ABSTRACT
Gastric cancer is the fifth most diagnosed cancer in the world. Infection by the bacteria Helicobacter pylori (HP) is associated with approximately 75% of gastric cancer cases. HP infection induces chronic gastric inflammation, damaging the stomach and fostering carcinogenesis. Most mechanistic studies on gastric cancer initiation are performed in mice and utilize either mouse-adapted strains of HP or the natural mouse pathogen Helicobacter felis (HF). Here, we identified the differences in gastric inflammation, atrophy, and metaplasia associated with HP and HF infection in mice. PMSS1 HP strain or the CS1 HF strain were co-cultured with mouse peritoneal macrophages to assess their immunostimulatory effects. HP and HF induced similar cytokine production from cultured mouse peritoneal macrophages revealing that both bacteria exhibit similar immunostimulatory effects in vitro. Next, C57BL/6J mice were infected with HP or HF and were assessed 2 months post-infection. HP-infected mice caused modest inflammation within both the gastric corpus and antrum, and did not induce significant atrophy within the gastric corpus. In contrast, HF induced significant inflammation throughout the gastric corpus and antrum. Moreover, HF infection was associated with significant atrophy of the chief and parietal cell compartments and induced the expression of pyloric metaplasia (PM) markers. HP is poorly immunogenic compared to HF. HF induces dramatic CD4+ T cell activation, which is associated with increased gastric cancer risk in humans. Thus, HP studies in mice are better suited for studies on colonization, while HF is more strongly suited for studies on the effects of gastric inflammation on tumorigenesis.
IMPORTANCE
Mouse infection models with Helicobacter species are widely used to study Helicobacter pathogenesis and gastric cancer initiation. However, Helicobacter pylori is not a natural mouse pathogen, and mouse-adapted H. pylori strains are poorly immunogenic. In contrast, Helicobacter felis is a natural mouse pathogen that induces robust gastric inflammation and is often used in mice to investigate gastric cancer initiation. Although both bacterial strains are widely used, their disease pathogenesis in mice differs dramatically. However, few studies have directly compared the pathogenesis of these bacterial species in mice, and the contrasting features of these two models are not clearly defined. This study directly compares the gastric inflammation, atrophy, and metaplasia development triggered by the widely used PMSS1 H. pylori and CS1 H. felis strains in mice. It serves as a useful resource for researchers to select the experimental model best suited for their studies.
KEYWORDS: Helicobacter pylori, Helicobacter felis, gastric cancer, CagA, stomach, inflammation
INTRODUCTION
Gastric cancer is the fifth most common cancer and the fourth leading cause of cancer deaths worldwide (1). Helicobacter pylori (HP) infection is the primary risk factor for gastric cancer, contributing to 75% of the global gastric cancer burden (2). HP infection is extremely prevalent, infecting approximately 50% of the world’s population and 1%–3% of infected individuals will develop cancer (3). Stomach infection by HP induces a well-established histopathological cascade, driving chronic inflammation of the gastric mucosa, which in turn causes widespread epithelial damage and metaplasia, eventually resulting in adenocarcinoma (2, 4). Despite the strong link between HP and gastric cancer, chronic inflammation is both necessary and sufficient for gastric cancer initiation. A host of previous studies have shown that Helicobacter-independent chronic gastric inflammation induced by the overexpression of proinflammatory cytokines, surgical depletion of anti-inflammatory hormones, or through autoimmune gastritis induces gastric atrophy, metaplasia, and tumorigenesis (5–9). Similarly, polymorphisms resulting in higher levels of the proinflammatory cytokines IL1B or TNF and lower levels of the anti-inflammatory cytokine IL10 are associated with increases in HP-related gastric cancer (10, 11). In contrast, multiple immune-deficient mouse models are protected from the gastric atrophy associated with Helicobacter infection (12–14). Although it is well established that chronic inflammation modifies gastric cancer risk, the inflammatory phenotypes that promote cancer initiation remain poorly defined.
The vast majority of mechanistic studies are performed in mouse models. However, studies in mice pose specific challenges. HP is not a natural mouse pathogen, and clinical isolates typically require serial passage through mice to enhance mouse colonization (15, 16). Mouse-adapted HP strains, such as the commonly used SS1 strain, have lost the expression of several virulence factors that increase disease severity in humans, and they rarely cause severe disease in mice (17). CagA is the best-described example of loss of virulence factors during mouse adaptation. The CagA pathogenicity island is the best-known correlate of cancer risk. Encoding a type IV secretion system (T4SS) and CagA oncoprotein, this system injects CagA and other bacterial components into host epithelial cells, exacerbating inflammation and inducing host cell mutations (18). In the SS1 strain, CagA translocation to host epithelial cells is lost due to rearrangements in CagY (19). Although the pre-mouse-adapted SS1 strain (PMSS1) maintains CagA expression and T4SS function in vitro, CagA translocation is lost after 3–4 months of mouse colonization (20, 21).
Helicobacter felis (HF) is a close relative to HP that was originally isolated from the gastric mucosa of a cat (22). The HF genome shares more than 60% homology with HP and encodes many orthologs of HP essential genes that are required for gastric colonization (23). HF readily colonizes mice and quickly triggers massive gastric inflammation and atrophic gastritis within 2 months post-infection (24). Long-term infection over 12–24 months induces dysplasia and occasionally non-invasive adenocarcinoma (25). Although HF does colonize the human stomach, it is not a gastric cancer risk factor (26, 27). Moreover, HF does not express the CagA pathogenicity island, potentially limiting its ability to promote tumorigenesis in experimental models (23). However, HF’s robust induction of gastric inflammation makes it well suited for studies on how severe inflammation affects the gastric mucosa.
Both HP and HF are widely used to study gastric cancer initiation, but the strengths and weaknesses of each bacteria species may not be readily apparent. Typically, studies have elected to use one species or the other, and few have directly compared either of the bacteria to one another or their effects on the host (28, 29). The goal of this study was to directly compare the effects of HP and HF on immune-cell activation in vitro and their effects on gastric inflammation, atrophy, and metaplasia development in mice. We infected C57BL/6J mice with the HP PMSS1 strain, the most widely used CagA+ strain, and the HF CS1 strain. Mice were assessed 2 months post-infection to ensure that HP still maintained CagA function. Our findings indicate that both bacterial species exhibit a similar ability to stimulate macrophages in vitro, but that HF induces significantly more intense and widespread gastric inflammation and atrophy in mice. Although both bacterial strains have specific utility, this study aims to serve as a resource to help researchers choose the bacterial model that best suits their experimental goals.
MATERIALS AND METHODS
Animal care and treatment
All mouse studies were performed with approval by the West Virginia University Animal Care and Use Committee. C57BL/6J mice were purchased from the Jackson Laboratories. Mice were administered standard chow and water ab libitum and maintained in a temperature- and humidity-controlled room with standard 12-hour light/dark cycles. Eight-week-old mice were mock infected or infected with HP or HF by oral gavage. Mock mice received 500-µL sterile Brucella broth, and infected mice were inoculated with 500 µL of Brucella broth containing 109 CFU of the respective bacteria two times 24 hours apart. For the HP infection studies, both male and female mice were used. While for the HF infection studies, only female mice were used as there are significant sex differences in the response to HF. For peritoneal macrophage isolation, mice received a single intraperitoneal (IP) injection of 1-mL sterile Brewers Thioglycollate media (Sigma-Aldrich). After 4 days, the peritoneal lavage was plated, and nonadherent cells were removed by washing with prewarmed 1x phosphate-buffered saline (PBS).
