Pathogenic mechanisms of the oncoprotein CagA in H. pylori-induced gastric cancer (Review)

  • Authors:
    • Shuai-Yin Chen
    • Rong-Guang Zhang
    • Guang-Cai Duan
  • View Affiliations

  • Published online on: October 4, 2016     https://doi.org/10.3892/or.2016.5145
  • Pages: 3087-3094
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Infection with Helicobacter pylori is the strongest risk factor for the development of chronic gastritis, gastric ulcer and gastric carcinoma. The majority of the H. pylori-infected population remains asymptomatic, and only 1% of individuals may progress to gastric cancer. The clinical outcomes caused by H. pylori infection are considered to be associated with bacterial virulence, genetic polymorphism of hosts as well as environmental factors. Most H. pylori strains possess a cytotoxin-associated gene (cag) pathogenicity island (cagPAI), encoding a 120-140 kDa CagA protein, which is the most important bacterial oncoprotein. CagA is translocated into host cells via T4SS system and affects the expression of signaling proteins in a phosphorylation-dependent and independent manner. Thus, this review summarizes the results of relevant studies, discusses the pathogenesis of CagA-mediated gastric cancer.

Introduction

Helicobacter pylori is a spiral-shaped, flagellated, micro-aerophilic Gram-negative bacillus first described in 1982 by Marshall and Warren. H. pylori is thought to colonize the gastric mucosa of >50% of the world's population, with the higher prevalence in the developing countries (1,2). However, the majority of H. pylori-infected population is asymptomatic, but resulting in chronic gastritis. Only 10% of infected individuals occurred symptomatic diseases. Furthermore, experimental and epidemiological studies have indicated that H. pylori infection indeed increase the risk of gastric cancer. Based on this evidence, the World Health Organization International Agency for Research on Cancer classified H. pylori as class I carcinogen in 1994. The estimated total of infection-attributable cancer is 1.9 million cases every year, which is 17.8% of the global cancer burden. The principal agent is H. pylori, ~63.4% of all stomach cancer or 5.5% of the global cancer burden would be attributable to H. pylori infection.

The interindividual differences in risk of H. pylori-induced gastric diseases involve significant heterogeneity of both host genetics and H. pylori strain virulence factors. In the H. pylori-associated diseases pathogenic mechanisms, several strain-specific virulence factors were reported, such as cagA (cytotoxin-associated gene A), vacA (vacuolating cytotoxin A), hpaA (Helicobacter pylori adhesin A), babA (blood group antigen binding adhesin), dupA (duodenal ulcer-promoting gene A), iceA (induced by contact with epithelium) genes. One of the main virulence factors is CagA, which is associated with higher risk of gastric cancer and peptic ulcer. CagA protein can interact with intercellular proteins and activate signaling pathways through both tyrosine phosphorylation-dependent or independent mechanisms. Here, we review the possible underlying pathogenic mechanisms of the oncoprotein CagA in H. pylori-induced gastric diseases.

Cytotoxin associated gene pathogenicity island (cagPAI) and cytotoxin-associated gene A (cagA)

The cag PAI is an ~40-kb DNA, which likely was acquired by horizontal gene transfer from another strain in the course of evolution. The cag PAI contains ~31 genes including cagA, cagB, cagC, cagL, cagM, cagI, cagY, which encode the CagA protein and functional components of a type IV secretion system (T4SS) (3,4). cag PAI is found in >95% East Asian strains, whereas 60% of Western strains isolated are cag PAI-positive (5). In some strains, cagPAI is split into a right segment (cagI) and a left segment (cagII) by an insertion sequence (IS605). IS605 was associated with gastric cancer that was higher in H. pylori isolated from patients with gastric carcinoma than in patients with duodenal ulcer or chronic gastritis (6).

The cagA gene is ~3,500–5,000 bp located in the beginning region of cag PAI, encoding 120–145 kDa CagA protein. It is a recognized marker for the entire cag locus. The size of the cagA gene and its protein varies in different strains due to structural diversity in its C-terminal region. In Western populations, cagA-positive strain is associated with enhanced induction of gastritis, peptic ulcer, and higher risk of gastric cancer. However, in East Asia cagA gene is not associated with an increased risk of gastric diseases where almost all strains are cagA-positive (7,8). Franco et al constructed the Mongolian gerbils infection model. They indicate that loss of CagA prevents the development of cancer in this model (9). Ohnishi et al provided first direct evidence for the role of CagA as a bacterium-derived oncoprotein by transgenic mouse model (10).

Translocation and phosphorylation of CagA protein

Translocation of CagA into host epithelial cells is the first step in the processes of CagA-induced diseases. Several different Cag proteins are involved in the translocation of CagA (11). CagL carries a RGD (arginine-glycine-aspartate) motif that is important for binding and interaction with integrin α5β1 receptor on gastric epithelial cells, and triggers CagA delivery into the target cells, as well as downstream signaling to active tyrosine kinases including FAK, Src and EGFR (12,13). CagM, along with CagX and CagT, forms an outer membrane-associated T4SS subcomplex (14). CagX and CagT interact directly, the C-terminal region of CagX is important for CagT interaction, and CagT depends on CagX for its stabilization (15). CagE is one of the energy providing components of CagA translocation. CagE is an inner membrane associated active NTPase and has multiple interacting partners including the inner membrane proteins CagV and Cagβ (16).

As reported, CagA also facilitates its translocation into host epithelial cells by T4SS-induced externalization of phosphatidylserine from inner leaflet of the plasma membrane. The protein binds to phosphatidylserine via Lys-Xn-Arg-X-Arg (K-Xn-R-X-R) motif present in the central region of CagA. The 2 arginine residues in K-Xn-R-X-R motif are highly conserved among CagA proteins derived from H. pylori strains (13,17). It has previously been reported that C-terminal CagA secretion signal and N-terminal CagA domain (D1) are crucial for efficient translocation (18). Collectively, these findings indicate that all components of this type IV secretion system, including the effector protein CagA, are encoded on the cag pathogenicity island.

