1
|
Lin S, Que Y, Que C, Li F, Deng M, Xu D. Exosome miR-3184-5p inhibits gastric cancer growth by targeting XBP1 to regulate the AKT, STAT3, and IRE1 signalling pathways. Asia Pac J Clin Oncol 2022; 19:e27-e38. [PMID: 35394683 DOI: 10.1111/ajco.13663] [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: 07/12/2021] [Revised: 07/12/2021] [Accepted: 08/09/2021] [Indexed: 11/30/2022]
Abstract
MicroRNAs can regulate the transcription of protein-coding genes associated with the development and progression of cancer. In this study, we explored the potential diagnostic function of exosome miR-3184-5p in gastric cancer. This exosome was isolated from the blood samples of 150 patients with gastric cancer and 60 healthy participants. The mean particle size and concentration of serum exosome in the patients with gastric cancer were 104.6 nm (93.97-115.84) and 6.21e+009 particles/ml (5.15e+009-7.12e+009), respectively. miR-3184-5p expression was substantially downregulated in the patients with gastric cancer compared with that in the healthy participants. The gastric cancer cell line HGC-27 was cultured and transfected with the mimic and an inhibitor to overexpress and inhibit miR-3184-5p expression. miR-3184-5p strongly suppressed cell proliferation, migration, and invasion but induced cell apoptosis. Luciferase reporter assay revealed that XBP1 was the target of miR-3184-5p. miR-3184-5p substantially downregulated the expression of CD44, cyclin D1, MMP2, p65, p-AKT, and p-STAT3 but upregulated that of GRP78, IRE1, p-JNK, and CHOP. Moreover, miR-3184-5p cleaved caspase-12 and inhibited BCL-2 expression. These results suggested that the downregulation of miR-3184-5p in patients with gastric cancer might regulate the AKT, STAT3, and IRE1 pathways to promote the vitality of gastric cancer cells.
Collapse
Affiliation(s)
- Shuangming Lin
- Department of Gastrointestinal and Anal Surgery, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, P.R. China
| | - Yonggu Que
- Department of Gastrointestinal and Anal Surgery, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, P.R. China
| | - Changrong Que
- Department of Gastrointestinal and Anal Surgery, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, P.R. China
| | - Fudi Li
- Department of Gastrointestinal and Anal Surgery, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, P.R. China
| | - Maoqing Deng
- Department of laboratory, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, P.R. China
| | - Dongbo Xu
- Department of Gastrointestinal and Anal Surgery, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, P.R. China
| |
Collapse
|
2
|
Zhu L, Wang H, Jiang C, Li W, Zhai S, Cai X, Wang X, Liao L, Tao F, Jin D, Chen G, Xia Y, Mao JH, Li B, Wang P, Hang B. Clinically applicable 53-Gene prognostic assay predicts chemotherapy benefit in gastric cancer: A multicenter study. EBioMedicine 2020; 61:103023. [PMID: 33069062 PMCID: PMC7569189 DOI: 10.1016/j.ebiom.2020.103023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/20/2020] [Accepted: 09/09/2020] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND We previously established a 53-gene prognostic signature for overall survival (OS) of gastric cancer patients. This retrospective multi-center study aimed to develop a clinically applicable gene expression detection assay and to investigate the prognostic value of this signature. METHODS A TCGA gastric adenocarcinoma cohort (TCGA-STAD) was used for comparing 53-gene signature with other gene signatures. A high-throughput mRNA hybridization gene expression assay was developed to quantify the expression of 53-genes in formalin-fixed paraffin-embedded tissues of 540 patients enrolled from three hospitals. 180 patents were randomly selected from two hospitals to build a prognostic prediction model based on the 53-gene signature using leave-p-out (one-third out) cross-validation method together with Cox regression and Kaplan-Meier analysis, and the model was assessed on three validation cohorts. FINDINGS In the evaluation phase, studies based on TCGA-STAD showed that the 53-gene signature was significantly superior to other three prognostic signatures and was independent of TCGA molecular subtypes and clinical factors. For clinical validation and utility, the prognostic scores were generated using the newly developed assay, which was reliable and sensitive, in 100 sampling training sets and were significantly associated with OS in 100 sampling validation sets. The scores were significantly associated with OS in three independent and combined validation cohorts, and in patients with stages II and III/IV. The multivariate Cox regression demonstrated that the prognostic power of the score was independent of clinical factors, consistent with those findings in the TCGA dataset. Finally, patients with good prognostic scores exhibited significantly a better 5-year OS rate from adjuvant FOLFOX chemotherapy after surgery than from other chemotherapies. INTERPRETATION The 53-gene prognostic score system is clinically applicable for predicting the OS of patients independent of clinical factors in gastric cancers, which could also be a promising predictive biomarker for FOLFOX regimen. FUNDING Chinese National Science and Technology, National Natural Science Foundation and Natural Science Foundation of Jiangsu Province.