Bacterial preparation
HF (ATCC 49179) was grown on tryptic soy agar plates (BD Biosciences) with 5% defibrinated sheep blood (Hemostat Labs) and 10 µg/mL vancomycin (Alfa Aesar) under microaerophilic conditions (5% O2 and 10% CO2) at 37°C for 2 days. HF was then harvested and transferred to Brucella broth (Research Products International) containing 5% fetal bovine serum (FBS) (R&D Systems) and 10 µg/mL vancomycin, and was grown overnight at 37°C under microaerophilic conditions with agitation. Bacteria were centrifuged and resuspended in fresh Brucella broth without antibiotics before spectrophotometry and mouse infection.
H. pylori PMSS1(30) (a gift from Manuel Amieva, Stanford University) was inoculated in Brucella broth containing 10% fetal bovine serum and 10 µg/mL vancomycin, and was grown overnight at 37°C under microaerophilic conditions with agitation. Bacteria were centrifuged and resuspended in fresh Brucella broth without antibiotics before spectrophotometry and mouse infection.
Tissue preparation
Mice were euthanized 2 months post-inoculation. Stomachs were removed and opened along the greater curvature and were washed in phosphate-buffered saline to remove gastric contents. One side of the stomach was fixed overnight in 4% paraformaldehyde at 4°C and cut into strips. The strips were either cryopreserved in 30% sucrose and embedded in optimal cutting temperature (OCT) media or transferred into 70% ethanol and submitted to the West Virginia University histology core for routine processing, embedding, sectioning, and H&E staining. For histology, strips were taken from the middle of the gastric corpus, avoiding the lesser curvature. A 2-mm biopsy was removed from the other side of the corpus for RNA isolation and was immediately snap-frozen in liquid nitrogen. The remainder of the corpus was disassociated into a single cell suspension for flow cytometry as described below.
Histology
Immunostaining was performed using standard methods. Briefly, 5-µm stomach cryosections were incubated with anti H+/K+ ATPase antibodies (clone 1H9, MBL Life Science), MIST1 (Cell Signaling Technologies), CD45 (clone 104; Biolegend), or CD44v9 (Cosmo Bio) for 1 hour at room temperature or overnight at 4°C. Sections were incubated in secondary antibodies for 1 hour at room temperature. Fluorescence-conjugated Griffonia simplicifolia lectin (GSII; Thermo Fisher Scientific) was added with secondary antibodies where indicated. Sections were mounted with Vectastain mounting media containing 4’,6-diamidino-2-phenylindole (DAPI) (Vector Laboratories). Images were obtained using a Zeiss 710 confocal laser-scanning microscope (Carl-Zeiss) and running Zen Black (Carl-Zeiss) imaging software.
RNA isolation and qRT-PCR
For gastric tissue, RNA was extracted in TRIzol (Thermo Fisher Scientific) and precipitated from the aqueous phase using an equal volume of 100% ethanol. The mixture was transferred to an RNA isolation column (Omega Bio-Tek), and the remaining steps were followed according to the manufacturer’s recommendations. For peritoneal macrophages, RNA was isolated using the MicroElute Total RNA kit (Omega Bio-Tek). RNA was treated with RNase-free DNase I (Omega Bio-Tek) as part of the isolation procedure. Reverse transcription followed by quantitative PCR was performed in the same reaction using the Universal Probes One-Step PCR kit (Bio-Rad Laboratories) and the TaqMan primers/probe mixtures (all from Thermo Fisher Scientific): Ppib (Mm00478295_m1), Wfdc2 (Mm00509434_m1), Il13 (Mm00434204_m1), Ifng (Mm01168134_m1), Cftr (Mm00445197_m1), Tnf (Mm00443258_m1), and Il1b (Mm00434228_m1). Relative gene expression was normalized to Ppib.
Flow cytometry
Corpus tissue from euthanized mice was washed in Hanks Balanced Salt Solution without Ca2+ or Mg2+ containing 5 mM HEPES, 5 mM EDTA, and 5% FBS at 37°C for 20 minutes. The tissue was then washed in Hanks Balanced Salt Solution with Ca2+ or Mg2+ briefly and then digested in 1 mg/mL collagenase (Worthington) for 30 minutes at 37°C. After digestion, the tissue fragments were pushed through a 100-µM strainer and then rinsed through a 40-µM strainer before debris was removed through an Optiprep (Serumwerk) density gradient. Fc receptors were blocked with TruStain (Biolegend) and then stained with antibodies for 20 minutes on ice. The following antibodies were used: CD45.2 (clone 104), CD3e (clone 145–2C11), B220 (clone RA3-6B2), CD4 (clone GK1.5), CD8a (clone 53–6.7), CD11b (clone M1/70), MHCII (clone M5/114.15.2), Ly6g (clone 1A8), F4/80 (clone BM8), and SiglecF (E50-2440). Actinomycin D (A1310, Invitrogen) was used to label dead cells. Cells were analyzed on a Cytek Aurora spectral flow cytometer (Cytek Biosciences). Flow cytometry analysis was performed using Cytobank (Beckman Coulter).
Peritoneal macrophage treatment and cytokine array
Peritoneal macrophages were treated with vehicle (sterile Brucella broth) or a 1:2 ratio of macrophages to HP or HF. Cells were washed and collected 3 hours posttreatment for RNA isolation. Media was collected 24 hours posttreatment for cytokine analysis. Cytokines were assessed using the Mouse C3 cytokine array (RayBiotech) following the manufacturer’s protocol. The arrays were imaged with an iBright 1500 (Thermo Fisher Scientific), and dot density was measured using the on-board analysis software. The cytokines were considered expressed if their pixel density was 1.5 times higher than the negative control dots printed on the cytokine array. A heatmap was generated using the Morpheus heat map tool (Broad Institute).
Statistical analysis
All error bars are ±SD of the mean. The sample size for each experiment is indicated in the figure legends. Experiments were repeated a minimum of two times. Statistical analyses were performed using a one-way analysis of variance with the post hoc Tukey t-test when comparing three or more groups, or by an unpaired t-test when comparing two groups. Statistical analysis was performed by GraphPad Prism 10 software. Statistical significance was set at P ≤ 0.05. Specific P values are listed in the figure legends.
