1
|
Liu Z, Huang H, Yu Y, Li L, Shi X, Wang F. Exploring the mechanism of ellagic acid against gastric cancer based on bioinformatics analysis and network pharmacology. J Cell Mol Med 2023; 27:3878-3896. [PMID: 37794689 PMCID: PMC10718161 DOI: 10.1111/jcmm.17967] [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: 04/27/2023] [Revised: 08/17/2023] [Accepted: 08/30/2023] [Indexed: 10/06/2023] Open
Abstract
Ellagic acid (EA) is a natural polyphenolic compound. Recent studies have shown that EA has potential anticancer properties against gastric cancer (GC). This study aims to reveal the potential targets and mechanisms of EA against GC. This study adopted methods of bioinformatics analysis and network pharmacology, including the weighted gene co-expression network analysis (WGCNA), construction of protein-protein interaction (PPI) network, receiver operating characteristic (ROC) and Kaplan-Meier (KM) survival curve analysis, Gene Ontology (GO) function and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, molecular docking and molecular dynamics simulations (MDS). A total of 540 EA targets were obtained. Through WGCNA, we obtained a total of 2914 GC clinical module genes, combined with the disease database for screening, a total of 606 GC-related targets and 79 intersection targets of EA and GC were obtained by constructing Venn diagram. PPI network was constructed to identify 14 core candidate targets; TP53, JUN, CASP3, HSP90AA1, VEGFA, HRAS, CDH1, MAPK3, CDKN1A, SRC, CYCS, BCL2L1 and CDK4 were identified as the key targets of EA regulation of GC by ROC and KM curve analysis. The enrichment analysis of GO and KEGG pathways of key targets was performed, and they were mainly enriched in p53 signalling pathway, PI3K-Akt signalling pathway. The results of molecular docking and MDS showed that EA could effectively bind to 13 key targets to form stable protein-ligand complexes. This study revealed the key targets and molecular mechanisms of EA against GC and provided a theoretical basis for further study of the pharmacological mechanism of EA against GC.
Collapse
Affiliation(s)
- Zhiyao Liu
- Department of Rehabilitation MedicineShandong University of Traditional Chinese MedicineJinanChina
| | - Hailiang Huang
- Department of Rehabilitation MedicineShandong University of Traditional Chinese MedicineJinanChina
| | - Ying Yu
- Innovative Institute of Chinese Medicine and PharmacyShandong University of Traditional Chinese MedicineJinanChina
| | - Lingling Li
- Department of Rehabilitation MedicineShandong University of Traditional Chinese MedicineJinanChina
| | - Xin Shi
- Department of Rehabilitation MedicineShandong University of Traditional Chinese MedicineJinanChina
| | - Fangqi Wang
- Department of Rehabilitation MedicineShandong University of Traditional Chinese MedicineJinanChina
| |
Collapse
|
2
|
Zhou F, Dou X, Li C. CKB affects human osteosarcoma progression by regulating the p53 pathway. Am J Cancer Res 2022; 12:4652-4665. [PMID: 36381321 PMCID: PMC9641398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 09/25/2022] [Indexed: 06/16/2023] Open
Abstract
This study aimed to explore the role of the creatine kinase B (CKB) gene in the development of human osteosarcoma (OS). Western blotting and qRT-PCR were performed to detect CKB expression in tissues and cells. CCK-8, colony formation, flow cytometry, Transwell, and cell scratch assays were performed to detect OS cell viability, proliferation, apoptosis, invasion, and migration. Gene set enrichment analysis (GSEA) was used to conduct signal pathway enrichment. CKB expression was higher in OS tissues and cells than that in normal tissues and cells. Silencing CKB expression reduced cell proliferation, migration, and invasion, and improved cell apoptosis in HOS cells, while overexpressing CKB increased cell proliferation, migration, and invasion, and decreased apoptosis in U2-OS cells. GSEA showed that CKB affected the p53 signaling pathway. Overexpression of CKB inhibited the protein expression of p53, p21, and Bax and promoted the expression of Bcl-2 and MDM2 in U2-OS cells. Conversely, silencing CKB promoted the protein expression of p53, p21, and Bax, and inhibited the expression of Bcl-2 and MDM2 in HOS cells. Silencing p53 could reverse the effect of the silencing CKB in HOS cells, and overexpressing p53 could reverse the effect of overexpressing CKB in U2-OS cells. Taken together, CKB affects the development of OS by regulating the activity of the p53 signaling pathway.
Collapse
Affiliation(s)
- Fengxin Zhou
- Department of Orthopedics, Tianjin Integrative Medicine Hospital (Tianjin Nankai Hospital)Tianjin 300100, China
| | - Xinli Dou
- Department of Oncology, Dagang HospitalBinhai New Area, Tianjin 300270, China
| | - Chenguang Li
- Department of Orthopedics, Tianjin Integrative Medicine Hospital (Tianjin Nankai Hospital)Tianjin 300100, China
| |
Collapse
|
3
|
Nisar A, Kayani MA, Nasir W, Mehmood A, Ahmed MW, Parvez A, Mahjabeen I. Fyn and Lyn gene polymorphisms impact the risk of thyroid cancer. Mol Genet Genomics 2022; 297:1649-1659. [PMID: 36058999 DOI: 10.1007/s00438-022-01946-7] [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: 06/27/2021] [Accepted: 08/11/2022] [Indexed: 10/14/2022]
Abstract
Thyroid cancer is the most common malignancy of the endocrine glands, and during last couple of decades, its incidence has risen alarmingly, across the globe. Etiology of thyroid cancer is still debatable. There are a few worth mentioning risk factors which contribute to initiation of abnormalities in thyroid gland leading to cancer. Genetic instability is major risk factors in thyroid carcinogenesis. Among the genetic factors, the Src family of genes (Src, Yes1, Fyn and Lyn) have been implicated in many cancers but there is little data regarding the association of these (Src, Yes1, Fyn and Lyn) genes with thyroid carcinogenesis. Fyn and Lyn genes of Src family found engaged in proliferation, migration, invasion, angiogenesis, and metastasis in different cancers. This study was planned to examine the effect of Fyn and Lyn SNPs on thyroid cancer risk in Pakistani population in 500 patients and 500 controls. Three polymorphisms of Fyn gene (rs6916861, rs2182644 and rs12910) and three polymorphisms of Lyn gene (rs2668011, rs45587541 and rs45489500) were analyzed using Tetra-primer ARMS-PCR followed by DNA sequencing. SNP rs6916861 of Fyn gene mutant genotype (CC) showed statistically significant threefold increased risk of thyroid cancer (P < 0.0001). In case of rs2182644 of Fyn gene, mutant genotype (AA) indicated statistically significant 17-fold increased risk of thyroid cancer (P < 0.0001). Statistically significant threefold increased risk of thyroid cancer was observed in genotype AC (P < 0.0001) of Fyn gene polymorphism rs12910. In SNP rs2668011 of Lyn gene, TT genotype showed statistically significant threefold increased risk of thyroid cancer (P < 0.0001). In case of rs45587541 of Lyn gene, GA genotypes showed statistically significant 11-fold increased risk in thyroid cancer (P < 0.0001). Haplotype analysis revealed that AAATAG*, AGACAG*, AGCCAA*, AGCCAG*, CAATAG*, CGCCAG* and CGCCGA* haplotypes of Fyn and Lyn polymorphisms are associated with increased thyroid cancer risk. These results showed that genotypes and allele distribution of Fyn and Lyn are significantly linked with increased thyroid cancer risk and could be genetic adjuster for said disease.