Collapse
Affiliation(s)
- Linghua Zhu
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Haifeng Wang
- Department of Gastrointestinal Surgery, Shaoxing People's Hospital (Shaoxing Hospital of Zhejiang University), Shaoxing, Zhejiang, China
| | - Chengfei Jiang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Wenhuan Li
- Department of Gastrointestinal Surgery, The First People's Hospital of Wenling, Wenling, Zhejiang, China
| | - Shuting Zhai
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiujun Cai
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xianfa Wang
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Linghong Liao
- Fujian Key Laboratory of TCM Health State, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Feng Tao
- Department of Gastrointestinal Surgery, Shaoxing People's Hospital (Shaoxing Hospital of Zhejiang University), Shaoxing, Zhejiang, China
| | - Dexi Jin
- Department of Gastrointestinal Surgery, The First People's Hospital of Wenling, Wenling, Zhejiang, China
| | - Guofu Chen
- Department of Gastrointestinal Surgery, The First People's Hospital of Wenling, Wenling, Zhejiang, China
| | - Yankai Xia
- School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jian-Hua Mao
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, United States
| | - Bin Li
- Nanjing KDRB Biotech Inc., Ltd, Jiangning District, Nanjing, Jiangsu, China.
| | - Pin Wang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.
| | - Bo Hang
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, United States.
| |
Collapse
|
3
|
Dai W, Li Q, Liu BY, Li YX, Li YY. Differential networking meta-analysis of gastric cancer across Asian and American racial groups. BMC SYSTEMS BIOLOGY 2018; 12:51. [PMID: 29745833 PMCID: PMC5998874 DOI: 10.1186/s12918-018-0564-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Gastric Carcinoma is one of the most lethal cancer around the world, and is also the most common cancers in Eastern Asia. A lot of differentially expressed genes have been detected as being associated with Gastric Carcinoma (GC) progression, however, little is known about the underlying dysfunctional regulation mechanisms. To address this problem, we previously developed a differential networking approach that is characterized by involving differential coexpression analysis (DCEA), stage-specific gene regulatory network (GRN) modelling and differential regulation networking (DRN) analysis. Result In order to implement differential networking meta-analysis, we developed a novel framework which integrated the following steps. Considering the complexity and diversity of gastric carcinogenesis, we first collected three datasets (GSE54129, GSE24375 and TCGA-STAD) for Chinese, Korean and American, and aimed to investigate the common dysregulation mechanisms of gastric carcinogenesis across racial groups. Then, we constructed conditional GRNs for gastric cancer corresponding to normal and carcinoma, and prioritized differentially regulated genes (DRGs) and gene links (DRLs) from three datasets separately by using our previously developed differential networking method. Based on our integrated differential regulation information from three datasets and prior knowledge (e.g., transcription factor (TF)-target regulatory relationships and known signaling pathways), we eventually generated testable hypotheses on the regulation mechanisms of two genes, XBP1 and GIF, out of 16 common cross-racial DRGs in gastric carcinogenesis. Conclusion The current cross-racial integrative study from the viewpoint of differential regulation networking provided useful clues for understanding the common dysfunctional regulation mechanisms of gastric cancer progression and discovering new universal drug targets or biomarkers for gastric cancer. Electronic supplementary material The online version of this article (10.1186/s12918-018-0564-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wentao Dai
- Shanghai Center for Bioinformation Technology, 1278 Keyuan Road, Shanghai, 201203, People's Republic of China.,Shanghai Engineering Research Center of Pharmaceutical Translation & Shanghai Industrial Technology Institute, 1278 Keyuan Road, Shanghai, 201203, People's Republic of China
| | - Quanxue Li
