Abstract
The goal of this review is to highlight literature which demonstrates how cytokines made by T lymphocytes impact the gastric epithelium, especially during Helicobacter pylori infection. These cytokines effect many of the diverse functions of the epithelium and the epithelium’s interactions with Helicobacter pylori. Focal point of this review will be on how T cell cytokines impact antimicrobial function and barrier function and how T cell cytokines influence the development and progression of cancer. Further, the modulation of epithelial-derived chemokines by H. pylori infection will be discussed.
Keywords: Helicobacter pylori, T cells, cytokines, gastric mucosa, gastric cancer
Introduction
The health and function of the stomach and the intestines are essential for interacting with the external environment and with the internal microbiome through the epithelial cell layer. Specifically, the function of the stomach is to store food, provide early digestion of food by producing acid and enzymes and macerating that food with its muscular lining. There are several specialized epithelial cells which aid in the stomach performing its necessary functions. These include chief cells, enteroendocrine cells, parietal cells, stem cells and foveolar cells (both surface mucous cells and mucous neck cells)(1)(Figure 1). Chief cells are found in the fundic glands, and most common, deep in the glands closer to the muscularis mucosae. The digestive enzymes of the stomach, pepsins, are secreted by chief cells. Located deep in the gastric glands are the enteroendocrine cells including G cells, D cells, and enterochromaffin-like cells (ECL cells), which are responsible for production of gastrin, somatostatin (SOM), and histamine, respectively. Gastrin and histamine impact parietal cell and chief cell function. Gastrin stimulates ECL cells to release histamine and then synergistically these hormones activate parietal cells to produce hydrochloric acid and chief cells to release pepsin. Foveolar cells are simple columnar epithelial cells which line the stomach. They are also found in gastric pits and at the top of the gastric glands (necks) where they are referred to as surface mucous cells and mucous neck cells, respectively. Surface mucous cells contain large amounts of mucin at their apical surface. The mucus and bicarbonate ions produced by the foveolar cells play an important role in modulating the impact of acids on the gastric epithelium. Gastric mucous neck cells are most common in the upper region of the glands.
Figure 1.
Illustration of the Stomach. The architecture of the human stomach is similar to the mouse with the exception of the presence of a nonglandular forestomach in the mouse. In the stomach epithelium, gastric pits lead to gastric glands that secrete gastric juice. The cellular composition of the gastric glands and pits varies based on localization in the corpus verses the antrum. Enlarged gastric pits and glands are shown containing different localization specialized epithelial cells including foveolar cells (both surface mucous cells and mucous neck cells), cells that that secrete hydrochloride acid, gastrin, somatostatin and activate pepsin (parietal cells, enteroendocrine cells, and chief cells), and gastric stem cells. H. pylori colonization is denser in the antrum of the stomach than the corpus.
The maintenance of the epithelium functions is vital for a body’s health. The gastric epithelium can be disrupted under several conditions including use of pharmacological reagents, alcohol or infection (2–4). Helicobacter pylori is the most common bacterial infection of the gastric mucosa. H. pylori’s prevalence is greater than 50% of the population, but there is significant variation in the prevalence between regions of the world (5, 6). Long term colonization by H. pylori is the most consequential risk factor for the development of gastric cancer (GC)(7), which imposes a significant global burden with over 1 million new stomach cancers diagnosed in 2018 (8). Like many bacterial pathogens, H. pylori must overcome natural defenses and the immune response to colonize and cause disease. H. pylori overcomes natural defenses of the stomach through production of urease which neutralizes the local acidic environment and through its flagella which provides the ability to traverse the mucus layer and interact with epithelial cells. Other bacterial factors which aid survival of H. pylori include its adherence to the epithelium, its ability to produce of catalase to neutralize hydrogen peroxide and its ability to acquire nutrients. Virulence factors, including the vacuolating toxin (vacA), the Cytotoxin A Type 4 secretion system (CagA T4SS), adhesins, among others, target the epithelial cells disrupting cell-cell communication, tight junctions and cellular signaling of the epithelium (9–11). Both bacterial interactions with GECs and the ongoing chronic inflammatory response to H. pylori contribute to carcinogenesis. This review will focus on how the chronic inflammatory process, especially T cell activation, impacts the epithelial cell response.
The Importance of the T cell Response during Helicobacter pylori infection
The innate immune response to H. pylori is activated through direct interactions of H. pylori or its products with the GECs. Both humoral and cellular immunity become chronically active, but this response is not effective in bacterial clearance. The cellular immune infiltrate in response to H. pylori is dominated by innate immune cells and CD4+ T cells (Figure 2). The CD4+ T cells response is instrumental during infection in mice and humans for the development of gastritis and control of infection. T and B cell deficient mice (recombination-activating gene or severe combined-immunodeficient mice (SCID)) do not control H. pylori colonization, nor do they exhibit inflammation like wild-type (WT) mice (12, 13). Reconstituting these immune deficient mice with CD4+ T cells, results in severe gastritis suggesting that CD4+ T cells are both necessary and sufficient for disease. MHC Class II−/− mice, which lack CD4+ T cells responses, are unable to control H. pylori colonization as well as WT mice and they do not develop a protective response to immunization (14). Utilizing antigen-specific adoptive transfer models has confirmed the importance of the CD4+ T cell on driving gastritis through epithelial cell damage, associated proliferative and metaplastic responses. Peterson et al. reconstituted SCID mice with congenic splenocytes from H. pylori infected or naïve WT mice, and found significant increases in GEC apoptosis and proliferation in infected recipient and donor, compared with non-recipient and uninfected mice at 3 months post transfer (15). These data suggest that splenocytes from H. pylori infected mice and the factors they produce contribute to GEC turnover by inducing apoptosis and proliferation.
Figure 2.
CD4+ T lymphocytes subsets migrate to the gastric mucosa in response to Helicobacter pylori infection and where they produce cytokines which impact epithelial cell function. In this figure, the immune cell filtrate in the lamina propria is represented. Green cells represent CD4+ T cells; blue cells represent B lymphocytes; purple cells represent dendritic cells and macrophages; while pink cells represent neutrophils. Th1 and Th17 cells dominate the response during gastritis producing IFNγ, IL-17, IL-21 and IL-22 (possibly produced by Th22 cells). Th2 lymphocytes are not commonly activated during H. pylori infection, but when present do impact epithelial cell responses. Finally, T regulatory cells are activated during H. pylori and their presence can influence not only other T cells but also epithelial cell biology through production of IL-10 and TFGβ.
It is important to recognize the double-edged sword of a T helper (Th) cell response. T cell derived cytokines are required for activating antimicrobial responses antibody responses against pathogens; but on the other hand, there is collateral damage chronic, exacerbated or unregulated pro-inflammatory responses. The balance between pro-inflammatory Th cells and Tregs cells effect H. pylori immune responses and gastric disease (16–18) whereby low Treg response leads to increased inflammation and gastric disease. CD4+ T cell cytokines are often categorized by the function of the T cell which produces them (19). For example, Th1 cells produce IFNγ; Th2 cells produce IL-4, IL-5 and IL-13; Th17 cells produce IL-17a, IL-17f, IL-21 and IL-22; Th22 cells produce IL-22; Tregs produce IL-10 and TGFβ. Many of these cytokines can play a role in activating antimicrobial responses, wound healing, proliferation and carcinogenesis through their signaling in epithelial cells.
The Epithelial Cell and Antigen Presenting Cell Response to H. pylori triggers Immune Activation
Gastritis is the culmination of innate and adaptive immune response which is initiated at the intersection of H. pylori contact with GECs. H. pylori activates the gastric epithelium to produce CXCL8 (IL-8, a neutrophil chemokine), IL-6, IL-1β, granulocytes-macrophage colony stimulating factor (GM-CSF), monocyte chemoattractant protein-1 (CCL2), macrophage migration inhibitory factor (MIF), tumor necrosis factor alpha (TNFα), and tumor growth factor beta (TGFβ) (20, 21). Many reviews have been written on the direct interaction between H. pylori and epithelial cells, so we will only briefly focus on the characteristics of H. pylori that lead to epithelial cell activation and strongly influence the T cell response.