RESULTS
H. felis and H. pylori exhibit similar immunostimulatory effects in vitro
HP has evolved to avoid the host immune system (30). Mutations in HP lipopolysaccaride (LPS) and flagellar proteins avoid binding of TLR4 and TLR5, respectively (28, 31). The enhanced immunogenicity of HF in vivo raises the possibility that HF is intrinsically more immunostimulatory than HP. We isolated thioglycolate-induced peritoneal macrophages from C57Bl6/J mice to examine the immunostimulatory effects of HP and HF. The cell cultures were challenged with a 2:1 cell ratio of HP or HF for 3 hours before assessment of proinflammatory cytokine expression by qRT-PCR. Challenge with either bacterium induced similar levels of the Tnf, while Il6 and Il1b induction was significantly higher in HF-challenged macrophages (Fig. 1A). Next, we used the same experimental system to measure bacterial induction of cytokines. Macrophages were co-cultured with either HF or HP for 24 hours, and the cell culture media was assessed by a mouse 62 cytokine dot plot array (Table 1). For this assay, a cytokine was considered detectable if the densitometry was 1.5-fold above the negative control background. Twenty-two cytokines were detected in the tissue culture media. Several cytokines were dramatically induced by bacterial co-culture, but surprisingly, there were no notable differences between the cytokine expression profiles of HP or HF-stimulated macrophages (Fig. 1B and C). These data demonstrate that HP and HF have a similar propensity to stimulate macrophages in vitro, suggesting that both strains are similarly immunogenic.
Fig 1.
H. pylori and H. felis elicit similar activation of macrophages in vitro. (A) Quantitative RT-PCR of the indicated genes using RNA isolated from cultured peritoneal macrophages 3 hours after mock stimulation or stimulation with a 1:1 ratio of H. pylori or H. felis. n ≥ 5. **P ≤ 0.01 ****P ≤ 0.0001. (B) Representative dot plot cytokine arrays. Arrays were probed with cell culture media from peritoneal macrophages stimulated for 24 hours with a 1:1 ratio of H. pylori or H. felis. Dot labels are found in Table 1. (C) Heatmap of the cytokine arrays is shown in (B). Cytokines were considered expressed if the dot density was 1.5-fold above the background. n ≥ 3.
TABLE 1.
Cytokine array dot plot key
| A | B | C | D | E | F | G | H | I | J | K | L | M | N | |
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| 1 | Pos | Pos | Neg | Neg | Blank | AXL | CXCL13 | TNFSF8 | TNFRSF8 | TNFRSF5 | CRG2 | CCL27 | CXCL16 | CCL11 |
| 2 | ||||||||||||||
| 3 | CCL24 | TNFSF6 | CX3CL1 | GCSF | GMCSF | IFNɣ | IGFBP3 | IGFBP5 | IGFBP6 | IL1⍺ | IL1β | IL2 | IL3 | IL3Rβ |
| 4 | ||||||||||||||
| 5 | IL4 | IL5 | IL6 | IL9 | IL10 | IL12 p40/p70 | IL12 p70 | IL13 | IL17A | CXCL1 | Leptin R | Leptin | LIX | CD62L |
| 6 | ||||||||||||||
| 7 | XCL1 | CCL2 | MCP5 | MCSF | CXCL9 | CCL3 | MIP1ɣ | MIP2 | CCL19 | CCL20 | CXCL4 | P-Selectin | CCL5 | SCF |
| 8 | ||||||||||||||
| 9 | SDF1 | CCL17 | CCL1 | CCL25 | TIMP1 | TNF⍺ | TNFRSF1A | TNFRSF1B | TPO | CD106 | VEGFA | Blank | Blank | POS |
| 10 |
H. felis infection induces severe inflammation of the gastric corpus
Chronic inflammation associated with Helicobacter infection is a primary risk factor for gastric cancer initiation. Our studies demonstrate that HP and HF elicit robust macrophage activation in vitro. Next, we assessed the gastric immune-cell landscape 2 months post-challenge to determine how these bacterial species affect gastric inflammation. Immunostaining of the gastric corpus for the common leukocyte antigen CD45 revealed relatively few immune cells in the mock-challenged stomach, which is consistent with previous reports that the healthy stomach harbors relatively few resident leukocytes (Fig. 2A) (7, 32). HP infection induced modest leukocyte infiltration within the gastric corpus. In contrast, HF infection induced severe leukocyte infiltration throughout the entire gastric corpus (Fig. 2A). Next, we utilized spectral flow cytometry to investigate the specific immune-cell populations that respond to Helicobacter infection. For these studies, we disassociated the entire gastric corpus. Compared to mock controls, both HP and HF infection induced a statistically significant increase in gastric immune infiltration (Fig. 2B). However, the HF-infected stomach had significantly more inflammation than the HP-infected stomach. Both CD3+ T cells and B220+ B cells were significantly increased in HF-infected mice, with T cells being the most abundant immune-cell population (Fig. 2B). Although T cells were increased in HP-infected stomachs, the increfase was not statistically significant. Macrophages were not significantly changed by either Helicobacter infection. Neutrophils and eosinophils were only significantly increased in HF-infected mice.
Fig 2.
H. felis-infected mice develop more extensive gastric inflammation than H. pylori. Cryosections (A), cells (B, C), and RNA (D) were collected from the gastric corpus 2 months after mock infection or challenge with H. pylori or H. felis. (A) Representative immunostaining. Sections were probed with antibodies against the common leukocyte antigen CD45 (green). Nuclei were stained with DAPI. n ≥ 6. Scale bars = 100 µm. (B, C) Flow cytometry of the indicated cell types from disassociated gastric corpus. (C) CD4+ and CD8+ T cell ratios. n ≥ 8. (D) Quantitative RT-PCR of RNA from the gastric corpus for the indicated genes. n ≥ 5. *P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.001, ****P ≤ 0.0001.
Gastric-infiltrating T cells are linked to the bulk of the gastric atrophy associated with Helicobacter infection in mice (12). Gastric T cells are rare in the murine stomach during steady-state conditions (Fig. 2B), but those present in mock-infected mice were almost entirely CD4+ Helper T cells. Interestingly, although HP induced only a modest increase in total gastric T cells, each bacterial strain differentially affected the CD4/CD8 T cell ratios. HP induced a CD8+ biased T cell response, while HF induced a CD4+ biased T cell response (Fig. 2C). Finally, we assessed the expression of inflammatory genes within the gastric corpus by qRT-PCR. Tnf and Il13 transcript levels were not significantly increased in response to HP infection, while both cytokines were significantly increased by HF infection (Fig. 2D). In contrast, Infg levels were significantly increased in both the HP- and HF-infected groups but were highest in HP-infected mice. Together, these results demonstrate that although the HP PMSS1 strain induces gastric inflammation, it is modest when compared to HF.