- Shanghai Center for Bioinformation Technology, 1278 Keyuan Road, Shanghai, 201203, People's Republic of China.,School of biotechnology, East China University of Science and Technology, Shanghai, 200237, China
| | - Bing-Ya Liu
- Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Yi-Xue Li
- Shanghai Center for Bioinformation Technology, 1278 Keyuan Road, Shanghai, 201203, People's Republic of China. .,School of biotechnology, East China University of Science and Technology, Shanghai, 200237, China. .,Shanghai Engineering Research Center of Pharmaceutical Translation & Shanghai Industrial Technology Institute, 1278 Keyuan Road, Shanghai, 201203, People's Republic of China. .,Key Lab of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Yuan-Yuan Li
- Shanghai Center for Bioinformation Technology, 1278 Keyuan Road, Shanghai, 201203, People's Republic of China. .,School of biotechnology, East China University of Science and Technology, Shanghai, 200237, China. .,Shanghai Engineering Research Center of Pharmaceutical Translation & Shanghai Industrial Technology Institute, 1278 Keyuan Road, Shanghai, 201203, People's Republic of China. .,Key Lab of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
4
|
Gastric Cancer Associated Genes Identified by an Integrative Analysis of Gene Expression Data. BIOMED RESEARCH INTERNATIONAL 2017; 2017:7259097. [PMID: 28232943 PMCID: PMC5292384 DOI: 10.1155/2017/7259097] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 12/29/2016] [Accepted: 01/04/2017] [Indexed: 01/13/2023]
Abstract
Gastric cancer is one of the most severe complex diseases with high morbidity and mortality in the world. The molecular mechanisms and risk factors for this disease are still not clear since the cancer heterogeneity caused by different genetic and environmental factors. With more and more expression data accumulated nowadays, we can perform integrative analysis for these data to understand the complexity of gastric cancer and to identify consensus players for the heterogeneous cancer. In the present work, we screened the published gene expression data and analyzed them with integrative tool, combined with pathway and gene ontology enrichment investigation. We identified several consensus differentially expressed genes and these genes were further confirmed with literature mining; at last, two genes, that is, immunoglobulin J chain and C-X-C motif chemokine ligand 17, were screened as novel gastric cancer associated genes. Experimental validation is proposed to further confirm this finding.
Collapse
|
5
|
Shi L, Resaul J, Owen S, Ye L, Jiang WG. Clinical and Therapeutic Implications of Follistatin in Solid Tumours. Cancer Genomics Proteomics 2017; 13:425-435. [PMID: 27807065 DOI: 10.21873/cgp.20005] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 10/05/2016] [Indexed: 12/20/2022] Open
Abstract
Follistatin (FST), as a single-chain glycosylated protein, has two major isoforms, FST288 and FST315. The FST315 isoform is the predominant form whilst the FST288 variant accounts for less than 5% of the encoded mRNA. FST is differentially expressed in human tissues and aberrant expression has been observed in a variety of solid tumours, including gonadal, gastric and lung cancer, hepatocellular carcinoma, basal cell carcinoma and melanoma. Based on the current evidence, FST is an antagonist of transforming growth factor beta family members, such as activin and bone morphogenetic proteins (BMPs). FST plays a role in tumourigenesis, metastasis and angiogenesis of solid tumours through its interaction with activin and BMPs, thus resulting in pathophysiological function. In terms of diagnosis, prognosis and therapy, FST has shown strong promise. Through a better understanding of its biological functions, potential clinical applications may yet emerge.
Collapse
Affiliation(s)
- Lei Shi
- Urology Department, Yantai Yu Huang Ding Hospital, Yantai, Shandong Province, P.R. China.,Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, U.K
| | - Jeyna Resaul
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, U.K
| | - Sioned Owen
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, U.K
| | - Lin Ye
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, U.K
| | - Wen G Jiang
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, U.K.
