scholarly journals A Novel Autophagy-Related Prognostic Risk Model and a Nomogram for Survival Prediction of Oral Cancer Patients

2022 ◽  
Vol 2022 ◽  
pp. 1-13
Author(s):  
Hongjun Fei ◽  
Xiongming Chen

Background. This study is aimed at constructing a risk signature to predict survival outcomes of ORCA patients. Methods. We identified differentially expressed autophagy-related genes (DEARGs) based on the RNA sequencing data in the TCGA database; then, four independent survival-related ARGs were identified to construct an autophagy-associated signature for survival prediction of ORCA patients. The validity and robustness of the prognostic model were validated by clinicopathological data and survival data. Subsequently, four independent prognostic DEARGs that composed the model were evaluated individually. Results. The expressions of 232 autophagy-related genes (ARGs) in 127 ORCA and 13 control tissues were compared, and 36 DEARGs were filtered out. We performed functional enrichment analysis and constructed protein–protein interaction network for 36 DEARGs. Univariate and multivariate Cox regression analyses were adopted for searching prognostic ARGs, and an autophagy-associated signature for ORCA patients was constructed. Eventually, 4 desirable independent survival-related ARGs (WDR45, MAPK9, VEGFA, and ATIC) were confirmed and comprised the prognostic model. We made use of multiple ways to verify the accuracy of the novel autophagy-related signature for survival evaluation, such as receiver-operator characteristic curve, Kaplan–Meier plotter, and clinicopathological correlational analyses. Four independent prognostic DEARGs that formed the model were also associated with the prognosis of ORCA patients. Conclusions. The autophagy-related risk model can evaluate OS for ORCA patients independently since it is accurate and stable. Four prognostic ARGs that composed the model can be studied deeply for target treatment.

BMC Cancer ◽  
2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Yang Zhai ◽  
Bin Zhao ◽  
Yuzhen Wang ◽  
Lina Li ◽  
Jingjin Li ◽  
...  

Abstract Background Lung adenocarcinoma (LUAD) is the most common pathology subtype of lung cancer. In recent years, immunotherapy, targeted therapy and chemotherapeutics conferred a certain curative effects. However, the effect and prognosis of LUAD patients are different, and the efficacy of existing LUAD risk prediction models is unsatisfactory. Methods The Cancer Genome Atlas (TCGA) LUAD dataset was downloaded. The differentially expressed immune genes (DEIGs) were analyzed with edgeR and DESeq2. The prognostic DEIGs were identified by COX regression. Protein-protein interaction (PPI) network was inferred by STRING using prognostic DEIGs with p value< 0.05. The prognostic model based on DEIGs was established using Lasso regression. Immunohistochemistry was used to assess the expression of FERMT2, FKBP3, SMAD9, GATA2, and ITIH4 in 30 cases of LUAD tissues. Results In total,1654 DEIGs were identified, of which 436 genes were prognostic. Gene functional enrichment analysis indicated that the DEIGs were involved in inflammatory pathways. We constructed 4 models using DEIGs. Finally, model 4, which was constructed using the 436 DEIGs performed the best in prognostic predictions, the receiver operating characteristic curve (ROC) was 0.824 for 3 years, 0.838 for 5 years, 0.834 for 10 years. High levels of FERMT2, FKBP3 and low levels of SMAD9, GATA2, ITIH4 expression are related to the poor overall survival in LUAD (p < 0.05). The prognostic model based on DEIGs reflected infiltration by immune cells. Conclusions In our study, we built an optimal prognostic signature for LUAD using DEIGs and verified the expression of selected genes in LUAD. Our result suggests immune signature can be harnessed to obtain prognostic insights.


BMC Cancer ◽  
2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Hongjun Fei ◽  
Songchang Chen ◽  
Chenming Xu

Abstract Background Little data is available on prognostic biomarkers and effective treatment options for Kidney Renal Papillary Cell Carcinoma (KIRP) patients, to find potential prognostic biomarkers and new targets was an urgent mission for KIRP therapy. Methods The differentially expressed autophagy-related genes (DEARGs) were screened out according to the RNA sequencing data in The Cancer Genome Atlas database, then identified survival-related DEARGs to establish a prognostic model for survival predicting of KIRP patients. Then we verified the robustness and validity of the prognostic risk model through clinicopathological data. At last, we evaluate the prognostic value of genes that formed the prognostic risk model individually. Results We analyzed the expression of 232 autophagy-related genes (ARGs) in 289 KIRP and 32 non-tumor tissue cases, and 40 mRNAs were screened out as DEARGs. The functional and pathway enrichment analysis was done and protein–protein interaction network was constructed for all DEARGs. To sift candidate DEARGs associated with KIRP patients’ survival and create an autophagy-related risk prognostic model, univariate and multivariate Cox regression analysis were did separately. Eventually 3 desirable independent prognostic DEARGs (P4HB, NRG1, and BIRC5) were picked out and used for construct the autophagy-related risk model. The accuracy of the prognostic risk model for survival prediction was assessed by Kaplan–Meier plotter, receiver-operator characteristic curve, and clinicopathological correlational analyses. The prognostic value of above 3 genes was verified individually by survival analysis and expression analysis on mRNA and protein level. Conclusions The autophagy-related prognostic model is accurate and applicable, it can predict OS independently for KIRP patients. Three independent prognostic DEARGs can benefit for facilitate personalized target treatment too.