One of the most important virulence factors of H. pylori is the the Cytotoxin A Type 4 Secretion system (Cag T4SS). It is encoded by the cag pathogenicity island (cagPAI), and this secretion system is responsible for translocating the effector protein CagA, peptidoglycan metabolites and DNA directly into epithelial cells. The functioning of this T4SS has been shown to be vital for carcinogenesis and has been referred to as an oncogenic factor. The biosynthesis of heptose-1,7-bisphosphate (HBP), an intermediate metabolite of LPS, contributes to cagPAI-dependent activation of epithelial cells (22). OipA is a phase-variable outer membrane protein which is postulated to bind to an integrin (23). Experimental analysis of phase on/off mutants of H. pylori demonstrate that OipA is necessary but not sufficient for CagA translocation into host epithelial cells (24). The significance of the T4SS dependent activation of epithelial cells is activation of proinflammatory signaling that induces the transcriptional activation and secretion of chemokines such as CXCL8 (IL-8) which recruits neutrophils to the site of infection. Polarization of the Th cell response is influenced in H. pylori infected humans both by whether the patient is infected with a CagA-positive strain and their stage of progression to gastric carcinogenesis (25). Patients colonized with CagA+ strains had Th1-mediated cellular immunity in earlier stages of gastric carcinogenesis. Patients colonized with CagA+ strains in advanced stages were dominated by Th2-mediated humoral immunity and negatively associated with a large number of Treg cells.
Several soluble or secreted proteins of H. pylori also directly impact the function of gastric epithelial cells and antigen presenting cells. VacA, a pore-forming toxin, induces mitochondrial damage, and contributes to epithelial cell death through apoptosis and programmed necrosis (26, 27). Large amounts of urease produced by H. pylori to accommodate its survival in the stomach’s acidic environment, induce production of IL-6 and TNFα in primary GECs and in the human GEC line MKN-45 (20). In addition, urease has been shown to contribute to apoptosis and inflammation through its interaction with host cells. The interaction of urease with CD74 on host cells contributes to CXCL8 production by GECs (28); while urease binding to MHC Class II contributes to apoptosis of GECs (29). These are relevant findings because they impact antigen uptake by recruited phagocytes and expression of T cell skewing cytokines.
The epithelial cell layer is intended to be a major defensive part of the innate immune response setting off alarms through toll-like receptors and other pattern recognition receptors (PRR). Disruption of the epithelial cell barrier by virulence factors leads to the innate activation of the immune response (reviewed in (23)) and trigger innate activation of antigen presenting cells (APCs) (30, 31). For example, high temperature regulator A (HtrA) disrupts epithelial barrier through cleavage of the ectodomain of E-cadherin (32, 33), permitting H. pylori, outer membrane vesicles, and H. pylori soluble factors to invade spaces between GECs (34).
As an extracellular bacterium that produces several secreted factors, H. pylori activates a strong CD4+ T cell response (31, 35). Activation, proliferation and differentiation of CD4+ T cells requires several signals (35–39). The first and second signals are mediated through the T cell’s receptor with its cognate peptide loaded MHC molecule on the APC (40); and through co-stimulatory molecule interactions between the T cell expressed CD28 molecule and B7.1 or B7.2 molecules on the APC (41). These two signals result in activation, clonal expansion and proliferation of the T cells. A third signal is required for T cell differentiation, and is provided by the cytokine microenvironment (42). Although it is widely accepted that H. pylori does not activate APCs as strongly as other GI pathogens, APC produce several T cell differentiating cytokines including (but not limited to) IL-1β, IL-10, IL-12, IL-18, IL-23, and TGFβ in response to H. pylori (43–45). These cytokines are induced through PRRs and inflammasome activation (46–48) triggering differentiation of Th1 and Th17 responses. Interestingly, it has been demonstrated that H. pylori factors including VacA and gamma-glutamyl transpeptidase (GGT) downregulate the magnitude of the proinflammatory T cell response. Specifically, GGT converts glutamine into glutamate and ammonia and converts glutathione into glutamate and cysteinylglycine (reviewed in (49)). H. pylori GGT induces immune tolerance through the inhibition of T cell-mediated immunity and antigen presenting cell cytokine production. VacA impacts several cell types as a pore-forming toxin. It can directly inhibit T cell proliferation and IL-2 production, but also indirectly impacts T cell differentiation to targeting myeloid cells especially in young mouse models to induce tolerance (reviewed in (50)) VacA and GGT similarly impact dendritic cell activity (promoting Treg differentiation) and inhibit T cell proliferation.
Th1-lymphocyte derived cytokines
IFNγ-producing CD4+ T cells, Th1 cells, are activated during H. pylori infection. Increased presence of Th1 cells (increased IFNγ) are associated with more detrimental outcomes of H. pylori infection (51, 52). IFNγ acts synergistically with TNFα to activate macrophages and NK cells, but most mucosal epithelial cells also express the IFNγ receptor at the basolateral membrane (53). IFNγ signals activation and nuclear translocation of signal transducer and activator of transcription 1 (STAT1). IFNγ induces several chemokines which regulate mucosal T cell trafficking. These CXC chemokines, CXCL9, CXCL10 and CXCL11, specifically attract CXCR3+CD4+ T cells. IFNγ and TNFα strongly induced secretion of these chemokines in GECs (Kato III, NCI, and AGS cells)(54). Interestingly, in some settings H. pylori may inhibit STAT1 activation by IFNγ (55); and soluble or membrane fractions of H. pylori have been shown to prevent the chemokine expression in the above mentioned GECs (54). Conversely, in another study cagPAI+ H. pylori strains augmented GEC responses to IFNγ and specifically increased CXCL9, CXCL10 and CXCL11 expression (56). Other chemokines are also induced by H. pylori and IFNγ. The murine GEC line, GSM06, responded synergistically to H. pylori and IFNγ to induce CXCL2 and iNOS expression supporting inflammation (57). Results from H. pylori infection of IFNγ−/− mice suggest that IFNγ plays a primary role in driving inflammation with minimal effect on colonization levels (58).
The turnover of mucosal epithelial cells is vital for homeostasis when the epithelial cell layer is colonized with inflammation-inducing bacterial colonizers or virulent bacterium. Homeostasis is maintained through a process of apoptosis balanced with proliferation. Inflammation and cytokines can disrupt this balance. Specifically, IFNγ enhances apoptosis during H. pylori infection. This has been demonstrated in organoid cultures (53) and in cell culture assays with human GECs (AGS cells and Kato III cells)(59). Mechanistically, IFNγ directly enhances caspase 3 activation and indirectly induces increased expression of MHC Class II molecules on the surface of GECs to which H. pylori can bind and trigger apoptosis through Bax activation. IFNγ clearly has a pro-apoptotic role on GECs, but it is also vital for enhancing gastritis through activation of chemotactic gradients which enhance immune cell infiltration to the gastric mucosa.
There is also evidence that IFNγ can alter the lineage, and therefore, the function of GECs. The human GEC line, NCI-N87 increased it expression of mucus, mucin 6 (Muc6), trefoil factor 2 (Tff2) and pepsinogen II in response to IFNγ, suggesting NCI-N87 cells became more like a mucous neck cell with this treatment (60). Infusion of IFNγ (in mice) expanded the mucous neck cell compartment and led to the proliferation of a gastric progenitor cell in the antral gland which can give rise to all gastric lineages in those glands (61). These were key experiments for understanding connections between inflammation and carcinogenesis.
Th17-lymphocyte derived cytokines
The Th17 subset also contributes to the immunopathogenic response to H. pylori. Th17 cells are differentiated in the presence of IL-1β, IL-6 and TGFβ; the subset is transcriptionally stabilized and expanded in the presence of IL-23 and IL-21. Once differentiated, Th17 cells make several cytokines including IL-17a, IL-17f, IL-21 and IL-22.