| |
Collapse
|
6
|
Hüneburg R, Marwitz T, van Heteren P, Weismüller TJ, Trebicka J, Adam R, Aretz S, Perez Bouza A, Pantelis D, Kalff JC, Nattermann J, Strassburg CP. Chromoendoscopy in combination with random biopsies does not improve detection of gastric cancer foci in CDH1 mutation positive patients. Endosc Int Open 2016; 4:E1305-E1310. [PMID: 27995193 PMCID: PMC5161122 DOI: 10.1055/s-0042-112582] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 06/13/2016] [Indexed: 02/06/2023] Open
Abstract
Background and study aims: Hereditary diffuse gastric cancer (HGGC), an autosomal dominant tumor-syndrome, accounts for 1 % to 3 % of gastric cancers worldwide. Presumably 30 % to 40 % of all patients fulfilling the clinical guidelines for HDGC are carriers of a pathogenic mutation in the CDH1 gene. Patients often show multiple foci of signet ring cell carcinoma at early age and are advised to undergo prophylactic total gastrectomy (PTG). Our aim was to improve the endoscopic detection of HDGC by using an enhanced endoscopic protocol. Patient and methods: Patients with a proven CDH1 germline mutation identified in our institute were prospectively included. Patients were advised to undergo PTG and offered a baseline endoscopic examination prior surgery. Examination was performed by using high-resolution white-light endoscopy and pan-gastric chromoendoscopy with indigo carmine as dye combined with targeted and multiple random biopsies assessed by an expert histopathologist. Postoperative histopathology was compared with results from endoscopic biopsies. Results: Between September 2012 and November 2014 8 patients with a proven CDH1 germline mutation were included. We conducted 44 targeted (6.3/patient) and 225 random (32.1/patient) biopsies in 7 patients. We detected 1 gastric cancer by random biopsy (14 %). All other examinations showed no signs of cancer. Histopathology of gastrectomy specimen revealed multiple foci of gastric carcinoma in 6 patients (86 %) with a total number of 27 cancer foci. Conclusions: Examination with targeted and random biopsies combined with chromoendoscopy is not able to detect small foci of gastric cancer in CDH1 mutation carriers. Therefore PTG is advocated in these patients.
Collapse
Affiliation(s)
- Robert Hüneburg
- Department of Internal Medicine I, University
Hospital Bonn, Bonn, Germany,Center for Hereditary Tumor Syndromes,
University Hospital Bonn, Bonn, Germany,Corresponding author Robert Hüneburg,
MD Department of Internal Medicine I
University of
BonnSigmund-Freud Straße
25D-53115 Bonn,
Germany+49-228-287-16043+49-228-287-19763
| | - Tim Marwitz
- Department of Internal Medicine I, University
Hospital Bonn, Bonn, Germany
| | - Peer van Heteren
- Department of Internal Medicine I, University
Hospital Bonn, Bonn, Germany,Center for Hereditary Tumor Syndromes,
University Hospital Bonn, Bonn, Germany
| | - Tobias J. Weismüller
- Department of Internal Medicine I, University
Hospital Bonn, Bonn, Germany,Center for Hereditary Tumor Syndromes,
University Hospital Bonn, Bonn, Germany
| | - Jonel Trebicka
- Department of Internal Medicine I, University
Hospital Bonn, Bonn, Germany
| | - Ronja Adam
- Institute of Human Genetics, University
Hospital Bonn, Bonn, Germany,Center for Hereditary Tumor Syndromes,
University Hospital Bonn, Bonn, Germany
| | - Stefan Aretz
- Institute of Human Genetics, University
Hospital Bonn, Bonn, Germany,Center for Hereditary Tumor Syndromes,
University Hospital Bonn, Bonn, Germany
| | - Alberto Perez Bouza
- Institute of Pathology, University Hospital
Bonn, Bonn, Germany,Center for Hereditary Tumor Syndromes,
University Hospital Bonn, Bonn, Germany
| | - Dimitrios Pantelis
- Department of Surgery, University Hospital
Bonn, Bonn, Germany,Center for Hereditary Tumor Syndromes,
University Hospital Bonn, Bonn, Germany
| | - Jörg C. Kalff
- Department of Surgery, University Hospital
Bonn, Bonn, Germany,Center for Hereditary Tumor Syndromes,
University Hospital Bonn, Bonn, Germany
| | - Jacob Nattermann
- Department of Internal Medicine I, University
Hospital Bonn, Bonn, Germany,Center for Hereditary Tumor Syndromes,
University Hospital Bonn, Bonn, Germany
| | - Cristian P. Strassburg
- Department of Internal Medicine I, University
Hospital Bonn, Bonn, Germany,Center for Hereditary Tumor Syndromes,
University Hospital Bonn, Bonn, Germany
| |
Collapse
|
7
|
Epithelial Regeneration After Gastric Ulceration Causes Prolonged Cell-Type Alterations. Cell Mol Gastroenterol Hepatol 2016; 2:625-647. [PMID: 27766298 PMCID: PMC5042868 DOI: 10.1016/j.jcmgh.2016.05.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 05/06/2016] [Indexed: 12/21/2022]