Once the protein has entered these target cells, CagA localizes to the inner surface of the cellular membrane, once again by the interaction between the K-Xn-R-X-R motif with phosphatidylserine (13). Then parts of CagA proteins undergo tyrosine phosphorylation at the C-terminal Glu-Pro-Ile-Tyr-Ala (EPIYA) motifs by Src family kinases and Abl kinase, while other CagA molecules remain unphosphorylated (1921).

CagA can be tyrosine phosphorylated at EPIYA motifs, which is found as part of repetitive region in the C-terminal of CagA. Based on the amino acid sequences surrounding each of the EPIYA motifs, four distinct EPIYA motifs (EPIYA-A, -B, -C, -D) have been classified. EPIYA-A and EPIYA-B are present throughout the world, EPIYA-C is found in strains isolated from Western countries. In contrast, East Asian strains carry East Asian CagA, which contains EPIYA-D (22,23) (Fig. 1). Through database searching and in silico analysis, Zhang et al revealed a strong non-random distribution of the EPIYA-B motif polymorphisms (including EPIYT and EPIYA) in Western H. pylori isolates, and provide evidence that the EPIYT are significantly less associated with gastric cancer than the EPIYA. CagA B-motif phosphorylation status is essential for its interaction with host PI3-kinase during colonization and that CagA with an EPIYT B-motif had significantly attenuated induction of interleukin-8 and the hummingbird phenotype, had higher affinity with PI3-kinase, and enhanced induction of AKT compared to the EPIYA (24). It was reported that the two SH2 domains from SHP-2 (Src homology 2-containing protein tyrosine phosphatase-2) bind to highly related sequences pY-(S/T/A/V/I)-X-(V/I/L)-X-(W/F). Intriguingly, the consensus motif perfectly matches the SHP-2-binding site of EPIYA-D (pY-A-T-I-D-F). Furthermore, EPIYA-C (pY-A-T-I-D-D) replacement of the pY + 5 position in Western CagA, reduces the binding affinity to SHP-2. This East Asian-specific sequence conferred stronger SHP-2 binding and morphologically transforming activities to Western CagA. So the East Asian CagA or larger numbers of EPIYA-C in Western CagA was associated with atrophic gastritis and increased the risk of gastric cancer. Several recent reports have demonstrated that the ABCCC type can induce the intestinal metaplasia, IL-8, perturbation of Crk adaptor proteins, anti-apoptotic effect and carcinogenic effect more significantly than ABC type (2528) (Fig. 2).

CagA phosphorylation-dependent or independent effects

Perturb host cell functions by CagA phosphorylation-dependent effects

Once within the host cells, CagA undergoes tyrosine phosphorylation at EPIYA motifs by Src family kinases (SFKs) and Abl kinase. Along with the report that c-Src only phosphorylated EPIYA-C or EPIYA-D, whereas c-Abl phosphorylated all EPIYA motifs. CagA proteins were phosphorylated on 1 or 2 EPIYA motifs, but never simultaneously on 3 motifs. Furthermore, none of the phosphorylated EPIYA motifs alone was sufficient for deregulation of cell growth and motility. Western CagA EPIYA-A and EPIYA-C were preferred combination phosphorylation, either across two CagA molecules or simultaneously on one (21,29).

CagA proteins were probably from a dimer in host cell by a 16-amino-acid CagA multimerization (CM) sequence, which is located downstream of the EPIYA-C motif in C-terminal region (30). After phosphorylated CagA (pCagA) from a homodimer through the CM sequence, the pCagA dimer can bind with a single SHP-2 molecule via its two homologous SH2 domains (31). The pCagA-SHP-2 complex triggers the phosphatase activity of SHP-2, which in return causes the dephosphorylation of focal adhesion kinase (FAK) and activation of Ras/MAPK/ERK signaling pathway (3234).

In addition to SHP-2, the phosphorylated EPIYA-A, -B can specifically bind to the C-terminal Src kinase (Csk) and activate Csk, and then the inhibitory tyrosine residues of SFKs are phosphorylated by Csk. So Csk is characterized as a negative regulator of SFKs, which results in reduced EPIYA phosphorylation of CagA. Therefore, the CagA-Csk interaction may establish a negative feedback mechanism that prevents damage to the host cells from H. pylori. This could be harmful to the long-term colonization of H. pylori in stomach (35).

CagA can associate with Crk adaptor proteins (Crk-I, Crk-II, Crk-L) EPIYA-phosphorylation-dependently. CagA-Crk interaction plays a critical role in promoting cell scattering by inducing several downstream signaling pathways, such as SoS1/H-Ras-Raf-MEK and C3G-Rap1/B-Raf-MEK (36). The human β-defensins (hβDs) are antimicrobial peptides that are highly active against H. pylori during early infection via EGFR-dependent activation of MAP kinase and JAK/STAT signaling pathways. However, during prolonged infection, hβD1 and hβD3 is subsequently downregulated by phosphorylated CagA (37,38) (Fig. 3).

Disruption of epithelial cells by CagA phosphorylation-independent effects

CagA also exerts numerous effects within host cells in a tyrosine phosphorylation-independent manner. A CRPIA (conserved repeat responsible for phosphorylation-independent activity) motif, FPLKRHDKVDDLSKVG, in the C-terminal region of CagA, which is distinct from the EPIYA motifs used for phosphorylation. The CRPIA motif in non-phosphorylated CagA was involved in interacting with the hepatocyte growth factor scatter factor receptor c-Met, which is involved in invasive growth of tumor cells. CagA binds c-Met and could represent an adaptor protein, which associates with phospholipase Cγ (PLCγ) and activates phosphatidylinositol 3-kinase/Akt signaling. This in turn led to the activation of β-catenin and NF-κB signaling, which promotes proliferation and inflammation (39,40).