H. felis induces more advanced atrophic gastritis than H. pylori in mice
Because HF induced significantly more overall immune-cell infiltration and gastric T cell recruitment, we hypothesized that gastric epithelial damage would also be more severe in HF-infected mice. The morphology of the gastric corpus was assessed by H&E. Compared to mock-infected controls, HP-infected mice exhibited a slight increase in inflammatory infiltrate, similar to our findings above in Figure 2. In addition, although the overall gland length appeared unchanged, there was a noticeable foveolar hyperplasia, with the pit cells extending down to the gland neck (Fig. 3A). However, the parietal cells and chief cell compartment appeared grossly normal. In stark contrast, HF induced significant changes to the gastric epithelium throughout the entire corpus with a massive increase of immune cells and thickening of the gastric mucosa (Fig. 3A). Moreover, there was a dramatic expansion of the gastric pit cells and mucous neck cells, as well as a total absence of parietal and chief cells. Next, we immunostained with the mucous neck cell marker GSII lectin, the mature chief cell marker MIST1 (BHLHA15), and the parietal cell-specific hydrogen–potassium ATPase to better visualize changes to the major gastric cell lineages. HP-infected mice appeared similar to mock-infected controls, with normal proportions of mucous neck, chief, and parietal cells (Fig. 3B). Moreover, qRT-PCR for the cell lineage markers Tff2 (mucous neck cells), Gif (chief cells), and Atp4b (parietal cells) demonstrated that the relative expression of these major gastric lineages did not change 2 months post-HP challenge (Fig. 3C). In contrast, HP-infected mice exhibited dramatically increased GSII immunostaining, complete absence of MIST1+ cells, and only a few scattered parietal cells (Fig. 3B). Similarly, qRT-PCR demonstrated a significant expansion of Tff2 and a significant 8.9-fold and 4.3-fold loss of Gif and Atp4b, respectively (Fig. 3C). These results demonstrate that HP infection induces non-atrophic gastritis in the gastric corpus within 2 months post-challenge, but that HF quickly induces atrophic gastritis denoted by massive damage to the gastric epithelium, leading to dramatic changes to the major gastric epithelial cell lineages.
Fig 3.
H. felis-infected mice develop more extensive atrophic gastritis than H. pylori. Tissues (A, B) and RNA (C) were collected from the gastric corpus 2 months after mock infection or challenge with H. pylori or H. felis. (A) Representative H&E micrographs. (B) Representative immunostaining. Sections were probed with antibodies against the MIST1 (mature chief cells, green), the H+/K+ ATPase (parietal cells, red), and the Griffonia simplicifolia lectin (mucous neck cells, gray). Nuclei were stained with DAPI. (A, B) n ≥ 6. Scale bars = 100 µM. (C) Quantitative RT-PCR of RNA from the gastric corpus for the indicated genes. n ≥ 5. **P ≤ 0.01, ****P ≤ 0.0001.
H. felis infection induces pyloric metaplasia
The gastric corpus responds to inflammation and glandular damage by modifying cellular differentiation, leading to the development of pyloric metaplasia (PM), a putative preneoplastic lesion (33). To assess PM development, we immunostained with the de novo PM marker CD44v9 (34). Mock-infected mice did not exhibit any detectable CD44v9 expression within the glands of the gastric corpus (Fig. 4A). Similarly, CD44v9 staining was largely absent within the corpus glands of HP-infected mice, although there were occasionally positive cells within the gland base. In contrast, HF-infected mice exhibited widespread CD44v9-positive glands throughout the gastric corpus glands. Next, we assessed the expression of the PM marker transcripts Wfdc2 and Cftr. Curiously, these transcripts were significantly increased in both HP- and HF-infected mice (Fig. 4B), and Cftr expression was significantly higher in HP-infected mice compared to HF-infected mice. Overall, HP-infected mice do not have gross morphological features of PM (Fig. 4A and B) and lack expression of CD44v9 (Fig. 4A) at 2 months post-infection.
Fig 4.
Pyloric metaplasia is more extensive in H. felis-infected mice. Tissues (A) and RNA (B) were collected from the gastric corpus 2 months after mock infection or challenge with H. pylori or H. felis. (A) Immunostaining for the pyloric metaplasia marker CD44v9. n ≥ 5. Scale bar = 100 µM. (B) Quantitative RT-PCR of RNA from the gastric corpus for the indicated genes. n ≥ 6. **P ≤ 0.01, ****P ≤ 0.0001.
H. felis drives more extensive inflammation in the gastric antrum
In humans, HP typically initially colonizes the gastric antrum and gradually follows a front of inflammation and atrophy into the gastric corpus (33). PMSS1 follows a similar colonization pattern, although colonization patterns are strain specific (35). Next, we used H&E micrographs to assess the gross morphology of the gastric antrum in HP- and HF-infected mice. Compared to mock-infected controls, the antrum of HP- and HF-infected mice developed modest leukocyte infiltration (Fig. 5). However, leukocyte infiltration was more extensive in HF-infected mice, and the glands were thickened. These results indicate that HF drives more extensive inflammation throughout the entire stomach, affecting both the corpus and pylorus.
Fig 5.
H. felis induces more extensive inflammation within the gastric antrum. H&E micrographs of the gastric antrum collected 2 months after mock infection or infection with H. pylori or H. felis. Scale bar = 50 µM. N ≥ 5.
DISCUSSION
Gastric-colonizing Helicobacter species have a complex relationship with the host immune response. Mild inflammation likely benefits the pathogen by causing oxyntic atrophy, allowing further colonization of the gastric corpus (33, 36) and increasing the expression of adhesion molecules by damaged and metaplastic gastric glands (37, 38). Severe inflammation is linked to lower bacterial loads due to increased pathogen clearance by the immune system and increased competition due to stomach colonization by intestinal flora as a result of gastric achlorhydria (39, 40). Severe inflammation ultimately may lead to Helicobacter eradication by the host. Therefore, HP has evolved to evade the host immune system (30). HP LPS lipid A is tetra-acetylated and poorly activates TLR4 (28, 41). HP flagella mutations at the TLR5 binding site evade TLR5 activation (31), and HP DNA is modified to avoid TLR9 activation (42). Rather, HP and HF are potent activators of TLR2 (28, 43). HF LPS has a similar effect on TLR4 activation, but unlike HP, it is reported to activate TLR9 to induce gastric inflammation (44). Our RNAseq analysis revealed that HF expressed higher levels of LPS synthesis genes and flagellar genes than HP. Moreover, HF induced significantly more robust inflammatory responses in mice. We, therefore, hypothesized that HF is innately more immunogenic than HP. Surprisingly, although HF induced higher mRNA levels of Il6 and Il1b, both bacterial species induced similar cytokine responses when assessed by a semi-quantitative dot plot array. Similar findings were reported by Mandell et al., where both HP SS1 and HF elicited similar activation of TLR4 and TRL2 (28). These data suggest that the enhanced inflammation observed in HF-colonized mouse stomachs is not due to enhanced immunogenicity of HF and may result from other mechanisms such as enhanced HF colonization or HP-mediated suppression of inflammatory pathways.
