Molecular and Cellular Mechanisms of Cancers: Gastric Cancer

A special issue of Cells (ISSN 2073-4409). This special issue belongs to the section "Cellular Pathology".

Deadline for manuscript submissions: closed (30 June 2020) | Viewed by 40733

Special Issue Editors


E-Mail Website
Guest Editor
Department of Pathology, Faculty of Medicine of the University of Porto, 4200-319 Porto, Portugal
Interests: Helicobacter pylori; gastric carcinoma

E-Mail Website
Guest Editor
i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto; Ipatimup – Institute of Molecular Pathology and Immunology of the University of Porto; and Faculty of Medicine of the University of Porto, Porto, Portugal
Interests: gastric cancer; molecular genetics of cancer; cancer immunology

Special Issue Information

Dear Colleagues,

Gastric cancer remains highly common and deadly cancer worldwide. Continuous research efforts have revealed that gastric cancer has a multifactorial etiology and represents a morphologically, biologically, and genetically heterogeneous disease.

This Special Issue aims to address the role of etiological factors, such as Helicobacter pylori infection, the microbiome, and other environmental factors in gastric cancer, emphasizing their molecular mechanisms of action. It also aims to explore the distinct molecular profiles associated with gastric carcinogenesis, including that associated with Epstein–Barr virus infection, and their potential as targets for therapeutic interventions and in the identification of biomarkers for screening, predicting treatment response, and monitoring disease progression. Finally, this Special Issue also intends to cover the latest findings related to hereditary gastric cancer.

It is our hope that the papers included in this Special Issue will be helpful in providing an overview of the field and in advancing new knowledge in basic and clinical aspects of gastric cancer.

Prof. Céu Figueiredo
Prof. Jose Carlos Machado
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Cells is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • gastric cancer
  • Helicobacter pylori
  • microbiome
  • molecular mechanisms of carcinogenesis
  • molecular profiles
  • Epstein–Barr virus
  • hereditary gastric cancer

Published Papers (8 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

21 pages, 4874 KiB  
Article
The Dysfunctional Immune System in Common Variable Immunodeficiency Increases the Susceptibility to Gastric Cancer
by Irene Gullo, Catarina Costa, Susana L. Silva, Cristina Ferreira, Adriana Motta, Sara P. Silva, Rúben Duarte Ferreira, Pedro Rosmaninho, Emília Faria, José Torres da Costa, Rita Câmara, Gilza Gonçalves, João Santos-Antunes, Carla Oliveira, José C. Machado, Fátima Carneiro and Ana E. Sousa
Cells 2020, 9(6), 1498; https://doi.org/10.3390/cells9061498 - 19 Jun 2020
Cited by 9 | Viewed by 3999
Abstract
Gastric carcinoma (GC) represents the most common cause of death in patients with common variable immunodeficiency (CVID). However, a limited number of cases have been characterised so far. In this study, we analysed the clinical features, bacterial/viral infections, detailed morphology and immune microenvironment [...] Read more.
Gastric carcinoma (GC) represents the most common cause of death in patients with common variable immunodeficiency (CVID). However, a limited number of cases have been characterised so far. In this study, we analysed the clinical features, bacterial/viral infections, detailed morphology and immune microenvironment of nine CVID patients with GC. The study of the immune microenvironment included automated digital counts of CD20+, CD4+, CD8+, FOXP3+, GATA3+ and CD138+ immune cells, as well as the evaluation of PD-L1 expression. Twenty-one GCs from non-CVID patients were used as a control group. GC in CVID patients was diagnosed mostly at early-stage (n = 6/9; 66.7%) and at younger age (median-age: 43y), when compared to non-CVID patients (p < 0.001). GC pathogenesis was closely related to Helicobacter pylori infection (n = 8/9; 88.9%), but not to Epstein-Barr virus (0.0%) or cytomegalovirus infection (0.0%). Non-neoplastic mucosa (non-NM) in CVID-patients displayed prominent lymphocytic gastritis (100%) and a dysfunctional immune microenvironment, characterised by higher rates of CD4+/CD8+/Foxp3+/GATA3+/PD-L1+ immune cells and the expected paucity of CD20+ B-lymphocytes and CD138+ plasma cells, when compared to non-CVID patients (p < 0.05). Changes in the immune microenvironment between non-NM and GC were not equivalent in CVID and non-CVID patients, reflecting the relevance of immune dysfunction for gastric carcinogenesis and GC progression in the CVID population. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Cancers: Gastric Cancer)
Show Figures

