Review Open Access
Copyright ©The Author(s) 2018. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Clin Oncol. Sep 14, 2018; 9(5): 83-89
Published online Sep 14, 2018. doi: 10.5306/wjco.v9.i5.83
Role of polymorphisms in genes that encode cytokines and Helicobacter pylori virulence factors in gastric carcinogenesis
Breno Bittencourt de Brito, Filipe Antônio França da Silva, Fabrício Freire de Melo, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Brazil
ORCID number: Breno Bittencourt de Brito (0000-0002-1831-7909); Filipe Antônio França da Silva (0000-0002-0550-1109); Fabrício Freire de Melo (0000-0002-5680-2753).
Author contributions: All authors equally contributed to this paper with conception and design of the study, literature review and analysis, drafting and critical revision and editing, and final approval of the final version.
Conflict-of-interest statement: No potential conflicts of interest. No financial support.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Fabrício Freire de Melo, PhD, Professor, Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Rua Hormindo Barros, 58, Quadra 17, Lote 58, Vitória da Conquista 45029-094, Brazil. freiremelo@yahoo.com.br
Telephone: +55-77-991968134
Received: April 30, 2018
Peer-review started: April 30, 2018
First decision: May 17, 2018
Revised: June 23, 2018
Accepted: June 27, 2018
Article in press: June 28, 2018
Published online: September 14, 2018

Abstract

The Helicobacter pylori (H. pylori) infection is a determinant factor in gastric cancer (GC) development. However, the infection outcomes are variable and depend on both host and bacterial characteristics. Some host cytokines such as interleukin (IL)-1β, IL-1Ra, IL-8, IL-10 and tumor necrosis factor-α play important roles in the host immune system response to the pathogen, in the development of gastric mucosal lesions and in cell malignant transformation. Therefore, these host factors are crucial in neoplastic processes. Certain polymorphisms in genes that encode these cytokines have been associated with an increased risk of GC. On the other hand, various virulence factors found in distinct H. pylori bacterial strains, including cytotoxin-associated antigen A, vacuolating cytotoxin, duodenal ulcer promoting gene A protein, outer inflammatory protein and blood group antigen binding adhesin, have been associated with the pathogenesis of different gastric diseases. The virulent factors mentioned above allow the successful infection by the bacterium and play crucial roles in gastric mucosa lesions, including malignant transformation. Moreover, the role of host polymorphisms and bacterial virulence factors in gastric carcinogenesis seems to vary among different countries and populations. The identification of host and bacterium factors that are associated with an increased risk of GC development may be useful in determining the prognosis of infection in patients, what could help in clinical decision-making and in providing of an optimized clinical approach.

Key Words: Helicobacter pylori, Virulence factors, Cytokines, Gene polymorphisms, Gastric cancer

Core tip: Various polymorphisms in host genes that encode cytokines and Helicobacter pylori virulence factors have been associated with different tendencies of gastric diseases development. Several reviews have been written on the role of host and bacterial isolated factors in gastric carcinogenesis. However, only a small amount of reviews unites the important characteristics of both bacterium and host in carcinogenesis. General overviews about polymorphisms in genes that encode cytokines are also scarce. We aimed to join the main polymorphisms in genes that encode cytokines and bacterial virulent factors related to gastric carcinogenesis and to provide a broad overview about these themes.



INTRODUCTION

Helicobacter pylori (H. pylori) is a gram negative bacterium, which inhabits the gastric epithelial tissue of most people in the world[1], and it is considered a determinant factor in the initiation of gastric carcinogenesis[2]. Gastric cancer (GC) is one of the four most prevalent neoplasms and the second biggest cause of deaths in consequence of cancer worldwide[3]. Despite the importance of H. pylori in gastric carcinogenesis, the development of GC only occurs in a minority of infected people, demonstrating that the infection outcomes are variable. It is believed that multifactorial precancerous processes associated with both host mucosal inflammatory response and pathogen characteristics are determinant in the severity of the disease[4].

The host immune system response plays a crucial role in the outcomes of H. pylori infection. Polymorphisms in genes that encode cytokines have been reported and associated with the severity of gastric mucosa inflammation and GC development. Some of these determinant variations are present in genes that encode cytokines such as interleukin (IL)-1β, IL-1Ra, IL-8, IL-10 and tumor necrosis factor (TNF)-α[5-13]. These polymorphisms are important aspects in understanding gastric carcinogenesis, since chronic inflammation induced by the bacterium is critical in the emergence and evolution of GC precursor lesions (Figure 1)[14].

Figure 1
Figure 1 Potential functions of the host genetic polymorphisms in gastric carcinogenesis. IL: Interleukin; GC: Gastric cancer; TNF: Tumor necrosis factor.