CagA can interact with Grb2, which results in the activation of the Ras/MEK/ERK pathway and leads to cell scattering as well as proliferation. This ability of CagA is independent from the tyrosine phosphorylation. However, the EPIYA sequences are indispensable for the Grb2 binding and induction of the cellular responses (41).

Moreover, CagA associates with the epithelial tight-junction scaffolding protein ZO-1 and the transmembrane protein JAM (junctional adhesion molecule), causing an ectopic assembly of tight-junction components at sites of bacterial attachment, and altering the composition and function of the apical-junctional complex (42).

CagA physically interacts with E-cadherin independently of EPIYA motif phosphorylation. The CagA/E-cadherin interaction impairs the E-cadherin/β-catenin complex, causing cytoplasmic and nuclear accumulation of β-catenin (43). Then it leads to nuclear translocation of free β-catenin, where it binds to the transcriptional cofactors of the Wnt pathway and upregulates the transcription of targeted genes such as axin, cyclin and myc (44). H. pylori alters the E-cadherin/β-catenin complex, leading to formation of a multiproteic complex composed of CagA, c-Met, E-cadherin, and p120-catenin. This complex abrogates c-Met and p120-catenin tyrosine phosphorylation and suppresses the cell-invasive phenotype induced by H. pylori (45). CagA deregulation of β-catenin requires residues 1,009–1,086 and residues 908–1,012 of Western CagA and East Asian CagA, respectively, and is mediated by the CM motif of CagA (46).

CagA also disrupts the tight junction and causes loss of apical-basolateral polarity by interaction with PAR1/MARK kinase, which is a central regulator of cell polarity. Association of CagA inhibits PAR1 kinase activity and prevents atypical protein kinase C (aPKC)-mediated PAR1 phosphorylation. The PAR-aPKC system is the molecular machinery that converts initial polarity cues in the establishment of complementary membrane domains along the polarity axis. CagA-PAR1 complex dissociates PAR1 from the membrane, collectively causing junctional and polarity defects. The PAR1 also promotes CagA multimerization, which stabilizes the CagA-SHP2 interaction (4749). This interaction is also dependent on the CM motif of CagA. The CM sequence of CagA isolated from East Asian H. pylori binds PAR1b more strongly than that of CagA isolated from Western H. pylori. Within Western CagA species, the ability to bind PAR1b is proportional to the number of CM sequences (50).

Lu et al found that CagA binds not only PAR1b but also other PAR1 isoforms, with order of strength as follows: PAR1b > PAR1d > or = PAR1a > PAR1c. They also indicate that malfunctioning of microtubules and myosin II by CagA-mediated PAR1 inhibition cooperates with deregulated SHP-2 in the morphogenetic activity of CagA (51). In MDCK tissue culture model, association of CagA with MARK2 not only causes disruption of apical junctions, but also inhibition of tubulogenesis and cell differentiation (52). Furthermore inhibition of PAR1b kinase activity contributed to an increased hummingbird phenotype. CagA-mediated inhibition of PAR1b and then prevented PAR1b mediated phosphorylation, which inactivates a RhoA-specific GEF, GEF-H1 and thereby strengthens the hummingbird phenotype induced by CagA-stimulated SHP2 (53). Moreover, the interaction of CagA with PAR1 is associated with reduction of an inhibitor NF-κB, called IκB kinase, which regulates microtubule stability by phosphorylating microtubule-associated proteins (MAPs). Since microtubule destabilization leads to the activation of NF-κB by promoting IκBα degradation, impairment of the microtubule system by CagA-PAR1 interaction may give a cytoskeletal cue that stimulates IκB degradation (54). Additionally, CagA was described to affect activity of protein kinase C-related kinase 2 (PRK2), which acts downstream of Rho GTPases and is known to affect cytoskeletal rearrangements and cell polarity (55) (Fig. 3).

CagA destroys the host cells via epigenetic modifications

Epigenetics may be defined as the mechanisms that initiate and maintain heritable patterns of gene function and regulation in a heritable manner without affecting the sequence of the genome. Epigenetic modifications include DNA methylation, post-translational modifications of histone proteins, microRNA expression, genomic imprinting and chromatin remodeling (56,57) (Fig. 4).

DNA methylation

DNA methylation is an important epigenetic modification involved in the regulation of numerous biological processes. In mammals, DNA methylation mainly occurs in the context of cytosine-phosphate-guanine (CpG) dinucleotides (58). H. pylori infection potently induces methylation of CpG islands (CGIs) to various degrees. Methylation levels of specific CGIs seemed to reflect gastric cancer risk in H. pylori-negative individuals (OR 95% CI 2.2–32). The promoter CpG islands of FLNc, HAND1, THBD, p41ARC, HRASLS and LOX gene were reportedly altered by H. pylori infection (5962). In vitro experiments showed significant CagA aberrant epigenetic silencing of let-7 expression leading to Ras upregulation by histone and DNA methylation (63). A significantly increased risk of RUNX3 methylation (OR, 4.28; 95% CI, 1.19–15.49) was observed with a high consumption of nuts in patients with CagA-positive H. pylori infection (64).

MicroRNAs

miRNAs are small, non-coding RNAs, which regulate gene expression in a sequence-specific manner. miRNAs have been implicated in the etiology, progression and prognosis of diseases, and many studies have shown that profiles of miRNA expression differ between infection and non-infection with H. pylori (65,66). miRNAs are involved in H. pylori-related pathology via the regulation of the transcription and expression of various genes, playing an important role in inflammation, cell proliferation, apoptosis and differentiation (67,68).

