microenvironment (TME) serves a pivotal role in affecting the prognosis and efficacy of immunotherapy. Given the poor prognosis of GC patients and the limitation of immunotherapy, we urged to identify new prognostic and immunotherapeutic biomarkers. Methods The transcriptome data were downloaded from the TCGA, GEO, and GEPIA databases, and performed differential analysis of AFF3 in tumor samples and normal samples. The UALCAN, Kaplan–Meier plotter and GEPIA databases were employed to assess the correlation of AFF3 with clinicopathological characteristics and prognosis. The potential mechanism of AFF3 was explored by the GO and KEGG enrichment. The potential role of AFF3 on tumor‐infiltrating immune cells (TIICs) was explored by TIMER2.0 and TISIDB. TIMER2.0 and SangerBox3.0 databases were, respectively, used to determine the correlation of AFF3 with immune checkpoint (ICs), tumor mutational burden (TMB), and microsatellite instability (MSI) in GC. Results We found significant downregulation of AFF3 in GC tissues as compared with normal tissues. However, GC patients having a higher expression of AFF3 were found to have worse clinicopathological characteristics and prognosis. Moreover, the GO enrichment analysis illustrated that AFF3 might regulate the immune cells in the TME. In addition, the AFF3 was positively correlated with TIICs, ICs, TMB, and MSI. Conclusion Here, we conclude that AFF3 may be a promising potential marker for the diagnosis and prognosis of GC patients, and may influence response to ICIs by affecting TIICs and ICs expression in the TME.
Collapse
Affiliation(s)
- Yuling Zeng
- Department of Blood Transfusion, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou City, China
| | - Xueping Zhang
- Department of Hepatobiliary Surgery, Zhengzhou Central Hospital Affiliated of Zhengzhou University, Zhengzhou City, China
| | - Fazhan Li
- Marshall Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou City, China
| | - Ying Wang
- Department of Blood Transfusion, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou City, China
| | - Ming Wei
- Department of Blood Transfusion, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou City, China
| |
Collapse
|
13
|
Dou M, Ding C, Zheng B, Deng G, Zhu K, Xu C, Xue W, Ding X, Zheng J, Tian P. Immune-Related Genes for Predicting Future Kidney Graft Loss: A Study Based on GEO Database. Front Immunol 2022; 13:859693. [PMID: 35281025 PMCID: PMC8913884 DOI: 10.3389/fimmu.2022.859693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 02/08/2022] [Indexed: 11/22/2022] Open
Abstract
Objective We aimed to identify feature immune-related genes that correlated with graft rejection and to develop a prognostic model based on immune-related genes in kidney transplantation. Methods Gene expression profiles were obtained from the GEO database. The GSE36059 dataset was used as a discovery cohort. Then, differential expression analysis and a machine learning method were performed to select feature immune-related genes. After that, univariate and multivariate Cox regression analyses were used to identify prognosis-related genes. A novel Riskscore model was built based on the results of multivariate regression. The levels of these feature genes were also confirmed in an independent single-cell dataset and other GEO datasets. Results 15 immune-related genes were expressed differently between non-rejection and rejection kidney allografts. Those differentially expressed immune-related genes (DE-IRGs) were mainly associated with immune-related biological processes and pathways. Subsequently, a 5-immune-gene signature was constructed and showed favorable predictive results in the GSE21374 dataset. Recipients were divided into the high-risk and low-risk groups according to the median value of RiskScore. The GO and KEGG analysis indicated that the differentially expressed genes (DEGs) between high-risk and low-risk groups were mainly involved in inflammatory pathways, chemokine-related pathways, and rejection-related pathways. Immune infiltration analysis demonstrated that RiskScore was potentially related to immune infiltration. Kaplan-Meier survival analysis suggested that recipients in the high-risk group had poor graft survival. AUC values of 1- and 3-year graft survival were 0.804 and 0.793, respectively. Conclusion Our data suggest that this immune-related prognostic model had good sensitivity and specificity in predicting the 1- and 3-year kidney graft survival and might act as a useful tool for predicting kidney graft loss.
Collapse
Affiliation(s)
- Meng Dou
- Department of Kidney Transplantation, Hospital of Nephropathy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Chenguang Ding
- Department of Kidney Transplantation, Hospital of Nephropathy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Bingxuan Zheng
- Department of Kidney Transplantation, Hospital of Nephropathy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Ge Deng
- Department of Kidney Transplantation, Hospital of Nephropathy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Kun Zhu
- Department of Kidney Transplantation, Hospital of Nephropathy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Cuixiang Xu
- Center of Shaanxi Provincial Clinical Laboratory, Shaanxi Provincial People’s Hospital, Xi’an, China
| | - Wujun Xue
- Department of Kidney Transplantation, Hospital of Nephropathy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xiaoming Ding
- Department of Kidney Transplantation, Hospital of Nephropathy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jin Zheng
- Department of Kidney Transplantation, Hospital of Nephropathy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Puxun Tian
- Department of Kidney Transplantation, Hospital of Nephropathy, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Puxun Tian,
| |
Collapse
|