On the other hand, the virulence factors of H. pylori are determinant in the interaction with host cells. Cytotoxin associated antigen A (CagA), vacuolating cytotoxin (VacA), duodenal ulcer promoting gene A protein (DupA), outer inflammatory protein (OipA) and blood group antigen binding adhesin (BabA) are some virulent factors that seem to be associated with different risks of GC development[15]. Furthermore, H. pylori with EPIYA-D or more than one EPIYA-C segment in its CagA gene have been associated with a higher risk of gastric carcinogenesis[16-20].

POLYMORPHISMS IN GENES THAT ENCODE CYTOKINES AND GASTRIC CARCINOGENESIS

Gastric carcinogenesis is a process in which chronic inflammatory status plays a crucial role. The increase of inflammatory cytokine levels, due to H. pylori infection, seems to be determinant in the initiation and progression of GC[12]. The intensity of the expression of cytokines can be modified by functional polymorphisms in the promoter regions of the genes, which has the potential to alter the affinity of transcription factors, interfering in the expression levels of the messenger ribonucleic acid (mRNA) of specific inflammatory mediators related to the susceptibility of GC initiation[21].

IL-1

IL-1 is a family of cytokines that possesses 11 described members, among which IL-1β and IL-1 receptor antagonist (IL-1Ra), combined with H. pylori infection, seem to be key factors in GC development[22-24]. Signaling through the IL-1 receptor is a necessary event for the beginning and sustenance of various responses of the immune system[25].

The promoter regions of IL1B and IL1RN genes, which encode IL-1β and IL-1Ra respectively, have SNPs that modify the expression of the genes and affect the inflammatory response[26]. These SNPs increase the IL-1β/IL-1Ra ratio, which unleashes processes that result in gastric hypochlorhydria, favoring GC development[15,27].

IL-1β is an important cytokine for host-response to pathogens; however, this mediator can exacerbate damage during chronic diseases[28]. High levels of IL-1β in H. pylori infections lead to gastrin overexpression, increased gastric inflammation, hypochlorhydria, and gastric atrophy[29]. Moreover, IL-1β might promote neoplastic growth[30]. The IL1B gene can be composed by three different SNPs: C-T base transition at IL-1B-511 (rs16944), T-C base transition at IL-1B-31 (rs1143627) and IL-1B-3954 (rs1143634), and all of them are strongly associated with increased production of proinflammatory cytokines, hypochlorhydria and increased GC risk, mainly intestinal type, among Caucasians, but not among Asians or Hispanics[31-34].

IL-1Ra inhibits IL-1α and IL-1β by means of binding to IL-1 receptors. IL1RN possesses a changeable number of tandem repeats in intron 2, forming long alleles (IL1RN1) with 3-6 repeats or a short allele (IL1RN2) with 2 repeats[35]. The IL1RN2 allele is associated with severe gastric lesions and higher risk for GC, besides raised IL-1β expression in Caucasians[33-36].

IL-8

IL-8 is a potent cytokine that induces the directed migration of cells to inflammatory sites, acting as a chemoattractant[37]. IL-8 secretion can be increased by different stimuli, such as live bacteria (including H. pylori) and lipopolysaccharides (LPS), besides others inflammatory cytokines, including IL-1 and TNF[38]. The association of IL-8 with angiogenesis, adhesion and tumorigenesis have been related[39,40].

The gene CXCL8, which encodes IL-8, is located on 4q12-21 chromosome and possesses four exons and three introns[41]. An A/T SNP in the -251 position of this gene (rs2227532) has been associated with the development of various inflammatory diseases and cancer, including GC in Asians, but not in Europeans[42,43]. Furthermore, the IL-8-251 A allele was related to increased levels of IL-8[41].

IL-10

In opposition to the cytokines mentioned above, IL-10 is an anti-inflammatory cytokine, and it is involved in the cytotoxic response of inflammation and in cell downregulation. Moreover, this mediator prevents the production of pro-inflammatory cytokines, including TNF-α and IL-8[44]. Some studies have demonstrated that SNPs, particularly IL-10-592 (rs1800872) and IL-10-1082 (rs3021097) alleles, might modulate transcriptional activation and affect IL-10 production in vitro. These IL-10 polymorphisms are related to lower mRNA expression of this cytokine and it have been associated with GC development in Asians[45-48].

TNF-α

TNF-α composes the TNF/TNFR cytokine superfamily and it is involved in maintenance and homeostasis of the immune system and host defense[49]. However, this cytokine is related to various pathologic processes, including autoimmunity, chronic inflammatory processes and malignant disease[50]. According to studies, TNF-α signaling through TNFR1 (TNF-α receptor) is important for gastric tumor development[51,52].

Some SNPs in the TNF-α gene are related to increased expression of this cytokine. Among these polymorphisms, TNF-α-857 C/T (rs1799724), TNF-α-308 G/A (rs1800629) and TNF-α-238 G/A (rs361525) are the most studied ones. TNF-α-308 G/A was significantly associated with GC only in Caucasians, while TNF-α-857 and TNF-α-238 were related to an increased risk of gastric tumorigenesis in Asians, but not in Caucasians[53-55].