IL-17a is the cytokine which has the greatest affinity for IL-17RA/RC on epithelial cells and its canonical activation of the IL-17R leads to activation of several transcription factors including NFκB, AP-1 and C/EBP (62). The major consequence of IL-17 signaling is induction of a chemotactic response recruiting PMNs (through activation of CXCL1, CXCL2, CCL2 and CCL5). This tight association between IL-17A expression and CXCL8 induction by epithelial cells has been investigated in H. pylori infected patients. Increased levels of IL-17A and CXCL8 are reported during H. pylori infection compared to uninfected patients (63, 64). Moreover, expression of IL-17A and CXCL8 correlates with PMN infiltration in the H. pylori infected patients. Further, it has been demonstrated that isolated lamina propria mononuclear cells from H. pylori infected patients can induce the production of CXCL8 in an IL-17A dependent fashion for inhibition of IL-17 using an anti-IL-17A antibody lead to decreased CXCL8 expression (63). Interestingly, IL-17 alone is not a strong inducer of NFκB, but it is its synergistic response with IFNγ, lymphotoxin and TNFα leads to a strong cellular response (65). In the case of co-stimulation with TNFα, IL-17 signaling leads to increases in mRNA stability of IL-17 target genes (66, 67).
Central to regulation of IL-17 signaling is the Act1 protein and several TNF associated factors (TRAFs). Act1, a U-box E3 ubiquitin ligase, is recruited to the IL-17R (and ubiquitinates TRAF6 (68)). These events are critical for NFκB activation; but not required for the mRNA stability processes mediated by IL-17 signaling (67). On the other hand, TRAF4 competes for the same binding site on Act1 as TRAF6. While TRAF4 restricts the level of NFκB activation as a result of IL-17 signaling (69), it can also activate signaling in a MEKK3 dependent pathway. TRAF4 mediated signaling has been studied extensively in keratinocytes (70) and while the importance of these signaling events in GECs is yet to be determined, they may have implications in ulcer-healing or carcinogenesis. In keratinocytes, TRAF4 can also mediate MEKK3-dependent extracellular signal-regulated kinase 5 (ERK5) activation inducing Steap4 which is critical for cellular metabolism and proliferation (70). Moreover, TRAF4 can also transactivate the epidermal growth factor receptor (EGFR). Chen et al. demonstrate that Act1–TRAF4–ERK5 signaling is a result of IL-17a induced EGFR transactivation and critical for stimulating epidermal hyperplasia and tumor growth in keratinocytes (71). Another regulator, TRAF3, interferes with Act1 binding to IL-17R providing a proximal regulation of IL-17 signaling (72). TRAF3 is also regulated by Nuclear Dbf2‐related kinase 1 (NDR1). NDR1 can bind to TRAF3 preventing it from binding to IL-17R, which allows for the formation of the IL-17R-Act1-TRAF6- signaling complex formation and canonical activation of IL-17 signaling.
IL-17a and IL-22 act synergistically to activate antimicrobial responses in epithelial cells (reviewed in (73)). It is difficult to separate the importance of Th17-mediated induction of neutrophil recruitment from the importance of activation of antimicrobial proteins (AMPs). Nevertheless, there is a clear trend toward the need for a Th17 response to control H. pylori; both PMNs and AMP production are likely contributing factors. IL-17a and IL-22 together enhanced the ability of GECs to upregulate expression of calprotectin, lipocalin and some β-defensins (74). Further, activating GECs with IL-17a and IL-22 increased their ability to kill H. pylori in vitro compared to treatment with each cytokine alone (74).
IL-17f also signals through the IL-17RA/RC complex, but research on mechanisms by which IL-17f acts on GECs or in the tissue during H. pylori infection is minimal. Since IL-17a appears to be the predominately produced cytokine compared to IL-17f during H. pylori infection (75), an effect of IL-17f may be minimal and/or some IL-17R signaling may be redundant. Several studies have investigated whether polymorphism in the IL-17f gene are linked to gastric cancer, but there is no consistency in their findings (76). Still, there is no profound understanding of expression of these receptors on GEC subsets and regulation of these receptors by inflammatory stimuli.
IL-22 is another T cell-derived cytokine which can activate antimicrobial and inflammatory epithelial cell functions, especially in the context of H. pylori infection(77). While we have placed the IL-22 under the Th17 cytokine umbrella for this review, this cytokine may also be produced by Th22 cells. In fact, during H. pylori infection, while IL-22 expression has been reported, it is not clear if the cells producing the cytokine are Th17 or Th22 cells. IL-22 signals through a heterodimeric receptor of IL-22RA1 and IL-10Rβ. Since IL-10Rβ is constitutively expressed, regulation of IL-22 signaling begins with regulated expression of IL-22RA1. IL-22 activates STAT3 signaling and enhances gene transcription of select genes involved in AMPs. Increased expression of IL-22 is reported in H. pylori infected mice and humans (74, 77), and its expression correlates with the level of H. pylori colonization and level of gastritis (77). Remarkably, GECs upregulated the IL-22RA1 in response to H. pylori in a CagA dependent manner (77). The influence of IL-22 on GEC is not clear cut. In one report, IL-22 downregulated CCL20 expression of H. pylori induced AGS cells, suggesting IL-22 may play a role protecting the gastric mucosa from damage induced by inflammation (78). On the other hand, IL-22−/− mice have reduced expression of CXCL2 in response to H. pylori infection compared to WT Balb/c mice (77). This reduced CXCL2 expression correlates with reduced influx of myeloid derived suppressor cells, reduced expression of calprotectin, and increased Th1 responses (77) suggesting a proinflammatory role for IL-22 in this mouse model.
IL-22 has also been shown to induce expression of matrix metalloprotease-10 (MMP-10) in GECs activating host defense pathways (79). MMP-10 expression is increased in parietal cells and chief cells in the H. pylori infected gastric mucosa, and IL-22 (not IL-17a or IFNγ) enhanced H. pylori’s activation of MMP-10 expression in AGS cells in an ERK dependent pathway (79). MMP-10 activates GECs and H. pylori induced inflammation. MMP-10−/− mice as well as MMP-10−/−/IL-22−/− mice have reduced gastritis associated with decreased expression of CXCL16 and reduced CD8+ T cell migration (79). Interestingly, the authors of this study also found that Reg3a, ZO-1 and occludin expression was inhibited by MMP-10, and they hypothesize that MMP-10 may facilitate persistence and damage to the epithelial cell layer through these pathways (79). Overall, these studies suggest that IL-22 dependent GEC activation contributes to regulating the gastritis response to H. pylori.
Th2-lymphocyte derived cytokines
Th2 cells are typically thought of as the CD4+ T cells which produce IL-4, IL-5 and IL-13 with the goal of helping to activate the humoral response and mast cells. Binding of IL-4 or of IL-13 initiates Jak-dependent tyrosine phosphorylation of the IL-4Rα-chain and STAT6. Activation of STAT6 in GECs is required for the gastrointestinal response to nematode infections or parasites (80). Th2 responses are not strongly activated during H. pylori infection, relative to Th1 or Th17 responses in humans, rhesus macaques, gerbils or mice (81–85); but the cytokines produced by Th2 cells may indirectly downregulate gastritis. In fact, even though C57Bl/6 WT mice do not demonstrate an increase in IL-4 expression in response to H. pylori infection, IL-4−/− mice do exhibit an increase in IFNγ expression and increased gastritis (82). Therefore, understanding how IL-4 might regulate IFNγ responses may increase our understanding of how these cytokines inhibit or activate GEC responses. During chronic gastritis, atrophy of gastric glands has been reported resulting in a decrease in the number of D cells (SOM secreting cells) and an increase in the number of G cells (gastrin secreting cells) (86, 87). The balance of Th1 and Th2 cytokines, specifically IFNγ and IL-4, may influence the turnover and function of these neuroendocrine cell populations in the gastric mucosa of chronic Helicobacter infection; for IL-4 provides a stimulus for SOM release from D cells and therefore, provides protection from Th1 mediated inflammation in the H. felis model (88).