Figure 1

21 pages, 3887 KiB  
Article
TAZ Controls Helicobacter pylori-Induced Epithelial–Mesenchymal Transition and Cancer Stem Cell-Like Invasive and Tumorigenic Properties
by Camille Tiffon, Julie Giraud, Silvia Elena Molina-Castro, Sara Peru, Lornella Seeneevassen, Elodie Sifré, Cathy Staedel, Emilie Bessède, Pierre Dubus, Francis Mégraud, Philippe Lehours, Océane C.B. Martin and Christine Varon
Cells 2020, 9(6), 1462; https://doi.org/10.3390/cells9061462 - 13 Jun 2020
Cited by 30 | Viewed by 4121
Abstract
Helicobacter pylori infection, the main risk factor for gastric cancer (GC), leads to an epithelial–mesenchymal transition (EMT) of gastric epithelium contributing to gastric cancer stem cell (CSC) emergence. The Hippo pathway effectors yes-associated protein (YAP) and transcriptional co-activator with PDZ binding motif (TAZ) [...] Read more.
Helicobacter pylori infection, the main risk factor for gastric cancer (GC), leads to an epithelial–mesenchymal transition (EMT) of gastric epithelium contributing to gastric cancer stem cell (CSC) emergence. The Hippo pathway effectors yes-associated protein (YAP) and transcriptional co-activator with PDZ binding motif (TAZ) control cancer initiation and progression in many cancers including GC. Here, we investigated the role of TAZ in the early steps of H. pylori-mediated gastric carcinogenesis. TAZ implication in EMT, invasion, and CSC-related tumorigenic properties were evaluated in three gastric epithelial cell lines infected by H. pylori. We showed that H. pylori infection increased TAZ nuclear expression and transcriptional enhancer TEA domain (TEAD) transcription factors transcriptional activity. Nuclear TAZ and zinc finger E-box-binding homeobox 1 (ZEB1) were co-overexpressed in cells harboring a mesenchymal phenotype in vitro, and in areas of regenerative hyperplasia in gastric mucosa of H. pylori-infected patients and experimentally infected mice, as well as at the invasive front of gastric carcinoma. TAZ silencing reduced ZEB1 expression and EMT phenotype, and strongly inhibited invasion and tumorsphere formation induced by H. pylori. In conclusion, TAZ activation in response to H. pylori infection contributes to H. pylori-induced EMT, invasion, and CSC-like tumorigenic properties. TAZ overexpression in H. pylori-induced pre-neoplastic lesions and in GC could therefore constitute a biomarker of early transformation in gastric carcinogenesis. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Cancers: Gastric Cancer)
Show Figures