Abstract
BACKGROUND & AIMS The peptic ulcer heals through a complex process, although the ulcer relapse often occurs several years later after healing. Our hypothesis is that even after visual evidence of healing of gastric ulceration, the regenerated epithelium is aberrant for an extended interval, increasing susceptibility of the regenerated epithelium to damage and further diseases. METHODS Gastric ulcers were induced in mice by serosal topical application of acetic acid. RESULTS Gastric ulcers induced by acetic acid visually healed within 30 days. However, regenerated epithelial architecture was poor. The gene profile of regenerated tissue was abnormal, indicating increased stem/progenitor cells, deficient differentiated gastric cell types, and deranged cell homeostasis. Despite up-regulation of PDX1 in the regenerated epithelium, no mature antral cell type was observed. Four months after healing, the regenerated epithelium lacks parietal cells, trefoil factor 2 (TFF2) and (sex-determining region Y)-box 9 (SOX9) remain up-regulated deep in the gastric gland, and the Na/H exchanger 2 (a TFF2 effector in gastric healing) remains down-regulated. Gastric ulcer healing was strongly delayed in TFF2 knockout mice, and re-epithelialization was accompanied with mucous metaplasia. After Helicobacter pylori inoculum 30 days after ulceration, we observed that the gastric ulcer selectively relapses at the same site where it originally was induced. Follow-up evaluation at 8 months showed that the relapsed ulcer was not healed in H pylori-infected tissues. CONCLUSIONS These findings show that this macroscopically regenerated epithelium has prolonged abnormal cell distribution and is differentially susceptible to subsequent damage by H pylori.
Collapse
Key Words
- CXCR4, C-X-C chemokine receptor type 4
- DCLK1, doublecortin-like kinase 1
- GAPDH, glyceraldehyde-3-phosphate dehydrogenase
- GIF, gastric intrinsic factor
- GSII, Griffonia simplicifolia lectin II
- Gastric Ulcer Healing
- H pylori
- HK-ATPase, hydrogen potassium exchanger adenosine triphosphatase
- KO, knockout
- Lgr5, Leucine-rich repeat-containing G protein-coupled receptor5
- MUC, Mucin
- Metaplasia
- NHE2
- NHE2, sodium hydrogen exchanger 2
- PCR, polymerase chain reaction
- PDX1, pancreatic and duodenal homeobox 1
- SOX2, (sex-determining region Y)-box 2
- SOX9
- SPEM, spasmolytic polypeptide-expressing metaplasia
- TFF2
- TFF2, trefoil factor 2
- UEA-1, ulex europaeus
- WT, wild type
- cDNA, complementary DNA
- mRNA, messenger RNA
Collapse
|
8
|
Lin X, Zhao Y, Song WM, Zhang B. Molecular classification and prediction in gastric cancer. Comput Struct Biotechnol J 2015; 13:448-58. [PMID: 26380657 PMCID: PMC4556804 DOI: 10.1016/j.csbj.2015.08.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 07/23/2015] [Accepted: 08/01/2015] [Indexed: 12/19/2022] Open
Abstract
Gastric cancer, a highly heterogeneous disease, is the second leading cause of cancer death and the fourth most common cancer globally, with East Asia accounting for more than half of cases annually. Alongside TNM staging, gastric cancer clinic has two well-recognized classification systems, the Lauren classification that subdivides gastric adenocarcinoma into intestinal and diffuse types and the alternative World Health Organization system that divides gastric cancer into papillary, tubular, mucinous (colloid), and poorly cohesive carcinomas. Both classification systems enable a better understanding of the histogenesis and the biology of gastric cancer yet have a limited clinical utility in guiding patient therapy due to the molecular heterogeneity of gastric cancer. Unprecedented whole-genome-scale data have been catalyzing and advancing the molecular subtyping approach. Here we cataloged and compared those published gene expression profiling signatures in gastric cancer. We summarized recent integrated genomic characterization of gastric cancer based on additional data of somatic mutation, chromosomal instability, EBV virus infection, and DNA methylation. We identified the consensus patterns across these signatures and identified the underlying molecular pathways and biological functions. The identification of molecular subtyping of gastric adenocarcinoma and the development of integrated genomics approaches for clinical applications such as prediction of clinical intervening emerge as an essential phase toward personalized medicine in treating gastric cancer.