IL-13 is also produced by Th2 cells, but there is little data on the direct role of IL-13 in GEC function. The IL-13R has been detected on chief cells and in models of induced spasmolytic polypeptide-expressing metaplasia. IL-13 was shown to be necessary and sufficient for chief cell transdifferentiation into metaplasia following parietal cell loss (89). In a small study of 23 H. pylori positive subjects, IL-13 localized to the inflammatory infiltrate (90). More specifically, in cases where the patients had intestinal metaplasia or intestinal type gastric cancer, IL-13 also localized to the GECs (90). The impact of IL-13 on intestinal epithelial cells has been studied, but its impact on GECs is largely undescribed in the literature.
Treg-derived cytokines
The presence of Tregs reduces the pathological outcomes of H. pylori infection in mice and human (91–95). Generally, the presence of Tregs is correlated with reduced gastritis, but at the expense of greater levels of H. pylori colonization. Mouse models demonstrate that depletion of Tregs leads to increased levels of IFNγ and increased inflammation (96). Tregs may be recruited to the H. pylori colonized mucosa via CCL20/CCR6 mediated chemotaxis orchestrated by the GEC response. CCL20 is upregulated in gastric biopsies of H. pylori infected patients (97); and further, CCL20 induction in GECs is cagPAI dependent. CCL20 expression was shown to induce Treg migration; for instance, CD4+Foxp3+ T cells (Tregs) isolated from H. pylori infected patients migrated to recombinant CCL20 in vitro while CD4+Foxp3− T cells did not (97).
Tregs are defined by their ability to regulate pro-inflammatory T cell responses through direct activation of apoptosis, through secretion of inhibitory cytokines, or by depleting the microenvironment of extracellular ATP and IL-2 (98, 99). Tregs not only target T cells, but also may target GECs through production of IL-10 and TGFβ. While IL-10 expression may be protective during inflammation, once cancer develops IL-10 be detrimental. In fact, increased IL-10 in the serum is an unfavorable prognostic marker for GC (100). IL-10 in the tumor microenvironment may be produced by a number of cell types. One potential mechanism by which IL-10 could stimulate GECs in the tumor is through activating c-Met-STAT3. GEC lines responded to culture supernatants cancer-associated macrophages (CAMs) in an IL-10 dependent way to induce proliferation and migration, while it also suppressed apoptosis (101).
TGF-β is also produced by Treg cells (and other immune cells), and may act as a tumor suppressor in early stages but a tumor promoter in later stages (102). TGFβ signals through many cell types including fibroblasts and mesothelial cells, inducing extracellular matrix protein synthesis and contributing to fibrogenesis (103). TGFβ1 and TGFβ2 bind to TGFβ receptor I through TGFβ receptor II (TGFβRII) to activate its serine/threonine kinase. The activated dimeric receptor then activates a complex of Smad proteins which facilitates transcription of target genes (104). To investigate the role of TGFβ signaling during H. pylori infection, mice which overexpress the dominant negative mutant of the TGFβ RII in their stomach were generated (pS2-dnRII) (105). When these mice and WT littermates were infected with H. pylori, the mice lacking TGFβ signaling developed gastric adenocarcinoma with high cell proliferation index in their epithelial cells compared to their WT littermates, suggesting TGFβ provides a protective effect suppressing carcinogenesis (105). In vitro studies performed on GEC lines (BGC823 and MKN-45) demonstrated that TGFβ treatment downregulated expression of E-cadherin significantly, while it upregulated expression of epithelial mesenchymal transition-related proteins, snail and vimentin (106); interestingly, upstream of TGFβ signaling, MFAP2 is activated and promotes this activity. Increased levels of TGFβ in the serum and gastric tissue is associated with advanced cancer (107–109) and expression appears to be higher in the tissues of intestinal type GC than diffuse type GC (110). Tumor growth may be promoted by TGFβ by inducing epithelial cells to undergo epithelial-mesenchymal transition through upregulating long noncoding RNA urothelial carcinoma associated 1 (LncRNA UCA1), which when silenced in GC cell lines reduced the levels of snail and vimentin and promoted expression of E-cadherin and ZO-1 (111). Moreover, erythrocyte membrane protein band 4.1-like 5 is also a target of TGFβ signaling and regulates GC cell metastasis (112). So, while Tregs may play a role downregulating the inflammatory response to H. pylori, Tregs may be detrimental during GC since Tregs and their cytokines activate pro-carcinogenic pathways in the tumor microenvironment.
Conclusions
The inflammatory response to H. pylori significantly influences the outcome of infection, and while this response involves the innate and the adaptive immune response, T lymphocytes play a major role in orchestrating the gastritis. CD4+ T cell-derived cytokines amplify the innate response by recruiting PMNs and macrophages to the site of infection, but they can also directly impact epithelial cell biology and carcinogenesis. Our understanding of how many of the T cell-derived cytokines impact GEC functions has increased in the last decade, but there are many gaps in knowledge to address. We lack understanding of the temporal activation of the epithelial cell response. Pro-inflammatory responses are necessary for control of the bacterial colonization, but chronically can lead to carcinogenesis; however, sustaining pro-inflammatory responses may allow for anti-tumor responses and a favorable prognosis. Secondly, many studies that have investigated the GEC response to H. pylori and/or T cell-derived cytokines have been performed in a reductionist manner when in fact, there appears to be significant cross-regulation of epithelial cell responses by cytokines. Further, there is evidence that increased numbers and mislocalized stem cell presence in gastric pits is associated with carcinogenesis. The links between inflammation and T cell- derived cytokines in the gastric tissue has largely been unexplored. Finally, the increased use of gastroids is vital for our increased understanding of the consequence of expression and signaling of these cytokines and H. pylori on the specialized cells of the gastric mucosa. Gastroids combined with single cell analyses will inform which cells respond to the stimuli and how. Research in this area will help identify appropriate promising therapeutic targets for persons with detrimental outcomes of H. pylori infection from chronic gastritis to the different types of GC.
Acknowledgments
I acknowledge the work of my Research Specialist, Beverly R.E.A. Dixon, M.S., for her continued support of our research program and for her contributions editing this brief review.
This work was supported by Office of Medical Research, Veterans Affairs Merit Review grant IBX000915A (to H.M.S.A.). The content is solely the responsibility of the authors and does not necessarily represent the official views of the Department of Veteran’s Affairs.
Abbreviations and acronyms:
- CCL or CXCL
Chemokine
- Cag T4SS
Cytotoxin-associated Type IV secretion system
- CagA
Cytotoxin A
- ECL
enteroendocrine cells
- GC
gastric cancer
- GECs
gastric epithelial cells
- H. pylori
Helicobacter pylori
- IL-
Interleukin
- IFNγ
Interferon gamma
- NFκB
Nuclear Factor kappa-light-chain-enhancer of activated B cells
- Treg
Regulatory CD4+ T cell
- STAT
Signal Transducer And Activator Of Transcription
- SOM
Somatostatin
- Th
T helper cell
- TNFα
Tumor Necrosis Factor alpha
- VacA
Vacuolating toxin A
- WT
Wild type
- knockout
−/−
Footnotes
Disclosures
No financial conflicts of interest.