Collapse
Affiliation(s)
- Asif Nisar
- Cancer Genetics and Epigenetics Lab, Department of Biosciences, COMSATS University Islamabad, Park Road Tarlai Kalan, Islamabad, Pakistan
| | - Mahmood Akhtar Kayani
- Cancer Genetics and Epigenetics Lab, Department of Biosciences, COMSATS University Islamabad, Park Road Tarlai Kalan, Islamabad, Pakistan
| | - Wajiha Nasir
- Department of Radiation, Nuclear Oncology Radiation Institute, Islamabad, Pakistan
| | - Azhar Mehmood
- Cancer Genetics and Epigenetics Lab, Department of Biosciences, COMSATS University Islamabad, Park Road Tarlai Kalan, Islamabad, Pakistan
| | - Malik Waqar Ahmed
- Cancer Genetics and Epigenetics Lab, Department of Biosciences, COMSATS University Islamabad, Park Road Tarlai Kalan, Islamabad, Pakistan.,Pakistan Institute of Rehabilitation Sciences (PIRS), Isra University Islamabad Campus, Islamabad, Pakistan
| | - Aamir Parvez
- Cancer Genetics and Epigenetics Lab, Department of Biosciences, COMSATS University Islamabad, Park Road Tarlai Kalan, Islamabad, Pakistan
| | - Ishrat Mahjabeen
- Cancer Genetics and Epigenetics Lab, Department of Biosciences, COMSATS University Islamabad, Park Road Tarlai Kalan, Islamabad, Pakistan.
| |
Collapse
|
4
|
Xie LY, Huang HY, Fang T, Liang JY, Hao YL, Zhang XJ, Xie YX, Wang C, Tan YH, Zeng L. A Prognostic Survival Model of Pancreatic Adenocarcinoma Based on Metabolism-Related Gene Expression. Front Genet 2022; 13:804190. [PMID: 35664305 PMCID: PMC9158121 DOI: 10.3389/fgene.2022.804190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 03/22/2022] [Indexed: 11/13/2022] Open
Abstract
Accurately predicting the survival prospects of patients suffering from pancreatic adenocarcinoma (PAAD) is challenging. In this study, we analyzed RNA matrices of 182 subjects with PAAD based on public datasets obtained from The Cancer Genome Atlas (TCGA) as training datasets and those of 63 subjects obtained from the Gene Expression Omnibus (GEO) database as the validation dataset. Genes regulating the metabolism of PAAD cells correlated with survival were identified. Furthermore, LASSO Cox regression analyses were conducted to identify six genes (XDH, MBOAT2, PTGES, AK4, PAICS, and CKB) to create a metabolic risk score. The proposed scoring framework attained the robust predictive performance, with 2-year survival areas under the curve (AUCs) of 0.61 in the training cohort and 0.66 in the validation cohort. Compared with the subjects in the low-risk cohort, subjects in the high-risk training cohort presented a worse survival outcome. The metabolic risk score increased the accuracy of survival prediction in patients suffering from PAAD.
Collapse
Affiliation(s)
- Lin-Ying Xie
- Bethune Institute of Epigenetic Medicine, The First Hospital of Jilin University, Changchun, China
| | - Han-Ying Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Tian Fang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Jia-Ying Liang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Yu-Lei Hao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Xue-Jiao Zhang
- Department of Endocrinology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yi-Xin Xie
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Chang Wang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Ye-Hui Tan
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Lei Zeng
- Bethune Institute of Epigenetic Medicine, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
5
|
Song S, Zhou J, Li Y, Liu J, Li J, Shu P. Network pharmacology and experimental verification based research into the effect and mechanism of Aucklandiae Radix-Amomi Fructus against gastric cancer. Sci Rep 2022; 12:9401. [PMID: 35672352 PMCID: PMC9174187 DOI: 10.1038/s41598-022-13223-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 05/23/2022] [Indexed: 12/19/2022] Open
Abstract
To investigate the mechanism of the Aucklandiae Radix–Amomi Fructus (AR–AF) herb pair in treating gastric cancer (GC) by using network pharmacology and experimental verification. Using the traditional Chinese medicine system pharmacology database and analysis platform (TCMSP), the major active components and their corresponding targets were estimated and screened out. Using Cytoscape 3.7.2 software, a visual network was established using the active components of AR–AF and the targets of GC. Based on STRING online database, the protein interaction network of vital targets was built and analyzed. With the Database for Annotation, Visualization, and Integrated Discovery (DAVID) server, the gene ontology (GO) biological processes and the Kyoto Encyclopedia of Genes and Genomes (KEGG) signaling pathways of the target enrichment were performed. AutoDock Vina was used to perform molecular docking and calculate the binding affinity. The mRNA and protein expression levels of the hub targets were analyzed by the Oncomine, GEPIA, HPA databases and TIMER online tool, and the predicted targets were verified by qRT–PCR in vitro. Eremanthin, cynaropicrin, and aceteugenol were identified as vital active compounds, and AKT1, MAPK3, IL6, MAPK1, as well as EGFR were considered as the major targets. These targets exerted therapeutic effects on GC by regulating the cAMP signaling pathway, and PI3K-Akt signaling pathway. Molecular docking revealed that these active compounds and targets showed good binding interactions. The validation in different databases showed that most of the results were consistent with this paper. The experimental results confirmed that eremanthin could inhibit the proliferation of AGS by reducing the mRNA expression of hub targets. As predicted by network pharmacology and validated by the experimental results, AR–AF exerts antitumor effects through multiple components, targets, and pathways, thereby providing novel ideas and clues for the development of preparations and the treatment of GC.
Collapse
Affiliation(s)
- Siyuan Song
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China.,Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China.,Jiangsu Provincial Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Jiayu Zhou
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China.,Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China.,Jiangsu Provincial Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Ye Li
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China.,Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China.,Jiangsu Provincial Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Jiatong Liu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China.,Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Jingzhan Li
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China.,Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China.,Jiangsu Provincial Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Peng Shu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China. .,Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China. .,Jiangsu Provincial Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
6
|
Ribeiro HF, de Castro Sant' Anna C, de Jesus Oliveira Kato V, de Sousa Brasil RM, Bona AB, da Costa DF, Lima IK, Soares PC, Guimarães APA, de Assumpção PP, Burbano RR. CDC25B Inhibition by menadione: a potential new therapeutical approach. Anticancer Agents Med Chem 2022; 22:2927-2932. [PMID: 35440317 DOI: 10.2174/1871520622666220418131935] [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: 11/19/2021] [Revised: 01/28/2022] [Accepted: 02/24/2022] [Indexed: 12/24/2022]
Abstract
Gastric cancer (GC) is the fifth most common type of tumor and the third leading cause of cancer death worldwide. The evolution of gastric carcinogenesis is still poorly understood and, for this reason, preclinical research protocols were established that included the development of gastric cancer cell lines and the establishment of models of gastric carcinogenesis in non-human primate Sapajus apella. A comprehensive literature search was performed in relevant databases such as PubMed, ResearchGate and Google Scholar to identify studies related to the topic. After an in-depth study of these reports, significant data/data were collected and compiled under appropriate headings. The main result of the studies carried out by the group on GC is the demonstration of the MYC gene overexpression as a common phenomenon in stomach carcinogenesis. Furthermore, we revealed that reducing the expression of the CDC25B gene, regulated by the MYC protein, is a therapeutic strategy against stomach tumors. This review article reveals preclinical evidence that treatment with menadione in experimental models of gastric tumorigenesis, in vivo and in vitro, inhibits the action of the phosphatase CDC25B and, consequently, prevents cell proliferation, invasion and migration.
Collapse
|
7
|
Jung M, Lee C, Han D, Kim K, Yang S, Nikas IP, Moon KC, Kim H, Song MJ, Kim B, Lee H, Ryu HS. Proteomic-Based Machine Learning Analysis Reveals PYGB as a Novel Immunohistochemical Biomarker to Distinguish Inverted Urothelial Papilloma From Low-Grade Papillary Urothelial Carcinoma With Inverted Growth. Front Oncol 2022; 12:841398. [PMID: 35402263 PMCID: PMC8987228 DOI: 10.3389/fonc.2022.841398] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/24/2022] [Indexed: 11/13/2022] Open
Abstract
Background The molecular biology of inverted urothelial papilloma (IUP) as a precursor disease of urothelial carcinoma is poorly understood. Furthermore, the overlapping histology between IUP and papillary urothelial carcinoma (PUC) with inverted growth is a diagnostic pitfall leading to frequent misdiagnoses. Methods To identify the oncologic significance of IUP and discover a novel biomarker for its diagnosis, we employed mass spectrometry-based proteomic analysis of IUP, PUC, and normal urothelium (NU). Machine learning analysis shortlisted candidate proteins, while subsequent immunohistochemical validation was performed in an independent sample cohort. Results From the overall proteomic landscape, we found divergent 'NU-like' (low-risk) and 'PUC-like' (high-risk) signatures in IUP. The latter were characterized by altered metabolism, biosynthesis, and cell-cell interaction functions, indicating oncologic significance. Further machine learning-based analysis revealed SERPINH1, PKP2, and PYGB as potential diagnostic biomarkers discriminating IUP from PUC. The immunohistochemical validation confirmed PYGB as a specific biomarker to distinguish between IUP and PUC with inverted growth. Conclusion In conclusion, we suggest PYGB as a promising immunohistochemical marker for IUP diagnosis in routine practice.