H. PYLORI VIRULENCE FACTORS AND CARCINOGENESIS

The capacity of H. pylori bacteria to trigger a carcinogenic process is not limited to the intense immune response that they unleash, but it also depends on various bacterial factors that can start and modulate neoplastic processes[56]. Different virulent factors found in distinct bacterial strains have been closely associated with the emergence of gastric carcinogenesis. However, genetic variations in genes that encode these virulence factors as well as geographic differences can influence the role of these proteins in GC emergence[15].

CagA

CagA is encoded by the cagA gene, present in a DNA segment containing 30 genes called cag pathogenicity island (cag PAI). Infections by strains containing CagA are more capable to induce carcinogenic processes, mainly those with EPIYA-D or more than one EPIYA-C segment[57]. Various cag PAI genes are involved in the codification of elements of a pilus structure named type IV secretion system (TFSS), which has the function of transporting CagA from bacterium to the cytoplasm of the cells from gastric epithelium[58].

After being injected into host cells by TFSS, CagA suffers tyrosine phosphorylation at a carboxi-terminal segment compound by distinct number of EPIYA (Glu-Pro-Ile-Tyr-Ala) regions. There are different EPIYA segments -A, B, C and D-, which contain distinct amino acids in their structure[20]. EPIYA A and B segments are present in most CagA proteins and are followed either by 0-3 EPIYA-C segments in H. pylori strains from Occidental countries or by EPIYA-D segments in Eastern countries[59].

Following EPIYA-C or EPIYA-D phosphorylation, an interaction between these segments and SHP-2 possessing SH2 domain occurs, unleashing SHP-2/mitogen-activated protein kinases (MAPK), ERK1, 2-JAK and STAT3 pathways[20]. Cytotoxin associated antigen containing EPIYA-D or more than one EPIYA-C segment ties to SHP-2 more strongly, being more effective in the activation of the pathways mentioned above[60]. This process, activated by CagA, leads to dysfunction of cell growth and of cell-to-cell contact inhibition, cell migration, epithelial cell elongation, and increase of epithelial cell turnover, increasing the propensity of acquirement of precancerous genetic changes by damaged cells[61]. Furthermore, it was demonstrated that relatives of GC patients are more often infected by H. pylori strains with more than one EPIYA-C segment in CagA structure[62]. Another study carried out by this same group, performed in a Brazilian population, showed that the host signal transducer and activator of transcription protein 3 (STAT3) rs7744166 polymorphism, as well as being infected by H. pylori with CagA containing more than one EPIYA-C segment, are independent predisposing factors for GC[20].

VacA

VacA is another determinant virulence factor in H. pylori infection and in gastric carcinogenesis. Patients infected with VacA-positive H. pylori strains have a higher propensity for GC development when compared with patients colonized by VacA-negative strains, either in American or in Asian people[63]. Particularly, individuals infected with H. pylori strains VacA s1, m1 and s1m1 had an increased risk for gastric carcinogenic unleash in Middle East, Africa and Latin America populations[64]. The peptide mentioned above has only two functional domains in its structure. One of them, the p55-58 domain, has the function of binding to receptors of gastric epithelial cells. The other functional domain, p33-37, produces the cytotoxic effect[65].

VacA is a 90 kDa exotoxin that is activated in a low pH environment[66]. This toxin promotes the generation of numerous acidic vacuoles in gastric epithelial cell cytoplasm[67]. In this process, VacA affects the structure and function of the membrane, the endoplasmic reticulum, the Golgi apparatus and the mitochondria, which can lead to cell death. Furthermore, vacuolating cytotoxin also plays an important role in the activation and suppression of the immune response[68]. This peptide induces a powerful inhibition over T lymphocyte proliferation by means of an interaction with dendritic cells, which are reprogrammed to a tolerogenic genotype[69]. The damage and the immunomodulation performed by this toxin contributes to the increase of gastric mucosa inflammation, ulceration and carcinogenesis in mammals[68].

DupA

Unlike the other virulence factors mentioned in this article, DupA seems to be a protective condition for GC. The dupA gene is constituted by two homologue genes of virB4, jhp0917 and jhp091, which constitute a continuous gene. The real function of the protein encoded by dupA is still obscure, however, its mechanisms seems to be related to the increase of the production of IL-8 in the gastric antrum, contributing to the development of gastritis that predominates in that gastric region, a process that leads to duodenal ulcer formation[70]. DupA has been significantly associated with duodenal ulcer formation in Asian countries, but this relation was not observed in the Western population[71]. Furthermore, DupA-positive H. pylori has been associated with eradication failure[72].

OipA

OipA constitutes a group of peptides described as outer membrane proteins (OMPs), a H. pylori protein family composed of 32 components[73]. OipA has been described as a better marker for severe clinical outcomes than CagA, since the infection by strains possessing OipA is an independent determinant risk factor of GC vs gastritis in Americans[74,75]. OipA enhances IL-8 production and leads to an increased inflammation status of gastric epithelium. Moreover, it was observed that OipA could inhibit the maturation of dendritic cells in vitro, which might contribute to the immunomodulatory processes performed by H. pylori[76].