References
- 1.OpenStax. 2016. The Digestive System In Anatomy & Physiology. OpenStax CNX. [Google Scholar]
- 2.Fromm D 1987. How do non-steroidal anti-inflammatory drugs affect gastric mucosal defenses? Clinical and investigative medicine. Medecine clinique et experimentale 10: 251–258. [PubMed] [Google Scholar]
- 3.Loguercio C, Tuccillo C, Federico A, Fogliano V, Del Vecchio Blanco C, and Romano M. 2009. Alcoholic beverages and gastric epithelial cell viability: effect on oxidative stress-induced damage. Journal of physiology and pharmacology : an official journal of the Polish Physiological Society 60 Suppl 7: 87–92. [PubMed] [Google Scholar]
- 4.Wroblewski LE, and Peek RM Jr. 2011. Targeted disruption of the epithelial-barrier by Helicobacter pylori. Cell communication and signaling : CCS 9: 29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Kotilea K, Bontems P, and Touati E. 2019. Epidemiology, Diagnosis and Risk Factors of Helicobacter pylori Infection. Advances in experimental medicine and biology 1149: 17–33. [DOI] [PubMed] [Google Scholar]
- 6.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, and Ng SC. 2017. Global Prevalence of Helicobacter pylori Infection: Systematic Review and Meta-Analysis. Gastroenterology 153: 420–429. [DOI] [PubMed] [Google Scholar]
- 7.Kato S, Matsukura N, Tsukada K, Matsuda N, Mizoshita T, Tsukamoto T, Tatematsu M, Sugisaki Y, Naito Z, and Tajiri T. 2007. Helicobacter pylori infection-negative gastric cancer in Japanese hospital patients: incidence and pathological characteristics. Cancer science 98: 790–794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, and Jemal A. 2018. Global Cancer Statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries In CA Cancer J Clin. I. A. f. R. o. C. (IARC), ed, The online GLOBOCAN 2018 database is accessible at http://gco.iarc.fr/. [DOI] [PubMed] [Google Scholar]
- 9.Chmiela M, and Kupcinskas J. 2019. Review: Pathogenesis of Helicobacter pylori infection. Helicobacter 24 Suppl 1: e12638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Alzahrani S, Lina TT, Gonzalez J, Pinchuk IV, Beswick EJ, and Reyes VE. 2014. Effect of Helicobacter pylori on gastric epithelial cells. World journal of gastroenterology 20: 12767–12780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Javed S, Skoog EC, and Solnick JV. 2019. Impact of Helicobacter pylori Virulence Factors on the Host Immune Response and Gastric Pathology. Current topics in microbiology and immunology 421: 21–52. [DOI] [PubMed] [Google Scholar]
- 12.Gray BM, Fontaine CA, Poe SA, and Eaton KA. 2013. Complex T cell interactions contribute to Helicobacter pylori gastritis in mice. Infection and immunity 81: 740–752. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Eaton KA, Ringler SR, and Danon SJ. 1999. Murine splenocytes induce severe gastritis and delayed-type hypersensitivity and suppress bacterial colonization in Helicobacter pylori-infected SCID mice. Infection and immunity 67: 4594–4602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Pappo J, Torrey D, Castriotta L, Savinainen A, Kabok Z, and Ibraghimov A. 1999. Helicobacter pylori infection in immunized mice lacking major histocompatibility complex class I and class II functions. Infection and immunity 67: 337–341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Peterson RA 2nd, Hoepf T, and Eaton KA. 2003. Adoptive transfer of splenocytes in SCID mice implicates CD4+ T cells in apoptosis and epithelial proliferation associated with Helicobacter pylori-induced gastritis. Comparative medicine 53: 498–509. [PubMed] [Google Scholar]
- 16.Serrano C, Wright SW, Bimczok D, Shaffer CL, Cover TL, Venegas A, Salazar MG, Smythies LE, Harris PR, and Smith PD. 2013. Downregulated Th17 responses are associated with reduced gastritis in Helicobacter pylori-infected children. Mucosal immunology 6: 950–959. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Jafarzadeh A, Larussa T, Nemati M, and Jalapour S. 2018. T cell subsets play an important role in the determination of the clinical outcome of Helicobacter pylori infection. Microbial pathogenesis 116: 227–236. [DOI] [PubMed] [Google Scholar]
- 18.Bagheri N, Salimzadeh L, and Shirzad H. 2018. The role of T helper 1-cell response in Helicobacter pylori-infection. Microbial pathogenesis 123: 1–8. [DOI] [PubMed] [Google Scholar]
- 19.Raphael I, Nalawade S, Eagar TN, and Forsthuber TG. 2015. T cell subsets and their signature cytokines in autoimmune and inflammatory diseases. Cytokine 74: 5–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Tanahashi T, Kita M, Kodama T, Yamaoka Y, Sawai N, Ohno T, Mitsufuji S, Wei YP, Kashima K, and Imanishi J. 2000. Cytokine expression and production by purified Helicobacter pylori urease in human gastric epithelial cells. Infection and immunity 68: 664–671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Beswick EJ, Pinchuk IV, Suarez G, Sierra JC, and Reyes VE. 2006. Helicobacter pylori CagA-dependent macrophage migration inhibitory factor produced by gastric epithelial cells binds to CD74 and stimulates procarcinogenic events. Journal of immunology 176: 6794–6801. [DOI] [PubMed] [Google Scholar]
- 22.Stein SC, Faber E, Bats SH, Murillo T, Speidel Y, Coombs N, and Josenhans C. 2017. Helicobacter pylori modulates host cell responses by CagT4SS-dependent translocation of an intermediate metabolite of LPS inner core heptose biosynthesis. PLoS pathogens 13: e1006514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Posselt G, Backert S, and Wessler S. 2013. The functional interplay of Helicobacter pylori factors with gastric epithelial cells induces a multi-step process in pathogenesis. Cell communication and signaling : CCS 11: 77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Horridge DN, Begley AA, Kim J, Aravindan N, Fan K, and Forsyth MH. 2017. Outer inflammatory protein a (OipA) of Helicobacter pylori is regulated by host cell contact and mediates CagA translocation and interleukin-8 response only in the presence of a functional cag pathogenicity island type IV secretion system. Pathogens and disease 75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Wang SK, Zhu HF, He BS, Zhang ZY, Chen ZT, Wang ZZ, and Wu GL. 2007. CagA+ H. pylori infection is associated with polarization of T helper cell immune responses in gastric carcinogenesis. World journal of gastroenterology 13: 2923–2931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Yamasaki E, Wada A, Kumatori A, Nakagawa I, Funao J, Nakayama M, Hisatsune J, Kimura M, Moss J, and Hirayama T. 2006. Helicobacter pylori vacuolating cytotoxin induces activation of the proapoptotic proteins Bax and Bak, leading to cytochrome c release and cell death, independent of vacuolation. The Journal of biological chemistry 281: 11250–11259. [DOI] [PubMed] [Google Scholar]
- 27.Radin JN, Gonzalez-Rivera C, Ivie SE, McClain MS, and Cover TL. 2011. Helicobacter pylori VacA induces programmed necrosis in gastric epithelial cells. Infection and immunity 79: 2535–2543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Beswick EJ, Pinchuk IV, Minch K, Suarez G, Sierra JC, Yamaoka Y, and Reyes VE. 2006. The Helicobacter pylori urease B subunit binds to CD74 on gastric epithelial cells and induces NF-kappaB activation and interleukin-8 production. Infection and immunity 74: 1148–1155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Fan X, Gunasena H, Cheng Z, Espejo R, Crowe SE, Ernst PB, and Reyes VE. 2000. Helicobacter pylori urease binds to class II MHC on gastric epithelial cells and induces their apoptosis. Journal of immunology 165: 1918–1924. [DOI] [PubMed] [Google Scholar]