Collapse
Affiliation(s)
- Minsun Jung
- Department of Pathology, Seoul National University College of Medicine, Seoul, South Korea
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Cheol Lee
- Department of Pathology, Seoul National University Hospital, Seoul, South Korea
| | - Dohyun Han
- Transdisciplinary Department of Medicine and Advanced Technology, Seoul National University Hospital, Seoul, South Korea
- Proteomics Core Facility, Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea
| | - Kwangsoo Kim
- Transdisciplinary Department of Medicine and Advanced Technology, Seoul National University Hospital, Seoul, South Korea
| | - Sunah Yang
- Transdisciplinary Department of Medicine and Advanced Technology, Seoul National University Hospital, Seoul, South Korea
| | - Ilias P. Nikas
- School of Medicine, European University Cyprus, Nicosia, Cyprus
| | - Kyung Chul Moon
- Department of Pathology, Seoul National University College of Medicine, Seoul, South Korea
- Department of Pathology, Seoul National University Hospital, Seoul, South Korea
- Kidney Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Hyeyoon Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul, South Korea
- Proteomics Core Facility, Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea
| | - Min Ji Song
- Center for Medical Innovation, Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea
| | - Bohyun Kim
- Department of Pathology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, South Korea
| | - Hyebin Lee
- Department of Radiation Oncology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Han Suk Ryu
- Department of Pathology, Seoul National University College of Medicine, Seoul, South Korea
- Department of Pathology, Seoul National University Hospital, Seoul, South Korea
- Center for Medical Innovation, Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea
| |
Collapse
|
8
|
CKB inhibits epithelial-mesenchymal transition and prostate cancer progression by sequestering and inhibiting AKT activation. Neoplasia 2021; 23:1147-1165. [PMID: 34706306 PMCID: PMC8551525 DOI: 10.1016/j.neo.2021.09.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 09/17/2021] [Indexed: 12/26/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) contributes to tumor invasion, metastasis and drug resistance. AKT activation is key in a number of cellular processes. While many positive regulators for either EMT or AKT activation have been reported, few negative regulators are established. Through kinase cDNA screen, we identified brain-type creatine kinase (CKB or BCK) as a potent suppressor for both. As a ubiquitously expressed kinase in normal tissues, CKB is significantly downregulated in several solid cancer types. Lower CKB expression is significantly associated with worse prognosis. Phenotypically, CKB overexpression suppresses, while its silencing promotes, EMT and cell migration, xenograft tumor growth and metastasis of prostate cancer cells. AKT activation is one of the most prominent signaling events upon CKB silencing in prostate cancer cells, which is in line with prostate cancer TCGA data. EMT enhanced by CKB silencing is abolished by AKT inhibition. Mechanistically, CKB interacts with AKT and sequestrates it from activation by mTOR. We further elucidated that an 84aa fragment at C-terminus of CKB protein interacts with AKT's PH domain. Ectopic expression of the 84aa CKB fragment inhibits AKT activation, EMT and cell proliferation. Interestingly, molecular dynamics simulation on crystal structures of AKT and CKB independently demonstrates that AKT's PH domain and CKB's 84aa fragment establish their major interaction interface. In summary, we have discovered CKB as a negative regulator of EMT and AKT activation, revealing a new mode of their regulation . We have also demonstrated that CKB downregulation is a poor prognosticator, which is sufficient to promote prostate cancer progression.
Collapse
|
9
|
Liang Y, Ma B, Jiang P, Yang HM. Identification of Methylation-Regulated Differentially Expressed Genes and Related Pathways in Hepatocellular Carcinoma: A Study Based on TCGA Database and Bioinformatics Analysis. Front Oncol 2021; 11:636093. [PMID: 34150612 PMCID: PMC8209385 DOI: 10.3389/fonc.2021.636093] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 05/11/2021] [Indexed: 12/21/2022] Open
Abstract
Background In recent years, DNA methylation modification has been shown to be a critical mechanism in the field of epigenetics. Methods Hepatocellular carcinoma (HCC) data were obtained from The Cancer Genome Atlas project, including RNA expression profiles, Illumina Human Methylation 450K BeadChip data, clinical information, and pathological features. Then, differentially expressed genes (DEGs) and differentially methylated genes were identified using R software. Methylation-regulated DEGs (MeDEGs) were further analyzed using Spearman’s correlation analysis. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed using the DAVID database and ClueGO in Cytoscape software. Kaplan–Meier survival analysis explored the relationship between methylation, expression of MeDEGs, and survival time. Gene set enrichment analysis (GSEA) was conducted to predict the function of prognosis-related MeDEGs. Results A total of nine up-regulated and 72 down-regulated MeDEGs were identified. GO and KEGG pathway analyses results indicated that multiple cancer-related terms were enriched. Kaplan–Meier survival analysis showed that the methylation status of four MeDEGs (CTF1, FZD8, PDK4, and ZNF334) was negatively associated with overall survival. Moreover, the methylation status of CDF1 and PDK4 was identified as an independent prognostic factor. According to GSEA, hypermethylation of prognosis-related MeDEGs was enriched in pathways that included “Spliceosome”, “Cell cycle”, “RNA degradation”, “RNA polymerase”, “DNA replication”, “Mismatch repair”, “Base excision repair”, “Nucleotide excision repair”, “Homologous recombination”, “Protein export”, and “Pyrimidine metabolism”. Conclusions Aberrant DNA methylation plays a critical role in malignant progression of HCC. Prognosis-related MeDEGs identified in this research may be potential biomarkers and targets in diagnosis and treatment.
Collapse
Affiliation(s)
- Yu Liang
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Bin Ma
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Peng Jiang
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Hong-Mei Yang
- Department of Internal Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| |
Collapse
|
10
|
Mining the plasma-proteome associated genes in patients with gastro-esophageal cancers for biomarker discovery. Sci Rep 2021; 11:7590. [PMID: 33828156 PMCID: PMC8027878 DOI: 10.1038/s41598-021-87037-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 03/15/2021] [Indexed: 12/22/2022] Open
Abstract
Gastro-esophageal (GE) cancers are one of the major causes of cancer-related death in the world. There is a need for novel biomarkers in the management of GE cancers, to yield predictive response to the available therapies. Our study aims to identify leading genes that are differentially regulated in patients with these cancers. We explored the expression data for those genes whose protein products can be detected in the plasma using the Cancer Genome Atlas to identify leading genes that are differentially regulated in patients with GE cancers. Our work predicted several candidates as potential biomarkers for distinct stages of GE cancers, including previously identified CST1, INHBA, STMN1, whose expression correlated with cancer recurrence, or resistance to adjuvant therapies or surgery. To define the predictive accuracy of these genes as possible biomarkers, we constructed a co-expression network and performed complex network analysis to measure the importance of the genes in terms of a ratio of closeness centrality (RCC). Furthermore, to measure the significance of these differentially regulated genes, we constructed an SVM classifier using machine learning approach and verified these genes by using receiver operator characteristic (ROC) curve as an evaluation metric. The area under the curve measure was > 0.9 for both the overexpressed and downregulated genes suggesting the potential use and reliability of these candidates as biomarkers. In summary, we identified leading differentially expressed genes in GE cancers that can be detected in the plasma proteome. These genes have potential to become diagnostic and therapeutic biomarkers for early detection of cancer, recurrence following surgery and for development of targeted treatment.
