stomach adenocarcinoma
Recently Published Documents


TOTAL DOCUMENTS

158
(FIVE YEARS 112)

H-INDEX

9
(FIVE YEARS 3)

Author(s):  
Ruoxi Xiao ◽  
Shasha Wang ◽  
Jing Guo ◽  
Shihai Liu ◽  
Aiping Ding ◽  
...  

2022 ◽  
Vol 23 (1) ◽  
Author(s):  
Zixuan Wang ◽  
Zixin Tian ◽  
Xi Song ◽  
Jun Zhang

Abstract Background Formin-binding protein 1/17 (FNBP1/FBP17), as a membrane-bound protein, is wildly expressed in eukaryotic cells and performs a critical role in tumor tumorigenesis and progression. However, the relationship between FNBP1 and immune infiltrating cells, prognostic value in patients still require comprehensive understanding. We purposed to explore the correlations of FNBP1 expression, prognosis and immune infiltration levels in various cancers. Method The expression and survival data of FNBP1 were collected from Oncomine, TIMER, GEPIA, Kaplan–Meier Plotter and PrognoScan databases. Correlations between FNBP1 and immune infiltrates were analyzed in TIMER and GEPIA databases. Results Compared with normal tissues, FNBP1 is significantly differentially expressed in a variety of tumor tissues. FNBP1 has significant and complex effects on the prognosis of kinds of cancers. High-expression was obviously correlated with better prognosis in breast carcinoma and lung adenocarcinoma, while worse prognosis in stomach adenocarcinoma. Besides, FNBP1 had a correlation with various immune infiltrating cells and diverse immune gene markers in breast invasive carcinoma (BRCA), lung adenocarcinoma (LUAD), and stomach adenocarcinoma (STAD). FNBP1 was also positively correlated with the adjustment of CD8+ cells, T cells, M2 macrophage, neutrophils, monocyte, Th1 cells, T regulatory cells (Treg) and Tumor-associated macrophages (TAMs). The expression level of FNBP1 is closely positively correlated with the expression level of multiple immune checkpoints in the three cancers. In addition, FNBP1 is significantly positively correlated with the expression levels of a variety of immunosuppressive molecules. Conclusion Our findings reveal FNBP1 can serve as a significant biomarker to influence the prognosis and the immune infiltrating levels in different cancers. The differential expression of FNBP1 might not only contribute to the judgment of metastatic and non-metastatic tumors but also in the immune escape by upregulating the expression of immune checkpoints.


2021 ◽  
Author(s):  
Chenxi Yuan ◽  
Qingwei Wang ◽  
Yipeng Song ◽  
Jinming Yu

Abstract Background: Stomach adenocarcinoma (STAD) is a common cancer type around the world. The prognosis in advanced patients is poor. Since NLRP3 was not extensively studied in the field of tumor, so we aimed to identify the impact of NLRP3 on STAD by bioinformatics analyses and in vitro experiments. Methods: TCGA , kaplan-Meier Plotter and TIMER database were utilized in this study. We compared the expression of NLRP3 in different cancers and evaluated its influence on survival of gastric carcinoma patients. The correlations between clinical information and NLRP3 expression were analyzed using logistic regression. Clinicopathologic characteristics associated with overall survival in were analyzed by Cox regression. In addition, we explored the correlation between NLRP3 and immune infiltrates. GSEA and co-expressed gene with NLRP3 were also done in this study. Finally, we verified the NLRP3 expression in paired gastric cancer tissues and adjacent normal tissues by western blot and q-PCR. We also constructed NLRP3 overexpression model in gastric cancer cell line and observed cell proliferation ability.Results: NLRP3 expressed disparately in gastric tumor tissues and normal tissues. Cox regression analysis indicated that up-regulated NLRP3 was an independent prognostic factor for bad prognosis in STAD. Logistic regression analysis showed increased NLRP3 expression was significantly correlated with unfavorable clinicopathologic parameters such as higher T stage, higher histologic grade and worse survival outcome. Specifically, a positive correlation between increased NLRP3 expression and immune infiltrating level of various immune cells was observed. Conclusion: Together with all these findings, increased NLRP3 expression correlates with poor prognosis, unfavorable clinicopathologic parameters and increased proportion of immune cells in STAD. In vitro analysis revealed that cell proliferation ability was enhanced in gastric cancer cells trasnfected with NLRP3 overexpression. These conclusions indicate that NLRP3 has great potential to serve as a biomarker for evaluating prognosis in patients diagnosed with gastric carcinoma.