- 30.Krauss-Etschmann S, Gruber R, Plikat K, Antoni I, Demmelmair H, Reinhardt D, and Koletzko S. 2005. Increase of antigen-presenting cells in the gastric mucosa of Helicobacter pylori-infected children. Helicobacter 10: 214–222. [DOI] [PubMed] [Google Scholar]
- 31.Suzuki T, Kato K, Ohara S, Noguchi K, Sekine H, Nagura H, and Shimosegawa T. 2002. Localization of antigen-presenting cells in Helicobacter pylori-infected gastric mucosa. Pathology international 52: 265–271. [DOI] [PubMed] [Google Scholar]
- 32.Lower M, Weydig C, Metzler D, Reuter A, Starzinski-Powitz A, Wessler S, and Schneider G. 2008. Prediction of extracellular proteases of the human pathogen Helicobacter pylori reveals proteolytic activity of the Hp1018/19 protein HtrA. PloS one 3: e3510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Hoy B, Lower M, Weydig C, Carra G, Tegtmeyer N, Geppert T, Schroder P, Sewald N, Backert S, Schneider G, and Wessler S. 2010. Helicobacter pylori HtrA is a new secreted virulence factor that cleaves E-cadherin to disrupt intercellular adhesion. EMBO reports 11: 798–804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Chmiela M, Walczak N, and Rudnicka K. 2018. Helicobacter pylori outer membrane vesicles involvement in the infection development and Helicobacter pylori-related diseases. Journal of biomedical science 25: 78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Algood HM, Gallo-Romero J, Wilson KT, Peek RM Jr., and Cover TL. 2007. Host response to Helicobacter pylori infection before initiation of the adaptive immune response. FEMS immunology and medical microbiology 51: 577–586. [DOI] [PubMed] [Google Scholar]
- 36.Odenbreit S, Puls J, Sedlmaier B, Gerland E, Fischer W, and Haas R. 2000. Translocation of Helicobacter pylori CagA into gastric epithelial cells by type IV secretion. Science 287: 1497–1500. [DOI] [PubMed] [Google Scholar]
- 37.Oertli M, Sundquist M, Hitzler I, Engler DB, Arnold IC, Reuter S, Maxeiner J, Hansson M, Taube C, Quiding-Jarbrink M, and Muller A. 2012. DC-derived IL-18 drives Treg differentiation, murine Helicobacter pylori-specific immune tolerance, and asthma protection. The Journal of clinical investigation 122: 1082–1096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Kranzer K, Sollner L, Aigner M, Lehn N, Deml L, Rehli M, and Schneider-Brachert W. 2005. Impact of Helicobacter pylori virulence factors and compounds on activation and maturation of human dendritic cells. Infection and immunity 73: 4180–4189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Niess JH, Brand S, Gu X, Landsman L, Jung S, McCormick BA, Vyas JM, Boes M, Ploegh HL, Fox JG, Littman DR, and Reinecker HC. 2005. CX3CR1-mediated dendritic cell access to the intestinal lumen and bacterial clearance. Science 307: 254–258. [DOI] [PubMed] [Google Scholar]
- 40.Burgdorf S, and Kurts C. 2008. Endocytosis mechanisms and the cell biology of antigen presentation. Current opinion in immunology 20: 89–95. [DOI] [PubMed] [Google Scholar]
- 41.Rudd CE, Taylor A, and Schneider H. 2009. CD28 and CTLA-4 coreceptor expression and signal transduction. Immunological reviews 229: 12–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Curtsinger JM, and Mescher MF. 2010. Inflammatory cytokines as a third signal for T cell activation. Current opinion in immunology 22: 333–340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Guiney DG, Hasegawa P, and Cole SP. 2003. Helicobacter pylori preferentially induces interleukin 12 (IL-12) rather than IL-6 or IL-10 in human dendritic cells. Infection and immunity 71: 4163–4166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Hafsi N, Voland P, Schwendy S, Rad R, Reindl W, Gerhard M, and Prinz C. 2004. Human dendritic cells respond to Helicobacter pylori, promoting NK cell and Th1-effector responses in vitro. Journal of immunology 173: 1249–1257. [DOI] [PubMed] [Google Scholar]
- 45.Yasmin S, Dixon B, Olivares-Villagomez D, and Algood HMS. 2019. IL-21 down-regulates dendritic cell cytokine responses to H. pylori and modulates T lymphocyte IL-17A expression in the Peyer’s patches during infection. Infection and immunity. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Pachathundikandi SK, Lind J, Tegtmeyer N, El-Omar EM, and Backert S. 2015. Interplay of the Gastric Pathogen Helicobacter pylori with Toll-Like Receptors. BioMed research international 2015: 192420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Li X, Liu S, Luo J, Liu A, Tang S, Liu S, Yu M, and Zhang Y. 2015. Helicobacter pylori induces IL-1beta and IL-18 production in human monocytic cell line through activation of NLRP3 inflammasome via ROS signaling pathway. Pathogens and disease 73. [DOI] [PubMed] [Google Scholar]
- 48.Semper RP, Mejias-Luque R, Gross C, Anderl F, Muller A, Vieth M, Busch DH, Prazeres da Costa C, Ruland J, Gross O, and Gerhard M. 2014. Helicobacter pylori-induced IL-1beta secretion in innate immune cells is regulated by the NLRP3 inflammasome and requires the cag pathogenicity island. Journal of immunology 193: 3566–3576. [DOI] [PubMed] [Google Scholar]
- 49.Ricci V, Giannouli M, Romano M, and Zarrilli R. 2014. Helicobacter pylori gamma-glutamyl transpeptidase and its pathogenic role. World journal of gastroenterology 20: 630–638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Djekic A, and Muller A. 2016. The Immunomodulator VacA Promotes Immune Tolerance and Persistent Helicobacter pylori Infection through Its Activities on T-Cells and Antigen-Presenting Cells. Toxins 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Bamford KB, Fan X, Crowe SE, Leary JF, Gourley WK, Luthra GK, Brooks EG, Graham DY, Reyes VE, and Ernst PB. 1998. Lymphocytes in the human gastric mucosa during Helicobacter pylori have a T helper cell 1 phenotype. Gastroenterology 114: 482–492. [DOI] [PubMed] [Google Scholar]
- 52.D’Elios MM, Manghetti M, De Carli M, Costa F, Baldari CT, Burroni D, Telford JL, Romagnani S, and Del Prete G. 1997. T helper 1 effector cells specific for Helicobacter pylori in the gastric antrum of patients with peptic ulcer disease. Journal of immunology 158: 962–967. [PubMed] [Google Scholar]
- 53.Osaki LH, Bockerstett KA, Wong CF, Ford EL, Madison BB, DiPaolo RJ, and Mills JC. 2019. Interferon-gamma directly induces gastric epithelial cell death and is required for progression to metaplasia. The Journal of pathology 247: 513–523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Kraft M, Riedel S, Maaser C, Kucharzik T, Steinbuechel A, Domschke W, and Luegering N. 2001. IFN-gamma synergizes with TNF-alpha but not with viable H. pylori in up-regulating CXC chemokine secretion in gastric epithelial cells. Clinical and experimental immunology 126: 474–481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Mitchell DJ, Huynh HQ, Ceponis PJ, Jones NL, and Sherman PM. 2004. Helicobacter pylori disrupts STAT1-mediated gamma interferon-induced signal transduction in epithelial cells. Infection and immunity 72: 537–545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Allison CC, Ferrand J, McLeod L, Hassan M, Kaparakis-Liaskos M, Grubman A, Bhathal PS, Dev A, Sievert W, Jenkins BJ, and Ferrero RL. 2013. Nucleotide oligomerization domain 1 enhances IFN-gamma signaling in gastric epithelial cells during Helicobacter pylori infection and exacerbates disease severity. Journal of immunology 190: 3706–3715. [DOI] [PubMed] [Google Scholar]