Collapse
|
11
|
Southekal S, Mishra NK, Guda C. Pan-Cancer Analysis of Human Kinome Gene Expression and Promoter DNA Methylation Identifies Dark Kinase Biomarkers in Multiple Cancers. Cancers (Basel) 2021; 13:cancers13061189. [PMID: 33801837 PMCID: PMC8001681 DOI: 10.3390/cancers13061189] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/04/2021] [Accepted: 03/08/2021] [Indexed: 11/16/2022] Open
Abstract
Kinases are a group of intracellular signaling molecules that play critical roles in various biological processes. Even though kinases comprise one of the most well-known therapeutic targets, many have been understudied and therefore warrant further investigation. DNA methylation is one of the key epigenetic regulators that modulate gene expression. In this study, the human kinome's DNA methylation and gene expression patterns were analyzed using the level-3 TCGA data for 32 cancers. Unsupervised clustering based on kinome data revealed the grouping of cancers based on their organ level and tissue type. We further observed significant differences in overall kinase methylation levels (hyper- and hypomethylation) between the tumor and adjacent normal samples from the same tissue. Methylation expression quantitative trait loci (meQTL) analysis using kinase gene expression with the corresponding methylated probes revealed a highly significant and mostly negative association (~92%) within 1.5 kb from the transcription start site (TSS). Several understudied (dark) kinases (PKMYT1, PNCK, BRSK2, ERN2, STK31, STK32A, and MAPK4) were also identified with a significant role in patient survival. This study leverages results from multi-omics data to identify potential kinase markers of prognostic and diagnostic importance and further our understanding of kinases in cancer.
Collapse
Affiliation(s)
| | | | - Chittibabu Guda
- Correspondence: (N.K.M.); (C.G.); Tel.: +1-402-559-5954 (C.G.)
| |
Collapse
|
12
|
Heitor da Silva Maués J, Ferreira Ribeiro H, de Maria Maués Sacramento R, Maia de Sousa R, Pereira de Tommaso R, Dourado Kovacs Machado Costa B, Cardoso Soares P, Pimentel Assumpção P, de Fátima Aquino Moreira-Nunes C, Mário Rodriguez Burbano R. Downregulated genes by silencing MYC pathway identified with RNA-SEQ analysis as potential prognostic biomarkers in gastric adenocarcinoma. Aging (Albany NY) 2020; 12:24651-24670. [PMID: 33351778 PMCID: PMC7803532 DOI: 10.18632/aging.202260] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 10/31/2020] [Indexed: 12/24/2022]
Abstract
MYC overexpression is a common phenomenon in gastric carcinogenesis. In this study, we identified genes differentially expressed with a downregulated profile in gastric cancer (GC) cell lines with silenced MYC. The TTLL12, CDKN3, CDC16, PTPRA, MZT2B, UBE2T genes were validated using qRT-PCR, western blot and immunohistochemistry in tissues of 213 patients with diffuse and intestinal GC. We identified high levels of TTLL12, MZT2B, CDC16, UBE2T, associated with early and advanced stages, lymph nodes, distant metastases and risk factors such as H. pylori. Our results show that in the diffuse GC the overexpression of CDC16 and UBE2T indicate markers of poor prognosis higher than TTLL12. That is, patients with overexpression of these two genes live less than patients with overexpression of TTLL12. In the intestinal GC, patients who overexpressed CDC16 had a significantly lower survival rate than patients who overexpressed MZT2B and UBE2T, indicating in our data a worse prognostic value of CDC16 compared to the other two genes. PTPRA and CDKN3 proved to be important for assessing tumor progression in the early and advanced stages. In summary, in this study, we identified diagnostic and prognostic biomarkers of GC under the control of MYC, related to the cell cycle and the neoplastic process.
Collapse
Affiliation(s)
- Jersey Heitor da Silva Maués
- Laboratory of Human Cytogenetics, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil
- Laboratory of Molecular Biology, Ophir Loyola Hospital, Belém, Belém 66063-240, PA, Brazil
| | - Helem Ferreira Ribeiro
- Laboratory of Human Cytogenetics, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil
- Center of Biological and Health Sciences, Department of Biomedicine, University of Amazon, Belém 66060-000, PA, Brazil
| | | | - Rafael Maia de Sousa
- Laboratory of Molecular Biology, Ophir Loyola Hospital, Belém, Belém 66063-240, PA, Brazil
| | | | | | - Paulo Cardoso Soares
- Laboratory of Molecular Biology, Ophir Loyola Hospital, Belém, Belém 66063-240, PA, Brazil
| | - Paulo Pimentel Assumpção
- Oncology Research Nucleus, University Hospital João de Barros Barreto, Federal University of Pará, Belém 66073-000, PA, Brazil
| | | | - Rommel Mário Rodriguez Burbano
- Laboratory of Human Cytogenetics, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil
- Laboratory of Molecular Biology, Ophir Loyola Hospital, Belém, Belém 66063-240, PA, Brazil
| |
Collapse
|
13
|
Activity-Based Protein Profiling Reveals Potential Dasatinib Targets in Gastric Cancer. Int J Mol Sci 2020; 21:ijms21239276. [PMID: 33291786 PMCID: PMC7729964 DOI: 10.3390/ijms21239276] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/30/2020] [Accepted: 12/02/2020] [Indexed: 12/28/2022] Open
Abstract
Dasatinib is a multi-target kinase inhibitor, whose targets include BCR-ABL, SRC family kinases, and various cancer kinases. The elevated SRC activity in gastric cancer (GC) has prompted the need for the therapeutic application of dasatinib in GC. We observed that the efficacy of dasatinib varied with the GC cell lines. The differential effect of dasatinib was not correlated with the basal SRC activity of each cell line. Moreover, the GC cell lines showing the strong antitumor effects of dasatinib were refractory to other SRC inhibitors, i.e., bosutinib and saracatinib, suggesting that unexpected dasatinib’s targets could exist. To profile the targets of dasatinib in GC, we performed activity-based protein profiling (ABPP) via mass spectrometry using a desthiobiotin-ATP probe. We identified 29 and 18 kinases as potential targets in dasatinib-sensitive (SNU-216, MKN-1) and -resistant (SNU-484, SNU-601) cell lines, respectively. The protein–protein interaction mapping of the differential drug targets in dasatinib-sensitive and -resistant GC using the STRING database suggested that dasatinib could target cellular energy homeostasis in the drug-sensitive GC. RNAi screening for identified targets indicated p90RSK could be a novel dasatinib target, which is important for maintaining the viability and motility of GC cells. Further functional validation of dasatinib off-target actions will provide more effective therapeutic options for GC.
Collapse
|
14
|
Wang L, Li C, Tian J, Liu J, Zhao Y, Yi Y, Zhang Y, Han J, Pan C, Liu S, Deng N, Xian Z, Li G, Zhang X, Liang A. Genome-wide transcriptional analysis of Aristolochia manshuriensis induced gastric carcinoma. PHARMACEUTICAL BIOLOGY 2020; 58:98-106. [PMID: 31957525 PMCID: PMC7006638 DOI: 10.1080/13880209.2019.1710219] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Context: Aristolochia manshuriensis Kom (Aristolochiaceae) (AMK) is known for toxicity and mutagenicity.Objective: The tumorigenic role of AMK has yet to be understood.Materials and methods: AMK extracts were extracted from root crude drug. SD (Sprague Dawley) rats underwent gavage with AMK (0.92 g/kg) every other day for 10 (AMK-10) or 20 (AMK-20) weeks. Stomach samples were gathered for histopathological evaluation, microarray and mRNA analysis.Results: The gastric weight to body weight ratio (GW/BW) is 1.7 in the AMK-10 cohort, and 1.8 in AMK-20 cohort compared to control (CTL) cohort. Liver function was damaged in AMK-10 and AMK-20 rats compared to CTL rats. There were no significant changes of CRE (creatinine) in AMK-10 and AMK-20 rats. Histopathological analysis revealed that rats developed dysplasia in the forestomach in AMK-10 rats, and became gastric carcinoma in AMK-20 rats. Genes including Mapk13, Nme1, Gsta4, Gstm1, Jun, Mgst2, Ggt6, Gpx2, Gpx8, Calml3, Rasgrp2, Cd44, Gsr, Dgkb, Rras, and Amt were found to be critical in AMK-10 and AMK-20 rats. Pik3cb, Plcb3, Tp53, Hras, Myc, Src, Akt1, Gnai3, and Fgfr3 worked in AMK-10 rats, and PDE2a and PDE3a played a pivotal role in AMK-20 rats.Discussion and conclusions: AMK induced benign or malignant gastric tumours depends on the period of AMK administration. Genes including Mapk13, Nme1, Gsta4, Gstm1, Jun, Mgst2, Ggt6, Gpx2, Gpx8, Calml3, Rasgrp2, Cd44, Gsr, Dgkb, Rras, Amt, Pik3cb, Plcb3, Tp53, Hras, Myc, Src, Akt1, Gnai3, Fgfr3, PDE2a, and PDE3a were found to be critical in aristolochic acid-induced gastric tumour process.