Graphical abstract

22 pages, 5185 KiB  
Article
Helicobacter Pylori Targets the EPHA2 Receptor Tyrosine Kinase in Gastric Cells Modulating Key Cellular Functions
by Marina Leite, Miguel S. Marques, Joana Melo, Marta T. Pinto, Bruno Cavadas, Miguel Aroso, Maria Gomez-Lazaro, Raquel Seruca and Ceu Figueiredo
Cells 2020, 9(2), 513; https://doi.org/10.3390/cells9020513 - 24 Feb 2020
Cited by 18 | Viewed by 4764
Abstract
Helicobacter pylori, a stomach-colonizing Gram-negative bacterium, is the main etiological factor of various gastroduodenal diseases, including gastric adenocarcinoma. By establishing a life-long infection of the gastric mucosa, H. pylori continuously activates host-signaling pathways, in particular those associated with receptor tyrosine kinases. Using [...] Read more.
Helicobacter pylori, a stomach-colonizing Gram-negative bacterium, is the main etiological factor of various gastroduodenal diseases, including gastric adenocarcinoma. By establishing a life-long infection of the gastric mucosa, H. pylori continuously activates host-signaling pathways, in particular those associated with receptor tyrosine kinases. Using two different gastric epithelial cell lines, we show that H. pylori targets the receptor tyrosine kinase EPHA2. For long periods of time post-infection, H. pylori induces EPHA2 protein downregulation without affecting its mRNA levels, an effect preceded by receptor activation via phosphorylation. EPHA2 receptor downregulation occurs via the lysosomal degradation pathway and is independent of the H. pylori virulence factors CagA, VacA, and T4SS. Using small interfering RNA, we show that EPHA2 knockdown affects cell–cell and cell–matrix adhesion, invasion, and angiogenesis, which are critical cellular processes in early gastric lesions and carcinogenesis mediated by the bacteria. This work contributes to the unraveling of the underlying mechanisms of H. pylori–host interactions and associated diseases. Additionally, it raises awareness for potential interference between H. pylori infection and the efficacy of gastric cancer therapies targeting receptors tyrosine kinases, given that infection affects the steady-state levels and dynamics of some receptor tyrosine kinases (RTKs) and their signaling pathways. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Cancers: Gastric Cancer)
Show Figures

Graphical abstract

18 pages, 5527 KiB  
Article
High Levels of Class I Major Histocompatibility Complex mRNA Are Present in Epstein–Barr Virus-Associated Gastric Adenocarcinomas
by Farhad Ghasemi, Steven F. Gameiro, Tanner M. Tessier, Allison H. Maciver and Joe S. Mymryk
Cells 2020, 9(2), 499; https://doi.org/10.3390/cells9020499 - 21 Feb 2020
Cited by 18 | Viewed by 2928
Abstract
Epstein–Barr virus (EBV) is responsible for approximately 9% of stomach adenocarcinomas. EBV-encoded microRNAs have been reported as reducing the function of the class I major histocompatibility complex (MHC-I) antigen presentation apparatus, which could allow infected cells to evade adaptive immune responses. Using data [...] Read more.
Epstein–Barr virus (EBV) is responsible for approximately 9% of stomach adenocarcinomas. EBV-encoded microRNAs have been reported as reducing the function of the class I major histocompatibility complex (MHC-I) antigen presentation apparatus, which could allow infected cells to evade adaptive immune responses. Using data from nearly 400 human gastric carcinomas (GCs), we assessed the impact of EBV on MHC-I heavy and light chain mRNA levels, as well as multiple other components essential for antigen processing and presentation. Unexpectedly, mRNA levels of these genes were as high, or higher, in EBV-associated gastric carcinomas (EBVaGCs) compared to normal control tissues or other GC subtypes. This coordinated upregulation could have been a consequence of the higher intratumoral levels of interferon γ in EBVaGCs, which correlated with signatures of increased infiltration by T and natural killer (NK) cells. These results indicate that EBV-encoded products do not effectively reduce mRNA levels of the MHC-I antigen presentation apparatus in human GCs. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Cancers: Gastric Cancer)
Show Figures