2020 ◽  
Vol 19 ◽  
pp. 153303382098417
Author(s):  
Ting-ting Liu ◽  
Shu-min Liu

Objective: The incidence of colorectal cancer is increasing every year, and autophagy may be related closely to the pathogenesis of colorectal cancer. Autophagy is a natural catabolic mechanism that allows the degradation of cellular components in eukaryotic cells. However, autophagy plays a dual role in tumorigenesis. It not only promotes normal cell survival and tumor growth but also induces cell death and suppresses tumors survival. In addition, the pathogenesis of various conditions, including inflammation, neurodegenerative diseases, or tumors, is associated with abnormal autophagy. The present work aimed to examine the significance of autophagy-related genes (ARGs) in prognosis prediction, to construct an autophagy prognostic model, and to identify independent prognostic factors for colorectal cancer (CRC). Methods: This study discovered a total of 36 ARGs in CRC cases using The Cancer Genome Atlas (TCGA) and Human Autophagy-dedicated (HADd) databases along with functional enrichment analysis. Then, an autophagy prognostic model was constructed using univariate Cox regression analysis, and the key prognostic genes were screened. Finally, independent prognostic markers were determined through independent prognostic analysis and clinical correlation analysis of key genes. Results: Of the 36 differentially expressed ARGs, 13 were related to prognosis, as determined by univariate Cox regression analysis. A total of 6 key genes were obtained by a multivariate Cox regression analysis. Independent prognostic values were shown by 3 genes, namely, microtubule-associated protein 1 light chain 3 (MAP1LC3C), small GTPase superfamily and Rab family (RAB7A), and WD-repeat domain phosphoinositide-interacting protein 2 (WIPI2) by independent prognostic analysis and clinical correlation. Conclusions: In this study, molecular bioinformatics technology was employed to determine and construct a prognostic model of autophagy for colon cancer patients, which revealed 3 autophagy-related features, namely, MAP1LC3C, WIPI2, and RAB7A.


Author(s):  
Yongmei Wang ◽  
Guimin Zhang ◽  
Ruixian Wang

Background: This study aims to explore the prognostic values of CT83 and CT83-related genes in lung adenocarcinoma (LUAD). Methods: We downloaded the mRNA profiles of 513 LUAD patients (RNA sequencing data) and 246 NSCLC patients (Affymetrix Human Genome U133 Plus 2.0 Array) from TCGA and GEO databases. According to the median expression of CT83, the TCGA samples were divided into high and low expression groups, and differential expression analysis between them was performed. Functional enrichment analysis of differential expression genes (DEGs) was conducted. Univariate Cox regression analysis and LASSO Cox regression analysis were performed to screen the optimal prognostic DEGs. Then we established the prognostic model. A Nomogram model was constructed to predict the overall survival (OS) probability of LUAD patients. Results: CT83 expression was significantly correlated to the prognosis of LUAD patients. A total of 59 DEGs were identified, and a predictive model was constructed based on six optimal CT83-related DEGs, including CPS1, RHOV, TNNT1, FAM83A, IGF2BP1, and GRIN2A, could effectively predict the prognosis of LUAD patients. The nomogram could reliably predict the OS of LUAD patients. Moreover, the six important immune checkpoints (CTLA4, PD1, IDO1, TDO2, LAG3, and TIGIT) were closely correlated with the Risk Score, which was also differentially expressed between the LUAD samples with high and low-Risk Scores, suggesting that the poor prognosis of LUAD patients with high-Risk Score might be due to the immunosuppressive microenvironments. Conclusion: A prognostic model based on six optimal CT83 related genes could effectively predict the prognosis of LUAD patients.


2020 ◽  
Author(s):  
Dai Zhang ◽  
Si Yang ◽  
Yiche Li ◽  
Meng Wang ◽  
Jia Yao ◽  
...  