Collapse
Affiliation(s)
- Lianmei Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Chunying Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Jingzhuo Tian
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Jing Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Yong Zhao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Yan Yi
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Yushi Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Jiayin Han
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Chen Pan
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Suyan Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Nuo Deng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Zhong Xian
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Guiqin Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Xin Zhang
- Blood Products Engineering Research and Development Center, Shenzhen, China
| | - Aihua Liang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
- CONTACT Aihua Liang Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| |
Collapse
|
15
|
Identification of metabolism-associated genes and construction of a prognostic signature in bladder cancer. Cancer Cell Int 2020; 20:538. [PMID: 33292266 PMCID: PMC7643334 DOI: 10.1186/s12935-020-01627-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 10/26/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Bladder cancer (BC) is a commonly diagnosed malignant tumor in the urinary system, with a high morbidity and a high recurrence rate. Current studies indicated that metabolism-associated genes (MAGs) having critical roles in the etiology of BC. The present study aims to identify differentially expressed MAGs and construct a MAGs based prognostic risk signature for BC by using The Cancer Genome Atlas (TCGA) database and proteomics data. METHODS RNA-sequence data from the TCGA database and proteomics data from our BC samples were used to identify differentially expressed MAGs and construct a MAGs based prognostic signature in BC. Subsequently, survival analysis and nomogram were used to evaluate the prognostic and predictive value of the MAGs based signature in BC. RNA isolation and reverse transcription‑quantitative PCR (RT-qPCR) were further performed to investigate the expression levels of MAGs in BC cells and explore the relationship between MAGs and M2 tumor associated macrophages (TAMs) secreted transforming growth factor-β1 (TGF-β1) in BC cells. RESULTS A total of 23 differentially expressed MAGs were identified and five MAGs were finally used to construct a MAGs based signature. Survival analysis revealed that the MAGs based signature was closely correlated with the survival outcomes of patients with BC. A nomogram with the MAGs based signature risk score and clinical features was also constructed to facilitate the individualized prediction of BC patients. RT-qPCR showed that five MAGs were significantly differentially expressed and the expression levels of three MAGs were positively correlated with M2 TAMs secreted TGF-β1 in T24 cells. CONCLUSIONS Our study identified novel prognostic MAGs and constructed a MAGs based signature, which can be used as an independent factor in evaluating the prognosis of patients with BC. Furthermore, M2 TAMs may promote the expression of MAGs via the TGF-β1 signaling pathway in the microenvironment of BC. Further clinical trials and experimental explorations are needed to validate our observations in BC.
Collapse
|
16
|
Exploration in the Mechanism of Kaempferol for the Treatment of Gastric Cancer Based on Network Pharmacology. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5891016. [PMID: 33145355 PMCID: PMC7596434 DOI: 10.1155/2020/5891016] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/07/2020] [Accepted: 10/09/2020] [Indexed: 12/11/2022]
Abstract
Background Kaempferol is a natural polyphenol in lots of Chinese herbs, which has shown promising treatment for gastric cancer (GC). However, the molecular mechanisms of its action have not been systematically revealed yet. In this work, a network pharmacology approach was used to elucidate the potential mechanisms of kaempferol in the treatment of GC. Methods The kaempferol was input into the PharmMapper and SwissTargetPrediction database to get its targets, and the targets of GC were obtained by retrieving the Online Mendelian Inheritance in Man (OMIM) database, MalaCards database, Therapeutic Target Database (TTD), and Coolgen database. The molecular docking was performed to assess the interactions between kaempferol and these targets. Next, the overlap targets of kaempferol and GC were identified for GO and KEGG enrichment analyses. Afterward, a protein-protein interaction (PPI) network was constructed to get the hub targets, and the expression and overall survival analysis of the hub target were investigated. Finally, the overall survival (OS) analysis of hub targets was performed using the Kaplan-Meier Plotter online tool. Results A total of 990 genes related to GC and 10 overlapping genes were determined through matching the 24 potential targets of kaempferol with disease-associated genes. The result of molecular docking indicated that kaempferol can bind with these hub targets with good binding scores. These targets were further mapped to 140 GO biological process terms and 11 remarkable pathways. In the PPI network analysis, 3 key targets were identified, including ESR1, EGFR, and SRC. The mRNA and protein expression levels of EGFR and SRC were obviously higher in GC tissues. High expression of these targets was related to poor OS in GC patients. Conclusions This study provided a novel approach to reveal the therapeutic mechanisms of kaempferol on GC, which will ease the future clinical application of kaempferol in the treatment of GC.
Collapse
|
17
|
Systematic Elucidation of the Mechanism of Quercetin against Gastric Cancer via Network Pharmacology Approach. BIOMED RESEARCH INTERNATIONAL 2020; 2020:3860213. [PMID: 32964029 PMCID: PMC7486643 DOI: 10.1155/2020/3860213] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 06/25/2020] [Indexed: 12/24/2022]
Abstract
This study was aimed at elucidating the potential mechanisms of quercetin in the treatment of gastric cancer (GC). A network pharmacology approach was used to analyze the targets and pathways of quercetin in treating GC. The predicted targets of quercetin against GC were obtained through database mining, and the correlation of these targets with GC was analyzed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. Next, the protein-protein interaction (PPI) network was constructed, and overall survival (OS) analysis of hub targets was performed using the Kaplan–Meier Plotter online tool. Finally, the mechanism was further analyzed via molecular docking of quercetin with the hub targets. Thirty-six quercetin-related genes were identified, 15 of which overlapped with GC-related targets. These targets were further mapped to 319 GO biological process terms and 10 remarkable pathways. In the PPI network analysis, six hub targets were identified, including AKT1, EGFR, SRC, IGF1R, PTK2, and KDR. The high expression of these targets was related to poor OS in GC patients. Molecular docking analysis confirmed that quercetin can bind to these hub targets. In conclusion, this study provided a novel approach to reveal the therapeutic mechanisms of quercetin on GC, which will ease the future clinical application of quercetin in the treatment of GC.