BabA

babA is a gene that encodes an adhesin whom allows the specific binding to the b and H-1 Lewis antigens, which are expressed in the surface of the gastric mucosa cells[77]. The adhesion of the H. pylori to the gastric epithelium mediated by blood group antigen binding adhesin (BabA) appears to play a critical function in the transference of bacterial virulence factors to the host cells. This process contributes to the development of tissue lesions, and a high correlation of babA-positive strains of H. pylori with GC has been described[78,79].

CONCLUSION

Despite the wide knowledge about host and H. pylori interaction developed since the discovery of its colonization in human stomach, many characteristics that contribute to the infection outcomes are still obscure. The understandings about host polymorphisms in genes that encode cytokines and bacterium virulence factors in GC development are important not only for the determination of patients’ prognosis, but it is also a potential way for the development of new preventive and therapeutic strategies.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Oncology

Country of origin: Brazil

Peer-review report classification

Grade A (Excellent): 0

Grade B (Very good): 0

Grade C (Good): C, C

Grade D (Fair): D

Grade E (Poor): 0

P- Reviewer: Barreto SG, Lee CL, Sun X S- Editor: Ji FF L- Editor: Filipodia E- Editor: Tan WW

References
1.  Alzahrani S, Lina TT, Gonzalez J, Pinchuk IV, Beswick EJ, Reyes VE. Effect of Helicobacter pylori on gastric epithelial cells. World J Gastroenterol. 2014;20:12767-12780.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 105]  [Cited by in F6Publishing: 105]  [Article Influence: 10.5]  [Reference Citation Analysis (6)]
2.  Shimizu T, Marusawa H, Watanabe N, Chiba T. Molecular Pathogenesis of Helicobacter pylori-Related Gastric Cancer. Gastroenterol Clin North Am. 2015;44:625-638.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
3.  Ang TL, Fock KM. Clinical epidemiology of gastric cancer. Singapore Med J. 2014;55:621-628.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 191]  [Cited by in F6Publishing: 252]  [Article Influence: 28.0]  [Reference Citation Analysis (0)]
4.  Wang MY, Liu XF, Gao XZ. Helicobacter pylori virulence factors in development of gastric carcinoma. Future Microbiol. 2015;10:1505-1516.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 11]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
5.  Gao Y, He Y, Ding J, Wu K, Hu B, Liu Y, Wu Y, Guo B, Shen Y, Landi D. An insertion/deletion polymorphism at miRNA-122-binding site in the interleukin-1alpha 3’ untranslated region confers risk for hepatocellular carcinoma. Carcinogenesis. 2009;30:2064-2069.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 120]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
6.  Kutikhin AG, Yuzhalin AE, Volkov AN, Zhivotovskiy AS, Brusina EB. Correlation between genetic polymorphisms within IL-1B and TLR4 genes and cancer risk in a Russian population: a case-control study. Tumour Biol. 2014;35:4821-4830.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 32]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
7.  Sun Z, Cui Y, Jin X, Pei J. Association between IL-4 -590C > T polymorphism and gastric cancer risk. Tumour Biol. 2014;35:1517-1521.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 12]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
8.  Zhang JZ, Liu CM, Peng HP, Zhang Y. Association of genetic variations in IL-6/IL-6R pathway genes with gastric cancer risk in a Chinese population. Gene. 2017;623:1-4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 18]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
9.  Cui X, Huang Q, Li X, Liu F, Wang D, Yan D, Wang B, Yang C, Mi J, Tian G. Relationship Between Interleukin-10 Gene C-819T Polymorphism and Gastric Cancer Risk: Insights From a Meta-Analysis. Med Sci Monit. 2016;22:2839-2845.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Qi WT, Gao JL, Zhang SS. Role of IL-17 gene polymorphisms in the susceptibility to gastric cancer. Genet Mol Res. 2015;14:13364-13369.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 11]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
11.  Cho YA, Kim J. Association of IL4, IL13, and IL4R polymorphisms with gastrointestinal cancer risk: A meta-analysis. J Epidemiol. 2017;27:215-220.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 20]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
12.  de Oliveira JG, Rossi AF, Nizato DM, Cadamuro AC, Jorge YC, Valsechi MC, Venâncio LP, Rahal P, Pavarino ÉC, Goloni-Bertollo EM, Silva AE. Influence of functional polymorphisms in TNF-α, IL-8, and IL-10 cytokine genes on mRNA expression levels and risk of gastric cancer. Tumour Biol. 2015;36:9159-9170.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 42]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
13.  Bockerstett KA, DiPaolo RJ. Regulation of Gastric Carcinogenesis by Inflammatory Cytokines. Cell Mol Gastroenterol Hepatol. 2017;4:47-53.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 68]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