Our mouse colonization studies demonstrated that HF induced significantly more extensive gastric inflammation and epithelial remodeling than the HP PMSS1 strain. Chronic inflammation is necessary and sufficient for gastric cancer initiation. Mice that lack mature B and T cells are protected from morphological changes to the gastric epithelium in response to infection by either HP or HF (12, 45), and myeloid cell activation is required for pyloric metaplasia initiation (7, 8, 32). Moreover, overexpression of the proinflammatory IL1B or IFN-gamma under control of the parietal cell-specific H+/K+ ATPase β promoter drives gastric neoplasia development without accompanying Helicobacter infection (5, 6). Although we found that both HP and HF induced similar macrophage activation in vitro, gastric inflammation 2 months post-infection was significantly different for these two bacterial species. HP elicited only mild inflammatory responses within the gastric corpus 2 months post-infection. In contrast, HF induced severe inflammation throughout the entire gastric corpus at the same time point. Although previous studies reported that HP triggered gastric inflammation, atrophy, and metaplasia by 2 months post-infection (20, 46, 47), our findings indicate that these events are mild and restricted to the gastric corpus lesser curvature. In humans, HP typically initially colonizes the gastric antrum and, over time, spreads through the corpus lesser curvature (48). Although the underlying mechanisms of this pattern of spread through the stomach are not fully defined, the lesser curvature is more susceptible to inflammation and consequential oxyntic atrophy (8, 33). In contrast, HF-induced metaplasia and inflammation were ubiquitous throughout the gastric corpus. Within the gastric antrum, both bacterial species induced inflammation but as within the corpus, HF triggered more extensive inflammation. Poor colonization by the PMSS1 strain may account for some of the poor inflammation. Sigal et al. found that HP PMSS1 is present in the antrum but does not colonize the gastric corpus glands of C57BL/6J mice 2 months post-challenge (49). Moreover, interactions with the microbiota may abrogate HP-induced inflammation as the same study reported that C57BL/6 mice from different vendors exhibit dramatic differences in corpus inflammation (49). It is important to note that HP colonization patterns vary significantly by strain. Other HP strains, such as X47, preferentially colonize the corpus, although this strain does not express a CagA (35).
T cells are required for the bulk of gastric epithelial damage and remodeling associated with Helicobacter infection in mice (12, 50). Elevated levels of Th1/Th17-associated cytokines are linked to increased cancer risk in humans (51, 52). Here, we found that HP does not induce a significant T cell response 2 months post-infection. This lack of response is consistent with the mild epithelial changes and lack of metaplasia development. HF induced dramatic T cell activation, particularly in the CD4+ T cell compartment. CD4+ T cells are reported to drive the bulk of gastric damage associated with HF infection (53). Interestingly, although HP only caused mild gastric T cell recruitment, it caused a shift in the CD4/CD8 T cell ratio, skewing toward an enhanced CD8 cytotoxic T cell response. This increase is likely due to the CagA pathogenicity island and functional T4SS that transfers bacterial components into host epithelial cells (54). We found that although HF inflammation was predominantly biased toward CD4+ T cells, there still was an overall increase in gastric CD8+ T cells. CD8 T cells have also been reported to induce gastric damage during HF infection, but their role is secondary to CD4 T cells (55). The exact role of these cytotoxic T cells is unclear. Although CD8+ T cell responses contribute to epithelial damage (54, 55), they undoubtedly also play important roles in responding to neoantigens to remove developing tumor cells. The hyper-inflamed environment induced by Helicobacter may promote cytotoxic T cell exhaustion and dysfunction, potentially explaining why more intense gastric inflammation is linked to increased cancer risk.
HP and HF are closely related Helicobacter species, sharing more than 60% of each other’s genome (23). HF’s genome encodes many HP orthologs that are required for gastric colonization, such as a urease cluster, flagellar and chemotactic genes, gastric epithelial adhesion proteins, RecA, collagenase, and iron uptake gene clusters. CagA and the vacuolating toxin VacA are the most well-known HP virulence factors associated with gastric cancer risk in humans and are notably absent from the HF genome (23). CagA is a bona fide bacterial oncoprotein that is injected into host epithelial cells through its T4SS (56). Once inside host cells, CagA attaches to the plasma membrane and promiscuously interacts with host proteins, inducing mutations, cellular reprogramming, and inflammation (57). Transgenic mice that ectopically express CagA from parietal cell-specific H+/K+ ATPase promoter spontaneously develop gastric tumors (58). Although CagA is sufficient to drive gastric inflammation and cancer initiation, our results indicate that inflammation induced by CagA+ HP still pales in comparison to HF. CagA expression is likely one of the most important rationales for using HP in mice infection studies. However, when HP was re-isolated from infected mice, it was found that they had lost their T4SS functionality by 3–4 months post-infection (20, 21). Thus, the selection of HP for mouse infection studies strains solely on their expression of CagA may not yield the desired phenotypes. Although we showed that HF induced more robust inflammation and epithelial remodeling than HP, it should be noted that we did not assess changes within gastric epithelial cells. CagA has multiple effects on epithelial cells, affecting cell signaling pathways, suppressing p53 and other tumor suppressors, and altering cellular proliferation (59–61). Thus, study goals and experimental endpoints should be carefully considered to select the appropriate Helicobacter model.
In summary, HP infection studies in mice mount specific challenges in modeling the inflammatory phenotypes and cancer initiation observed in humans. These problems stem from the fact that HP is not a natural mouse pathogen and common mouse-adapted strains such as PMSS1 suffer from weak immunogenicity and functional loss of virulence factors that promote cancer risk. HF has been used for decades to model pro-neoplastic gastric inflammation and cancer initiation, and quickly and consistently initiates inflammation in mouse models. Although HF is more strongly suited for studies on Helicobacter-induced gastric inflammation, metaplasia, and cancer initiation, its lack of CagA and VacA limits its utility for studies on the direct effects of the bacteria on gastric epithelial cells. Moreover, HP is likely better suited for studies on bacterial colonization and interactions with the gastric microbiota.
ACKNOWLEDGMENTS
This work was supported by West Virginia University start-up funds (J.T.B) and a grant from National Institutes of Health P20GM121322 (J.T.B.). The West Virginia University Microscope Imaging Facility and Flow Cytometry & Single Cell Core receive support from the National Institutes of Health grants P30GM103503 and S10 grant OD028605, respectively. The authors thank Richard Peek Jr. (Vanderbilt University Medical Center) for technical assistance.
S.R.D., S.V., S.K., B.C.D., M.T.M., and J.T.B. performed experiments. S.R.D., S.V., E.S.-K., and F.H.D. analyzed data. F.H.D., G.W.L., and J.T.B. planned the study. J.T.B. drafted the original manuscript and S.R.D., G.W.L., and J.T.B. revised the manuscript. All authors approved the final version of the manuscript.
Contributor Information
Jonathan T. Busada, Email: jonathan.busada@hsc.wvu.edu.
Yuan Pin Hung, Tainan Hospital, Ministry of Health and Welfare, Tainan, Taiwan.