Abstract Background: Ovarian cancer (OV) is deemed as the most lethal gynecological cancer in women. The aim of this study was construct an effective gene prognostic model for OV patients.Methods: The expression profiles of glycolysis-related genes (GRGs) and clinical data of patients with OV were extracted from The Cancer Genome Atlas (TCGA) database. Univariate, multivariate, and least absolute shrinkage and selection operator Cox regression analyses were conducted, and a prognostic signature based on GRGs was constructed. The predictive ability of the signature was analyzed in training and test sets.Results: Based on nine GRGs (ISG20, CITED2, PYGB, IRS2, ANGPTL4, TGFBI, LHX9, PC, and DDIT4), a gene risk signature was identified to predict the outcome of patients with OV. The signature showed a good prognostic ability for OV, particularly high-grade OV, in the TCGA dataset, with areas under the curve of 0.709, 0.762, and 0.808 for 3-, 5- and 10-year survival, respectively. Similar results were found in the test sets, and the signature was also an independent prognostic factor. Moreover, a nomogram combining the prediction model and clinical factors was constructed.Conclusion: Our study established a nine-GRG risk model and a nomogram to better perform on OV patients’ survival prediction. The risk model represents a promising and independent prognostic predictor for OV patients. Moreover, our study of GRGs could offer guidances for underlying mechanisms explorations in the future.


2021 ◽  
Author(s):  
Rui Feng ◽  
Jian Li ◽  
Weiling Xuan ◽  
Hanbo Liu ◽  
Dexin Cheng ◽  
...  

Abstract Background Hepatocellular carcinoma (HCC) is a prevalent primary liver cancer and the main cause of cancer mortality. Its high complexity and dismal prognosis bring dramatic difficulty to treatment. Due to the disclosed dual functions of autophagy in cancer development, understanding autophagy-related genes devotes into seeking novel biomarkers for HCC. Methods Differential expression of genes in normal and tumor groups was analyzed to acquire autophagy-related genes in HCC. GO and KEGG pathway analyses were conducted on these genes. Genes were then screened by univariate regression analysis. The screened genes were subjected to multivariate Cox regression analysis to build a prognostic model. The model was validated by ICGC validation set. Results Altogether, 42 autophagy-related differential genes were screened by differential expression analysis. Enrichment analysis showed that they were mainly enriched in pathways including regulation of autophagy and cell apoptosis. Genes were screened by univariate analysis and multivariate Cox regression analysis to build a prognostic model. The model was constituted by 6 feature genes: EIF2S1, BIRC5, SQSTM1, ATG7, HDAC1, FKBP1A. Validation confirmed the accuracy and independence of this model in predicting HCC patient’s prognosis. Conclusion A total of 6 feature genes were identified to build a prognostic risk model. This model is conducive to investigating interplay between autophagy-related genes and HCC prognosis.


PeerJ ◽  
2021 ◽  
Vol 9 ◽  
pp. e10884
Author(s):  
Xin Yu ◽  
Qian Yang ◽  
Dong Wang ◽  
Zhaoyang Li ◽  
Nianhang Chen ◽  
...  

Applying the knowledge that methyltransferases and demethylases can modify adjacent cytosine-phosphorothioate-guanine (CpG) sites in the same DNA strand, we found that combining multiple CpGs into a single block may improve cancer diagnosis. However, survival prediction remains a challenge. In this study, we developed a pipeline named “stacked ensemble of machine learning models for methylation-correlated blocks” (EnMCB) that combined Cox regression, support vector regression (SVR), and elastic-net models to construct signatures based on DNA methylation-correlated blocks for lung adenocarcinoma (LUAD) survival prediction. We used methylation profiles from the Cancer Genome Atlas (TCGA) as the training set, and profiles from the Gene Expression Omnibus (GEO) as validation and testing sets. First, we partitioned the genome into blocks of tightly co-methylated CpG sites, which we termed methylation-correlated blocks (MCBs). After partitioning and feature selection, we observed different diagnostic capacities for predicting patient survival across the models. We combined the multiple models into a single stacking ensemble model. The stacking ensemble model based on the top-ranked block had the area under the receiver operating characteristic curve of 0.622 in the TCGA training set, 0.773 in the validation set, and 0.698 in the testing set. When stratified by clinicopathological risk factors, the risk score predicted by the top-ranked MCB was an independent prognostic factor. Our results showed that our pipeline was a reliable tool that may facilitate MCB selection and survival prediction.