14.  Wang F, Meng W, Wang B, Qiao L. Helicobacter pylori-induced gastric inflammation and gastric cancer. Cancer Lett. 2014;345:196-202.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 375]  [Cited by in F6Publishing: 482]  [Article Influence: 48.2]  [Reference Citation Analysis (1)]
15.  Rocha GA, Guerra JB, Rocha AM, Saraiva IE, da Silva DA, de Oliveira CA, Queiroz DM. IL1RN polymorphic gene and cagA-positive status independently increase the risk of noncardia gastric carcinoma. Int J Cancer. 2005;115:678-683.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 52]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
16.  Zhang J, Wu J, Peng X, Song J, Wang J, Dong W. Associations between STAT3 rs744166 polymorphisms and susceptibility to ulcerative colitis and Crohn’s disease: a meta-analysis. PLoS One. 2014;9:e109625.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
17.  Blaser MJ, Perez-Perez GI, Kleanthous H, Cover TL, Peek RM, Chyou PH, Stemmermann GN, Nomura A. Infection with Helicobacter pylori strains possessing cagA is associated with an increased risk of developing adenocarcinoma of the stomach. Cancer Res. 1995;55:2111-2115.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Basso D, Zambon CF, Letley DP, Stranges A, Marchet A, Rhead JL, Schiavon S, Guariso G, Ceroti M, Nitti D. Clinical relevance of Helicobacter pylori cagA and vacA gene polymorphisms. Gastroenterology. 2008;135:91-99.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 283]  [Cited by in F6Publishing: 269]  [Article Influence: 16.8]  [Reference Citation Analysis (0)]
19.  Naito M, Yamazaki T, Tsutsumi R, Higashi H, Onoe K, Yamazaki S, Azuma T, Hatakeyama M. Influence of EPIYA-repeat polymorphism on the phosphorylation-dependent biological activity of Helicobacter pylori CagA. Gastroenterology. 2006;130:1181-1190.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 148]  [Cited by in F6Publishing: 144]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
20.  Batista SA, Rocha GA, Rocha AM, Saraiva IE, Cabral MM, Oliveira RC, Queiroz DM. Higher number of Helicobacter pylori CagA EPIYA C phosphorylation sites increases the risk of gastric cancer, but not duodenal ulcer. BMC Microbiol. 2011;11:61.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 73]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
21.  Palomo J, Dietrich D, Martin P, Palmer G, Gabay C. The interleukin (IL)-1 cytokine family--Balance between agonists and antagonists in inflammatory diseases. Cytokine. 2015;76:25-37.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 273]  [Cited by in F6Publishing: 300]  [Article Influence: 33.3]  [Reference Citation Analysis (0)]
22.  Oelmann E, Stein H, Berdel WE, Herbst H. Expression of Interleukin-1 and Interleukin-1 Receptors Type 1 and Type 2 in Hodgkin Lymphoma. PLoS One. 2015;10:e0138747.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 11]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
23.  Sakamoto K, Hikiba Y, Nakagawa H, Hayakawa Y, Yanai A, Akanuma M, Ogura K, Hirata Y, Kaestner KH, Omata M. Inhibitor of kappaB kinase beta regulates gastric carcinogenesis via interleukin-1alpha expression. Gastroenterology. 2010;139:226-238.e6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 42]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
24.  Ma J, Sawai H, Matsuo Y, Ochi N, Yasuda A, Takahashi H, Wakasugi T, Funahashi H, Sato M, Okada Y. Interleukin-1alpha enhances angiogenesis and is associated with liver metastatic potential in human gastric cancer cell lines. J Surg Res. 2008;148:197-204.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 38]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
25.  Ruzzo A, Graziano F, Pizzagalli F, Santini D, Battistelli V, Panunzi S, Canestrari E, Catalano V, Humar B, Ficarelli R. Interleukin 1B gene (IL-1B) and interleukin 1 receptor antagonist gene (IL-1RN) polymorphisms in Helicobacter pylori-negative gastric cancer of intestinal and diffuse histotype. Ann Oncol. 2005;16:887-892.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 53]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
26.  Raza Y, Khan A, Khan AI, Khan S, Akhter S, Mubarak M, Ahmed A, Kazmi SU. Combination of Interleukin 1 Polymorphism and Helicobacter pylori Infection: an Increased Risk of Gastric Cancer in Pakistani Population. Pathol Oncol Res. 2017;23:873-880.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 12]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
27.  Hwang IR, Kodama T, Kikuchi S, Sakai K, Peterson LE, Graham DY, Yamaoka Y. Effect of interleukin 1 polymorphisms on gastric mucosal interleukin 1beta production in Helicobacter pylori infection. Gastroenterology. 2002;123:1793-1803.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 276]  [Cited by in F6Publishing: 303]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
28.  Lopez-Castejon G, Brough D. Understanding the mechanism of IL-1β secretion. Cytokine Growth Factor Rev. 2011;22:189-195.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 626]  [Cited by in F6Publishing: 841]  [Article Influence: 64.7]  [Reference Citation Analysis (0)]