REFERENCES
- 1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. 2021. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 71:209–249. doi: 10.3322/caac.21660 [DOI] [PubMed] [Google Scholar]
- 2. Amieva M, Peek RM. 2016. Pathobiology of Helicobacter pylori-induced gastric cancer. Gastroenterology 150:64–78. doi: 10.1053/j.gastro.2015.09.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Hooi JKY, Lai WY, Ng WK, Suen MMY, Underwood FE, Tanyingoh D, Malfertheiner P, Graham DY, Wong VWS, Wu JCY, Chan FKL, Sung JJY, Kaplan GG, Ng SC. 2017. Global prevalence of Helicobacter pylori infection: systematic review and meta-analysis. Gastroenterology 153:420–429. doi: 10.1053/j.gastro.2017.04.022 [DOI] [PubMed] [Google Scholar]
- 4. Correa P. 1995. Helicobacter pylori and gastric carcinogenesis. Am J Surg Pathol 19 Suppl 1:S37–43. [PubMed] [Google Scholar]
- 5. Tu S, Bhagat G, Cui G, Takaishi S, Kurt-Jones EA, Rickman B, Betz KS, Penz-Oesterreicher M, Bjorkdahl O, Fox JG, Wang TC. 2008. Overexpression of interleukin-1beta induces gastric inflammation and cancer and mobilizes myeloid-derived suppressor cells in mice. Cancer Cell 14:408–419. doi: 10.1016/j.ccr.2008.10.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Syu LJ, El-Zaatari M, Eaton KA, Liu Z, Tetarbe M, Keeley TM, Pero J, Ferris J, Wilbert D, Kaatz A, Zheng X, Qiao X, Grachtchouk M, Gumucio DL, Merchant JL, Samuelson LC, Dlugosz AA. 2012. Transgenic expression of interferon-gamma in mouse stomach leads to inflammation, metaplasia, and dysplasia. Am J Pathol 181:2114–2125. doi: 10.1016/j.ajpath.2012.08.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Busada JT, Peterson KN, Khadka S, Xu X, Oakley RH, Cook DN, Cidlowski JA. 2021. Glucocorticoids and androgens protect from gastric metaplasia by suppressing group 2 innate lymphoid cell activation. Gastroenterology 161:637–652. doi: 10.1053/j.gastro.2021.04.075 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Busada JT, Ramamoorthy S, Cain DW, Xu X, Cook DN, Cidlowski JA. 2019. Endogenous glucocorticoids prevent gastric metaplasia by suppressing spontaneous inflammation. J Clin Invest 129:1345–1358. doi: 10.1172/JCI123233 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Bockerstett KA, Petersen CP, Noto CN, Kuehm LM, Wong CF, Ford EL, Teague RM, Mills JC, Goldenring JR, DiPaolo RJ. 2020. Interleukin 27 protects from gastric atrophy and metaplasia during chronic autoimmune gastritis. Cell Mol Gastroenterol Hepatol 10:561–579. doi: 10.1016/j.jcmgh.2020.04.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. El-Omar EM, Carrington M, Chow WH, McColl KE, Bream JH, Young HA, Herrera J, Lissowska J, Yuan CC, Rothman N, Lanyon G, Martin M, Fraumeni JF, Rabkin CS. 2001. The role of Interleukin-1 polymorphisms in the pathogenesis of gastric cancer. Nature 412:99. doi: 10.1038/35083631 [DOI] [PubMed] [Google Scholar]
- 11. El-Omar EM, Rabkin CS, Gammon MD, Vaughan TL, Risch HA, Schoenberg JB, Stanford JL, Mayne ST, Goedert J, Blot WJ, Fraumeni JF, Chow WH. 2003. Increased risk of noncardia gastric cancer associated with proinflammatory cytokine gene polymorphisms. Gastroenterology 124:1193–1201. doi: 10.1016/s0016-5085(03)00157-4 [DOI] [PubMed] [Google Scholar]
- 12. Roth KA, Kapadia SB, Martin SM, Lorenz RG. 1999. Cellular immune responses are essential for the development of Helicobacter felis-associated gastric pathology. J Immunol 163:1490–1497. doi: 10.4049/jimmunol.163.3.1490 [DOI] [PubMed] [Google Scholar]
- 13. Modi N, Chen Y, Dong X, Hu X, Lau GW, Wilson KT, Peek RM, Chen LF. 2024. BRD4 regulates glycolysis-dependent Nos2 expression in macrophages upon H pylori infection. Cell Mol Gastroenterol Hepatol 17:292–308. doi: 10.1016/j.jcmgh.2023.10.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. El-Zaatari M, Bass AJ, Bowlby R, Zhang M, Syu LJ, Yang Y, Grasberger H, Shreiner A, Tan B, Bishu S, Leung WK, Todisco A, Kamada N, Cascalho M, Dlugosz AA, Kao JY. 2017. Indoleamine 2,3-dioxygenase 1, increased in human gastric pre-neoplasia, promotes inflammation and metaplasia in mice and is associated with type II hypersensitivity/autoimmunity. Gastroenterology 154:140–153. doi: 10.1053/j.gastro.2017.09.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Ayraud S, Janvier B, Fauchère JL. 2002. Experimental colonization of mice by fresh clinical isolates of Helicobacter pylori is not influenced by the cagA status and the vacA genotype. FEMS Immunol Med Microbiol 34:169–172. doi: 10.1111/j.1574-695X.2002.tb00620.x [DOI] [PubMed] [Google Scholar]
- 16. Lee A, O’Rourke J, De Ungria MC, Robertson B, Daskalopoulos G, Dixon MF. 1997. A standardized mouse model of Helicobacter pylori infection: introducing the Sydney strain. Gastroenterology 112:1386–1397. doi: 10.1016/s0016-5085(97)70155-0 [DOI] [PubMed] [Google Scholar]
- 17. Amalia R, Panenggak NSR, Doohan D, Rezkitha YAA, Waskito LA, Syam AF, Lubis M, Yamaoka Y, Miftahussurur M. 2023. A comprehensive evaluation of an animal model for Helicobacter pylori-associated stomach cancer: fact and controversy. Helicobacter 28:e12943. doi: 10.1111/hel.12943 [DOI] [PubMed] [Google Scholar]
- 18. Cover TL, Lacy DB, Ohi MD. 2020. The Helicobacter pylori Cag type IV secretion system. Trends Microbiol 28:682–695. doi: 10.1016/j.tim.2020.02.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Barrozo RM, Cooke CL, Hansen LM, Lam AM, Gaddy JA, Johnson EM, Cariaga TA, Suarez G, Peek RM, Cover TL, Solnick JV. 2013. Functional plasticity in the type IV secretion system of Helicobacter pylori. PLoS Pathog 9:e1003189. doi: 10.1371/journal.ppat.1003189 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Arnold IC, Lee JY, Amieva MR, Roers A, Flavell RA, Sparwasser T, Müller A. 2011. Tolerance rather than immunity protects from Helicobacter pylori-induced gastric preneoplasia. Gastroenterology 140:199–209. doi: 10.1053/j.gastro.2010.06.047 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Dyer V, Brüggemann H, Sörensen M, Kühl AA, Hoffman K, Brinkmann V, Reines MDM, Zimmerman S, Meyer TF, Koch M. 2018. Genomic features of the Helicobacter pylori strain PMSS1 and its virulence attributes as deduced from its in vivo colonisation patterns. Mol Microbiol 110:761–776. doi: 10.1111/mmi.14123 [DOI] [PubMed] [Google Scholar]