Collapse
|
18
|
Src Family Kinases as Therapeutic Targets in Advanced Solid Tumors: What We Have Learned so Far. Cancers (Basel) 2020; 12:cancers12061448. [PMID: 32498343 PMCID: PMC7352436 DOI: 10.3390/cancers12061448] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 05/29/2020] [Accepted: 05/31/2020] [Indexed: 12/17/2022] Open
Abstract
Src is the prototypal member of Src Family tyrosine Kinases (SFKs), a large non-receptor kinase class that controls multiple signaling pathways in animal cells. SFKs activation is necessary for the mitogenic signal from many growth factors, but also for the acquisition of migratory and invasive phenotype. Indeed, oncogenic activation of SFKs has been demonstrated to play an important role in solid cancers; promoting tumor growth and formation of distant metastases. Several drugs targeting SFKs have been developed and tested in preclinical models and many of them have successfully reached clinical use in hematologic cancers. Although in solid tumors SFKs inhibitors have consistently confirmed their ability in blocking cancer cell progression in several experimental models; their utilization in clinical trials has unveiled unexpected complications against an effective utilization in patients. In this review, we summarize basic molecular mechanisms involving SFKs in cancer spreading and metastasization; and discuss preclinical and clinical data highlighting the main challenges for their future application as therapeutic targets in solid cancer progression
Collapse
|
19
|
Su R, Zhang J. Oncogenic role of LYN in human gastric cancer via the Wnt/β-catenin and AKT/mTOR pathways. Exp Ther Med 2020; 20:646-654. [PMID: 32509024 DOI: 10.3892/etm.2020.8672] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 01/30/2020] [Indexed: 01/16/2023] Open
Abstract
LYN kinase (LYN) is a member of the Src tyrosine kinase family, which plays an important role in multiple tumor-related functions. The current study demonstrated that LYN functions as a pro-oncogene in AGS gastric cancer cells. It was found that LYN expression levels were significantly raised in gastric cancer tissue and were significantly associated with the pathological grades of patients with gastric cancer. This was accomplished by knocking down LYN in AGS cells using short hairpin RNA (shRNA) plasmid transfection, with reverse transcription-quantitative PCR detection verifying the effectiveness of RNA interference. It was found that the cell proliferation and colony formation abilities of AGS cells were significantly inhibited, using CCK-8 and clone formation assays, respectively. Furthermore, LYN knockdown was found to induce apoptosis and inhibit both migration and invasion in AGS cells, using flow cytometry and Transwell assays, respectively. A mechanical investigation further suggested that LYN knockdown resulted in the activation of the mitochondrial apoptotic pathway. Likewise, the Wnt/β-catenin pathway was inactivated by LYN knockdown, including decreased levels of Wnt3a, β-catenin, snail family transcriptional repressor (Snail)1 and Snail2. Epithelial-mesenchymal transition mesenchymal markers (including N-cadherin and vimentin) were also found to be downregulated, and E-cadherin was upregulated in LYN-silenced AGS cells. Finally, the AKT/mTOR pathway was found to be downregulated by LYN knockdown in AGS cells, including decreased levels of phosphorylated (p)-AKT (Ser473), p-mTOR (Ser2448), and the down-stream effector p70S6 kinase (p70S6K). Furthermore, the AKT pathway activator, insulin like growth factor-1 (IGF-1), was found to reverse the inhibitory effects of LYN knockdown on the proliferation, migration and invasion of AGS cells. In conclusion, the current study demonstrated that LYN plays an oncogenic role in the proliferation, survival and movement of human gastric cancer cells by activating the mitochondrial apoptotic pathway, and downregulating the Wnt/β-catenin and AKT/mTOR pathways. The current research provides a comprehensive insight into the regulation of LYN in gastric cancer and may help with the development of new tumor treatment strategies.
Collapse
Affiliation(s)
- Rui Su
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China.,Department of Gastrointestinal Surgery, Affiliated Hospital of Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Jun Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| |
Collapse
|
20
|
Meng L, Chen Z, Jiang Z, Huang T, Hu J, Luo P, Zhang H, Huang M, Huang L, Chen Y, Lu M, Xu AM, Ying S. MiR-122-5p suppresses the proliferation, migration, and invasion of gastric cancer cells by targeting LYN. Acta Biochim Biophys Sin (Shanghai) 2020; 52:49-57. [PMID: 31828293 DOI: 10.1093/abbs/gmz141] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 11/05/2019] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer (GC) is one of malignant tumors with high mortality and morbidity in the world. MicroRNA-122 (miR-122) acts as a tumor suppressor in a variety of cancers and has been found to be dominant in gastric adenocarcinoma. However, the specific biological function of miR-122-5p in GC is not completely clear. In this study, we found that miR-122-5p was low-expressed in GC tissues and cell lines by using qRT-PCR. Overexpression of miR-122-5p inhibited the proliferation, migration, and invasion of GC cells by using CCK-8 and transwell assays. On the contrary, downregulation of miR-122-5p promoted the proliferation, migration, and invasion of GC cells. In addition, we found that the expression of LYN, an Src family tyrosine kinase, was inversely correlated with miR-122-5p expression in GC tissues by using western blot analysis, immunohistochemistry, and qRT-PCR assays. Meanwhile, luciferase assay results indicated that LYN is a direct target of miR-122-5p in GC cells. Moreover, silencing LYN expression by its siRNA inhibited the proliferation, migration, and invasion of GC cells. Importantly, overexpression of LYN restored miR-122-5p-mediated inhibition of the proliferation, migration, and invasion of GC cells. Taken together, our results indicated miR-122-5p inhibited the proliferation, migration, and invasion by targeting LYN in GC.
Collapse
Affiliation(s)
- Lei Meng
- Department of General Surgery, Fourth Affiliated Hospital of Anhui Medical University, Hefei 230012, China
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Zhangming Chen
- Department of General Surgery, Fourth Affiliated Hospital of Anhui Medical University, Hefei 230012, China
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Zhe Jiang
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Ting Huang
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Jie Hu
- Department of General Surgery, Fourth Affiliated Hospital of Anhui Medical University, Hefei 230012, China
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Panquan Luo
- Department of General Surgery, Fourth Affiliated Hospital of Anhui Medical University, Hefei 230012, China
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Hongli Zhang
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Mengqi Huang
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Lei Huang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Yu Chen
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Ming Lu
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - A-man Xu
- Department of General Surgery, Fourth Affiliated Hospital of Anhui Medical University, Hefei 230012, China
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Songcheng Ying
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
21
|
Chichirau BE, Diechler S, Posselt G, Wessler S. Tyrosine Kinases in Helicobacter pylori Infections and Gastric Cancer. Toxins (Basel) 2019; 11:toxins11100591. [PMID: 31614680 PMCID: PMC6832112 DOI: 10.3390/toxins11100591] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/02/2019] [Accepted: 10/09/2019] [Indexed: 12/11/2022] Open
Abstract
Helicobacter pylori (H. pylori) has been identified as a leading cause of gastric cancer, which is one of the most frequent and malignant types of tumor. It is characterized by its rapid progression, distant metastases, and resistance to conventional chemotherapy. A number of receptor tyrosine kinases and non-receptor tyrosine kinases have been implicated in H. pylori-mediated pathogenesis and tumorigenesis. In this review, recent findings of deregulated EGFR, c-Met, JAK, FAK, Src, and c-Abl and their functions in H. pylori pathogenesis are summarized.
Collapse
Affiliation(s)
- Bianca E Chichirau
- Department of Biosciences, Paris-Lodron University of Salzburg, 5020 Salzburg, Austria.
| | - Sebastian Diechler
- Department of Biosciences, Paris-Lodron University of Salzburg, 5020 Salzburg, Austria.
| | - Gernot Posselt
- Department of Biosciences, Paris-Lodron University of Salzburg, 5020 Salzburg, Austria.
| | - Silja Wessler
- Cancer Cluster Salzburg, Department of Biosciences, Paris-Lodron University of Salzburg, 5020 Salzburg, Austria.
| |
Collapse
|
22
|
Identification of DNA methylation-regulated differentially-expressed genes and related pathways using Illumina 450K BeadChip and bioinformatic analysis in gastric cancer. Pathol Res Pract 2019; 215:152570. [PMID: 31378454 DOI: 10.1016/j.prp.2019.152570] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 07/16/2019] [Accepted: 07/26/2019] [Indexed: 01/11/2023]
Abstract
In the current research, we aimed to identify and analyze methylation-regulated differentially-expressed genes (MeDEGs) and related pathways using bioinformatic methods. We downloaded RNA-seq, Illumina Human Methylation 450 K BeadChip and clinical information of gastric cancer (GC) from The Cancer Genome Atlas (TCGA) project. Differentially-expressed genes (DEGs) were identified using the edgeR package. Then, we performed Spearman's correlation analysis between DEG expression levels and methylation levels. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed in the DAVID database. We then conducted Kaplan-Meier survival analysis to explore the relationship between methylation, expression and prognosis. The protein-protein interaction networks were further analyzed using the STRING database. A total of 204 down-regulated DEGs and 164 up-regulated DEGs were identified as MeDEGs. GO and KEGG pathway analyses showed that MeDEGs were enriched in multiple cancer-related terms. Kaplan-Meier survival analysis showed that eight up-regulated MeDEGs (CAMKV, COMP, FGF3, FGF19, FOXL2, IGF2BP1, IGFBP1 and NPPB) and five down-regulated MeDEGs (ALDH3B2, CALML3, FLRT1, G6PC and HRASLS2) were associated with prognosis of GC patients. In addition, PPI networks and KEGG pathway analyses further confirmed the critical role of prognosis-related MeDEGs. In conclusion, methylation plays a critical role in GC progression. Multiple MeDEGs are related to prognosis, suggesting that they may be potential targets in tumor treatment.