CagA may be involved in cellular regulation of certain miRNAs in the gastric epithelium. miRNA expression patterns in H. pylori-infected gastric mucosa are determined by microarray. The results found that expression levels of let-7 family miRNAs are significantly altered following infection with CagA positive strain (69). CagA enhanced c-myc, DNA methyltransferase 3B (DNMT3B) and enhancer of zeste homologue 2 (EZH2) expression and attenuated miR-26a and miR-101 expression, which resulted in the attenuation of let-7 expression (63). Using mammalian miRNA profile microarrays, miR-1290 and miR-584 expressions are upregulated in CagA-transformed cells. miR-1290 is upregulated in an Erk1/2-dependent manner, and miR-584 is activated by NF-κB. Foxa1 is an important target of miR-584 and miR-1290, which promote the epithelial-mesenchymal transition significantly (70). In vitro, CagA inhibited miR-370 expression, which led to overexpression of FoxM1. The upregulated FoxM1 expression altered the expression of p27 (Kip1), and promoted proliferation in gastric cells (71). CagA may function as an initiator in the process of carcinogenesis by upregulating miR-222, which further participates in the progression of cancer by promoting proliferation and inhibiting RECK (reversion-inducing cysteine-rich protein with Kazal motifs) (72). Shortly after H. pylori infection, miR-372 and miR-373 synthesis is highly inhibited, leading to the post-transcriptional release of LATS2 expression and thus, to a cell cycle arrest at the G1/S transition. This downregulation of a specific cell cycle-regulating microRNA is dependent on the translocation of CagA into the host cells (73). The integrative analysis and immunohistochemistry staining validation indicated that miR-155 and miR-146b are upregulated in H. pylori-positive gastroduodenal ulcer. Further experiments in gastric epithelial cells revealed that CagA mediated upregulation of miR-155 and miR-146b, which decreases IL6 overexpression (74).

Histone modifications

Nucleosomes are represented by DNA wrapped around eight histone proteins, H2A, H2B, H3, and H4. Histones are primary protein components of eukaryotic chromatin and play a role in gene regulation. A histone modification is a covalent post-translational modification (PTM) to histone proteins which include methylation, phosphorylation, acetylation, ubiquitylation, and sumoylation (75,76).

Substantial research has investigated the effects of H. pylori infection on histone modification. Chromatin immunoprecipitation analysis of NCI-N87 and primary gastric cells revealed that H. pylori induce cell cycle control factor p21WAF1 overexpression. H. pylori is associated with hyperacetylation of histone H4 which can release HDAC-1 from the p21WAF1 promoter (77). cagPAI-dependent decreases of H3 phosphorylation levels at serine 10 (pH3Ser10) and threonine 3 (pH3Thr3) are observed. H. pylori causes a strong decrease of the cell division cycle 25 (CDC25C) phosphatase (78). Liang et al demonstrated that RBP2, a newly identified H3K4 demethylase, can be induced by CagA via PI3K/AKT-Sp1 pathway depending on AKT phosphorylation. Furthermore, the novel CagA-PI3K/AKT-Sp1-RBP2-Cyclin D1 pathway links chronic inflammation to tumor during GC development (79).

In conclusion, the CagA protein is encoded by cag PAI and delivered into the host cells during the T4SS system. Following translocation, Src and Abl kinases phosphorylate CagA on EPIYA motifs. Phosphorylated CagA can interact with SHP2, Csk, Crk and hβD, which trigger several intracellular signaling pathways resulting in epithelial cell gene expression. The unphosphorylated CagA directly interacts with certain intracellular proteins such as PARIb, E-cadherin/β-catenin, c-Met, Grb2 and ZO-1, then disrupts the cell-to-cell junctions and gastric epithelial cell polarity.

References

1 

Pacchiani N, Censini S, Buti L and Covacci A: Echoes of a distant past: The cag pathogenicity island of Helicobacter pylori. Cold Spring Harb Perspect Med. 3:a0103552013. View Article : Google Scholar : PubMed/NCBI

2 

Cavaleiro-Pinto M, Peleteiro B, Lunet N and Barros H: Helicobacter pylori infection and gastric cardia cancer: Systematic review and meta-analysis. Cancer Causes Control. 22:375–387. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Akopyants NS, Clifton SW, Kersulyte D, Crabtree JE, Youree BE, Reece CA, Bukanov NO, Drazek ES, Roe BA and Berg DE: Analyses of the cag pathogenicity island of Helicobacter pylori. Mol Microbiol. 28:37–53. 1998. View Article : Google Scholar : PubMed/NCBI

4 

Censini S, Lange C, Xiang Z, Crabtree JE, Ghiara P, Borodovsky M, Rappuoli R and Covacci A: cag, a pathogenicity island of Helicobacter pylori, encodes type I-specific and disease-associated virulence factors. Proc Natl Acad Sci USA. 93:14648–14653. 1996. View Article : Google Scholar : PubMed/NCBI

5 

Yamaoka Y: Helicobacter pylori typing as a tool for tracking human migration. Clin Microbiol Infect. 15:829–834. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Lai CH, Perng CL, Lan KH and Lin HJ: Association of IS605 and cag-PAI of Helicobacter pylori isolated from patients with gastrointestinal diseases in Taiwan. Gastroenterol Res Pract. 2013:3562172013. View Article : Google Scholar : PubMed/NCBI

7 

Torres J, Perez-Perez GI, Leal-Herrera Y and Munoz O: Infection with CagA+ Helicobacter pylori strains as a possible predictor of risk in the development of gastric adenocarcinoma in Mexico. Int J Cancer. 78:298–300. 1998. View Article : Google Scholar : PubMed/NCBI

8 

Batista SA, Rocha GA, Rocha AM, Saraiva IE, Cabral MM, Oliveira RC and Queiroz DM: Higher number of Helicobacter pylori CagA EPIYA C phosphorylation sites increases the risk of gastric cancer, but not duodenal ulcer. BMC Microbiol. 11:612011. View Article : Google Scholar : PubMed/NCBI