2021 ◽  
Author(s):  
Ru-Hong Tu ◽  
Jian-Xian Lin ◽  
Jian-Wei Xie ◽  
Jia-Bin Wang ◽  
Jun Lu ◽  
...  

Abstract Gastric cancer is a leading cause of death from malignant tumors worldwide. With the development of genome sequencing technology, an increasing number of key driver genes and tumor suppressors have been discovered. Some studies have suggested that Dynamin 3 (DNM3) is a novel tumor suppressor; however, the role of DNM3 in malignancy remains unclear. We performed a systematic analysis using The Cancer Genome Atlas Stomach Adenocarcinoma (TCGA-STAD) cohorts, and 160 patients with stomach adenocarcinoma at Fujian Medical University Union Hospital (FJMUUH) (48 quantitative PCR [qPCR] and 112 immunohistochemistry). DNM3 expression was found to be downregulated in gastric cancer compared to that in paraneoplastic tissue. Low expression of DNM3 was mainly associated with DNM3 promoter hypermethylation status. Low expression of DNM3 can upregulate the tumor cell cycle and oxidative phosphorylation process and downregulate immune regulation, and Th17 and Th2 immune cell infiltration was increased in patients with lower expression of DNM3. Patients with a lower DNM3 expression had a higher rate of lymph node metastasis and poor prognosis. In summary, DNM3 is a tumor suppressor and an independent predictive factor of poor prognosis that regulates the cell cycle and immunosuppression in the tumor microenvironment in gastric cancer via methylation.


2021 ◽  
Vol 8 ◽  
Author(s):  
Tong Wang ◽  
Weiwei Wen ◽  
Hongfei Liu ◽  
Jun Zhang ◽  
Xiaofeng Zhang ◽  
...  

Background: Stomach adenocarcinoma (STAD) is a significant global health problem. It is urgent to identify reliable predictors and establish a potential prognostic model.Methods: RNA-sequencing expression data of patients with STAD were downloaded from the Gene Expression Omnibus (GEO) and the Cancer Genome Atlas (TCGA) database. Gene expression profiling and survival analysis were performed to investigate differentially expressed genes (DEGs) with significant clinical prognosis value. Overall survival (OS) analysis and univariable and multivariable Cox regression analyses were performed to establish the prognostic model. Protein–protein interaction (PPI) network, functional enrichment analysis, and differential expression investigation were also performed to further explore the potential mechanism of the prognostic genes in STAD. Finally, nomogram establishment was undertaken by performing multivariate Cox regression analysis, and calibration plots were generated to validate the nomogram.Results: A total of 229 overlapping DEGs were identified. Following Kaplan–Meier survival analysis and univariate and multivariate Cox regression analysis, 11 genes significantly associated with prognosis were screened and five of these genes, including COL10A1, MFAP2, CTHRC1, P4HA3, and FAP, were used to establish the risk model. The results showed that patients with high-risk scores have a poor prognosis, compared with those with low-risk scores (p = 0.0025 for the training dataset and p = 0.045 for the validation dataset). Subsequently, a nomogram (including TNM stage, age, gender, histologic grade, and risk score) was created. In addition, differential expression and immunohistochemistry stain of the five core genes in STAD and normal tissues were verified.Conclusion: We develop a prognostic-related model based on five core genes, which may serve as an independent risk factor for survival prediction in patients with STAD.