Collapse
Affiliation(s)
- Xiandong Lin
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, NY 10029, USA
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fujian Provincial Cancer Hospital, No. 420 Fuma Road, Jinan District, Fuzhou, Fujian 350014, PR China
| | - Yongzhong Zhao
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, NY 10029, USA
| | - Won-min Song
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, NY 10029, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, NY 10029, USA
| |
Collapse
|
9
|
Xiao P, Ling H, Lan G, Liu J, Hu H, Yang R. Trefoil factors: Gastrointestinal-specific proteins associated with gastric cancer. Clin Chim Acta 2015; 450:127-34. [PMID: 26265233 DOI: 10.1016/j.cca.2015.08.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Revised: 08/07/2015] [Accepted: 08/07/2015] [Indexed: 12/11/2022]
Abstract
Trefoil factor family (TFF), composed of TFF1, TFF2, and TFF3, is a cluster of secreted peptides characterized by trefoil domain (s) and C-terminal dimerization domain. TFF1, a gastric tumor suppressor, is a single trefoil peptide originally detected in breast cancer cell lines but expressed mainly in the stomach; TFF2, a candidate of gastric cancer suppressor with two trefoil domains, is abundant in the stomach and duodenal Brunner's glands; and TFF3 is another single trefoil peptide expressed throughout the intestine which can promote the development of gastric carcinoma. According to multiple studies, TFFs play a regulatory function in the mammals' digestive system, namely in mucosal protection and epithelial cell reconstruction, tumor suppression or promotion, signal transduction and the regulation of proliferation and apoptosis. Action mechanisms of TFFs remain unresolved, but the recent demonstration of a GKN (gastrokine) 2-TFF1 heterodimer implicates structural and functional interplay with gastrokines. This review aims to encapsulate the structural and biological characteristics of TFF.
Collapse
Affiliation(s)
- Ping Xiao
- Key Laboratory of Tumor Cellular and Molecular Pathology, University of South China, College of Hunan Province, Cancer Research Institute, Hengyang, Hunan 421001, PR China; Center for Gastric Cancer Research of Hunan Province, University of South China, Hengyang, Hunan 421001, PR China
| | - Hui Ling
- Key Laboratory of Tumor Cellular and Molecular Pathology, University of South China, College of Hunan Province, Cancer Research Institute, Hengyang, Hunan 421001, PR China; Center for Gastric Cancer Research of Hunan Province, University of South China, Hengyang, Hunan 421001, PR China.
| | - Gang Lan
- Key Laboratory for Atherosclerology of Hunan Province, Cardiovascular Research Institute, University of South China, Hengyang, Hunan 421001, PR China
| | - Jiao Liu
- Key Laboratory of Tumor Cellular and Molecular Pathology, University of South China, College of Hunan Province, Cancer Research Institute, Hengyang, Hunan 421001, PR China; Center for Gastric Cancer Research of Hunan Province, University of South China, Hengyang, Hunan 421001, PR China
| | - Haobin Hu
- Key Laboratory of Tumor Cellular and Molecular Pathology, University of South China, College of Hunan Province, Cancer Research Institute, Hengyang, Hunan 421001, PR China; Center for Gastric Cancer Research of Hunan Province, University of South China, Hengyang, Hunan 421001, PR China
| | - Ruirui Yang
- Key Laboratory of Tumor Cellular and Molecular Pathology, University of South China, College of Hunan Province, Cancer Research Institute, Hengyang, Hunan 421001, PR China; Center for Gastric Cancer Research of Hunan Province, University of South China, Hengyang, Hunan 421001, PR China
| |
Collapse
|
10
|
Jin EH, Lee SI, Kim J, Seo EY, Lee SY, Hur GM, Shin S, Hong JH. Association between Promoter Polymorphisms of TFF1, TFF2, and TFF3 and the Risk of Gastric and Diffuse Gastric Cancers in a Korean Population. J Korean Med Sci 2015; 30:1035-41. [PMID: 26240479 PMCID: PMC4520932 DOI: 10.3346/jkms.2015.30.8.1035] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 04/17/2015] [Indexed: 01/26/2023] Open
Abstract
Gastric cancer is one of the most common cancers in the world. The aims of this study were to evaluate the association between polymorphisms in TFF gene family, TFF1, TFF2, and TFF3 and the risk of gastric cancer (GC) and GC subgroups in a Korean population via a case-control study. The eight polymorphisms in TFF gene family were identified by sequencing and genotyped with 377 GC patients and 396 controls by using TaqMan genotyping assay. The rs184432 TT genotype of TFF1 was significantly associated with a reduced risk of GC (odds ratio, [OR) = 0.45; 95% confidence interval, [CI] = 0.25-0.82; P = 0.009), more protective against diffuse-type GC (OR = 0.20; 95% CI = 0.05-0.89; P = 0.035) than GC (OR = 0.34; 95% CI = 0.14-0.82; P = 0.017) in subjects aged < 60 yr, and correlated with lymph node metastasis negative GC and diffuse-type GC (OR = 0.44; 95% CI = 0.23-0.86; P = 0.016 and OR = 0.20; 95% CI = 0.05-0.87; P = 0.031, respectively). In addition, a decreased risk of lymph node metastasis negative GC and diffuse-type GC was observed for rs225359 TT genotype of TFF1 (OR = 0.46, 95% CI = 0.24-0.88; P = 0.020 and OR = 0.21, 95% CI = 0.05-0.88; P = 0.033, respectively). These findings suggest that the rs184432 and rs225359 polymorphisms in TFF1 have protective effects for GC and contribute to the development of GC in Korean individuals.