- 22. Lee A, Hazell SL, O’Rourke J, Kouprach S. 1988. Isolation of a spiral-shaped bacterium from the cat stomach. Infect Immun 56:2843–2850. doi: 10.1128/iai.56.11.2843-2850.1988 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Arnold IC, Zigova Z, Holden M, Lawley TD, Rad R, Dougan G, Falkow S, Bentley SD, Müller A. 2011. Comparative whole genome sequence analysis of the carcinogenic bacterial model pathogen Helicobacter felis. Genome Biol Evol 3:302–308. doi: 10.1093/gbe/evr022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Lee A, Fox JG, Otto G, Murphy J. 1990. A small animal model of human Helicobacter pylori active chronic gastritis. Gastroenterology 99:1315–1323. doi: 10.1016/0016-5085(90)91156-z [DOI] [PubMed] [Google Scholar]
- 25. Cai X, Carlson J, Stoicov C, Li H, Wang TC, Houghton J. 2005. Helicobacter felis eradication restores normal architecture and inhibits gastric cancer progression in C57Bl/6 mice. Gastroenterology 128:1937–1952. doi: 10.1053/j.gastro.2005.02.066 [DOI] [PubMed] [Google Scholar]
- 26. Fritz EL, Slavik T, Delport W, Olivier B, van der Merwe SW. 2006. Incidence of Helicobacter felis and the effect of coinfection with Helicobacter pylori on the gastric mucosa in the African population. J Clin Microbiol 44:1692–1696. doi: 10.1128/JCM.44.5.1692-1696.2006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Kusters JG, Kuipers EJ. 1998. Non-pylori Helicobacter infections in humans. Eur J Gastroenterol Hepatol 10:239–241. doi: 10.1097/00042737-199803000-00009 [DOI] [PubMed] [Google Scholar]
- 28. Mandell L, Moran AP, Cocchiarella A, Houghton J, Taylor N, Fox JG, Wang TC, Kurt-Jones EA. 2004. Intact gram-negative Helicobacter pylori, Helicobacter felis, and Helicobacter hepaticus bacteria activate innate immunity via toll-like receptor 2 but not toll-like receptor 4. Infect Immun 72:6446–6454. doi: 10.1128/IAI.72.11.6446-6454.2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Court M, Robinson PA, Dixon MF, Crabtree JE. 2002. Gastric Helicobacter species infection in murine and gerbil models: comparative analysis of effects of H. pylori and H. felis on gastric epithelial cell proliferation. J Infect Dis 186:1348–1352. doi: 10.1086/344321 [DOI] [PubMed] [Google Scholar]
- 30. Zhang X, Arnold IC, Müller A. 2020. Mechanisms of persistence, innate immune activation and immunomodulation by the gastric pathogen Helicobacter pylori. Curr Opin Microbiol 54:1–10. doi: 10.1016/j.mib.2020.01.003 [DOI] [PubMed] [Google Scholar]
- 31. Kim JH, Namgung B, Jeon YJ, Song WS, Lee J, Kang SG, Yoon SI. 2018. Helicobacter pylori flagellin: TLR5 evasion and fusion-based conversion into a TLR5 agonist. Biochem Biophys Res Commun 505:872–878. doi: 10.1016/j.bbrc.2018.09.179 [DOI] [PubMed] [Google Scholar]
- 32. Busada JT, Khadka S, Peterson KN, Druffner SR, Stumpo DJ, Zhou L, Oakley RH, Cidlowski JA, Blackshear PJ. 2021. Tristetraprolin prevents gastric metaplasia in mice by suppressing pathogenic inflammation. Cell Mol Gastroenterol Hepatol 12:1831–1845. doi: 10.1016/j.jcmgh.2021.07.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Sáenz JB. 2021. Follow the metaplasia: characteristics and oncogenic implications of metaplasia’s pattern of spread throughout the stomach. Front Cell Dev Biol 9:741574. doi: 10.3389/fcell.2021.741574 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Wada T, Ishimoto T, Seishima R, Tsuchihashi K, Yoshikawa M, Oshima H, Oshima M, Masuko T, Wright NA, Furuhashi S, Hirashima K, Baba H, Kitagawa Y, Saya H, Nagano O. 2013. Functional role of CD44V-xCT system in the development of spasmolytic polypeptide-expressing metaplasia. Cancer Sci 104:1323–1329. doi: 10.1111/cas.12236 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Akada JK, Ogura K, Dailidiene D, Dailide G, Cheverud JM, Berg DE. 2003. Helicobacter pylori tissue tropism: mouse-colonizing strains can target different gastric niches. Microbiology (Reading) 149:1901–1909. doi: 10.1099/mic.0.26129-0 [DOI] [PubMed] [Google Scholar]
- 36. Kuipers EJ, Lundell L, Klinkenberg-Knol EC, Havu N, Festen HP, Liedman B, Lamers CB, Jansen JB, Dalenback J, Snel P, Nelis GF, Meuwissen SG. 1996. Atrophic gastritis and Helicobacter pylori infection in patients with reflux esophagitis treated with omeprazole or fundoplication. N Engl J Med 334:1018–1022. doi: 10.1056/NEJM199604183341603 [DOI] [PubMed] [Google Scholar]
- 37. Mahdavi J, Sondén B, Hurtig M, Olfat FO, Forsberg L, Roche N, Angstrom J, Larsson T, Teneberg S, Karlsson KA, Altraja S, Wadström T, Kersulyte D, Berg DE, Dubois A, Petersson C, Magnusson KE, Norberg T, Lindh F, Lundskog BB, Arnqvist A, Hammarström L, Borén T. 2002. Helicobacter pylori SabA adhesin in persistent infection and chronic inflammation. Science 297:573–578. doi: 10.1126/science.1069076 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Sáenz JB, Vargas N, Mills JC. 2019. Tropism for spasmolytic polypeptide-expressing metaplasia allows Helicobacter pylori to expand its intragastric niche. Gastroenterology 156:160–174. doi: 10.1053/j.gastro.2018.09.050 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Arnold IC, Zhang X, Urban S, Artola-Borán M, Manz MG, Ottemann KM, Müller A. 2017. NLRP3 controls the development of gastrointestinal CD11B(+) dendritic cells in the steady state and during chronic bacterial infection. Cell Rep 21:3860–3872. doi: 10.1016/j.celrep.2017.12.015 [DOI] [PubMed] [Google Scholar]