2021 ◽  
Vol 2021 ◽  
pp. 1-19
Author(s):  
Dongyun Xue ◽  
Dong Li ◽  
Cong Dou ◽  
Junshan Li

Background. Activation of NOTCH signaling pathways, which are key regulators of multiple cellular functions, has been frequently implicated in cancer pathogenesis, and NOTCH inhibitors have received much recent focus in the context of cancer therapeutics. However, the role and possible involvement of NOTCH pathways in stomach adenocarcinoma (STAD) are unclear. Here, putative regulatory mechanisms and functions of NOTCH pathways in STAD were investigated. Methods. Publicly available data from the TCGA-STAD database were utilized to explore the involvement of canonical NOTCH pathways in STAD by analyzing RNA expression levels of NOTCH receptors, ligands, and downstream genes. Statistical analysis of the data pertaining to cancer and noncancerous samples was performed using R software packages and public databases/webservers. Results. Significant differential gene expression between control and STAD samples was noted for all NOTCH receptors (NOTCH1, 2, 3, and 4), the delta-like NOTCH ligands (DLL-3 and 4), and typical downstream genes (HES1 and HEY1). Four genes (NOTCH1, NOTCH2, NOTCH3, and HEY1) presented prognostic values for the STAD outcome in terms of overall survival. Functional enrichment analysis indicated that NOTCH family genes-strongly correlated genes were mainly enriched in several KEGG signaling pathways such as the PI3K-Akt signaling pathway, human papillomavirus infection, focal adhesion, Rap1 signaling pathway, and ECM-receptor interaction. Gene set enrichment analysis (GSEA) results showed that NOTCH family genes-significantly correlated genes were mainly enriched in four signaling pathways, ECM (extracellular matrix), tumor angiogenesis, inflammatory response, and immune regulation. Conclusions. NOTCH family genes may play an essential role in the progression of STAD by modulating immune cells and mediating ECM synthesis, angiogenesis, focal adhesion, and PI3K-Akt signaling. Multiple NOTCH family genes are valuable candidate biomarkers or therapeutic targets for the management of STAD.


2021 ◽  
Author(s):  
Yi He ◽  
Haiyang Zhang ◽  
Yan Zhang ◽  
Peiyun Wang ◽  
Kegan Zhu ◽  
...  

Abstract Background: Stomach adenocarcinoma (STAD) is the common cancer and ranks third leading cause of cancer death worldwide. TGF‑β receptor 1 (TGFBR1), serving important roles in the TGF‑β family, the mechanisms whereby TGFβ2 governs tumor progression, immune cell infiltration and its correlation with tumor microenvironment (TME) in STAD remains unintelligible. Methods: First, we used the data in the TCGA, GEPIA, and HPA databases to explore the expression level of TGFBR1 in STAD, the correlation between TGFBR1 expression and the clinical features of STAD, its impact on the survival of STAD. Subsequently, a receiver operating characteristic (ROC) curve and nomogram were constructed and LASSO (the Least Absolute Shrinkage and Selection Operator)-selected features were used to build the TGFBR1 prognostic signature. Moreover, GSEA enrichment analysis is used to find the potential molecular mechanism of TGFBR1 to promote the malignant process of STAD. Finally, we further explored the influence of theTGFBR1 expression on the immune microenvironment of STAD patients through the TIMER2.0 and GEPIA database.Results: In our study, TGFBR1 expression was significantly elevated in patients with STAD and positively co-expression with pathologic stage, lymph node metastases (LNM) stage and histopathological grade of STAD. LASSO-selected features were used to build the TGFBR1 prognostic signature. 9 factors with non-zero coefficients were identified. The corresponding risk scores were computed, according to the following formula: Risk score = (-0.2914) *DIXDC1+ (0.1113) *STON1-GTF2A1L+(0.3092) *FERMT2+(-0.0146) *BHMT2+(0.1798) *ABCC9+(0.068) *MSRB3+(-0.1007) *SYNC+(-0.0891) *SORBS1+(0.0828) *TGFBR1.Survival analysis revealed that patients with high TGFBR1 had shorter OS, FP, and PPS. Multivariate Cox analysis revealed TGFBR1 was an independent prognostic factor for OS in STAD. The receiver operating characteristic (ROC) analysis suggested high diagnostic value with the area under curve (AUC) of TGFBR1 was 0.739, and a prognostic nomogram involving age, T, N, M classification, pathologic stage, primary therapy outcome, histologic grade and TGFBR1 to predict the 1, 3, 5-year OS was constructed. GSEA revealed that high TGFBR1 expression was correlated with pathway in cancer, MAPK signaling pathway, NOTCH signaling pathway, focal adhesion and VEGF-C production. ssGSEA showed that TGFBR1 is correlated with NK cells, Tem and Th17 cells. Furthermore, elevated TGFBR1 expression was found to be significantly correlated with several immune checkpoint and immune markers associated with immune cell subsets. Conclusion: In summary, TGFBR1 could be a prognostic biomarker and an important regulator of immune cell infiltration in STAD. The present study revealed the probable underlying molecular mechanisms of TGFBR1 in STAD and provided a potential target for improving the prognosis.