9 

Franco AT, Johnston E, Krishna U, Yamaoka Y, Israel DA, Nagy TA, Wroblewski LE, Piazuelo MB, Correa P and Peek RM Jr: Regulation of gastric carcinogenesis by Helicobacter pylori virulence factors. Cancer Res. 68:379–387. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Ohnishi N, Yuasa H, Tanaka S, Sawa H, Miura M, Matsui A, Higashi H, Musashi M, Iwabuchi K, Suzuki M, et al: Transgenic expression of Helicobacter pylori CagA induces gastrointestinal and hematopoietic neoplasms in mouse. Proc Natl Acad Sci USA. 105:1003–1008. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Backert S, Tegtmeyer N and Fischer W: Composition, structure and function of the Helicobacter pylori cag pathogenicity island encoded type IV secretion system. Future Microbiol. 10:955–965. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Tegtmeyer N, Hartig R, Delahay RM, Rohde M, Brandt S, Conradi J, Takahashi S, Smolka AJ, Sewald N and Backert S: A small fibronectin-mimicking protein from bacteria induces cell spreading and focal adhesion formation. J Biol Chem. 285:23515–23526. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Murata-Kamiya N, Kikuchi K, Hayashi T, Higashi H and Hatakeyama M: Helicobacter pylori exploits host membrane phosphatidylserine for delivery, localization, and pathophysiological action of the CagA oncoprotein. Cell Host Microbe. 7:399–411. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Fischer W: Assembly and molecular mode of action of the Helicobacter pylori Cag type IV secretion apparatus. FEBS J. 278:1203–1212. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Gopal GJ, Pal J, Kumar A and Mukhopadhyay G: C-terminal domain of CagX is responsible for its interaction with CagT protein of Helicobacter pylori type IV secretion system. Biochem Biophys Res Commun. 456:98–103. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Shariq M, Kumar N, Kumari R, Kumar A, Subbarao N and Mukhopadhyay G: Biochemical analysis of CagE: A VirB4 homologue of Helicobacter pylori Cag-T4SS. PLoS One. 10:e01426062015. View Article : Google Scholar : PubMed/NCBI

17 

Odenbreit S, Püls J, Sedlmaier B, Gerland E, Fischer W and Haas R: Translocation of Helicobacter pylori CagA into gastric epithelial cells by type IV secretion. Science. 287:1497–1500. 2000. View Article : Google Scholar : PubMed/NCBI

18 

Schindele F, Weiss E, Haas R and Fischer W: Quantitative analysis of CagA type IV secretion by Helicobacter pylori reveals substrate recognition and translocation requirements. Mol Microbiol. 100:188–203. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Stein M, Bagnoli F, Halenbeck R, Rappuoli R, Fantl WJ and Covacci A: c-Src/Lyn kinases activate Helicobacter pylori CagA through tyrosine phosphorylation of the EPIYA motifs. Mol Microbiol. 43:971–980. 2002. View Article : Google Scholar : PubMed/NCBI

20 

Tegtmeyer N and Backert S: Role of Abl and Src family kinases in actin-cytoskeletal rearrangements induced by the Helicobacter pylori CagA protein. Eur J Cell Biol. 90:880–890. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Tammer I, Brandt S, Hartig R, König W and Backert S: Activation of Abl by Helicobacter pylori: A novel kinase for CagA and crucial mediator of host cell scattering. Gastroenterology. 132:1309–1319. 2007. View Article : Google Scholar : PubMed/NCBI

22 

Zhu Y-L, Zheng S, Du Q, Qian K-D and Fang P-C: Characterization of CagA variable region of Helicobacter pylori isolates from Chinese patients. World J Gastroenterol. 11:880–884. 2005. View Article : Google Scholar : PubMed/NCBI

23 

Vaziri F, Peerayeh SN, Alebouyeh M, Maghsoudi N, Azimzadeh P, Siadat SD and Zali MR: Novel effects of Helicobacter pylori CagA on key genes of gastric cancer signal transduction: A comparative transfection study. Pathog Dis. 73:ftu0212015. View Article : Google Scholar : PubMed/NCBI

24 

Zhang XS, Tegtmeyer N, Traube L, Jindal S, Perez-Perez G, Sticht H, Backert S and Blaser MJ: A specific A/T polymorphism in Western tyrosine phosphorylation B-motifs regulates Helicobacter pylori CagA epithelial cell interactions. PLoS Pathog. 11:e10046212015. View Article : Google Scholar : PubMed/NCBI

25 

Ferreira RM, Machado JC, Leite M, Carneiro F and Figueiredo C: The number of Helicobacter pylori CagA EPIYA C tyrosine phosphorylation motifs influences the pattern of gastritis and the development of gastric carcinoma. Histopathology. 60:992–998. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Higashi H, Tsutsumi R, Fujita A, Yamazaki S, Asaka M, Azuma T and Hatakeyama M: Biological activity of the Helicobacter pylori virulence factor CagA is determined by variation in the tyrosine phosphorylation sites. Proc Natl Acad Sci USA. 99:14428–14433. 2002. View Article : Google Scholar : PubMed/NCBI

27 

Monstein H-J, Karlsson A, Ryberg A and Borch K: Application of PCR amplicon sequencing using a single primer pair in PCR amplification to assess variations in Helicobacter pylori CagA EPIYA tyrosine phosphorylation motifs. BMC Res Notes. 3:352010. View Article : Google Scholar : PubMed/NCBI

28 

De Souza D, Fabri LJ, Nash A, Hilton DJ, Nicola NA and Baca M: SH2 domains from suppressor of cytokine signaling-3 and protein tyrosine phosphatase SHP-2 have similar binding specificities. Biochemistry. 41:9229–9236. 2002. View Article : Google Scholar : PubMed/NCBI