- 57.Obonyo M, Guiney DG, Harwood J, Fierer J, and Cole SP. 2002. Role of gamma interferon in Helicobacter pylori induction of inflammatory mediators during murine infection. Infection and immunity 70: 3295–3299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Sawai N, Kita M, Kodama T, Tanahashi T, Yamaoka Y, Tagawa Y, Iwakura Y, and Imanishi J. 1999. Role of gamma interferon in Helicobacter pylori-induced gastric inflammatory responses in a mouse model. Infection and immunity 67: 279–285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Shimada M, Ina K, Kyokane K, Imada A, Yamaguchi H, Nishio Y, Hayakawa M, Iinuma Y, Ohta M, Ando T, and Kusugami K. 2002. Upregulation of mucosal soluble fas ligand and interferon-gamma may be involved in ulcerogenesis in patients with Helicobacter pylori-positive gastric ulcer. Scandinavian journal of gastroenterology 37: 501–511. [DOI] [PubMed] [Google Scholar]
- 60.Kang W, Rathinavelu S, Samuelson LC, and Merchant JL. 2005. Interferon gamma induction of gastric mucous neck cell hypertrophy. Laboratory investigation; a journal of technical methods and pathology 85: 702–715. [DOI] [PubMed] [Google Scholar]
- 61.Qiao XT, Ziel JW, McKimpson W, Madison BB, Todisco A, Merchant JL, Samuelson LC, and Gumucio DL. 2007. Prospective identification of a multilineage progenitor in murine stomach epithelium. Gastroenterology 133: 1989–1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.McGeachy MJ, Cua DJ, and Gaffen SL. 2019. The IL-17 Family of Cytokines in Health and Disease. Immunity 50: 892–906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Luzza F, Parrello T, Monteleone G, Sebkova L, Romano M, Zarrilli R, Imeneo M, and Pallone F. 2000. Up-regulation of IL-17 is associated with bioactive IL-8 expression in Helicobacter pylori-infected human gastric mucosa. Journal of immunology 165: 5332–5337. [DOI] [PubMed] [Google Scholar]
- 64.Mizuno T, Ando T, Nobata K, Tsuzuki T, Maeda O, Watanabe O, Minami M, Ina K, Kusugami K, Peek RM, and Goto H. 2005. Interleukin-17 levels in Helicobacter pylori-infected gastric mucosa and pathologic sequelae of colonization. World journal of gastroenterology 11: 6305–6311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Awane M, Andres PG, Li DJ, and Reinecker HC. 1999. NF-kappa B-inducing kinase is a common mediator of IL-17-, TNF-alpha-, and IL-1 beta-induced chemokine promoter activation in intestinal epithelial cells. Journal of immunology 162: 5337–5344. [PubMed] [Google Scholar]
- 66.Hartupee J, Liu C, Novotny M, Li X, and Hamilton T. 2007. IL-17 enhances chemokine gene expression through mRNA stabilization. Journal of immunology 179: 4135–4141. [DOI] [PubMed] [Google Scholar]
- 67.Hartupee J, Liu C, Novotny M, Sun D, Li X, and Hamilton TA. 2009. IL-17 signaling for mRNA stabilization does not require TNF receptor-associated factor 6. Journal of immunology 182: 1660–1666. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Liu C, Qian W, Qian Y, Giltiay NV, Lu Y, Swaidani S, Misra S, Deng L, Chen ZJ, and Li X. 2009. Act1, a U-box E3 ubiquitin ligase for IL-17 signaling. Science signaling 2: ra63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Zepp JA, Liu C, Qian W, Wu L, Gulen MF, Kang Z, and Li X. 2012. Cutting edge: TNF receptor-associated factor 4 restricts IL-17-mediated pathology and signaling processes. Journal of immunology 189: 33–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Wu L, Chen X, Zhao J, Martin B, Zepp JA, Ko JS, Gu C, Cai G, Ouyang W, Sen G, Stark GR, Su B, Vines CM, Tournier C, Hamilton TA, Vidimos A, Gastman B, Liu C, and Li X. 2015. A novel IL-17 signaling pathway controlling keratinocyte proliferation and tumorigenesis via the TRAF4-ERK5 axis. The Journal of experimental medicine 212: 1571–1587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Chen X, Cai G, Liu C, Zhao J, Gu C, Wu L, Hamilton TA, Zhang CJ, Ko J, Zhu L, Qin J, Vidimos A, Koyfman S, Gastman BR, Jensen KB, and Li X. 2019. IL-17R-EGFR axis links wound healing to tumorigenesis in Lrig1(+) stem cells. The Journal of experimental medicine 216: 195–214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Zhu S, Pan W, Shi P, Gao H, Zhao F, Song X, Liu Y, Zhao L, Li X, Shi Y, and Qian Y. 2010. Modulation of experimental autoimmune encephalomyelitis through TRAF3-mediated suppression of interleukin 17 receptor signaling. The Journal of experimental medicine 207: 2647–2662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Eyerich K, Dimartino V, and Cavani A. 2017. IL-17 and IL-22 in immunity: Driving protection and pathology. European journal of immunology 47: 607–614. [DOI] [PubMed] [Google Scholar]
- 74.Dixon BR, Radin JN, Piazuelo MB, Contreras DC, and Algood HM. 2016. IL-17a and IL-22 Induce Expression of Antimicrobials in Gastrointestinal Epithelial Cells and May Contribute to Epithelial Cell Defense against Helicobacter pylori. PloS one 11: e0148514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Algood HM, Allen SS, Washington MK, Peek RM Jr., Miller GG, and Cover TL. 2009. Regulation of gastric B cell recruitment is dependent on IL-17 receptor A signaling in a model of chronic bacterial infection. Journal of immunology 183: 5837–5846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Li XF, Shen M, Cai JW, Zeng YQ, Li M, Yang GL, Xu XM, and Hu YY. 2015. Association of interleukin-17 gene polymorphisms and Helicobacter pylori infection with gastric cancer susceptibility: a cumulative and comprehensive meta-analysis. International journal of clinical and experimental medicine 8: 17623–17633. [PMC free article] [PubMed] [Google Scholar]
- 77.Zhuang Y, Cheng P, Liu XF, Peng LS, Li BS, Wang TT, Chen N, Li WH, Shi Y, Chen W, Pang KC, Zeng M, Mao XH, Yang SM, Guo H, Guo G, Liu T, Zuo QF, Yang HJ, Yang LY, Mao FY, Lv YP, and Zou QM. 2015. A pro-inflammatory role for Th22 cells in Helicobacter pylori-associated gastritis. Gut 64: 1368–1378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Chen JP, Wu MS, Kuo SH, and Liao F. 2014. IL-22 negatively regulates Helicobacter pylori-induced CCL20 expression in gastric epithelial cells. PloS one 9: e97350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Lv YP, Cheng P, Zhang JY, Mao FY, Teng YS, Liu YG, Kong H, Wu XL, Hao CJ, Han B, Ma Q, Yang SM, Chen W, Peng LS, Wang TT, Zou QM, and Zhuang Y. 2019. Helicobacter pylori-induced matrix metallopeptidase-10 promotes gastric bacterial colonization and gastritis. Science advances 5: eaau6547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Madden KB, Whitman L, Sullivan C, Gause WC, Urban JF Jr., Katona IM, Finkelman FD, and Shea-Donohue T. 2002. Role of STAT6 and mast cells in IL-4- and IL-13-induced alterations in murine intestinal epithelial cell function. Journal of immunology 169: 4417–4422. [DOI] [PubMed] [Google Scholar]
- 81.Karttunen R, Karttunen T, Ekre HP, and MacDonald TT. 1995. Interferon gamma and interleukin 4 secreting cells in the gastric antrum in Helicobacter pylori positive and negative gastritis. Gut 36: 341–345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Smythies LE, Waites KB, Lindsey JR, Harris PR, Ghiara P, and Smith PD. 2000. Helicobacter pylori-induced mucosal inflammation is Th1 mediated and exacerbated in IL-4, but not IFN-gamma, gene-deficient mice. Journal of immunology 165: 1022–1029. [DOI] [PubMed] [Google Scholar]
- 83.Mattapallil JJ, Dandekar S, Canfield DR, and Solnick JV. 2000. A predominant Th1 type of immune response is induced early during acute Helicobacter pylori infection in rhesus macaques. Gastroenterology 118: 307–315. [DOI] [PubMed] [Google Scholar]