29.  Huang FY, Chan AO, Lo RC, Rashid A, Wong DK, Cho CH, Lai CL, Yuen MF. Characterization of interleukin-1β in Helicobacter pylori-induced gastric inflammation and DNA methylation in interleukin-1 receptor type 1 knockout (IL-1R1(-/-)) mice. Eur J Cancer. 2013;49:2760-2770.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 39]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
30.  Furuta T, El-Omar EM, Xiao F, Shirai N, Takashima M, Sugimura H. Interleukin 1beta polymorphisms increase risk of hypochlorhydria and atrophic gastritis and reduce risk of duodenal ulcer recurrence in Japan. Gastroenterology. 2002;123:92-105.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 258]  [Cited by in F6Publishing: 253]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
31.  Wang P, Xia HH, Zhang JY, Dai LP, Xu XQ, Wang KJ. Association of interleukin-1 gene polymorphisms with gastric cancer: a meta-analysis. Int J Cancer. 2007;120:552-562.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 64]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
32.  Chen A, Li CN, Hsu PI, Lai KH, Tseng HH, Hsu PN, Lo GH, Lo CC, Lin CK, Hwang IR. Risks of interleukin-1 genetic polymorphisms and Helicobacter pylori infection in the development of gastric cancer. Aliment Pharmacol Ther. 2004;20:203-211.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 33]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
33.  Chung HW, Lim JB. Role of the tumor microenvironment in the pathogenesis of gastric carcinoma. World J Gastroenterol. 2014;20:1667-1680.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 65]  [Cited by in F6Publishing: 68]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
34.  Xue H, Lin B, Ni P, Xu H, Huang G. Interleukin-1B and interleukin-1 RN polymorphisms and gastric carcinoma risk: a meta-analysis. J Gastroenterol Hepatol. 2010;25:1604-1617.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 99]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
35.  Chiurillo MA. Role of gene polymorphisms in gastric cancer and its precursor lesions: current knowledge and perspectives in Latin American countries. World J Gastroenterol. 2014;20:4503-4515.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 21]  [Cited by in F6Publishing: 19]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
36.  Figueiredo C, Machado JC, Pharoah P, Seruca R, Sousa S, Carvalho R, Capelinha AF, Quint W, Caldas C, van Doorn LJ. Helicobacter pylori and interleukin 1 genotyping: an opportunity to identify high-risk individuals for gastric carcinoma. J Natl Cancer Inst. 2002;94:1680-1687.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 463]  [Cited by in F6Publishing: 453]  [Article Influence: 20.6]  [Reference Citation Analysis (0)]
37.  Sun Q, Sun F, Wang B, Liu S, Niu W, Liu E, Peng C, Wang J, Gao H, Liang B. Interleukin-8 promotes cell migration through integrin αvβ6 upregulation in colorectal cancer. Cancer Lett. 2014;354:245-253.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 41]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
38.  Remick DG. Interleukin-8. Crit Care Med. 2005;33:S466-S467.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 156]  [Cited by in F6Publishing: 182]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
39.  Kim JH, Frantz AM, Anderson KL, Graef AJ, Scott MC, Robinson S, Sharkey LC, O’Brien TD, Dickerson EB, Modiano JF. Interleukin-8 promotes canine hemangiosarcoma growth by regulating the tumor microenvironment. Exp Cell Res. 2014;323:155-164.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 35]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
40.  Ju D, Sun D, Xiu L, Meng X, Zhang C, Wei P. Interleukin-8 is associated with adhesion, migration and invasion in human gastric cancer SCG-7901 cells. Med Oncol. 2012;29:91-99.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 39]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
41.  Vairaktaris E, Yapijakis C, Serefoglou Z, Derka S, Vassiliou S, Nkenke E, Vylliotis A, Wiltfang J, Avgoustidis D, Critselis E. The interleukin-8 (-251A/T) polymorphism is associated with increased risk for oral squamous cell carcinoma. Eur J Surg Oncol. 2007;33:504-507.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 53]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
42.  Ohyauchi M, Imatani A, Yonechi M, Asano N, Miura A, Iijima K, Koike T, Sekine H, Ohara S, Shimosegawa T. The polymorphism interleukin 8 -251 A/T influences the susceptibility of Helicobacter pylori related gastric diseases in the Japanese population. Gut. 2005;54:330-335.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 165]  [Cited by in F6Publishing: 175]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
43.  Zhang Y, Zeng X, Lu H, Li Y, Ji H. Association between Interleukin-8-251A/T polymorphism and gastric cancer susceptibility: a meta-analysis based on 5286 cases and 8000 controls. Int J Clin Exp Med. 2015;8:22393-22402.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Maeda H, Okabayashi T, Nishimori I, Sugimoto T, Namikawa T, Dabanaka K, Tsujii S, Onishi S, Kobayashi M, Hanazaki K. Clinicopathologic features of adenocarcinoma at the gastric cardia: is it different from distal cancer of the stomach? J Am Coll Surg. 2008;206:306-310.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 42]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