29 

Mueller D, Tegtmeyer N, Brandt S, Yamaoka Y, De Poire E, Sgouras D, Wessler S, Torres J, Smolka A and Backert S: c-Src and c-Abl kinases control hierarchic phosphorylation and function of the CagA effector protein in Western and East Asian Helicobacter pylori strains. J Clin Invest. 122:1553–1566. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Ren S, Higashi H, Lu H, Azuma T and Hatakeyama M: Structural basis and functional consequence of Helicobacter pylori CagA multimerization in cells. J Biol Chem. 281:32344–32352. 2006. View Article : Google Scholar : PubMed/NCBI

31 

Yamazaki S, Yamakawa A, Ito Y, Ohtani M, Higashi H, Hatakeyama M and Azuma T: The CagA protein of Helicobacter pylori is translocated into epithelial cells and binds to SHP-2 in human gastric mucosa. J Infect Dis. 187:334–337. 2003. View Article : Google Scholar : PubMed/NCBI

32 

Montagner A, Yart A, Dance M, Perret B, Salles JP and Raynal P: A novel role for Gab1 and SHP2 in epidermal growth factor-induced Ras activation. J Biol Chem. 280:5350–5360. 2005. View Article : Google Scholar : PubMed/NCBI

33 

Higashi H, Nakaya A, Tsutsumi R, Yokoyama K, Fujii Y, Ishikawa S, Higuchi M, Takahashi A, Kurashima Y, Teishikata Y, et al: Helicobacter pylori CagA induces Ras-independent morphogenetic response through SHP-2 recruitment and activation. J Biol Chem. 279:17205–17216. 2004. View Article : Google Scholar : PubMed/NCBI

34 

Tsutsumi R, Takahashi A, Azuma T, Higashi H and Hatakeyama M: Focal adhesion kinase is a substrate and downstream effector of SHP-2 complexed with Helicobacter pylori CagA. Mol Cell Biol. 26:261–276. 2006. View Article : Google Scholar : PubMed/NCBI

35 

Tsutsumi R, Higashi H, Higuchi M, Okada M and Hatakeyama M: Attenuation of Helicobacter pylori CagA × SHP-2 signaling by interaction between CagA and C-terminal Src kinase. J Biol Chem. 278:3664–3670. 2003. View Article : Google Scholar : PubMed/NCBI

36 

Suzuki M, Mimuro H, Suzuki T, Park M, Yamamoto T and Sasakawa C: Interaction of CagA with Crk plays an important role in Helicobacter pylori-induced loss of gastric epithelial cell adhesion. J Exp Med. 202:1235–1247. 2005. View Article : Google Scholar : PubMed/NCBI

37 

Bauer B, Pang E, Holland C, Kessler M, Bartfeld S and Meyer TF: The Helicobacter pylori virulence effector CagA abrogates human β-defensin 3 expression via inactivation of EGFR signaling. Cell Host Microbe. 11:576–586. 2012. View Article : Google Scholar : PubMed/NCBI

38 

Patel SR, Smith K, Letley DP, Cook KW, Memon AA, Ingram RJ, Staples E, Backert S, Zaitoun AM, Atherton JC, et al: Helicobacter pylori downregulates expression of human β-defensin 1 in the gastric mucosa in a type IV secretion-dependent fashion. Cell Microbiol. 15:2080–2092. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Churin Y, Al-Ghoul L, Kepp O, Meyer TF, Birchmeier W and Naumann M: Helicobacter pylori CagA protein targets the c-Met receptor and enhances the motogenic response. J Cell Biol. 161:249–255. 2003. View Article : Google Scholar : PubMed/NCBI

40 

Suzuki M, Mimuro H, Kiga K, Fukumatsu M, Ishijima N, Morikawa H, Nagai S, Koyasu S, Gilman RH, Kersulyte D, et al: Helicobacter pylori CagA phosphorylation-independent function in epithelial proliferation and inflammation. Cell Host Microbe. 5:23–34. 2009. View Article : Google Scholar : PubMed/NCBI

41 

Mimuro H, Suzuki T, Tanaka J, Asahi M, Haas R and Sasakawa C: Grb2 is a key mediator of Helicobacter pylori CagA protein activities. Mol Cell. 10:745–755. 2002. View Article : Google Scholar : PubMed/NCBI

42 

Amieva MR, Vogelmann R, Covacci A, Tompkins LS, Nelson WJ and Falkow S: Disruption of the epithelial apical-junctional complex by Helicobacter pylori CagA. Science. 300:1430–1434. 2003. View Article : Google Scholar : PubMed/NCBI

43 

Murata-Kamiya N, Kurashima Y, Teishikata Y, Yamahashi Y, Saito Y, Higashi H, Aburatani H, Akiyama T, Peek RM Jr, Azuma T, et al: Helicobacter pylori CagA interacts with E-cadherin and deregulates the β-catenin signal that promotes intestinal transdifferentiation in gastric epithelial cells. Oncogene. 26:4617–4626. 2007. View Article : Google Scholar : PubMed/NCBI

44 

Neal JT, Peterson TS, Kent ML and Guillemin K: H. pylori virulence factor CagA increases intestinal cell proliferation by Wnt pathway activation in a transgenic zebrafish model. Dis Model Mech. 6:802–810. 2013. View Article : Google Scholar : PubMed/NCBI

45 

Oliveira MJ, Costa AM, Costa AC, Ferreira RM, Sampaio P, Machado JC, Seruca R, Mareel M and Figueiredo C: CagA associates with c-Met, E-cadherin, and p120-catenin in a multiproteic complex that suppresses Helicobacter pylori-induced cell-invasive phenotype. J Infect Dis. 200:745–755. 2009. View Article : Google Scholar : PubMed/NCBI