Collapse
Affiliation(s)
- Eun-Heui Jin
- Clinical Trials Center, Chungnam National University Hospital, Daejeon, Korea
| | - Sang-Il Lee
- Department of Surgery, Chungnam National University Hospital, Daejeon, Korea
| | - JaeWoo Kim
- Clinical Trials Center, Chungnam National University Hospital, Daejeon, Korea
| | - Eun Young Seo
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Korea
| | - Su Yel Lee
- National Biobank of Korea, Chungnam National University Hospital, Daejeon, Korea
| | - Gang Min Hur
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, Korea
| | - Sanghee Shin
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, Korea
| | - Jang Hee Hong
- Clinical Trials Center, Chungnam National University Hospital, Daejeon, Korea
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, Korea
| |
Collapse
|
11
|
Blank S, Deck C, Dreikhausen L, Weichert W, Giese N, Falk C, Schmidt T, Ott K. Angiogenic and growth factors in gastric cancer. J Surg Res 2014; 194:420-429. [PMID: 25577146 DOI: 10.1016/j.jss.2014.11.028] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 10/10/2014] [Accepted: 11/19/2014] [Indexed: 12/31/2022]
Abstract
BACKGROUND Antiangiogenic treatment is at the horizon in the palliative treatment of gastric cancer (GC), but data on proangiogenic biomarkers are still limited. The aim of this study was to analyze five proteins with a function in tumor angiogenesis: vascular endothelial growth factor (VEGF), angiopoietin-2 (Ang-2), follistatin, leptin, and platelet endothelial cell adhesion molecule 1 (CD31) in peripheral blood and corresponding tumor tissue. MATERIAL AND METHODS From 2008-2010, tumor tissue (n = 76) and corresponding preoperative serum (n = 69) of patients with localized GC were collected; 45 had perioperative chemotherapy. Protein serum or tumor lysate levels of these factors were measured by an angiogenesis multiplex immunoassay and correlated with response and survival. RESULTS Serum Ang-2 had prognostic relevance in the whole study population (P = 0.027). In subgroup analysis, serum VEGF and Ang-2 had prognostic relevance in primarily resected patients (P = 0.028; P = 0.048) but no association was found in neoadjuvantly treated patients. Follistatin concentration in the tumor tissue was associated with prognosis in all patients (P = 0.019). Tumor VEGF concentrations were correlated with histopathologic response (P = 0.011), with patients showing >50% remaining tumor having higher VEGF concentrations. The tissue Ang-2/VEGF ratio was significantly correlated with both clinical and histopathologic response (P = 0.029, P = 0.009). Additionally, the level of leptin in the tissue was associated with clinical response: nonresponding patients had higher leptin levels than those of responding patients (P = 0.032). CONCLUSIONS Our results show the importance of angiogenetic factors in serum and tumor tissue in GC for prognosis and treatment response. Further trials in larger patient populations are warranted for a further evaluation of proangiogenetic factors as biomarkers in gastrointestinal cancer.