Collapse
|
23
|
Zhang Q, Meng X, Qin G, Xue X, Dang N. Lyn Kinase Promotes the Proliferation of Malignant Melanoma Cells through Inhibition of Apoptosis and Autophagy via the PI3K/Akt Signaling Pathway. J Cancer 2019; 10:1197-1208. [PMID: 30854129 PMCID: PMC6400685 DOI: 10.7150/jca.28908] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 01/05/2019] [Indexed: 01/02/2023] Open
Abstract
Melanoma is a malignant tumor of cutaneous melanocytes that is characterized by high grade malignancy, rapid progression and high mortality. Thus far, its specific etiological mechanism has been unclear. In this study, we discovered that Lyn kinase expression was up-regulated in melanoma tissues and cells. The function of Lyn was determined by knocking down its expression with a lentivirus containing Lyn shRNA and upregulating its expression with pcDNA3.1-Lyn in the melanoma cell lines M14 and A375. The results showed that Lyn knockdown could significantly inhibit the proliferation, migration and invasiveness through its inhibition of apoptosis and autophagy via the PI3K/Akt pathway in melanoma cell lines. This was further confirmed by treatment with PI3K inhibitor BEZ235. Up-regulation of Lyn promoted the expression of p-Akt and Cyclin D1. Additionally, we investigated the effects of Lyn inhibitor Bafetinib on melanoma cells and the results were consistent with Lyn knockdown. Collectively, our results indicated that Lyn plays a carcinogenic role in multiple cellular functions during melanoma development through regulating apoptosis and autophagy via the PI3K/Akt pathway and may be a valuable potential target for the clinical treatment of melanoma.
Collapse
Affiliation(s)
- Qianqian Zhang
- Department of Dermatology, Jinan Central Hospital affiliated to Shandong University, Jinan, Shandong Province, China
- Taishan Medical University, Taian, Shandong Province, China
- Department of Dermatology, No. 960 Hospital of The Chinese People's Liberation Army, Taian, Shandong Province, China
| | - Xianguang Meng
- Department of Dermatology, Jinan Central Hospital affiliated to Shandong University, Jinan, Shandong Province, China
| | - Guojing Qin
- Department of Dermatology, Jinan Central Hospital affiliated to Shandong University, Jinan, Shandong Province, China
| | - Xiaotong Xue
- Department of Dermatology, Jinan Central Hospital affiliated to Shandong University, Jinan, Shandong Province, China
- School of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Ningning Dang
- Department of Dermatology, Jinan Central Hospital affiliated to Shandong University, Jinan, Shandong Province, China
| |
Collapse
|
24
|
Xu Y, He Q, Lu Y, Tao F, Zhao L, Ou R. MicroRNA-218-5p inhibits cell growth and metastasis in cervical cancer via LYN/NF-κB signaling pathway. Cancer Cell Int 2018; 18:198. [PMID: 30524205 PMCID: PMC6278036 DOI: 10.1186/s12935-018-0673-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 10/31/2018] [Indexed: 01/09/2023] Open
Abstract
Background We are committed to investigate miR-218-5 effects on the progression of cervical cancer (CC) cell and find out the molecular mechanism. Methods GSE9750 was obtained from GEO database and R Limma package was applied to filter out dysregulated genes. The pathways were enriched by GSEA software, ClusterProfiler and enrichplot packages to predict the function of DEGs. The binding sites of LYN were detected by miRanda and TargetScan. The miR2Disease database was used to find miRNAs related with CC. The expression of miR-218-5p and LYN were quantified by qRT-PCR and that of LYN protein was measured by western blot. The targeted relationships between miR-218-5p and LYN were verified by dual-luciferase reporter assay. Colony formation assays, wound healing, transwell invasion assay and flow cytometer analysis were performed to investigate the roles that miR-218-5p and LYN played in migration, invasion and death of cervical carcinoma. Xenografts established in nude mice were used to assess tumor growth in vivo. Results The highly expressed mRNA LYN was selected by microarray analysis in GSE9750. NF-κB signaling pathway was enriched base on GSEA results. The expression of miR-218-5p was lower but LYN was higher in CC primary tumors compared with normal control. In addition, miR-218-5p could regulate the expression of LYN in HeLa cells negatively. Overexpression of LYN could promote cell migration and invasion, but inhibit cell death in vitro, and also promote tumor formation in vivo via activating NF-κB signaling pathway which could be reversed by miR-218-5p. Conclusions MiR-218-5p suppressed the progression of CC via LYN/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yunsheng Xu
- 1Laboratory for Advanced Interdisciplinary Research, Institutes of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang China.,2Department of Dermatovenereology, The Seventh Affiliated Hospital, Sun Yat-sen University, No. 628 Zhenyuan Road, Guangming District, Shenzhen, 518107 Guangdong China
| | - Qin He
- 3Department of Dermatovenereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang China
| | - Yiyi Lu
- 3Department of Dermatovenereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang China
| | - Fengxing Tao
- 3Department of Dermatovenereology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang China
| | - Liang Zhao
- 1Laboratory for Advanced Interdisciplinary Research, Institutes of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang China
| | - Rongying Ou
- 4Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang Street, Ouhai District, Wenzhou, 325000 Zhejiang China
| |
Collapse
|
25
|
Gastric Cancer Cell Lines Have Different MYC-Regulated Expression Patterns but Share a Common Core of Altered Genes. Can J Gastroenterol Hepatol 2018; 2018:5804376. [PMID: 30410872 PMCID: PMC6206580 DOI: 10.1155/2018/5804376] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 09/12/2018] [Accepted: 09/23/2018] [Indexed: 12/15/2022] Open
Abstract
MYC is an oncogene responsible for excessive cell growth in cancer, enabling transcriptional activation of genes involved in cell cycle regulation, metabolism, and apoptosis, and is usually overexpressed in gastric cancer (GC). By using siRNA and Next-Generation Sequencing (NGS), we identified MYC-regulated differentially expressed Genes (DEGs) in three Brazilian gastric cancer cell lines representing the histological subtypes of GC (diffuse, intestinal, and metastasis). The DEGs were picked using Sailfish software, followed by Gene Set Enrichment Analysis (GSEA) and Kyoto Encyclopedia of Gene and Genome (KEGG) pathway analysis using KEGG. We found 11 significantly enriched gene sets by using enrichment score (ES), False Discovery Rate (FDR), and nominal P-values. We identified a total of 5.471 DEGs with correlation over (80%). In diffuse-type and in metastatic GC cell lines, MYC-silencing caused DEGs downregulation, while the intestinal-type GC cells presented overall DEGs upregulation after MYC siRNA depletion. We were able to detect 11 significant gene sets when comparing our samples to the hallmark collection of gene expression, enriched mostly for the following hallmarks: proliferation, pathway, signaling, metabolic, and DNA damage response. When we analyzed our DEGs considering KEGG metabolic pathways, we found 12 common branches covering a wide range of biological functions, and three of them were common to all three cell lines: ubiquitin-mediated proteolysis, ribosomes, and system and epithelial cell signaling in Helicobacter pylori infection. The GC cell lines used in this study share 14 MYC-regulated genes, but their gene expression profile is different for each histological subtype of GC. Our results present a computational analysis of MYC-related signatures in GC, and we present evidence that GC cell lines representing distinct histological subtypes of this disease have different MYC-regulated expression profiles but share a common core of altered genes. This is an important step towards the understanding of MYC's role in gastric carcinogenesis and an indication of probable new drug targets in stomach cancer.
Collapse
|
26
|
Dai X, Wang LJ, Wu J, Shi YX, Li GP, Yang XQ. Src kinase inhibitor PP2 regulates the biological characteristics of A549 cells via the PI3K/Akt signaling pathway. Oncol Lett 2018; 16:5059-5065. [PMID: 30250573 PMCID: PMC6144924 DOI: 10.3892/ol.2018.9282] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 04/16/2018] [Indexed: 02/07/2023] Open
Abstract
Lung cancer is one of the most prevalent types of cancer worldwide, with a poor prognosis for patients and a concomitant financial burden on society. There are a number of different pathological subtypes, with non-small cell lung cancer (NSCLC) being the primary subtype. Although anticancer therapy has led to a marked improvement in the survival rate of patients in recent years, the survival rate remains poor. Potential reasons for this include a lack of early diagnosis and drug resistance, which is considered to be associated with mutations in components of signaling pathways, tumor suppressors and epidermal growth factor receptor, and certain other complex mechanisms to a certain extent. It is therefore imperative to develop novel therapies. In the present study, the pyrazolopyrimidine compound PP2 was used to inhibit Src family protein tyrosine kinases in A549 cells. It was demonstrated that PP2 was able to suppress cell viability, migration and invasion, and promote apoptosis via regulating the phosphoinositide 3-kinase/protein kinase B/B-cell lymphoma 2/caspase-3 signaling pathway. PP2 may therefore be useful in anti-NSCLC therapy in the future.