46 

Kurashima Y, Murata-Kamiya N, Kikuchi K, Higashi H, Azuma T, Kondo S and Hatakeyama M: Deregulation of beta-catenin signal by Helicobacter pylori CagA requires the CagA-multimerization sequence. Int J Cancer. 122:823–831. 2008. View Article : Google Scholar : PubMed/NCBI

47 

Saadat I, Higashi H, Obuse C, Umeda M, Murata-Kamiya N, Saito Y, Lu H, Ohnishi N, Azuma T, Suzuki A, et al: Helicobacter pylori CagA targets PAR1/MARK kinase to disrupt epithelial cell polarity. Nature. 447:330–333. 2007. View Article : Google Scholar : PubMed/NCBI

48 

Goldstein B and Macara IG: The PAR proteins: Fundamental players in animal cell polarization. Dev Cell. 13:609–622. 2007. View Article : Google Scholar : PubMed/NCBI

49 

Suzuki A and Ohno S: The PAR-aPKC system: Lessons in polarity. J Cell Sci. 119:979–987. 2006. View Article : Google Scholar : PubMed/NCBI

50 

Lu HS, Saito Y, Umeda M, Murata-Kamiya N, Zhang HM, Higashi H and Hatakeyama M: Structural and functional diversity in the PAR1b/MARK2-binding region of Helicobacter pylori CagA. Cancer Sci. 99:2004–2011. 2008.PubMed/NCBI

51 

Lu H, Murata-Kamiya N, Saito Y and Hatakeyama M: Role of partitioning-defective 1/microtubule affinity-regulating kinases in the morphogenetic activity of Helicobacter pylori CagA. J Biol Chem. 284:23024–23036. 2009. View Article : Google Scholar : PubMed/NCBI

52 

Zeaiter Z, Cohen D, Müsch A, Bagnoli F, Covacci A and Stein M: Analysis of detergent-resistant membranes of Helicobacter pylori infected gastric adenocarcinoma cells reveals a role for MARK2/Par1b in CagA-mediated disruption of cellular polarity. Cell Microbiol. 10:781–794. 2008. View Article : Google Scholar : PubMed/NCBI

53 

Yamahashi Y and Hatakeyama M: PAR1b takes the stage in the morphogenetic and motogenetic activity of Helicobacter pylori CagA oncoprotein. Cell Adhes Migr. 7:11–18. 2013. View Article : Google Scholar

54 

Suzuki N, Murata-Kamiya N, Yanagiya K, Suda W, Hattori M, Kanda H, Bingo A, Fujii Y, Maeda S, Koike K, et al: Mutual reinforcement of inflammation and carcinogenesis by the Helicobacter pylori CagA oncoprotein. Sci Rep. 5:100242015. View Article : Google Scholar : PubMed/NCBI

55 

Mishra JP, Cohen D, Zamperone A, Nesic D, Muesch A and Stein M: CagA of Helicobacter pylori interacts with and inhibits the serine-threonine kinase PRK2. Cell Microbiol. 17:1670–1682. 2015. View Article : Google Scholar : PubMed/NCBI

56 

Sandoval J and Esteller M: Cancer epigenomics: Beyond genomics. Curr Opin Genet Dev. 22:50–55. 2012. View Article : Google Scholar : PubMed/NCBI

57 

Verma M: The role of epigenomics in the study of cancer biomarkers and in the development of diagnostic tools. Adv Exp Med Biol. 867:59–80. 2015. View Article : Google Scholar : PubMed/NCBI

58 

Hernando-Herraez I, Garcia-Perez R, Sharp AJ and Marques-Bonet T: DNA methylation: Insights into human evolution. PLoS Genet. 11:e10056612015. View Article : Google Scholar : PubMed/NCBI

59 

Maekita T, Nakazawa K, Mihara M, Nakajima T, Yanaoka K, Iguchi M, Arii K, Kaneda A, Tsukamoto T, Tatematsu M, et al: High levels of aberrant DNA methylation in Helicobacter pylori-infected gastric mucosae and its possible association with gastric cancer risk. Clin Cancer Res. 12:989–995. 2006. View Article : Google Scholar : PubMed/NCBI

60 

Yoshida T, Kato J, Maekita T, Yamashita S, Enomoto S, Ando T, Niwa T, Deguchi H, Ueda K, Inoue I, et al: Altered mucosal DNA methylation in parallel with highly active Helicobacter pylori-related gastritis. Gastric Cancer. 16:488–497. 2013. View Article : Google Scholar : PubMed/NCBI

61 

Tomita H, Takaishi S, Menheniott TR, Yang X, Shibata W, Jin G, Betz KS, Kawakami K, Minamoto T and Tomasetto C: Inhibition of gastric carcinogenesis by the hormone gastrin is mediated by suppression of TFF1 epigenetic silencing. Gastroenterology. 140:879–891. 2011. View Article : Google Scholar : PubMed/NCBI

62 

Guo XB, Guo L, Zhi QM, Ji J, Jiang JL, Zhang RJ, Zhang JN, Zhang J, Chen XH, Cai Q, et al: Helicobacter pylori induces promoter hypermethylation and downregulates gene expression of IRX1 transcription factor on human gastric mucosa. J Gastroenterol Hepatol. 26:1685–1690. 2011. View Article : Google Scholar : PubMed/NCBI

63 

Hayashi Y, Tsujii M, Wang J, Kondo J, Akasaka T, Jin Y, Li W, Nakamura T, Nishida T, Iijima H, et al: CagA mediates epigenetic regulation to attenuate let-7 expression in Helicobacter pylori-related carcinogenesis. Gut. 62:1536–1546. 2013. View Article : Google Scholar : PubMed/NCBI