45.  Qi M, Liu DM, Pan LL, Lin YX. Interleukin-10 gene -592C>A polymorphism and susceptibility to gastric cancer. Genet Mol Res. 2014;13:8954-8961.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
46.  Pan F, Tian J, Pan YY, Zhang Y. Association of IL-10-1082 promoter polymorphism with susceptibility to gastric cancer: evidence from 22 case-control studies. Mol Biol Rep. 2012;39:7143-7154.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 12]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
47.  Zhu Y, Wang J, He Q, Zhang JQ. The association between interleukin-10-592 polymorphism and gastric cancer risk: a meta-analysis. Med Oncol. 2011;28:133-136.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 9]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
48.  Yu T, Lu Q, Ou XL, Cao DZ, Yu Q. Clinical study on gastric cancer susceptibility genes IL-10-1082 and TNF-α. Genet Mol Res. 2014;13:10909-10912.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
49.  Croft M, Benedict CA, Ware CF. Clinical targeting of the TNF and TNFR superfamilies. Nat Rev Drug Discov. 2013;12:147-168.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 341]  [Cited by in F6Publishing: 318]  [Article Influence: 28.9]  [Reference Citation Analysis (0)]
50.  Balkwill F. TNF-alpha in promotion and progression of cancer. Cancer Metastasis Rev. 2006;25:409-416.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 554]  [Cited by in F6Publishing: 622]  [Article Influence: 36.6]  [Reference Citation Analysis (0)]
51.  Oshima H, Ishikawa T, Yoshida GJ, Naoi K, Maeda Y, Naka K, Ju X, Yamada Y, Minamoto T, Mukaida N. TNF-α/TNFR1 signaling promotes gastric tumorigenesis through induction of Noxo1 and Gna14 in tumor cells. Oncogene. 2014;33:3820-3829.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 100]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
52.  Oguma K, Oshima H, Oshima M. Inflammation, tumor necrosis factor and Wnt promotion in gastric cancer development. Future Oncol. 2010;6:515-526.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 32]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
53.  Yang JP, Hyun MH, Yoon JM, Park MJ, Kim D, Park S. Association between TNF-α-308 G/A gene polymorphism and gastric cancer risk: a systematic review and meta-analysis. Cytokine. 2014;70:104-114.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 26]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
54.  Gorouhi F, Islami F, Bahrami H, Kamangar F. Tumour-necrosis factor-A polymorphisms and gastric cancer risk: a meta-analysis. Br J Cancer. 2008;98:1443-1451.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 83]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
55.  Yu JY, Li L, Ma H, Liu K, Cheng X, Li YL, Song XL. Tumor necrosis factor-α 238 G/A polymorphism and gastric cancer risk: a meta-analysis. Tumour Biol. 2013;34:3859-3863.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 19]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
56.  Odenbreit S, Püls J, Sedlmaier B, Gerland E, Fischer W, Haas R. Translocation of Helicobacter pylori CagA into gastric epithelial cells by type IV secretion. Science. 2000;287:1497-1500.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 954]  [Cited by in F6Publishing: 934]  [Article Influence: 38.9]  [Reference Citation Analysis (0)]
57.  Fischer W. Assembly and molecular mode of action of the Helicobacter pylori Cag type IV secretion apparatus. FEBS J. 2011;278:1203-1212.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 117]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
58.  Rocha GA, Rocha AM, Gomes AD, Faria CL Jr, Melo FF, Batista SA, Fernandes VC, Almeida NB, Teixeira KN, Brito KS, Queiroz DM. STAT3 polymorphism and Helicobacter pylori CagA strains with higher number of EPIYA-C segments independently increase the risk of gastric cancer. BMC Cancer. 2015;15:528.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 20]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
59.  Hatakeyama M. Oncogenic mechanisms of the Helicobacter pylori CagA protein. Nat Rev Cancer. 2004;4:688-694.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 535]  [Cited by in F6Publishing: 554]  [Article Influence: 27.7]  [Reference Citation Analysis (0)]
60.  Lee YS, Lee DY, Yu DY, Kim S, Lee YC. Helicobacter pylori induces cell migration and invasion through casein kinase 2 in gastric epithelial cells. Helicobacter. 2014;19:465-475.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 16]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
61.  Suzuki N, Murata-Kamiya N, Yanagiya K, Suda W, Hattori M, Kanda H, Bingo A, Fujii Y, Maeda S, Koike K. Mutual reinforcement of inflammation and carcinogenesis by the Helicobacter pylori CagA oncoprotein. Sci Rep. 2015;5:10024.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 46]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
62.  Queiroz DM, Silva CI, Goncalves MH, Braga-Neto MB, Fialho AB, Fialho AM, Rocha GA, Rocha AM, Batista SA, Guerrant RL. Higher frequency of cagA EPIYA-C phosphorylation sites in H. pylori strains from first-degree relatives of gastric cancer patients. BMC Gastroenterol. 2012;12:107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 22]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