BMC Cancer ◽  
2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Chen Han ◽  
Cong Zhang ◽  
Huixia Wang ◽  
Kexin Li ◽  
Lianmei Zhao

Abstract Background Stomach adenocarcinoma (STAD), which accounts for approximately 95% of gastric cancer types, is a malignancy cancer with high morbidity and mortality. Tumor angiogenesis plays important roles in the progression and pathogenesis of STAD, in which long noncoding RNAs (lncRNAs) have been verified to be crucial for angiogenesis. Our study sought to construct a prognostic signature of angiogenesis-related lncRNAs (ARLncs) to accurately predict the survival time of STAD. Methods The RNA-sequencing dataset and corresponding clinical data of STAD were acquired from The Cancer Genome Atlas (TCGA). ARLnc sets were obtained from the Ensemble genome database and Molecular Signatures Database (MSigDB, Angiogenesis M14493, INTegrin pathway M160). A ARLnc-related prognostic signature was then constructed via univariate Cox and multivariate Cox regression analysis in the training cohort. Survival analysis and Cox regression were performed to assess the performance of the prognostic signature between low- and high-risk groups, which was validated in the validation cohort. Furthermore, a nomogram that combined the clinical pathological characteristics and risk score conducted to predict the overall survival (OS) of STAD. In addition, ARLnc-mRNA coexpression pairs were constructed with Pearson’s correlation analysis and visualized to infer the functional annotation of the ARLncs by gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. The expression of four ARLncs in STAD and their correlation with the angiogenesis markers, CD34 and CD105, were also validated by RT–qPCR in a clinical cohort. Results A prognostic prediction signature including four ARLncs (PVT1, LINC01315, AC245041.1, and AC037198.1) was identified and constructed. The OS of patients in the high-risk group was significantly lower than that of patients in the low-risk group (p < 0.001). The values of the time-dependent area under the curve (AUC) for the ARLnc signature for 1-, 3-, and 5- year OS were 0.683, 0.739, and 0.618 in the training cohort and 0.671, 0.646, and 0.680 in the validation cohort, respectively. Univariate and multivariate Cox regression analyses indicated that the ARLnc signature was an independent prognostic factor for STAD patients (p < 0.001). Furthermore, the nomogram and calibration curve showed accurate prediction of the survival time based on the risk score. In addition, 262 mRNAs were screened for coexpression with four ARLncs, and GO analysis showed that mRNAs were mainly involved in biological processes, including angiogenesis, cell adhesion, wound healing, and extracellular matrix organization. Furthermore, correlation analysis showed that there was a positive correlation between risk score and the expression of the angiogenesis markers, CD34 and CD105, in TCGA datasets and our clinical sample cohort. Conclusion Our study constructed a prognostic signature consisting of four ARLnc genes, which was closely related to the survival of STAD patients, showing high efficacy of the prognostic signature. Thus, the present study provided a novel biomarker and promising therapeutic strategy for patients with STAD.


Sign in / Sign up

Export Citation Format

Share Document