64 

Zhang YW, Eom SY, Yim DH, Song YJ, Yun HY, Park JS, Youn SJ, Kim BS, Kim YD and Kim H: Evaluation of the relationship between dietary factors, CagA-positive Helicobacter pylori infection, and RUNX3 promoter hypermethylation in gastric cancer tissue. World J Gastroenterol. 19:1778–1787. 2013. View Article : Google Scholar : PubMed/NCBI

65 

He L and Hannon GJ: MicroRNAs: Small RNAs with a big role in gene regulation. Nat Rev Genet. 5:522–531. 2004. View Article : Google Scholar : PubMed/NCBI

66 

Xiao C and Rajewsky K: MicroRNA control in the immune system: Basic principles. Cell. 136:26–36. 2009. View Article : Google Scholar : PubMed/NCBI

67 

Libânio D, Dinis-Ribeiro M and Pimentel-Nunes P: Helicobacter pylori and microRNAs: Relation with innate immunity and progression of preneoplastic conditions. World J Clin Oncol. 6:111–132. 2015. View Article : Google Scholar : PubMed/NCBI

68 

Noto JM and Peek RM: The role of microRNAs in Helicobacter pylori pathogenesis and gastric carcinogenesis. Front Cell Infect Microbiol. 1:212012. View Article : Google Scholar : PubMed/NCBI

69 

Matsushima K, Isomoto H, Inoue N, Nakayama T, Hayashi T, Nakayama M, Nakao K, Hirayama T and Kohno S: MicroRNA signatures in Helicobacter pylori-infected gastric mucosa. Int J Cancer. 128:361–370. 2011. View Article : Google Scholar : PubMed/NCBI

70 

Zhu Y, Jiang Q, Lou X, Ji X, Wen Z, Wu J, Tao H, Jiang T, He W, Wang C, et al: MicroRNAs up-regulated by CagA of Helicobacter pylori induce intestinal metaplasia of gastric epithelial cells. PLoS One. 7:e351472012. View Article : Google Scholar : PubMed/NCBI

71 

Feng Y, Wang L, Zeng J, Shen L, Liang X, Yu H, Liu S, Liu Z, Sun Y, Li W, et al: FoxM1 is overexpressed in Helicobacter pylori-induced gastric carcinogenesis and is negatively regulated by miR-370. Mol Cancer Res. 11:834–844. 2013. View Article : Google Scholar : PubMed/NCBI

72 

Li N, Tang B, Zhu ED, Li BS, Zhuang Y, Yu S, Lu DS, Zou QM, Xiao B and Mao XH: Increased miR-222 in H. pylori-associated gastric cancer correlated with tumor progression by promoting cancer cell proliferation and targeting RECK. FEBS Lett. 586:722–728. 2012. View Article : Google Scholar : PubMed/NCBI

73 

Belair C, Baud J, Chabas S, Sharma CM, Vogel J, Staedel C and Darfeuille F: Helicobacter pylori interferes with an embryonic stem cell micro RNA cluster to block cell cycle progression. Silence. 2:72011. View Article : Google Scholar : PubMed/NCBI

74 

Cheng SF, Li L and Wang LM: miR-155 and miR-146b negatively regulates IL6 in Helicobacter pylori (cagA+) infected gastroduodenal ulcer. Eur Rev Med Pharmacol Sci. 19:607–613. 2015.PubMed/NCBI

75 

Harr JC, Gonzalez-Sandoval A and Gasser SM: Histones and histone modifications in perinuclear chromatin anchoring: From yeast to man. EMBO Rep. 17:139–155. 2016. View Article : Google Scholar : PubMed/NCBI

76 

Schones DE, Cui K, Cuddapah S, Roh TY, Barski A, Wang Z, Wei G and Zhao K: Dynamic regulation of nucleosome positioning in the human genome. Cell. 132:887–898. 2008. View Article : Google Scholar : PubMed/NCBI

77 

Xia G, Schneider-Stock R, Diestel A, Habold C, Krueger S, Roessner A, Naumann M and Lendeckel U: Helicobacter pylori regulates p21 (WAF1) by histone H4 acetylation. Biochem Biophys Res Commun. 369:526–531. 2008. View Article : Google Scholar : PubMed/NCBI

78 

Fehri LF, Rechner C, Janssen S, Mak TN, Holland C, Bartfeld S, Brüggemann H and Meyer TF: Helicobacter pylori-induced modification of the histone H3 phosphorylation status in gastric epithelial cells reflects its impact on cell cycle regulation. Epigenetics. 4:577–586. 2009. View Article : Google Scholar : PubMed/NCBI

79 

Liang X, Zeng J, Wang L, Shen L, Li S, Ma L, Ci X, Yu J, Jia M, Sun Y, et al: Histone demethylase RBP2 induced by Helicobactor pylori CagA participates in the malignant transformation of gastric epithelial cells. Oncotarget. 5:5798–5807. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2016
Volume 36 Issue 6

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen S, Zhang R and Duan G: Pathogenic mechanisms of the oncoprotein CagA in H. pylori-induced gastric cancer (Review). Oncol Rep 36: 3087-3094, 2016
APA
Chen, S., Zhang, R., & Duan, G. (2016). Pathogenic mechanisms of the oncoprotein CagA in H. pylori-induced gastric cancer (Review). Oncology Reports, 36, 3087-3094. https://doi.org/10.3892/or.2016.5145
MLA
Chen, S., Zhang, R., Duan, G."Pathogenic mechanisms of the oncoprotein CagA in H. pylori-induced gastric cancer (Review)". Oncology Reports 36.6 (2016): 3087-3094.
Chicago
Chen, S., Zhang, R., Duan, G."Pathogenic mechanisms of the oncoprotein CagA in H. pylori-induced gastric cancer (Review)". Oncology Reports 36, no. 6 (2016): 3087-3094. https://doi.org/10.3892/or.2016.5145