- 40. Noto JM, Peek RM. 2017. The gastric microbiome, its interaction with Helicobacter pylori, and its potential role in the progression to stomach cancer. PLoS Pathog 13:e1006573. doi: 10.1371/journal.ppat.1006573 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Moran AP, Lindner B, Walsh EJ. 1997. Structural characterization of the lipid A component of Helicobacter pylori rough- and smooth-form lipopolysaccharides. J Bacteriol 179:6453–6463. doi: 10.1128/jb.179.20.6453-6463.1997 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Varga MG, Piazuelo MB, Romero-Gallo J, Delgado AG, Suarez G, Whitaker ME, Krishna US, Patel RV, Skaar EP, Wilson KT, Algood HMS, Peek RM. 2016. TLR9 activation suppresses inflammation in response to Helicobacter pylori infection. Am J Physiol Gastrointest Liver Physiol 311:G852–G858. doi: 10.1152/ajpgi.00175.2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Obonyo M, Sabet M, Cole SP, Ebmeyer J, Uematsu S, Akira S, Guiney DG. 2007. Deficiencies of myeloid differentiation factor 88, toll-like receptor 2 (TLR2), or TLR4 produce specific defects in macrophage cytokine secretion induced by Helicobacter pylori. Infect Immun 75:2408–2414. doi: 10.1128/IAI.01794-06 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Tang K, McLeod L, Livis T, West AC, Dawson R, Yu L, Balic JJ, Chonwerawong M, Wray-McCann G, Oshima H, Oshima M, Deswaerte V, Ferrero RL, Jenkins BJ. 2022. Toll-like receptor 9 promotes initiation of gastric tumorigenesis by augmenting inflammation and cellular proliferation. Cell Mol Gastroenterol Hepatol 14:567–586. doi: 10.1016/j.jcmgh.2022.06.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Eaton KA, Ringler SR, Danon SJ. 1999. Murine splenocytes induce severe gastritis and delayed-type hypersensitivity and suppress bacterial colonization in Helicobacter pylori-infected SCID mice. Infect Immun 67:4594–4602. doi: 10.1128/IAI.67.9.4594-4602.1999 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Khurana SS, Riehl TE, Moore BD, Fassan M, Rugge M, Romero-Gallo J, Noto J, Peek RM, Stenson WF, Mills JC. 2013. The hyaluronic acid receptor CD44 coordinates normal and metaplastic gastric epithelial progenitor cell proliferation. J Biol Chem 288:16085–16097. doi: 10.1074/jbc.M112.445551 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Sierra JC, Asim M, Verriere TG, Piazuelo MB, Suarez G, Romero-Gallo J, Delgado AG, Wroblewski LE, Barry DP, Peek RM, Gobert AP, Wilson KT. 2018. Epidermal growth factor receptor inhibition downregulates Helicobacter pylori-induced epithelial inflammatory responses, DNA damage and gastric carcinogenesis. Gut 67:1247–1260. doi: 10.1136/gutjnl-2016-312888 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Graham DY. 2014. History of Helicobacter pylori, duodenal ulcer, gastric ulcer and gastric cancer. World J Gastroenterol 20:5191–5204. doi: 10.3748/wjg.v20.i18.5191 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Sigal M, Rothenberg ME, Logan CY, Lee JY, Honaker RW, Cooper RL, Passarelli B, Camorlinga M, Bouley DM, Alvarez G, Nusse R, Torres J, Amieva MR. 2015. Helicobacter pylori activates and expands Lgr5(+) stem cells through direct colonization of the gastric glands. Gastroenterology 148:1392–1404. doi: 10.1053/j.gastro.2015.02.049 [DOI] [PubMed] [Google Scholar]
- 50. Gray BM, Fontaine CA, Poe SA, Eaton KA. 2013. Complex T cell interactions contribute to Helicobacter pylori gastritis in mice. Infect Immun 81:740–752. doi: 10.1128/IAI.01269-12 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Dixon B, Hossain R, Patel RV, Algood HMS. 2019. Th17 cells in Helicobacter pylori infection: a dichotomy of help and harm. Infect Immun 87:e00363-19. doi: 10.1128/IAI.00363-19 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Ikuse T, Blanchard TG, Czinn SJ. 2019. Inflammation, immunity, and vaccine development for the gastric pathogen Helicobacter pylori. Curr Top Microbiol Immunol 421:1–19. doi: 10.1007/978-3-030-15138-6_1 [DOI] [PubMed] [Google Scholar]
- 53. McCracken VJ, Martin SM, Lorenz RG. 2005. The Helicobacter felis model of adoptive transfer gastritis. Immunol Res 33:183–194. doi: 10.1385/IR:33:2:183 [DOI] [PubMed] [Google Scholar]
- 54. Koch MRA, Gong R, Friedrich V, Engelsberger V, Kretschmer L, Wanisch A, Jarosch S, Ralser A, Lugen B, Quante M, Vieth M, Vasapolli R, Schulz C, Buchholz VR, Busch DH, Mejías-Luque R, Gerhard M. 2023. CagA-specific gastric CD8(+) tissue-resident T cells control Helicobacter pylori during the early infection phase. Gastroenterology 164:550–566. doi: 10.1053/j.gastro.2022.12.016 [DOI] [PubMed] [Google Scholar]
- 55. Fukui T, Nishio A, Okazaki K, Kasahara K, Saga K, Tanaka J, Uza N, Ueno S, Kido M, Ohashi S, Asada M, Nakase H, Watanabe N, Chiba T. 2007. Cross-primed CD8+ cytotoxic T cells induce severe Helicobacter-associated gastritis in the absence of CD4+ T cells. Helicobacter 12:486–497. doi: 10.1111/j.1523-5378.2007.00536.x [DOI] [PubMed] [Google Scholar]
- 56. Fischer W. 2011. Assembly and molecular mode of action of the Helicobacter pylori Cag type IV secretion apparatus. FEBS J 278:1203–1212. doi: 10.1111/j.1742-4658.2011.08036.x [DOI] [PubMed] [Google Scholar]
- 57. Hatakeyama M. 2014. Helicobacter pylori CagA and gastric cancer: a paradigm for hit-and-run carcinogenesis. Cell Host Microbe 15:306–316. doi: 10.1016/j.chom.2014.02.008 [DOI] [PubMed] [Google Scholar]
- 58. Ohnishi N, Yuasa H, Tanaka S, Sawa H, Miura M, Matsui A, Higashi H, Musashi M, Iwabuchi K, Suzuki M, Yamada G, Azuma T, Hatakeyama M. 2008. Transgenic expression of Helicobacter pylori CagA induces gastrointestinal and hematopoietic neoplasms in mouse. Proc Natl Acad Sci USA 105:1003–1008. doi: 10.1073/pnas.0711183105 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Suzuki M, Mimuro H, Kiga K, Fukumatsu M, Ishijima N, Morikawa H, Nagai S, Koyasu S, Gilman RH, Kersulyte D, Berg DE, Sasakawa C. 2009. Helicobacter pylori CagA phosphorylation-independent function in epithelial proliferation and inflammation. Cell Host Microbe 5:23–34. doi: 10.1016/j.chom.2008.11.010 [DOI] [PubMed] [Google Scholar]
- 60. Nagy TA, Frey MR, Yan F, Israel DA, Polk DB, Peek RM. 2009. Helicobacter pylori regulates cellular migration and apoptosis by activation of phosphatidylinositol 3-kinase signaling. J Infect Dis 199:641–651. doi: 10.1086/596660 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Buti L, Spooner E, Van der Veen AG, Rappuoli R, Covacci A, Ploegh HL. 2011. Helicobacter pylori cytotoxin-associated gene A (CagA) subverts the apoptosis-stimulating protein of p53 (ASPP2) tumor suppressor pathway of the host. Proc Natl Acad Sci USA 108:9238–9243. doi: 10.1073/pnas.1106200108 [DOI] [PMC free article] [PubMed] [Google Scholar]