Collapse
Affiliation(s)
- Susanne Blank
- Department of Surgery, University Hospital of Heidelberg, Heidelberg, Germany.
| | - Catrin Deck
- Department of Surgery, University Hospital of Heidelberg, Heidelberg, Germany
| | - Lena Dreikhausen
- Department of Surgery, University Hospital of Heidelberg, Heidelberg, Germany
| | - Wilko Weichert
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Natalia Giese
- Department of Surgery, University Hospital of Heidelberg, Heidelberg, Germany
| | - Christine Falk
- Institute of Transplant Immunology, IFB-Tx, Hannover Medical School, Hannover, Germany
| | - Thomas Schmidt
- Department of Surgery, University Hospital of Heidelberg, Heidelberg, Germany
| | - Katja Ott
- Department of Surgery, University Hospital of Heidelberg, Heidelberg, Germany
| |
Collapse
|
12
|
Jiang P, Yu G, Zhang Y, Xiang Y, Zhu Z, Feng W, Lee W, Zhang Y. Promoter hypermethylation and downregulation of trefoil factor 2 in human gastric cancer. Oncol Lett 2014; 7:1525-1531. [PMID: 24765170 PMCID: PMC3997660 DOI: 10.3892/ol.2014.1904] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 11/21/2013] [Indexed: 01/22/2023] Open
Abstract
Trefoil factor 2 (TFF2) plays a protective role in gastric mucosa and may be involved in the progression of gastric cancer, but the detailed functions and underlying molecular mechanisms are not clear. The present study used a combination of clinical observations and molecular methods to investigate the correlation between abnormal expression of TFF2 and gastric cancer progression. TFF2 expression was evaluated by reverse transcription polymerase chain reaction (RT-PCR), quantitative PCR (qPCR), and western blot and immunohistochemistry analyses. TFF2 methylation levels were analyzed by genomic bisulfite sequencing method. The results showed that TFF2 mRNA and protein expression were decreased in gastric cancer tissues compared with the matched non-cancerous mucosa, and the decreased level was associated with the differentiation and invasion of gastric cancer. Moreover, the average TFF2 methylation level of CpG sites in the promoter region was 70.4% in three gastric cancer tissues, while the level in associated non-neoplastic tissues was 41.0%. Furthermore, the promoter hypermethylation of TFF2 was also found in gastric cancer cell lines, AGS and N87, and gene expression was significantly increased following treatment with a demethylating agent, 5-Aza-2′-deoxycytidine. In conclusion, TFF2 expression was markedly decreased in gastric cancer and promoter hypermethylation was found to regulate the downregulation of TFF2. TFF2 has been suggested as a tumor suppressor in gastric carcinogenesis and metastasis.
Collapse
Affiliation(s)
- Ping Jiang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, P.R. China ; Department of Pathology and Pathophysiology, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Guoyu Yu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, P.R. China ; Department of Biochemistry, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Yong Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, P.R. China
| | - Yang Xiang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, P.R. China
| | - Zhu Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Weiyang Feng
- Department of Biochemistry, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Wenhui Lee
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, P.R. China
| | - Yun Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, P.R. China
| |
Collapse
|
13
|
Bornschein J, Rokkas T, Selgrad M, Malfertheiner P. Gastric cancer: clinical aspects, epidemiology and molecular background. Helicobacter 2011; 16 Suppl 1:45-52. [PMID: 21896085 DOI: 10.1111/j.1523-5378.2011.00880.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The validity and usefulness of the 7th edition of the UICC tumor node metastasis classification in the context of clinical management of gastric cancer are discussed. The most relevant new agent in gastric cancer therapy is trastuzumab for HER2-positive gastric carcinomas. This marks the success of continuous effort of translational research. Trastuzumab, initially applied in palliative settings, is currently being evaluated also in neoadjuvant treatment regimens. Several new meta-analyses support the carcinogenic effect of high salt intake and smoking in the context of Helicobacter pylori infection. Further data have become available on the efficacy of protective agents, acetyl salicylic acid/nonsteroidal anti-inflammatory drugs, and antioxidants. In search for a successful prevention strategy, the focus is on the identification of individuals at high risk who demand screening (testing) and surveillance. Serological assessment of gastric mucosal abnormalities with increased risk for gastric cancer development is extensively studied, and new data are presented from Asia as well as from Europe. New high-throughput techniques combined with bioinformatic vector analysis open the gate to the identification of new potential diagnostic and therapeutic targets. Furthermore, these approaches allow us to elucidate the interplay of bacterial virulence factors and the host's immune response as well as H. pylori-associated alterations of mucosal gene expression.
Collapse
Affiliation(s)
- Jan Bornschein
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke-University of Magdeburg, 39120 Magdeburg, Germany
| | | | | | | |
Collapse
|