2022 ◽  
Vol 11 ◽  
Author(s):  
Zehua Liu ◽  
Rongfang Pan ◽  
Wenxian Li ◽  
Yanjiang Li

This study aimed to identify critical cell cycle-related genes (CCRGs) in prostate cancer (PRAD) and to evaluate the clinical prognostic value of the gene panel selected. Gene set variation analysis (GSVA) of dysregulated genes between PRAD and normal tissues demonstrated that the cell cycle-related pathways played vital roles in PRAD. Patients were classified into four clusters, which were associated with recurrence-free survival (RFS). Moreover, 200 prognostic-related genes were selected using the Kaplan–Meier (KM) survival analysis and univariable Cox regression. The prognostic CCRG risk score was constructed using random forest survival and multivariate regression Cox methods, and their efficiency was validated in Memorial Sloan Kettering Cancer Center (MSKCC) and GSE70770. We identified nine survival-related genes: CCNL2, CDCA5, KAT2A, CHTF18, SPC24, EME2, CDK5RAP3, CDC20, and PTTG1. Based on the median risk score, the patients were divided into two groups. Then the functional enrichment analyses, mutational profiles, immune components, estimated half-maximal inhibitory concentration (IC50), and candidate drugs were screened of these two groups. In addition, the characteristics of nine hub CCRGs were explored in Oncomine, cBioPortal, and the Human Protein Atlas (HPA) datasets. Finally, the expression profiles of these hub CCRGs were validated in RWPE-1 and three PRAD cell lines (PC-3, C4-2, and DU-145). In conclusion, our study systematically explored the role of CCRGs in PRAD and constructed a risk model that can predict the clinical prognosis and immunotherapeutic benefits.


Author(s):  
Hanyi Zeng ◽  
Chengdong Liu ◽  
Xiaohan Zhou ◽  
Li Liu

Background: Hepatocellular carcinoma (HCC) is a malignant tumour with poor prognosis. The effect of DNA repair on prognosis cannot be ignored; and long non-coding RNA (lncRNA) can regulate the DNA repair process. Objective: : To obtain DNA repair-associated lncRNA (DR-lncRNA) prognostic signature for improved ability to prediction of HCC prognosis. Methods: Our study used the Cancer Genome Atlas database. Gene set variation analysis was performed to differentiate high and low levels of DNA repair to identify DR-lncRNAs. By performing univariate Cox regression, LASSO regression, and multivariate Cox regression analyses, we finally obtained a DR-lncRNA prognostic signature and constructed a nomogram prognostic model. Time-dependent receiver operating characteristic (ROC) curves, calibration plots, decision curve analysis (DCA), and clinical impact curves were used to assess predictive ability and clinical utility. Differentially expressed genes (DEGs) functional enrichment analysis was performed to further explore the underlying mechanisms that influence HCC prognosis. Results: We obtained a DR-lncRNA prognostic signature—AP002478.1, AC116351.1, LINC02580, and LINC00861. The ROC curves and calibration plots showed good discrimination and calibration properties. Combining the DR-lncRNA prognostic signature and tumour stages, we established a nomogram prognostic model. DCA and clinical impact curves showed the clinical utility of the nomogram prognostic model. DEGs of high-risk and low-risk groups predicted by the DR-lncRNA prognostic were significantly associated with cell cycle and various metabolic pathways and biological processes such as the oxidation-reduction process and cell division. Conclusion: We identified a DR-lncRNA prognostic signature and constructed a nomogram prognostic model, which could be a beneficial prognostic strategy for HCC.


2020 ◽  
Vol 19 ◽  
pp. 153303382096357
Author(s):  
Xiaoyong Gong ◽  
Bobin Ning

Prostate cancer (PCa) is a highly malignant tumor, with increasing incidence and mortality rates worldwide. The aim of this study was to identify the prognostic lncRNAs and construct an lncRNA signature for PCa diagnosis by the interaction network between lncRNAs and protein-coding genes (PCGs). The differentially expressed lncRNAs (DElncRNAs) and PCGs (DEPCGs) between PCa and normal prostate tissues were screened from The Cancer Genome Atlas (TCGA) database. The DEPCGs were functionally annotated in terms of the enriched pathways. Weighted gene co-expression network analysis (WGCNA) of 104 PCa samples identified 15 co-expression modules, of which the Turquoise module was negatively correlated with cancer and included 5 key lncRNAs and 47 PCGs. KEGG pathway analyses of the core 47 PCGs showed significant enrichment in classic PCa-related pathways, and overlapped with the enriched pathways of the DEPCGs. LINC00857, LINC00900, LINC00908, LINC00900, SNHG3 and FENDRR were significantly associated with the survival of PCa and have not been reported previously. Finally, Multivariable Cox regression analysis was used to establish a prognostic risk formula, and the patients were accordingly stratified into the low- and high-risk groups. The latter had significantly worse OS compared to the low-risk group (P < 0.01), and the area under the receiver operating characteristic curve (ROC) of 14-year OS was 0.829. The accuracy of our prediction model was determined by calculating the corresponding concordance index (C-index) and risk curves. In conclusion, we established a 5-lncRNA prognostic signature that provides insights into the biological and clinical relevance of lncRNAs in PCa.


Sign in / Sign up

Export Citation Format

Share Document