Figure 1

14 pages, 3747 KiB  
Article
Impact of Truncated O-glycans in Gastric-Cancer-Associated CD44v9 Detection
by Inês B. Moreira, Filipe Pinto, Catarina Gomes, Diana Campos and Celso A. Reis
Cells 2020, 9(2), 264; https://doi.org/10.3390/cells9020264 - 21 Jan 2020
Cited by 8 | Viewed by 3586
Abstract
CD44 variant isoforms are often upregulated in cancer and associated with increased aggressive tumor phenotypes. The CD44v9 is one of the major protein splice variant isoforms expressed in human gastrointestinal cancer cells. Immunodetection of CD44 isoforms like CD44v9 in tumor tissue is almost [...] Read more.
CD44 variant isoforms are often upregulated in cancer and associated with increased aggressive tumor phenotypes. The CD44v9 is one of the major protein splice variant isoforms expressed in human gastrointestinal cancer cells. Immunodetection of CD44 isoforms like CD44v9 in tumor tissue is almost exclusively performed by using specific monoclonal antibodies. However, the structural variability conferred by both the alternative splicing and CD44 protein glycosylation is disregarded. In the present work, we have evaluated the role of O-glycosylation using glycoengineered gastric cancer models in the detection of CD44v9 by monoclonal antibodies. We demonstrated, using different technical approaches, that the presence of immature O-glycan structures, such as Tn and STn, enhance CD44v9 protein detection. These findings can have significant implications in clinical applications mainly at the detection and targeting of this cancer-related CD44v9 isoform and highlight the utmost importance of considering glycan structures in cancer biomarker detection and in therapy targeting. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Cancers: Gastric Cancer)
Show Figures

Figure 1

17 pages, 2367 KiB  
Article
Comprehensive Multi-Omics Analysis Reveals Aberrant Metabolism of Epstein–Barr-Virus-Associated Gastric Carcinoma
by Sang Jun Yoon, Jun Yeob Kim, Nguyen Phuoc Long, Jung Eun Min, Hyung Min Kim, Jae Hee Yoon, Nguyen Hoang Anh, Myung Chan Park, Sung Won Kwon and Suk Kyeong Lee
Cells 2019, 8(10), 1220; https://doi.org/10.3390/cells8101220 - 8 Oct 2019
Cited by 25 | Viewed by 4820
Abstract
The metabolic landscape of Epstein–Barr-virus-associated gastric cancer (EBVaGC) remains to be elucidated. In this study, we used transcriptomics, metabolomics, and lipidomics to comprehensively investigate aberrant metabolism in EBVaGC. Specifically, we conducted gene expression analyses using microarray-based data from gastric adenocarcinoma epithelial cell lines [...] Read more.
The metabolic landscape of Epstein–Barr-virus-associated gastric cancer (EBVaGC) remains to be elucidated. In this study, we used transcriptomics, metabolomics, and lipidomics to comprehensively investigate aberrant metabolism in EBVaGC. Specifically, we conducted gene expression analyses using microarray-based data from gastric adenocarcinoma epithelial cell lines and tissue samples from patients with clinically advanced gastric carcinoma. We also conducted complementary metabolomics and lipidomics using various mass spectrometry platforms. We found a significant downregulation of genes related to metabolic pathways, especially the metabolism of amino acids, lipids, and carbohydrates. The effect of dysregulated metabolic genes was confirmed in a survival analysis of 3951 gastric cancer patients. We found 57 upregulated metabolites and 31 metabolites that were downregulated in EBVaGC compared with EBV-negative gastric cancer. Sixty-nine lipids, mainly ether-linked phospholipids and triacylglycerols, were downregulated, whereas 45 lipids, mainly phospholipids, were upregulated. In total, 15 metabolisms related to polar metabolites and 15 lipid-associated pathways were involved in alteration of metabolites by EBV in gastric cancer. In this work, we have described the metabolic landscape of EBVaGC at the multi-omics level. These findings could help elucidate the mechanism of EBVaGC oncogenesis. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Cancers: Gastric Cancer)
Show Figures