- 84.Yamaoka Y, Yamauchi K, Ota H, Sugiyama A, Ishizone S, Graham DY, Maruta F, Murakami M, and Katsuyama T. 2005. Natural history of gastric mucosal cytokine expression in Helicobacter pylori gastritis in Mongolian gerbils. Infection and immunity 73: 2205–2212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Eskandari-Nasab E, Sepanjnia A, Moghadampour M, Hadadi-Fishani M, Rezaeifar A, Asadi-Saghandi A, Sadeghi-Kalani B, Manshadi MD, Pourrajab F, and Pourmasoumi H. 2013. Circulating levels of interleukin (IL)-12 and IL-13 in Helicobacter pylori-infected patients, and their associations with bacterial CagA and VacA virulence factors. Scandinavian journal of infectious diseases 45: 342–349. [DOI] [PubMed] [Google Scholar]
- 86.el-Omar EM, Penman ID, Ardill JE, Chittajallu RS, Howie C, and McColl KE. 1995. Helicobacter pylori infection and abnormalities of acid secretion in patients with duodenal ulcer disease. Gastroenterology 109: 681–691. [DOI] [PubMed] [Google Scholar]
- 87.Martinez V, Curi AP, Torkian B, Schaeffer JM, Wilkinson HA, Walsh JH, and Tache Y. 1998. High basal gastric acid secretion in somatostatin receptor subtype 2 knockout mice. Gastroenterology 114: 1125–1132. [DOI] [PubMed] [Google Scholar]
- 88.Zavros Y, Rathinavelu S, Kao JY, Todisco A, Del Valle J, Weinstock JV, Low MJ, and Merchant JL. 2003. Treatment of Helicobacter gastritis with IL-4 requires somatostatin. Proceedings of the National Academy of Sciences of the United States of America 100: 12944–12949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Petersen CP, Meyer AR, De Salvo C, Choi E, Schlegel C, Petersen A, Engevik AC, Prasad N, Levy SE, Peebles RS, Pizarro TT, and Goldenring JR. 2018. A signalling cascade of IL-33 to IL-13 regulates metaplasia in the mouse stomach. Gut 67: 805–817. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Marotti B, Rocco A, De Colibus P, Compare D, de Nucci G, Staibano S, Tatangelo F, Romano M, and Nardone G. 2008. Interleukin-13 mucosal production in Helicobacter pylori-related gastric diseases. Digestive and liver disease : official journal of the Italian Society of Gastroenterology and the Italian Association for the Study of the Liver 40: 240–247. [DOI] [PubMed] [Google Scholar]
- 91.Raghavan S, Fredriksson M, Svennerholm AM, Holmgren J, and Suri-Payer E. 2003. Absence of CD4+CD25+ regulatory T cells is associated with a loss of regulation leading to increased pathology in Helicobacter pylori-infected mice. Clinical and experimental immunology 132: 393–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Rad R, Brenner L, Bauer S, Schwendy S, Layland L, da Costa CP, Reindl W, Dossumbekova A, Friedrich M, Saur D, Wagner H, Schmid RM, and Prinz C. 2006. CD25+/Foxp3+ T cells regulate gastric inflammation and Helicobacter pylori colonization in vivo. Gastroenterology 131: 525–537. [DOI] [PubMed] [Google Scholar]
- 93.Jang TJ 2010. The number of Foxp3-positive regulatory T cells is increased in Helicobacter pylori gastritis and gastric cancer. Pathology, research and practice 206: 34–38. [DOI] [PubMed] [Google Scholar]
- 94.Kandulski A, Malfertheiner P, and Wex T. 2010. Role of regulatory T-cells in H. pylori-induced gastritis and gastric cancer. Anticancer research 30: 1093–1103. [PubMed] [Google Scholar]
- 95.Raghavan S, and Quiding-Jarbrink M. 2012. Immune modulation by regulatory T cells in Helicobacter pylori-associated diseases. Endocrine, metabolic & immune disorders drug targets 12: 71–85. [DOI] [PubMed] [Google Scholar]
- 96.Raghavan S, Suri-Payer E, and Holmgren J. 2004. Antigen-specific in vitro suppression of murine Helicobacter pylori-reactive immunopathological T cells by CD4CD25 regulatory T cells. Scandinavian journal of immunology 60: 82–88. [DOI] [PubMed] [Google Scholar]
- 97.Cook KW, Letley DP, Ingram RJ, Staples E, Skjoldmose H, Atherton JC, and Robinson K. 2014. CCL20/CCR6-mediated migration of regulatory T cells to the Helicobacter pylori-infected human gastric mucosa. Gut 63: 1550–1559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Vignali DA, Collison LW, and Workman CJ. 2008. How regulatory T cells work. Nature reviews. Immunology 8: 523–532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Sojka DK, Huang YH, and Fowell DJ. 2008. Mechanisms of regulatory T-cell suppression - a diverse arsenal for a moving target. Immunology 124: 13–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Szaflarska A, Szczepanik A, Siedlar M, Czupryna A, Sierzega M, Popiela T, and Zembala M. 2009. Preoperative plasma level of IL-10 but not of proinflammatory cytokines is an independent prognostic factor in patients with gastric cancer. Anticancer research 29: 5005–5012. [PubMed] [Google Scholar]
- 101.Chen L, Shi Y, Zhu X, Guo W, Zhang M, Che Y, Tang L, Yang X, You Q, and Liu Z. 2019. IL10 secreted by cancer associated macrophages regulates proliferation and invasion in gastric cancer cells via cMet/STAT3 signaling. Oncology reports 42: 595–604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Batlle E, and Massague J. 2019. Transforming Growth Factor-beta Signaling in Immunity and Cancer. Immunity 50: 924–940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Bonnans C, Chou J, and Werb Z. 2014. Remodelling the extracellular matrix in development and disease. Nature reviews. Molecular cell biology 15: 786–801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Sun N, Taguchi A, and Hanash S. 2016. Switching Roles of TGF-beta in Cancer Development: Implications for Therapeutic Target and Biomarker Studies. Journal of clinical medicine 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Hahm KB, Lee KM, Kim YB, Hong WS, Lee WH, Han SU, Kim MW, Ahn BO, Oh TY, Lee MH, Green J, and Kim SJ. 2002. Conditional loss of TGF-beta signalling leads to increased susceptibility to gastrointestinal carcinogenesis in mice. Alimentary pharmacology & therapeutics 16 Suppl 2: 115–127. [DOI] [PubMed] [Google Scholar]
- 106.Wang JK, Wang WJ, Cai HY, Du BB, Mai P, Zhang LJ, Ma W, Hu YG, Feng SF, and Miao GY. 2018. MFAP2 promotes epithelial-mesenchymal transition in gastric cancer cells by activating TGF-beta/SMAD2/3 signaling pathway. OncoTargets and therapy 11: 4001–4017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Nakamura M, Katano M, Kuwahara A, Fujimoto K, Miyazaki K, Morisaki T, and Mori M. 1998. Transforming growth factor beta1 (TGF-beta1) is a preoperative prognostic indicator in advanced gastric carcinoma. British journal of cancer 78: 1373–1378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Maehara Y, Kakeji Y, Kabashima A, Emi Y, Watanabe A, Akazawa K, Baba H, Kohnoe S, and Sugimachi K. 1999. Role of transforming growth factor-beta 1 in invasion and metastasis in gastric carcinoma. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 17: 607–614. [DOI] [PubMed] [Google Scholar]
- 109.Ebert MP, Yu J, Miehlke S, Fei G, Lendeckel U, Ridwelski K, Stolte M, Bayerdorffer E, and Malfertheiner P. 2000. Expression of transforming growth factor beta-1 in gastric cancer and in the gastric mucosa of first-degree relatives of patients with gastric cancer. British journal of cancer 82: 1795–1800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Pak KH, Kim DH, Kim H, Lee DH, and Cheong JH. 2016. Differences in TGF-beta1 signaling and clinicopathologic characteristics of histologic subtypes of gastric cancer. BMC cancer 16: 60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Zuo ZK, Gong Y, Chen XH, Ye F, Yin ZM, Gong QN, and Huang JS. 2017. TGFbeta1-Induced LncRNA UCA1 Upregulation Promotes Gastric Cancer Invasion and Migration. DNA and cell biology 36: 159–167. [DOI] [PubMed] [Google Scholar]
- 112.Jeong MH, Park SY, Lee SH, Seo J, Yoo JY, Park SH, Kim MJ, Lee S, Jang S, Choi HK, Lee JE, Shin SJ, Choi KC, Cheong JH, and Yoon HG. 2019. EPB41L5 Mediates TGFbeta-Induced Metastasis of Gastric Cancer. Clinical cancer research : an official journal of the American Association for Cancer Research 25: 3617–3629. [DOI] [PubMed] [Google Scholar]