Collapse
Affiliation(s)
- Xi Dai
- State Key laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR 999056, P.R. China.,Respiratory Medicine Department, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Li-Jiao Wang
- Respiratory Medicine Department, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Juan Wu
- Respiratory Medicine Department, The Second Chinese Medicine Hospital of Sichuan Province, Chengdu, Sichuan 610031, P.R. China
| | - Ya-Xu Shi
- Respiratory Medicine Department, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Guo-Ping Li
- Department of Respiratory Medicine, The Third People's Hospital of Chengdu/Affiliated Hospital of Southwest Jiaotong University, Chengdu, Sichuan 610000, P.R. China
| | - Xiao-Qiong Yang
- Respiratory Medicine Department, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
27
|
Liu S, Hao X, Ouyang X, Dong X, Yang Y, Yu T, Hu J, Hu L. Tyrosine kinase LYN is an oncotarget in human cervical cancer: A quantitative proteomic based study. Oncotarget 2018; 7:75468-75481. [PMID: 27690342 PMCID: PMC5342753 DOI: 10.18632/oncotarget.12258] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 09/02/2016] [Indexed: 01/05/2023] Open
Abstract
Cervical cancer is one of the most common malignant tumor in women. The mechanisms of cervical cancer are intricate and have not been fully understood. Therefore, we employed iTRAQ to obtain novel proteins profile which participates in the tumor oncogenesis of cervical cancer. 3300 proteins were identified aberrantly expressed in cervical cancer, and western bolt was performed to validate the results of iTRAQ. Then, we selected LYN for further study. Immunohistochemistry identified that LYN expression was significantly increased in cervical cancer tissues than that in cancer adjacent normal cervical tissues and normal cervical tissues. The increased LYN expression was significantly correlated with cancer differentiation and FIGO stage. Silencing LYN inhibited cell proliferation, migration and invasion, conversely, overexpression LYN promoted cell proliferation, migration and invasion. In terms of mechanism, LYN could also promote cervical cancer cells metastasis through activating IL-6/STAT3 pathway. In vivo study, overexpression LYN promoted tumor growth, meanwhile knockdown LYN inhibited tumor growth. These results indicate that LYN tyrosine kinase is an oncogenic gene and can serve as a novel target for cervical cancer research and therapy.
Collapse
Affiliation(s)
- Shuaibin Liu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Xiaoming Hao
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Xiaolan Ouyang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Xiaojing Dong
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Yixuan Yang
- Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Tinghe Yu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Jianguo Hu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Lina Hu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| |
Collapse
|
28
|
Liao Z, Wang X, Liang H, Yu A, ur Rehman U, Fan Q, Hu Y, Wang C, Zhou Z, Wang T. miR-1 suppresses the proliferation and promotes the apoptosis of esophageal carcinoma cells by targeting Src. Cancer Med 2017; 6:2957-2965. [PMID: 29034995 PMCID: PMC5727306 DOI: 10.1002/cam4.1214] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 08/29/2017] [Accepted: 08/31/2017] [Indexed: 11/23/2022] Open
Abstract
Nonreceptor tyrosine kinase c-Src, also known as Src, is a potent oncogene involved in a series of biological processes including cell growth, differentiation, and apoptosis; however, its expression pattern and function in esophageal cancer is poorly addressed. In this study, abnormal overexpression of Src protein was observed in esophageal cancer tissues, which fuelled the speculation that microRNA-mediated posttranscriptional regulatory mechanism might be involved. Bioinformatic analyses were applied to identify miRNAs that could potentially target Src. miR-1 was predicted and further validated as a direct repressor of Src. Moreover, we manipulated knockdown and overexpression experiment on TE-1 and TE-10 cells to demonstrate miR-1 suppressed proliferation and promoted apoptosis in esophageal cancer cells by inhibiting Src. Taken together, this study underlines a negative regulatory mechanism in which miR-1 serves as a suppressor of Src in esophageal cancer cells and may provide insights into novel therapeutic approaches for esophageal cancer.
Collapse
Affiliation(s)
- Zhicong Liao
- Department of Thoracic and Cardiovascular SurgeryNanjing Drum Tower Hospitalthe Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsu210008China
| | - Xiaojun Wang
- Nanjing Medical University Affiliated Cancer HospitalNanjingJiangsu210009China
| | - Hongwei Liang
- State Key Laboratory of Pharmaceutical BiotechnologyNanjing Advanced Institute of Life SciencesJiangsu Engineering Research Center for MicroRNA Biology and BiotechnologyNanjingJiangsu210093China
| | - Ao Yu
- Department of Thoracic and Cardiovascular SurgeryNanjing Drum Tower Hospitalthe Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsu210008China
| | - Uzair ur Rehman
- State Key Laboratory of Pharmaceutical BiotechnologyNanjing Advanced Institute of Life SciencesJiangsu Engineering Research Center for MicroRNA Biology and BiotechnologyNanjingJiangsu210093China
| | - Qian Fan
- Department of LymphomaTianjin Medical University Cancer Institute and HospitalNational Clinical Research Center of CancerKey Laboratory of Cancer Prevention and TherapyTianjin300060China
| | - Yue Hu
- Nanjing Multicenter BiobankBiobank of Nanjing Drum Tower Hospitalthe Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsu210008China
| | - Chen Wang
- State Key Laboratory of Pharmaceutical BiotechnologyNanjing Advanced Institute of Life SciencesJiangsu Engineering Research Center for MicroRNA Biology and BiotechnologyNanjingJiangsu210093China
| | - Zhen Zhou
- State Key Laboratory of Pharmaceutical BiotechnologyNanjing Advanced Institute of Life SciencesJiangsu Engineering Research Center for MicroRNA Biology and BiotechnologyNanjingJiangsu210093China
| | - Tao Wang
- Department of Thoracic and Cardiovascular SurgeryNanjing Drum Tower Hospitalthe Affiliated Hospital of Nanjing University Medical SchoolNanjingJiangsu210008China
| |
Collapse
|
29
|
Zhou ZR, Huang P, Song GH, Zhang Z, An K, Lu HW, Ju XL, Ding W. Comparative proteomic analysis of rats subjected to water immersion and restraint stress as an insight into gastric ulcers. Mol Med Rep 2017; 16:5425-5433. [PMID: 28849061 PMCID: PMC5647087 DOI: 10.3892/mmr.2017.7241] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 06/15/2017] [Indexed: 12/13/2022] Open
Abstract
In the present study, comparative proteomic analysis was performed in rats subjected to water immersion-restraint stress (WRS). A total of 26 proteins were differentially expressed and identified using matrix-assisted laser desorption/ionization time of flight mass spectrometry. Among the 26 differentially expressed protein spots identified, 13 proteins were significantly upregulated under WRS, including pyruvate kinase and calreticulin, which may be closely associated with energy metabolism. In addition, 12 proteins were downregulated under WRS, including hemoglobin subunit β-2 and keratin type II cytoskeletal 8, which may be important in protein metabolism and cell death. Gene Ontology analysis revealed the cellular distribution, molecular function and biological processes of the identified proteins. The mRNA levels of certain differentially expressed proteins were analyzed using fluorescence quantitative polymerase chain reaction analysis. The results of the present study aimed to offer insights into proteins, which are differentially expressed in gastric ulcers in stress, and provide theoretical evidence of a radical cure for gastric ulcers in humans.
Collapse
Affiliation(s)
- Zheng-Rong Zhou
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Pan Huang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Guang-Hao Song
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Zhuang Zhang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Ke An
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Han-Wen Lu
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Xiao-Li Ju
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Wei Ding
- Department of Animal Husbandry and Veterinary Medicine, Jiangsu Polytechnic College of Agriculture and Forestry, Jurong, Jiangsu 212499, P.R. China
| |
Collapse
|