Graphical abstract

13 pages, 2248 KiB  
Article
ARID1A Mutations Are Associated with Increased Immune Activity in Gastrointestinal Cancer
by Lin Li, Mengyuan Li, Zehang Jiang and Xiaosheng Wang
Cells 2019, 8(7), 678; https://doi.org/10.3390/cells8070678 - 4 Jul 2019
Cited by 77 | Viewed by 5855
Abstract
Because traditional treatment strategies for advanced gastrointestinal (GI) cancers often have a limited therapeutic effect, immunotherapy could be a viable approach for the therapy of advanced GI cancers, considering the recent success of immunotherapy in treating various refractory malignancies, including the DNA mismatch [...] Read more.
Because traditional treatment strategies for advanced gastrointestinal (GI) cancers often have a limited therapeutic effect, immunotherapy could be a viable approach for the therapy of advanced GI cancers, considering the recent success of immunotherapy in treating various refractory malignancies, including the DNA mismatch repair-deficient GI cancers. However, only a subset of cancer patients currently respond to immunotherapy. Thus, it is important to identify useful biomarkers for predicting cancer immunotherapy response. The tumor suppressor gene ARID1A has a high mutation rate in GI cancers and its deficiency is correlated with the microsatellite instability (MSI) genomic feature of cancer. We investigated the correlation between ARID1A mutations and tumor immunity using three GI cancer genomics datasets by the bioinformatic approach, and found that diverse antitumor immune signatures were more highly enriched in ARID1A-mutated GI cancers than in ARID1A-wildtype GI cancers. The elevated immune activity in ARID1A-mutated GI cancers was associated with the higher tumor mutation burden and lower tumor aneuploidy level, as well as a higher proportion of MSI cancers in this GI cancer subtype. Moreover, we found that ARID1A-mutated GI cancers more highly expressed PD-L1 than ARID1A-wildtype GI cancers. The elevated antitumor immune signatures and PD-L1 expression could contribute to the more active immunotherapeutic responsiveness and better survival prognosis in ARID1A-mutated GI cancers than in ARID1A-wildtype GI cancers in the immunotherapy setting, as evidenced in three cancer cohorts receiving immunotherapy. Thus, the ARID1A mutation could be a useful biomarker for identifying GI cancer patients responsive to immunotherapy. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Cancers: Gastric Cancer)
Show Figures

Graphical abstract

Review

Jump to: Research

23 pages, 1178 KiB  
Review
The Extracellular Matrix: An Accomplice in Gastric Cancer Development and Progression
by Ana Margarida Moreira, Joana Pereira, Soraia Melo, Maria Sofia Fernandes, Patrícia Carneiro, Raquel Seruca and Joana Figueiredo
Cells 2020, 9(2), 394; https://doi.org/10.3390/cells9020394 - 8 Feb 2020
Cited by 62 | Viewed by 9991
Abstract
The extracellular matrix (ECM) is a dynamic and highly organized tissue structure, providing support and maintaining normal epithelial architecture. In the last decade, increasing evidence has emerged demonstrating that alterations in ECM composition and assembly strongly affect cellular function and behavior. Even though [...] Read more.
The extracellular matrix (ECM) is a dynamic and highly organized tissue structure, providing support and maintaining normal epithelial architecture. In the last decade, increasing evidence has emerged demonstrating that alterations in ECM composition and assembly strongly affect cellular function and behavior. Even though the detailed mechanisms underlying cell-ECM crosstalk are yet to unravel, it is well established that ECM deregulation accompanies the development of many pathological conditions, such as gastric cancer. Notably, gastric cancer remains a worldwide concern, representing the third most frequent cause of cancer-associated deaths. Despite increased surveillance protocols, patients are usually diagnosed at advanced disease stages, urging the identification of novel diagnostic biomarkers and efficient therapeutic strategies. In this review, we provide a comprehensive overview regarding expression patterns of ECM components and cognate receptors described in normal gastric epithelium, pre-malignant lesions, and gastric carcinomas. Important insights are also discussed for the use of ECM-associated molecules as predictive biomarkers of the disease or as potential targets in gastric cancer. Full article
(This article belongs to the Special Issue Molecular and Cellular Mechanisms of Cancers: Gastric Cancer)
Show Figures

Figure 1

Back to TopTop