63.  Li Q, Liu J, Gong Y, Yuan Y. Serum VacA antibody is associated with risks of peptic ulcer and gastric cancer: A meta-analysis. Microb Pathog. 2016;99:220-228.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 15]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
64.  Matos JI, de Sousa HA, Marcos-Pinto R, Dinis-Ribeiro M. Helicobacter pylori CagA and VacA genotypes and gastric phenotype: a meta-analysis. Eur J Gastroenterol Hepatol. 2013;25:1431-1441.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 81]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
65.  Liu C, Wang YM, Li ZX, Zhang L, Ma JL, Zhou T, You WC, Pan KF. [Serological assessment of Helicobacter pylori-specific antibodies and their association with gastric lesions in a high-risk population]. Zhonghua Zhongliu Zazhi. 2013;35:547-551.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Hotchin NA, Cover TL, Akhtar N. Cell vacuolation induced by the VacA cytotoxin of Helicobacter pylori is regulated by the Rac1 GTPase. J Biol Chem. 2000;275:14009-14012.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 50]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
67.  Isomoto H, Moss J, Hirayama T. Pleiotropic actions of Helicobacter pylori vacuolating cytotoxin, VacA. Tohoku J Exp Med. 2010;220:3-14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 57]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
68.  Boquet P, Ricci V. Intoxication strategy of Helicobacter pylori VacA toxin. Trends Microbiol. 2012;20:165-174.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 74]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
69.  Gebert B, Fischer W, Weiss E, Hoffmann R, Haas R. Helicobacter pylori vacuolating cytotoxin inhibits T lymphocyte activation. Science. 2003;301:1099-1102.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 404]  [Cited by in F6Publishing: 395]  [Article Influence: 18.8]  [Reference Citation Analysis (0)]
70.  Lu H, Hsu PI, Graham DY, Yamaoka Y. Duodenal ulcer promoting gene of Helicobacter pylori. Gastroenterology. 2005;128:833-848.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 216]  [Cited by in F6Publishing: 207]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
71.  Yamaoka Y. Roles of the plasticity regions of Helicobacter pylori in gastroduodenal pathogenesis. J Med Microbiol. 2008;57:545-553.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 73]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
72.  Shiota S, Nguyen LT, Murakami K, Kuroda A, Mizukami K, Okimoto T, Kodama M, Fujioka T, Yamaoka Y. Association of helicobacter pylori dupA with the failure of primary eradication. J Clin Gastroenterol. 2012;46:297-301.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 32]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
73.  Yamaoka Y, Kwon DH, Graham DY. A M(r) 34,000 proinflammatory outer membrane protein (oipA) of Helicobacter pylori. Proc Natl Acad Sci U S A. 2000;97:7533-7538.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 292]  [Cited by in F6Publishing: 305]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
74.  Graham DY, Opekun AR, Osato MS, El-Zimaity HM, Lee CK, Yamaoka Y, Qureshi WA, Cadoz M, Monath TP. Challenge model for Helicobacter pylori infection in human volunteers. Gut. 2004;53:1235-1243.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 145]  [Cited by in F6Publishing: 141]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
75.  Liu J, He C, Chen M, Wang Z, Xing C, Yuan Y. Association of presence/absence and on/off patterns of Helicobacter pylori oipA gene with peptic ulcer disease and gastric cancer risks: a meta-analysis. BMC Infect Dis. 2013;13:555.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 47]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
76.  Dabiri H, Maleknejad P, Yamaoka Y, Feizabadi MM, Jafari F, Rezadehbashi M, Nakhjavani FA, Mirsalehian A, Zali MR. Distribution of Helicobacter pylori cagA, cagE, oipA and vacA in different major ethnic groups in Tehran, Iran. J Gastroenterol Hepatol. 2009;24:1380-1386.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 46]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
77.  Shahi H, Reiisi S, Sadeghiani M, Mahsa M, Bahreini R, Moghni M, damavandi M, fatollahi F, Shahverdi E, ramezani G. Prevalence of cagA and babA2 genes in Helicobacter Pylori strains Isolated from Iranian gastrointestinal disorder patients and their gastritis classification. J Biol Today’s World. 2014;3:256-260.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
78.  Rad R, Gerhard M, Lang R, Schöniger M, Rösch T, Schepp W, Becker I, Wagner H, Prinz C. The Helicobacter pylori blood group antigen-binding adhesin facilitates bacterial colonization and augments a nonspecific immune response. J Immunol. 2002;168:3033-3041.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 127]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
79.  Talebi Bezmin Abadi A, Taghvaei T, Mohabbati Mobarez A, Vaira G, Vaira D. High correlation of babA 2-positive strains of Helicobacter pylori with the presence of gastric cancer. Intern Emerg Med. 2013;8:497-501.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 39]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]